Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Hematol Oncol ; 43(3): e358-e364, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31815885

RESUMEN

Japanese patients with neuroblastoma completing induction therapy and high-dose chemotherapy received antidisialoganglioside antibody dinutuximab 17.5 mg/m2 for 4 days during each of 5 consecutive 28-day cycles. Patients also received macrophage colony-stimulating factor (M-CSF) or granulocyte colony-stimulating factor (G-CSF) during cycles 1, 3, and 5 combined with interleukin-2 teceleukin during cycles 2 and 4. A total of 25 patients (11 in the M-CSF group and 14 in the G-CSF group) were enrolled, and dose-limiting toxicity was assessed in the first 12 patients (6 in each group). The recommended doses of dinutuximab, M-CSF, and G-CSF were determined to be 17.5 mg/m2, 6.0×106 U/m2, and 5 µg/kg/d, respectively, whereas that of teceleukin was 0.75×106 IU/m2 during week 1 and 1×106 IU/m2 during week 2. The most common grade 3 or 4 adverse events in both groups were neutrophil count decreased, platelet count decreased, pyrexia, and alanine aminotransferase increased. Four patients (2 in each group) discontinued the treatment because of adverse events. At the end of the study, survival was confirmed in 22 patients (9 in the M-CSF group and 13 in the G-CSF group). From these results, we concluded that this combination regimen is a feasible treatment for Japanese patients with neuroblastoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Adolescente , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos Inmunológicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Femenino , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Interleucina-2/efectos adversos , Interleucina-2/uso terapéutico , Japón/epidemiología , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Neuroblastoma/epidemiología , Resultado del Tratamiento
2.
Mol Ther ; 22(9): 1580-92, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24962162

RESUMEN

We have produced an Fc conjugate of colony-stimulating factor (CSF) 1 with an improved circulating half-life. CSF1-Fc retained its macrophage growth-promoting activity, and did not induce proinflammatory cytokines in vitro. Treatment with CSF1-Fc did not produce adverse effects in mice or pigs. The impact of CSF1-Fc was examined using the Csf1r-enhanced green fluorescent protein (EGFP) reporter gene in MacGreen mice. Administration of CSF1-Fc to mice drove extensive infiltration of all tissues by Csf1r-EGFP positive macrophages. The main consequence was hepatosplenomegaly, associated with proliferation of hepatocytes. Expression profiles of the liver indicated that infiltrating macrophages produced candidate mediators of hepatocyte proliferation including urokinase, tumor necrosis factor, and interleukin 6. CSF1-Fc also promoted osteoclastogenesis and produced pleiotropic effects on other organ systems, notably the testis, where CSF1-dependent macrophages have been implicated in homeostasis. However, it did not affect other putative CSF1 targets, notably intestine, where Paneth cell numbers and villus architecture were unchanged. CSF1 has therapeutic potential in regenerative medicine in multiple organs. We suggest that the CSF1-Fc conjugate retains this potential, and may permit daily delivery by injection rather than continuous infusion required for the core molecule.


Asunto(s)
Hepatocitos/metabolismo , Hepatomegalia/inducido químicamente , Fragmentos Fc de Inmunoglobulinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Esplenomegalia/inducido químicamente , Porcinos/inmunología , Animales , Células CHO , Proliferación Celular , Cricetulus , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Semivida , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Medicina Regenerativa
4.
J Natl Cancer Inst ; 86(1): 39-45, 1994 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-8271281

RESUMEN

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.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Femenino , Humanos , Infusiones Intravenosas , Recuento de Leucocitos/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Metástasis de la Neoplasia , Recuento de Plaquetas/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Resultado del Tratamiento
5.
Cancer Res ; 55(19): 4339-46, 1995 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-7671245

RESUMEN

In a Phase II study, 14 patients with metastatic gastrointestinal cancer received the mAb D612 (40 mg/m2, days 4, 7, and 11) in combination with recombinant human monocyte colony-stimulating factor [(rhM-CSF) 80 micrograms/kg/days 1-14]. The combined treatment was well tolerated and resulted in characteristic biological activity associated with each of the agents. Thus, 10 of 14 patients experienced D612-associated secretory diarrhea, which responded to the prostaglandin inhibitor Indomethacin in 5 of 7 patients. rhM-CSF therapy was associated with peripheral monocytosis (peak absolute monocyte count, 1444 +/- 394/mm3) and thrombocytopenia (nadir count, 78 +/- 10/mm3). Monocyte surface marker analysis revealed a high baseline expression of CD16+ cells in our patient population with an additional increase with rhM-CSF therapy. We observed a correlation between the degree of thrombocytopenia and the pretreatment CD16+ monocyte count. Of the plasma cytokines assayed, serum Neopterin demonstrated the most consistent increase during rhM-CSF therapy. There was a significant difference in the half-life of the first and last dose of D612 (35.8 +/- 2 versus 27 +/- 2.9 h; P < 0.05). Eleven of fourteen patients developed low-moderate levels of anti-D612 antibody. Despite the observed biological activity of both rhM-CSF and D612 and the previously described in vitro synergy, no clinical antitumor responses were observed in this Phase II study.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Gastrointestinales/terapia , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Adulto , Anciano , Animales , Anticuerpos Antiidiotipos/sangre , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Recuento de Células Sanguíneas , Citocinas/sangre , Femenino , Humanos , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Receptores de IgG/análisis , Proteínas Recombinantes/administración & dosificación
6.
Cancer Res ; 54(15): 4084-90, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8033141

RESUMEN

Macrophage colony-stimulating factor (M-CSF) is a known inducer of proliferation and differentiation of cells of the mononuclear phagocyte lineage, and gamma-interferon (gamma-IFN) is a known activator of mononuclear phagocytes. In this Phase I clinical trial of combined therapy with M-CSF and gamma-IFN, 36 patients were treated with 14-day continuous infusions of M-CSF at doses ranging from 10 to 140 micrograms/kg/day. In all but five patients, gamma-IFN was administered by daily s.c. injection on days 8-14 of the M-CSF infusion at doses of 0.05 or 0.1 mg/m2/day. A total of 73 courses of M-CSF and 66 courses of gamma-IFN were administered. The maximally tolerated dose combination was 120 micrograms/kg/day M-CSF, 0.1 mg/m2/day gamma-IFN. The addition of gamma-IFN did not alter the maximally tolerated dose of M-CSF therapy, although some additional toxicities were noted with combined therapy. At the 140-micrograms/kg/day M-CSF dose level, grade 4 thrombocytopenia occurred in 2 of 3 patients, with a median platelet count nadir of 26,000/mm3 after 7-10 days of M-CSF infusion. At this dose level, there was one reversible grade 3 hepatic toxicity, and one grade 3 exacerbation of underlying chronic obstructive lung disease. Peripheral blood monocytosis was observed at all M-CSF dose levels exceeding 40 micrograms/kg/day, approaching 3-fold elevations at the 100-micrograms/kg/day M-CSF dose level. The induction of monocytosis was correlated with the development of thrombocytopenia. At the conclusion of therapy with 100 micrograms/kg/day M-CSF, 0.1 mg/m2/day gamma-IFN, 78% of peripheral blood monocytes expressed the low affinity Fc gamma receptor for aggregated immunoglobulin, Fc gamma RIII (CD16), and CD14 was expressed by only 36% of the cells. This phenotype has been shown previously to be associated with cellular activation. In contrast, 35% of monocytes from patients treated with M-CSF therapy alone at the same dose expressed CD16 and 88% expressed CD14. A partial clinical response was noted in a patient with metastatic renal cell carcinoma, and minor clinical responses were observed in patients with a diffuse/follicular lymphoma, metastatic renal cell carcinoma, and metastatic thymoma. At M-CSF doses exceeding 20 micrograms/kg/day within the maximally tolerated dose range, gamma-IFN did not modulate the ability of M-CSF to reliably induce peripheral blood monocytosis. This study shows that M-CSF and gamma-IFN therapy induces the proliferation and differentiation of circulating mononuclear phagocytes.


Asunto(s)
Interferón gamma/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neoplasias/terapia , Adulto , Anciano , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Interferón gamma/efectos adversos , Leucocitosis/etiología , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Masculino , Persona de Mediana Edad , Monocitos , Neoplasias/sangre , Fenotipo , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Trastornos Respiratorios/etiología , Trombocitopenia/etiología
7.
J Clin Oncol ; 10(10): 1643-9, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1403042

RESUMEN

PURPOSE: Recombinant human macrophage colony-stimulating factor (M-CSF) has been shown to stimulate specifically macrophage lineage differentiation in vitro and to induce cells capable of antitumor activity alone or in combination with an antibody. The administration of M-CSF to mice has demonstrated antitumor therapeutic effects in vivo. Therefore, a phase I trial of M-CSF administration to patients with metastatic cancer was undertaken. PATIENTS AND METHODS: M-CSF was given by intermittent intravenous bolus infusion every 8 hours for 7 days; the treatment cycle was repeated once after a week of rest. Cohorts of three patients underwent dose escalation from 10 to 100,000 micrograms/m2/d; 23 patients received 27 courses of M-CSF administration. All patients had metastatic solid tumors refractory to conventional therapy, including renal cell carcinoma (RCC) (nine), melanoma (seven), and colorectal carcinoma (seven). RESULTS: Treatment-related toxicity was minimal; five patients developed transient signs of ocular or periorbital inflammation, with iridocyclitis as the most severe manifestation. At the highest doses, platelet counts decreased with therapy (but remained > 100,000/mm3) and the absolute monocyte count increased during the course of therapy. Only at 30,000 and 100,000 micrograms/m2/d was treatment limited because of toxicity (iritis and malaise). Pharmacokinetic studies demonstrated up to a 1,000-fold increase in circulating serum M-CSF after bolus infusion; half-life varied from 1 to 6 hours. Complete regression of mediastinal adenopathy and multiple pulmonary metastases were observed in one patient with RCC. CONCLUSION: Recombinant M-CSF can be administered safely to patients with metastatic cancer at doses that demonstrate biologic activity.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Femenino , Humanos , Infusiones Intravenosas , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Resultado del Tratamiento
8.
J Clin Oncol ; 15(8): 2954-65, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9256140

RESUMEN

PURPOSE: To determine whether macrophage colony-stimulating factor (M-CSF) reduces the incidence and duration of febrile neutropenia during three courses of intensive consolidation therapy and whether it shortens time to complete consolidation therapy. PATIENTS AND METHODS: In 198 adult patients with acute myeloid leukemia (AML) in complete remission (CR), M-CSF (8 x 10(6) U/d) or placebo was administered from 1 day after the end of each consolidation chemotherapy for 14 days. RESULTS: The duration and incidence of febrile neutropenia was significantly reduced by 34% (P = .00285) and 17% (P = .02065), respectively, in 88 assessable patients in the M-CSF group compared with those in 94 assessable patients in the placebo group. Patients in the M-CSF group had 565 days and 133 episodes of febrile neutropenia during 7,901 days at risk, while patients in the placebo group had 977 days and 185 episodes during 9,077 days at risk. The median period required to finish the three courses of consolidation therapy was 93 days in the M-CSF group, which was significantly shorter than 110 days in placebo group (P = .0050). In the M-CSF group, the recovery of neutrophils and platelets was significantly faster (P = .0348 and P = 0.0364, respectively), the administration of systemic antimicrobial agents tended to be less (P = .0839), and the frequency of platelet transfusion (P = .0259) and the total volume of transfused platelets (P = .0292) were significantly less. However, there was no significant difference in the disease-free survival. CONCLUSION: M-CSF significantly reduced the incidence and duration of febrile neutropenia during the intensive consolidation therapy, and shortened the time to complete consolidation chemotherapy in AML.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Fiebre/terapia , Leucemia Mieloide/tratamiento farmacológico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neutropenia/terapia , Enfermedad Aguda , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Método Doble Ciego , Femenino , Fiebre/inducido químicamente , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Leucemia Mieloide/sangre , Recuento de Leucocitos , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/orina , Masculino , Persona de Mediana Edad , Neutropenia/sangre , Neutropenia/inducido químicamente , Neutrófilos , Recuento de Plaquetas , Factores de Tiempo
9.
J Clin Oncol ; 12(1): 97-106, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8270990

RESUMEN

PURPOSE: Recombinant human macrophage colony-stimulating factor (rM-CSF) has been demonstrated to control the growth, differentiation, and function of mononuclear phagocytes. Preclinical studies have indicated antitumor effects, and therefore a phase I trial of rM-CSF in patients with malignancy was initiated. The toxicity and hematologic and immunologic effects were investigated. PATIENTS AND METHODS: rM-CSF was administered as a subcutaneous injection on days 1 through 5 and 8 through 12. Cycles were repeated every 28 days. Cohorts of four to seven patients received rM-CSF at dose levels from 0.1 to 25.6 mg/m2/d. Forty-two patients received 88 cycles of rM-CSF. All patients had metastatic solid tumors refractory to standard therapy. RESULTS: The toxicity of rM-CSF was mild. Dose-limiting toxicity included thrombocytopenia (two patients) and iritis (one patient) occurring at a dose of 25.6 mg/m2/d. Hematologic studies demonstrated dose-related monocytosis occurring routinely at doses > or = 3.2 mg/m2/d, and thrombocytopenia. Immunologic studies demonstrated enhanced secretion of tumor necrosis factor-alpha (TNF-alpha) and interleukin-1-beta (IL-1 beta) by monocytes after in vitro stimulation with lipopolysaccharide, and increased expression of TNF-alpha mRNA at higher rM-CSF dose levels. Pharmacokinetic studies demonstrated that the systemic clearance rate of M-CSF increases during week 1 of therapy, resulting in lower blood levels of M-CSF during the second week of therapy. CONCLUSION: rM-CSF can be safely administered to patients, and has biologic activity on peripheral-blood monocytes.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Monocitos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Adulto , Anciano , Citocinas/biosíntesis , Citocinas/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Subcutáneas , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/farmacocinética , Masculino , Persona de Mediana Edad , Modelos Biológicos , Monocitos/inmunología , Neoplasias/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico
10.
Leukemia ; 6 Suppl 3: 123S-125S, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1602807

RESUMEN

Exposure of 3 month old SJL/J mice to a single dose of 300 r yielded 15-30% acute myelomonocytic leukemia (AML) development at a mean latency of 1 year. Additional treatment with dexamethasone shortly after irradiation increased leukemia incidence to 50%. All tumors were characterized by a partial deletion of one allele of chromosome 2 and the same deletion was detected in bone marrow and spleen cells of most irradiated mice, irrespective of the development of the disease. The presence of potential leukemic cells (PLC) in mice 4 months after the leukemogenic treatment was confirmed by transplantation studies. In these experiments PLC transition into overt AML seemed to be dependent on their transfer into irradiated recipients. Thus, exposure to 300 r results in the initiation of potential leukemic cells. Experiments were conducted in order to explore the possible role of radiation, cytokines and different hemopoietic growth factors on PLC promotion to overt leukemia. Exposure to 300 r, beside PLC initiation, was found to trigger the production of IL-6 and CSF-1; the additional administration of dexamethasone further increased CSF-1 levels. In vivo administration of CSF-1 into mice carrying radiation-induced PLC was most effective in PLC promotion to overt AML development.


Asunto(s)
Leucemia Mielomonocítica Aguda/etiología , Leucemia Inducida por Radiación/etiología , Animales , Citocinas/efectos adversos , Dexametasona/farmacología , Femenino , Sustancias de Crecimiento/efectos adversos , Interleucina-6/metabolismo , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones , Preleucemia/etiología
11.
Clin Cancer Res ; 2(2): 295-302, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9816172

RESUMEN

Macrophage colony-stimulating factor (M-CSF) is a lineage-specific, homodimeric growth factor that supports the proliferation and maturation of bone marrow progenitors and the survival and function of mononuclear/macrophage cells. In vitro studies have demonstrated antitumor activity of macrophage colony-stimulating factor-treated monocytes against melanoma target cells. A Phase I study was conducted by administering the glycosylated form of the protein to patients with metastatic melanoma as two 7-day continuous i.v. infusions separated by a 2-week rest. Cohorts of three patients per dose level received escalating doses of 10-160 microgram/kg/day. Safety, clinical, and biological effects were evaluated. The infusions were well tolerated with occasional maximum grade 2 nonhematological toxicity. Rapidly reversible thrombocytopenia was the major hematological adverse effect. Its etiology may in part be explained by proliferation and activation of monocyte/macrophage cells in bone marrow samples. Evidence for a biological effect on tumors was suggested by the delayed, complete disappearance of multiple lesions in one patient and a decrease in the size of one marker lesion in a second patient with a mixed response. Fasting serum cholesterol levels decreased during the infusions and may represent an additional therapeutic application for this growth factor.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/efectos adversos , Melanoma/terapia , Adulto , Anciano , Femenino , Humanos , Infusiones Intravenosas , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/farmacocinética , Masculino , Melanoma/secundario , Persona de Mediana Edad , Proteínas Recombinantes/efectos adversos
12.
J Immunother (1991) ; 12(1): 50-4, 1992 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1637782

RESUMEN

Fourteen patients were entered into a phase I dose-escalation trial of macrophage colony-stimulating factor (M-CSF). M-CSF was administered to inpatients by rapid 15 min i.v. infusion every 8 h x 5 days, repeated after a 9-day rest. Dose levels evaluated were 20, 40, 80, 330, and 1,100 micrograms/m2. Monitoring of patients every 4 h included vital signs, daily complete blood count (CBC), and serum chemistries (SGOT, creatinine, and bilirubin) while receiving M-CSF. No clinical or laboratory evidence of toxicity was seen. The average serum t1/2 varied with dose level. At 330 and 1,100 micrograms/m2, the serum t1/2 was 25 and 84 min, respectively, implying a saturable mechanism of clearance. After 5 days of treatment, the t1/2 decreased by twofold, consistent with enhancement of the saturable mechanism. Monocyte cytotoxicity against the A375 melanoma cell line was evaluated pretreatment and day 5 of each cycle. No consistent enhancement of monocyte cytotoxicity was seen. No effect on peripheral blood monocyte number was seen until the 1,100 micrograms/m2 dose level. At this dose level, the mean monocyte number on day 5 was increased compared to baseline (1,300 mm3 vs. 300/mm3). Clinical activity was seen in two patients with previously progressive leiomyosarcoma metastatic to the liver. A partial response (PR) lasting 7 months occurred at the 330 micrograms/m2 dose level while a patient treated at 1,100 micrograms/m2 has had stable disease for 20+ months. The maximum tolerated dose (MTD) of M-CSF was not determined. Based on clinical responses, a phase II trial is warranted in patients with metastatic soft tissue sarcoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Neoplasias/tratamiento farmacológico , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Biopterinas/análogos & derivados , Biopterinas/sangre , Pruebas Inmunológicas de Citotoxicidad , Evaluación de Medicamentos , Humanos , Infusiones Intravenosas , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/farmacocinética , Masculino , Monocitos/inmunología , Neopterin , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/farmacocinética , Factores de Tiempo
13.
Blood Rev ; 8(3): 169-78, 1994 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7529606

RESUMEN

The use of the recombinant hematopoietic growth factors G-CSF and GM-CSF have shortened the period of neutropenia, or avoided this problem, in many cancer patients who have received cytotoxic therapy. Although these benefits have been particularly striking in the autologous bone marrow and/or autologous peripheral blood progenitor cell transplant setting, most data suggest that the use of G-CSF and GM-CSF only marginally enhance recovery of the neutrophil count when administered after allogeneic bone marrow infusion. Furthermore, in the allograft setting these expensive agents have not provided benefit in the form of enhanced platelet count recovery, lessening the incidence of graft-versus-host disease, or improvement in overall survival. These data do not justify routine widespread use of G-CSF and GM-CSF and suggest that these agents should be reserved for patients who experience delay in engraftment after allogeneic bone marrow infusion. Administration of erythropoietin, on the other hand, may reduce the need for homologous red blood cell transfusions, and may increase the safety margin for both the allogeneic bone marrow recipient and as well as the donor. Recombinant hematopoietic growth factors targetted specifically to enhance platelet recovery after transplantation (such as interleukin-3, interleukin-6, and interleukin-11) have shown promise after autotransplantation and after conventional dose chemotherapy, and likely will be evaluated in the allogeneic transplant patient.


Asunto(s)
Trasplante de Médula Ósea , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Hematopoyesis/efectos de los fármacos , Ensayos Clínicos como Asunto , Eritropoyetina/uso terapéutico , Supervivencia de Injerto/efectos de los fármacos , Enfermedad Injerto contra Huésped/prevención & control , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Humanos , Depleción Linfocítica , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neoplasias/sangre , Neoplasias/terapia , Neutropenia/prevención & control , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Trasplante Homólogo , Resultado del Tratamiento
14.
Bone ; 16(3): 315-24, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7540405

RESUMEN

It has recently been shown that following treatment with colony-stimulating factor-1 (CSF-1) the osteopetrotic condition in toothless (tl) rats greatly improves and growth is accelerated. We have examined the effects of such treatment on the microvasculature of the distal femoral chondro-osseous junction, a site where bone growth in length is coordinated with angiogenesis. Vascular casts and ultrastructural analyses of this region showed that, compared to untreated normal rats, untreated mutants showed little bone growth or angiogenesis. When mutants were treated with CSF-1 angiogenesis was markedly accelerated. These data show a remarkable effect of this growth factor on angiogenesis in this osteopetrotic mutation. Whether this effect of CSF-1 on angiogenesis is direct or indirect is not known and indicates that its effects on the normal microvasculature deserve further study.


Asunto(s)
Fémur/irrigación sanguínea , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Neovascularización Patológica/inducido químicamente , Osteopetrosis/tratamiento farmacológico , Animales , Arteriolas/efectos de los fármacos , Arteriolas/crecimiento & desarrollo , Arteriolas/ultraestructura , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Capilares/ultraestructura , Cartílago/citología , Cartílago/efectos de los fármacos , Cartílago/ultraestructura , Molde por Corrosión , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/ultraestructura , Femenino , Fémur/efectos de los fármacos , Fémur/ultraestructura , Factor Estimulante de Colonias de Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Mutación/genética , Osteopetrosis/genética , Ratas , Ratas Mutantes
15.
Drug Saf ; 13(6): 371-406, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8652081

RESUMEN

A number of cytokines are used as haemopoietic growth factors and this review focuses on toxicities associated with granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), interleukin (IL)-1, IL-3, IL-4, IL-6 and macrophage colony-stimulating factor (M-CSF). Both GM-CSF and G-CSF, currently approved for clinical use, are generally well tolerated by the majority of patients during short term administration. Constitutional symptoms and bone pain are the most frequently reported adverse effects, but they are rarely treatment-limiting. Reactivation of rheumatoid symptoms, and exacerbation of autoimmune thyroiditis or autoimmune haematological disorders have sometimes been described. Severe cardiovascular complications include the possibility for arterial thromboses and the vascular leak syndrome, which is more specifically observed with GM-CSF. Reports of several cases and small series of patients have suggested that growth factors might increase the pulmonary toxicity of chemotherapy, a possibility that remains debated and requires further attention. Generalised or local cutaneous reactions are frequently noted with GM-CSF. Leukocytoclastic vasculitis was observed with both growth factors, while neutrophilic dermatoses have been mostly described with G-CSF. Exacerbation of psoriasis and isolated anaphylactic reactions have appeared with GM-CSF and G-CSF. The hepatotoxic potential of the growth factors is not clearly established, but the occurrence of coagulation abnormalities has recently been reported. Renal and biological disturbances are usually transient. Long term treatment with GM-CSF and G-CSF also seems to be well tolerated, but the possible occurrence of several adverse events, i.e. bone disorders, leukaemia, unmasking or acceleration of underlying disease, require further investigation in patients receiving prolonged treatment, as in myelodysplasia. Finally, antibodies against growth factors have been reported only with GM-CSF. Other cytokines are still under investigation. Flu-like and constitutional symptoms, sometimes dose-limiting, have been reported with IL-1, IL-3, IL-4 and IL-6, while M-CSF was occasionally associated with such adverse effects. More specific adverse events, also frequently considered as dose-limiting toxicities, include hypotension with IL-1, severe headache or skin rash with IL-3, and nasal congestion and gastroduodenal lesions with IL-4. Severe capillary leak syndrome has been reported only with IL-4. M-CSF toxicity is minimal and limited to reversible but sometimes dose-limiting thrombocytopenia and ophthalmological symptoms with the recombinant product. Again, the safety of long term administration of these cytokines has not yet been determined, and IL-3-induced disease progression in myelodysplastic patients has been suggested.


Asunto(s)
Citocinas/efectos adversos , Factores de Crecimiento de Célula Hematopoyética/efectos adversos , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Humanos , Interleucina-1/efectos adversos , Interleucina-3/efectos adversos , Interleucina-4/efectos adversos , Interleucina-6/efectos adversos , Factor Estimulante de Colonias de Macrófagos/efectos adversos
16.
Toxicol Sci ; 43(1): 61-7, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9629620

RESUMEN

The safety of M-CSF was assessed in cynomolgus monkeys in an intravenous dosing regimen. Exposure (AUC0-24) multiples (monkey vs human) were calculated using the no observable adverse effect level (NOAEL) observed in this study and correlated with known M-CSF-induced toxicities in a previous continuous intravenous infusion (civ) study in monkeys. M-CSF was administered by daily intravenous infusion (2 h) to cynomolgus monkeys (2/sex/ group) at 0.1, 0.3, 0.7, and 1.0 mg/kg/day, for 28 consecutive days. Control animals (2/sex) received placebo. The 0.7 mg/kg/day group was held for an additional 4-week recovery period. Criteria evaluated included physical observations, ophthalmoscopy, electrocardiography, body weight, food consumption, clinical pathology, antibody formation, pharmacokinetics, necropsy, organ weights, and histopathology. The only effect previously seen in monkeys after intravenously administered M-CSF occurred in animals in the 0.7 and 1.0 mg/kg/day groups. They exhibited a slight decrease in platelets between days 4 and 12 with subsequent recovery. No effects related to M-CSF administration were evident in macroscopic or microscopic evaluations and there was no evidence of anti-M-CSF antibody production. M-CSF at all dose levels was completely eliminated within each dosing interval with no accumulation. Clearance of M-CSF was enhanced during the first week of dosing, but returned to baseline clearance levels by day 27. This dosing regimen was shown to be remarkably free of toxicities noted in a previous monkey study where M-CSF was given by civ at similar daily doses. At the high dose, which was considered to be the NOAEL, the AUC0-24 was 40-fold greater than the AUC0-24 in clinical trials where 2.0 mg/m2 was administered by a daily 2-h infusion.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/efectos adversos , Animales , Anticuerpos/inmunología , Área Bajo la Curva , Evaluación de Medicamentos , Femenino , Semivida , Macaca fascicularis , Factor Estimulante de Colonias de Macrófagos/inmunología , Factor Estimulante de Colonias de Macrófagos/farmacocinética , Masculino , Nivel sin Efectos Adversos Observados , Placebos , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética
17.
Leuk Lymphoma ; 15(3-4): 347-9, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7866285

RESUMEN

A patient is presented who developed cutaneous papular histiocytic infiltrates after treatment with monocyte colony stimulating factor (rhM-CSF). This is the first reported complication of this type after treatment with this new cytokine.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/efectos adversos , Enfermedades de la Piel/inducido químicamente , Humanos , Infusiones Intravenosas , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Enfermedades de la Piel/patología
18.
Oncol Rep ; 10(1): 127-31, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12469157

RESUMEN

We performed a randomized double blind study between 1992 and 1995 in which 214 patients with FIGO stage I to III ovarian cancers received administration of 10(6) units (low dose group) or 8x10(6) units (high dose group) of macrophage colony-stimulating factor (M-CSF) after cyclophosphamide/adriamycin/cisplatin (CAP) therapy. The period required to finish a set of intensive chemotherapy, which was the primary endpoint, was significantly shortened (p=0.0004), and the incidence of febrile neutropenia significantly decreased (p=0.04). In this study, we followed the patients for a prolonged period. The patients were divided into two groups: patients with complete tumor excision and those with incomplete excision, then the relapse rate and survival rate 5 years after initiation of the clinical study were compared. The relapse rate tended to be lower in the high dose group than in the low dose group in patients with no residual tumor (p=0.0750). However, there was no difference in the relapse rate between the two dose groups in patients with residual tumor. Although there were no significant differences in the survival rate between the high and low dose groups in patients with or without residual tumor, the survival rate in mucinous adenocarcinoma patients with no residual tumor was 64.3% in the low dose group (n=14) and 92.3% in the high dose group (n=14), showing a significantly higher rate (p=0.0436), and the survival rate tended to be higher in the high dose group in patients with serous adenocarcinoma (p=0.0786). Furthermore, in patients aged 40 years or younger with no residual tumor, the survival rates were 73.9 and 100% in the low and high dose groups, respectively, showing a significantly higher rate in the high dose group (p=0.0310). Our results suggest that administration of M-CSF can improve the long-term prognosis of ovarian cancer patients with no residual tumor, but further prospective randomized trials with a primary endpoint of relapse-preventing effect are needed.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/administración & dosificación , Ciclofosfamida/administración & dosificación , Método Doble Ciego , Doxorrubicina/administración & dosificación , Femenino , Humanos , Incidencia , Inyecciones Intraperitoneales , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Tasa de Supervivencia , Factores de Tiempo
19.
Br J Haematol ; 121(4): 614-22, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12752103

RESUMEN

To characterize recombinant human macrophage-colony stimulating factor (rhM-CSF)-associated thrombocytopenia (TCP), in vivo studies were performed in dogs, including the biodistributions and recoveries of radiolabelled autologous and allogeneic platelets. rhM-CSF induced a reversible, dose-dependent decrease in platelet counts. The number of megakaryocytes in spleen and marrow of rhM-CSF-treated dogs was increased two to threefold. Recoveries of allogeneic platelets transfused from rhM-CSF-treated donors into tolerized recipients (n = 3) were not significantly different from allogeneic baseline studies (93 +/- 10% of baseline values at 24 h and 90 +/- 1% at 40 h), whereas autologous platelets infused back into rhM-CSF-treated donors had decreased recoveries (45 +/- 2% of baseline values at 24 h, P = 0.03 and 20 +/- 4% at 40 h, P = 0.001). Platelet biodistribution studies showed increased accumulation of radiolabelled platelets over the spleens and livers of rhM-CSF-treated dogs. Histochemistry showed increased levels of platelet-specific antigen (CD41; glycoprotein IIb) associated with Kupffer cells. The sensitivity of platelets from rhM-CSF-treated dogs to activation from thrombin, as measured by expression of P-selectin (CD62P), was not significantly different when compared with baseline studies (P = 0.18; n = 4). These results support the concept that rhM-CSF induces an activation of the monocyte-macrophage system (MMS), which causes a reversible TCP in a dog model.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/efectos adversos , Trombocitopenia/inducido químicamente , Animales , Plaquetas/fisiología , Supervivencia Celular , Perros , Relación Dosis-Respuesta a Droga , Macrófagos del Hígado/fisiología , Hígado , Recuento de Plaquetas , Bazo
20.
Biotechnol Ther ; 2(3-4): 231-9, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1845128

RESUMEN

rhM-CSF has diverse in vitro and in vivo hematologic and metabolic effects. Clinical trials with this new biopharmaceutical are now in progress and early results have confirmed preclinical findings. The major demonstrated effects of rhM-CSF include an increase in number and activation of monocytes and macrophages, the ability to enhance antibody-dependent cellular cytotoxicity, enhancement of macrophage microbial phagocytic and killing activity, cholesterol lowering, and platelet lowering. The range of potential indications for the use of rhM-CSF makes the further clinical development of this molecule both a challenge and an opportunity. Investigation of combination therapies with other cytokines or monoclonal antibodies will be an important aspect of future investigations.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Trasplante de Médula Ósea , Colesterol/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Técnicas In Vitro , Infecciones/terapia , Leucemia/terapia , Factor Estimulante de Colonias de Macrófagos/efectos adversos , Neoplasias/terapia , Proteínas Recombinantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA