Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Blood Cancer J ; 6: e378, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26745852

RESUMEN

In a phase 3 trial of denosumab vs zoledronic acid in patients (n=1776) with bone metastases and solid tumors or multiple myeloma, denosumab was superior to zoledronic acid for the primary end point of prevention of skeletal-related events. There was no difference in overall survival between the two groups; however, an ad hoc overall survival analysis in the multiple myeloma subset of patients (n=180) favored zoledronic acid (hazard ratio (HR) 2.26; 95% confidence interval (CI) 1.13-4.50; P=0.014). In the present analysis, we found imbalances between the groups with respect to baseline risk characteristics. HRs with two-sided 95% CIs were estimated using the Cox model. After adjustment in a covariate analysis, the CI crossed unity (HR 1.86; 95% CI 0.90-3.84; P=0.0954). Furthermore, we found a higher rate of early withdrawals for the reasons of lost to follow-up and withdrawal of consent in the zoledronic acid group; after accounting for these, the HR was 1.31 (95% CI 0.80-2.15; P=0.278). In conclusion, the survival results in multiple myeloma patients in this trial were confounded and will eventually be resolved by an ongoing phase 3 trial.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Denosumab/uso terapéutico , Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Denosumab/administración & dosificación , Denosumab/efectos adversos , Difosfonatos/administración & dosificación , Difosfonatos/efectos adversos , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Imidazoles/administración & dosificación , Imidazoles/efectos adversos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias/patología , Trasplante Autólogo , Resultado del Tratamiento , Ácido Zoledrónico
2.
Eur J Cancer ; 53: 75-83, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26693901

RESUMEN

BACKGROUND: Analyses of phase III trials showed that denosumab was superior to zoledronic acid (ZA) in preventing skeletal-related events (SREs) irrespective of age, history of SREs, or baseline pain status. This analysis assessed the risk of SREs across additional baseline characteristics. PATIENTS AND METHODS: Patients (N = 5543) from three phase III trials who had breast cancer, prostate cancer, or other solid tumours and one or more bone metastasis were included. Superiority of denosumab versus ZA in reducing risk of first SRE and first and subsequent SREs was assessed in subgroups defined by the Eastern Cooperative Oncology Group performance status (ECOG PS), bone metastasis location, bone metastasis number, visceral metastasis presence/absence, and urinary N-telopeptide (uNTx) level using Cox proportional hazards and Anderson-Gill models. Subgroups except bone metastasis location were also assessed for each solid tumour type. RESULTS: Compared with ZA, denosumab significantly reduced the risk of first SRE across all subgroups (hazard ratio [HR] ranges: ECOG PS, 0.79-0.84; bone metastasis location, 0.78-0.83; bone metastasis number, 0.78-0.84; visceral metastasis presence/absence, 0.80-0.82; uNTx level, 0.73-0.86) and reduced the risk of first and subsequent SREs in all subgroups (HR ranges: ECOG PS, 0.76-0.83; bone metastasis location, 0.78-0.84; bone metastasis number, 0.79-0.81; visceral metastasis presence/absence, 0.79-0.81; uNTx level, 0.74-0.83). Similar results were observed in subgroups across tumour types. CONCLUSION: Denosumab was superior to ZA in preventing SREs in patients with bone metastases from advanced cancer, regardless of ECOG PS, bone metastasis number, baseline visceral metastasis presence/absence, and uNTx level.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Denosumab/administración & dosificación , Difosfonatos/administración & dosificación , Imidazoles/administración & dosificación , Administración Cutánea , Enfermedades Óseas/prevención & control , Neoplasias Óseas/secundario , Femenino , Humanos , Infusiones Intravenosas , Masculino , Resultado del Tratamiento , Ácido Zoledrónico
3.
Ann Oncol ; 23(12): 3045-3051, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22851406

RESUMEN

BACKGROUND: Patients with metastatic bone disease are living longer in the metastatic stage due to improvements in cancer therapy, making strategies to prevent the aggravation of bone disease and its complications, such as skeletal-related events (SREs) and pain, increasingly important. PATIENTS AND RESULTS: In this phase 3 trial in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma, denosumab reduced the risk of radiation to bone by 22% relative to zoledronic acid (P = 0.026), prevented worsening of pain and pain interference (2-point increase in Brief Pain Inventory score; P < 0.05 versus zoledronic acid), and reduced the frequency of a shift from no/weak opioid analgesic use to strong opioids (P < 0.05 versus zoledronic acid at months 3-5). Denosumab delayed the time to moderate-to-severe pain compared with zoledronic acid in patients with mild or no pain at the baseline (P = 0.04), supporting early treatment. Health-related quality-of-life scores were similar in both groups. The number needed to treat to avoid one SRE for denosumab was 3 patient-years versus placebo and 10 patient-years versus zoledronic acid. CONCLUSION: The use of denosumab was associated with better prevention of the complications of metastatic bone disease secondary to solid tumors or multiple myeloma versus zoledronic acid.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Dolor/prevención & control , Neoplasias Óseas/secundario , Denosumab , Método Doble Ciego , Humanos , Dolor/tratamiento farmacológico , Dolor/etiología , Calidad de Vida , Ligando RANK/antagonistas & inhibidores , Resultado del Tratamiento , Ácido Zoledrónico
4.
Ann Oncol ; 23(6): 1640-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22015451

RESUMEN

BACKGROUND: Tumor lysis syndrome (TLS) is a life-threatening disorder characterized by hyperuricemia and metabolic derangements. The efficacy of rasburicase, administered daily for 5 days, has been well established. However, the optimal duration of therapy is unknown in adults. PATIENTS AND METHODS: We evaluated the efficacy of rasburicase (0.15 mg/kg) administered as single dose followed by as needed dosing (maximum five doses) versus daily dosing for 5 days in adult patients at risk for TLS. RESULTS: Eighty of the 82 patients enrolled received rasburicase; 40 high risk [median uric acid (UA) 8.5 mg/dl; range, 1.5-19.7] and 40 potential risk (UA = 5.6 mg/dl; range, 2.4-7.4). Seventy-nine patients (99%) experienced normalization in their UA within 4 h after the first dose; 84% to an undetectable level (<0.7 mg/dl). Thirty-nine of 40 (98%) patients in the daily-dose arm and 34 of 40 (85%) patients in single-dose arm showed sustained UA response. Six high-risk patients within the single-dose arm required second dose for UA >7.5 mg/dl. Rasburicase was well tolerated; one patient with glucose-6-phosphate dehydrogenase deficiency developed methemoglobinemia and hemolysis. CONCLUSIONS: Rasburicase is highly effective for prevention and management of hyperuricemia in adults at risk for TLS. Single-dose rasburicase was effective in most patients; only a subset of high-risk patients required a second dose.


Asunto(s)
Supresores de la Gota/administración & dosificación , Síndrome de Lisis Tumoral/prevención & control , Urato Oxidasa/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Femenino , Supresores de la Gota/uso terapéutico , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Factores de Riesgo , Resultado del Tratamiento , Síndrome de Lisis Tumoral/etiología , Urato Oxidasa/uso terapéutico , Ácido Úrico/sangre
5.
Oncology (Williston Park) ; 15(7 Suppl 8): 35-8, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11497230

RESUMEN

Recombinant human thrombopoietin (rhTPO) is a full-length glycosylated molecule that has been under evaluation in the setting of chemotherapy-induced myelosuppression. It has been shown to be a potent stimulator of platelet production in cancer patients when administered prior to chemotherapy. The peak platelet response to a single dose of rhTPO is observed around day 12, and is accompanied by a significant increase in the number of mature megakaryocytes in bone marrow. Consistent with this biologic effect, rhTPO administered postchemotherapy has been shown to be effective in attenuating severe thrombocytopenia induced by carboplatin, which produces a late platelet nadir. Early clinical experience with a regimen that produces an early nadir, however, such as AI (doxorubicin [Adriamycin] and ifosfamide [Ifex]), suggests that administration of rhTPO both prior to and following chemotherapy might be important to reduce thrombocytopenia severity. Treatment with rhTPO in these clinical trials has been well tolerated with a favorable safety profile. Randomized clinical trials have been initiated to determine further the importance of schedule in the prevention and treatment of severe thrombocytopenia in cancer patients.


Asunto(s)
Antineoplásicos/efectos adversos , Médula Ósea/efectos de los fármacos , Carboplatino/efectos adversos , Trombocitopenia/tratamiento farmacológico , Trombopoyetina/uso terapéutico , Antineoplásicos/uso terapéutico , Protocolos Antineoplásicos , Carboplatino/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/inducido químicamente , Trombopoyetina/administración & dosificación
6.
Bone Marrow Transplant ; 27(6): 647-52, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11319596

RESUMEN

Lymphocytes play a major role in host defense against Aspergillus, but little is known about the contribution of dendritic cells (DC) to antifungal immunity in humans. We have observed that DC derived from normal volunteers phagocytose heat-killed A. fumigatus conidia. Following 24 h of exposure to the fungus, DC displayed an increase in the mean fluorescence intensity of HLA-DR, CD80, and CD86, and an increase in the percentage of CD54(+) cells. These DC also displayed increased production of IL-12. DC derived from CD34(+) progenitors or monocytes stimulated autologous lymphocytes to proliferate and produce high levels of interferon-gamma, but not interleukin-10, in response to fungal antigen. DC generated from CD34(+) progenitors collected prior to autologous or allogeneic stem cell transplantation also partially restored the in vitro antifungal proliferative response of lymphocytes obtained from patients 1 month after transplantation. These results suggest that DC are important to host-response to A. fumigatus, and that ex vivo-generated DC might be useful in restoring or enhancing the antifungal immunity after hematopoietic stem cell transplantation.


Asunto(s)
Aspergillus fumigatus/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo , Fagocitosis/inmunología , Trasplante Autólogo
7.
Semin Hematol ; 37(2 Suppl 4): 28-34, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10831286

RESUMEN

Since the identification and cloning of c-Mpl ligand, two forms of recombinant human thrombopoietin have undergone clinical development. Both the full-length molecule, known as rhTPO, and the truncated version of the molecule, known as pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF), have been evaluated in phase I/II clinical trials in cancer patients receiving myelosuppressive chemotherapy. Early clinical trials with PEG-rHuMGDF in cancer patients demonstrated its clinical safety and platelet-stimulating activity. However, the development of neutralizing antibodies and clinically significant thrombocytopenia in some patients and normal donors who received PEG-rHuMGDF have led to discontinuation of clinical trials with this molecule in the United States. Clinical experience with rhTPO so far indicates that this full-length glycosylated molecule is remarkably well tolerated and has a favorable safety profile. In these studies, rhTPO exhibited dose-dependent increases in circulating platelet counts and bone marrow megakaryocytes before chemotherapy. In addition, there was an increase in the frequency and proliferation of bone marrow progenitor cells and mobilization of progenitors into the peripheral blood. Early results also showed that rhTPO can attenuate chemotherapy-induced severe thrombocytopenia and reduce the need for platelet transfusions. However, in this setting, the optimal schedule of rhTPO administration may depend on the length of the regimen and anticipated timing of the platelet nadir. These initial results indicate that rhTPO is a safe and potentially useful agent in the prevention and management of chemotherapy-induced thrombocytopenia. Results of larger randomized clinical trials will determine the therapeutic potential of this novel growth factor in various clinical settings.


Asunto(s)
Antineoplásicos/efectos adversos , Trombocitopenia/tratamiento farmacológico , Trombopoyetina/uso terapéutico , Ensayos Clínicos como Asunto , Glicosilación , Humanos , Recuento de Plaquetas/efectos de los fármacos , Polietilenglicoles/metabolismo , Proteínas Recombinantes de Fusión , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/inducido químicamente
8.
Ann Intern Med ; 132(5): 364-8, 2000 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-10691586

RESUMEN

BACKGROUND: Thrombocytopenia is a significant problem in the treatment of cancer. OBJECTIVE: To assess the clinical safety of therapy with recombinant human thrombopoietin (rhTPO) and its ability to ameliorate chemotherapy-induced severe thrombocytopenia. DESIGN: Phase I/II clinical cohort study. SETTING: The University of Texas M.D. Anderson Cancer Center, Houston, Texas. PATIENTS: 29 patients with gynecologic cancer. INTERVENTION: Recombinant human thrombopoietin was given before chemotherapy and after a second cycle of carboplatin therapy. MEASUREMENTS: Peripheral blood counts and platelet transfusions. RESULTS: Administration of rhTPO after chemotherapy significantly reduced the degree and duration of thrombocytopenia and enhanced platelet recovery. In patients who received the optimal biological dose of rhTPO (1.2 microg/kg of body weight) in cycle 2 (carboplatin plus rhTPO), the mean platelet count nadir was higher (44x10(9) cells/L and 20x10(9) cells/L; P = 0.002) and the duration of thrombocytopenia was shorter (days with a platelet count <20x10(9) cells/L, 1 and 4 [P = 0.002]; days with a platelet count <50x10(9) cells/L, 4 and 7 [P = 0.006]) than in cycle 1 (carboplatin only). The need for platelet transfusion in this group was reduced from 75% of patients in cycle 1 to 25% of patients in cycle 2 (P = 0.013). CONCLUSIONS: Therapy with rhTPO seems to be safe and may attenuate chemotherapy-induced severe thrombocytopenia and reduce the need for platelet transfusions.


Asunto(s)
Antineoplásicos/efectos adversos , Carboplatino/efectos adversos , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Trombocitopenia/inducido químicamente , Trombocitopenia/prevención & control , Trombopoyetina/uso terapéutico , Análisis de Varianza , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Recuento de Plaquetas , Transfusión de Plaquetas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/terapia , Trombopoyetina/administración & dosificación
9.
Br J Haematol ; 104(2): 403-11, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10050726

RESUMEN

Chemotherapy-induced thrombocytopenia represents a significant clinical problem in the management of patients with malignancy. Recombinant human thrombopoietin (rhTPO) is a potent stimulator of platelet production in vivo. The ability to cryopreserve rhTPO-derived platelets would enable the use of autologous platelets during the period of thrombocytopenia. ThromboSol is a platelet-stabilizing formulation consisting of second messenger effectors that inhibit specific activation pathways endogenous to platelets. To investigate the effect of ThromboSol cryopreservation, platelets from rhTPO-treated patients (n = 23) and normal donors were treated with ThromboSol and 2% DMSO and cryopreserved for up to 6 months. The platelets were thawed at different intervals and tested for retention of platelet functional activity in vitro. Following a short-term storage (1 week), the cryopreserved platelets from patients treated with rhTPO exhibited significantly higher retention of functional activities including discoid morphology (70% v 57%), extent of shape change (19% v 13%) stirring shape change (15% v 11%) and hypotonic shock response (56% v 25%), as compared to the cryopreserved platelets from controls. Furthermore, there was no further significant loss of functional activity following cryopreservation for up to 6 months. These findings suggest that cryopreservation of platelets from rhTPO-treated donors may provide a useful novel strategy for autologous or allogeneic donation for subsequent transfusions to manage treatment-related thrombocytopenia.


Asunto(s)
Plaquetas/fisiología , Criopreservación/métodos , Crioprotectores/uso terapéutico , Dimetilsulfóxido/uso terapéutico , Neoplasias Ováricas/terapia , Sarcoma/terapia , Trombocitopenia/terapia , Trombopoyetina/uso terapéutico , Plaquetas/ultraestructura , Femenino , Humanos , Microscopía Electrónica , Proteínas Recombinantes , Trombocitopenia/etiología
10.
Oncology (Williston Park) ; 12(11): 1597-604, 1607-8; discussion 1611-4, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9834938

RESUMEN

After an almost 40-year search for a primary regulatory of platelet production, thrombopoietin has recently been purified and cloned. Thrombopoietin regulates all stages in the production of platelets by promoting both the proliferation of megakaryocyte progenitors and their maturation into platelet-producing megakaryocytes. In preclinical studies in normal mice and non-human primates, administration of thrombopoietin resulted in a rapid rise in platelet counts to levels previously unattainable with other thrombopoietic cytokines. In myelosuppressed animal models, use of thrombopoietin following chemotherapy, radiation, or stem-cell transplantation accelerated megakaryocyte and platelet recovery. Thrombopoietin has rapidly moved from the laboratory to the clinic in the last 3 years. Preliminary results of clinical trials using truncated or full-length forms of the molecule indicate that thrombopoietin is a powerful stimulus in the production of megakaryocytes and normal platelets in humans and enhances platelet recovery following chemotherapy. Although the peripheral effect is selective on platelet lineage, thrombopoietin mediates stimulatory effects on progenitors of multiple cell lineages at the bone marrow level and mobilizes progenitor cells into the peripheral blood. These biological effects suggest that thrombopoietin holds promise as a useful agent for the prevention and treatment of thrombocytopenia in cancer patients and for other disorders of thrombocytopenia.


Asunto(s)
Plaquetas/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Trombocitopenia/prevención & control , Trombopoyetina/farmacología , Animales , Ensayos Clínicos como Asunto , Humanos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/etiología , Trombopoyetina/uso terapéutico
11.
Semin Hematol ; 35(3): 261-8, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9685172

RESUMEN

Thrombopoietin (TPO) has rapidly moved from the laboratory to the clinic in less than 2 years since its cloning. Two forms of recombinant TPO have been developed for clinical use. The full-length molecule (Genentech Inc, San Francisco, CA) is referred to as recombinant human thrombopoietin, and the truncated version of the molecule (Amgen, Thousand Oaks, CA) is referred to as pegylated recombinant human megakaryocyte growth and development factor. Both of these forms have been evaluated in phase I/II clinical trials. Administration of either of these agents elicits an increase in circulating platelet counts by several-fold in patients who have normal hematopoiesis before chemotherapy. The response in platelets is accompanied by a significant increase in bone marrow megakaryocytes, an increase in frequency and the number of bone marrow progenitor cells of multiple cell lineages, and a marked mobilization of progenitor cells into the peripheral blood. The results of early trials suggest that TPO attenuates thrombocytopenia and enhances platelet recovery after chemotherapy. Several clinical trials are investigating the potential of this cytokine to enhance platelet recovery after high-dose chemotherapy with or without progenitor cell support, bone marrow transplantation, and leukemia treatment, and for increasing the apheresis yield of platelets in normal donors and cancer patients. Based on the encouraging results of the initial clinical trials, TPO holds promise as a useful therapeutic agent in the prevention and treatment of thrombocytopenia in cancer patients and other disorders.


Asunto(s)
Trombopoyetina/uso terapéutico , Enfermedad Aguda , Antineoplásicos/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucemia/terapia , Proteínas Recombinantes , Trombocitopenia/inducido químicamente , Trombocitopenia/tratamiento farmacológico , Trombopoyetina/efectos adversos , Trombopoyetina/genética
12.
Am J Clin Oncol ; 21(3): 317-21, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9626808

RESUMEN

The authors evaluate the efficacy and feasibility of dose-intensive doxorubicin and ifosfamide combination chemotherapy in patients with sarcomas. From January 1995 to April 1996, 33 evaluable patients with either metastatic sarcoma or primary sarcomas with a high-risk for metastases (all except one was previously untreated with chemotherapy) were treated on two consecutive protocols. The median age was 45 years (range, 15-68 years). The first protocol included doxorubicin at 75 mg/m2 given as a 72-hour infusion on days 1 to 3 along with ifosfamide at 2 g/m2/d over 2 hours x 5, days 1 to 5 (protocol AI 75/10). Granulocyte colony-stimulating factor (G-CSF) was used only if indicated according to American Society of Clinical Oncology guidelines. The second protocol included doxorubicin at 90 mg/m2 as a 72-hour continuous infusion and ifosfamide at 2.5 g/m2/d for 4 days (protocol AI 90/10) with prophylactic G-CSF. A median of four cycles were administered (range, 1-6). Three patients achieved a pathologic complete response (CR) and 18 patients achieved a partial response (PR) for a response rate (RR) of 64% (95% confidence interval (CI), 45-80%). Response rate for the subset of patients with soft-tissue sarcomas was 66% (95% CI, 46-82%). Only three patients progressed on therapy. Febrile neutropenia was noted in 31% of cycles at AI 75/10 and in 56% of cycles at AI 90/10. One patient developed reversible grade 3 central nervous system (CNS) toxicity. There was one treatment-related death on AI 90/10 secondary to doxorubicin cardiac toxicity at a cumulative dose of 435 mg/m2. Dose-intensive doxorubicin plus ifosfamide is feasible in appropriately selected patients and appears to be a very active regimen in patients with sarcomas. The authors are currently testing this regimen with G-CSF and thrombopoietin.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sarcoma/tratamiento farmacológico , Adolescente , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Supervivencia sin Enfermedad , Doxorrubicina/administración & dosificación , Femenino , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Ifosfamida/administración & dosificación , Masculino , Persona de Mediana Edad , Sarcoma/patología
13.
Exp Hematol ; 26(3): 207-16, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9502616

RESUMEN

Thrombopoietin (TPO), the primary regulator of megakaryocytopoiesis, also mediates biologic effects in vitro on hematopoietic cells more primitive than those committed to the megakaryocyte (MK) lineage. To assess the spectrum of hematopoietic effects of recombinant human (rh)TPO in vivo, we evaluated its proliferative effect on bone marrow (BM) progenitor cells, its maturation effect on BM MKs, and its mobilizing effect on peripheral blood (PB) progenitor cells during a phase I clinical laboratory investigation in which rhTPO was administered to cancer patients with normal hematopoiesis. Twelve patients received a single dose of rhTPO (0.3, 0.6, 1.2, or 2.4 microg/kg of body weight) prior to chemotherapy. BM and PB samples from these patients were analyzed 1 to 2 days before (baseline) and 7 days after rhTPO administration. At higher doses (1.2-2.4 microg/kg), rhTPO produced increased concentrations of primitive CD34+Thy-1+Lin-cells (mean 2.1-fold), CD34+mpl+ cells (mean 5.2-fold), CD34+CD41+CD14- promegakaryoblasts (mean 2.9-fold), and myeloerythroid colony-forming cells (mean threefold) in BM. No significant increases in the frequency of BM colony-forming unit (CFU)-MK were observed. Elevated numbers of both immature (2N-8N) and more mature (64N and 128N) CD41+ MKs were detected in BM, with modal ploidy remaining at 16N. Higher doses of rhTPO (1.2-2.4 microg/kg) also induced increased concentrations of CD34+ cell subsets in PB, including both primitive CD34+Thy-1+Lin- (mean 8.8-fold) and MK lineage-committed CD34+CD41+CD14- cells (mean 14.6-fold) as well as various myeloerythroid colony-forming cells (mean 3.6- to 5.5-fold). These results demonstrate that rhTPO given as a single dose not only promotes proliferation and maturation of cells of the MK lineage, but also expands the pool of BM primitive hematopoietic cells. In addition, rhTPO induces mobilization of hematopoietic progenitors into peripheral circulation. The extent to which such multilineage effects on human progenitor cells will contribute to clinical efficacy remains to be determined.


Asunto(s)
Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Hematopoyesis , Células Madre Hematopoyéticas/efectos de los fármacos , Megacariocitos/citología , Sarcoma/sangre , Trombopoyetina/farmacología , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recuento de Plaquetas/efectos de los fármacos , Ploidias , Proteínas Recombinantes
14.
Int J Radiat Oncol Biol Phys ; 40(2): 331-6, 1998 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9457817

RESUMEN

PURPOSE: Recent studies document the value of early combined modality therapy of small cell lung cancer, but also indicate that early thoracic radiation adds to myelosuppression and can complicate further chemotherapy. Other studies indicate that simultaneous use of growth factors with thoracic radiation may be deleterious. However, temporal separation of growth factor use from cytotoxic therapy may allow dose intensity to be maintained/enhanced during combined modality treatment. We sought to integrate filgrastim into a novel chemoradiation regimen for patients with limited small cell lung cancer using an approach that separated growth factor administration from both chemotherapy and thoracic radiation. METHODS AND MATERIALS: Twenty-seven patients with limited disease small cell lung cancer were enrolled in a Phase I trial of cisplatin, ifosfamide/mesna, oral etoposide, and thoracic radiation (1.5 Gy b.i.d. x 30 fractions days 1-19 cycle 1) +/- filgrastim (5 microg/kg/day). Filgrastim was given on days 20-25 of cycle 1 after completion of radiation and following completion of oral etoposide in subsequent cycles. The primary end point was determination of maximum tolerated dose (MTD) of chemotherapy. Serial cohorts were treated with and without filgrastim. RESULTS: Because of dose-limiting thrombocytopenia, primarily, and nonhematologic toxicity, the MTDs with and without filgrastim were identical (cisplatin 20 mg/m2 i.v. and ifosfamide 1200 mg/m2 i.v., both given days 1-3, and etoposide 40 mg/m2 p.o. days 1-14). Filgrastim use shortened the duration of neutropenia at the MTD (median 4 vs. 7 days), but was not associated with a reduction in febrile neutropenia. Although growth factor administration did not allow dose escalation of this regimen, it did allow chemotherapy doses to be maintained at the MTD more frequently through four cycles of therapy. In the 24 evaluable patients, the overall response rate was 100% (71% partial and 29% complete). CONCLUSIONS: Despite careful attention to the timing of growth factor with chemoradiation, the administration of filgrastim with this regimen did not allow dose escalation. As in many other recent studies of hematopoietic growth factors given prophylactically with chemotherapy, the duration of neutropenia at the MTD was shortened and the need for dose reduction throughout treatment was reduced in patients receiving filgrastim at the MTD.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/radioterapia , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Neutropenia/prevención & control , Adulto , Anciano , Carcinoma de Células Pequeñas/patología , Cisplatino/administración & dosificación , Estudios de Cohortes , Terapia Combinada , Supervivencia sin Enfermedad , Etopósido/administración & dosificación , Femenino , Fiebre/etiología , Filgrastim , Humanos , Ifosfamida/administración & dosificación , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Neutropenia/etiología , Proteínas Recombinantes , Trombocitopenia/etiología , Trombocitopenia/prevención & control
15.
Stem Cells ; 16 Suppl 2: 199-206, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-11012192

RESUMEN

Patients undergoing anticancer therapy are often at risk for developing severe and/or prolonged posttreatment thrombocytopenia. This can be associated with significant bleeding; currently, it is treated with supportive platelet transfusions. Frequent platelet transfusions can cause alloimmunization which requires HLA-matched donors and more frequent blood transfusions, and transmission of both viral and bacterial infections via platelet transfusions remains a concern. Furthermore, thrombocytopenia can mandate a decrease in the dose intensity of cytotoxic therapy by causing either delays or dose reductions in therapy administration. An intervention that reduces the risk or shortens the duration of severe thrombocytopenia would represent an important medical advance. Thrombopoietin (TPO), a naturally occurring, glycosylated polypeptide that was cloned by Genentech in 1994, is capable of inducing differentiation of stem cells into megakaryocytes and accelerating the maturation of megakaryocytes, thereby increasing the platelet count. Recombinant human TPO (rHuTPO) is currently undergoing testing in phase 1 and 2 studies in patients receiving myelosuppressive or myeloablative therapy. For the purposes of illustration, preliminary safety and activity data from one ongoing phase 1 myelosuppression trial (rHuTPO in women with advanced gynecologic malignancies receiving carboplatin) and one ongoing phase 1 myeloablation trial (rHuTPO for peripheral blood progenitor cell mobilization prior to myeloablative chemotherapy for high risk breast cancer) will be presented.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Radioterapia/efectos adversos , Proteínas Recombinantes/administración & dosificación , Trombocitopenia/tratamiento farmacológico , Trombopoyetina/administración & dosificación , Adolescente , Adulto , Anciano , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Quimioterapia Combinada , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Humanos , Persona de Mediana Edad , Proteínas Recombinantes/efectos adversos , Trombopoyetina/efectos adversos
16.
Anticancer Drugs ; 9(6): 577-9, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9877247

RESUMEN

Platinum-based chemotherapy is the standard treatment for advanced ovarian cancer, with response rates of 40-60%. In patients who fail platinum treatment, paclitaxel has resulted in response rates of 10-48%. Docetaxel has partial non-cross-resistance with and is twice as potent in vitro as paclitaxel in inhibiting microtubule disaggregation. The combination of docetaxel and cyclophosphamide is synergistic in pre-clinical studies and clinically active in breast cancer. We present the case of a patient with platinum and paclitaxel refractory ovarian cancer who achieved a remission with docetaxel and cyclophosphamide.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/tratamiento farmacológico , Cistadenocarcinoma Papilar/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Taxoides , Antígeno Ca-125/sangre , Carcinoma/sangre , Carcinoma/patología , Ciclofosfamida/administración & dosificación , Cistadenocarcinoma Papilar/sangre , Cistadenocarcinoma Papilar/patología , Docetaxel , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación , Paclitaxel/análogos & derivados
17.
J Clin Oncol ; 15(6): 2378-84, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9196153

RESUMEN

PURPOSE: To evaluate the efficacy and feasibility of high-dose ifosfamide (HDI) at a total dose of 14 g/m2 per cycle with mesna in combination with granulocyte colony-stimulating factor (G-CSF) in adult patients with sarcomas. PATIENTS AND METHODS: Between July 1991 and February 1994, 74 patients with sarcomas (37 bone and 37 soft tissue) were treated on two simultaneous phase II studies that evaluated HDI given as a continuous infusion over 74 hours. G-CSF was started on day 5 at 5 microg/kg/d until recovery of granulocyte count. Additionally, between March 1993 and March 1994, 15 similar patients with previously treated bone or soft tissue sarcomas were treated on a pilot study in which the same total dose of ifosfamide was administered by a bolus schedule, along with mesna and G-CSF. Patients were treated until maximal response, and where possible, surgical resection of gross disease was performed. RESULTS: Seventy-two patients from the phase II study using continuous infusion are assessable for response. Four complete responses (CRs) and 17 partial responses (PRs) were noted, for an overall response rate of 29% (95% confidence interval [CI], 19% to 39%). The response rate was 40% (95% CI, 24% to 56%) for bone sarcomas and 19% (95% CI, 6% to 32%) for soft tissue sarcomas. Fourteen patients from the pilot study that used a bolus schedule are assessable for response. One CR and seven PRs were noted, for an overall response rate of 57% (95% CI, 31% to 83%) and a response rate of 45% for soft tissue sarcomas. Two patients developed grade 3 to 4 renal toxicity, three developed grade 3 CNS toxicity, one had possible grade 3 cardiac toxicity, and two developed severe painful peripheral neuropathy. There were no treatment-related deaths. CONCLUSION: HDI at 14 g/m2 with mesna and G-CSF is an active salvage regimen for patients with bone and soft tissue sarcomas. There is a definite positive dose-response curve, and bolus administration appears to be more active than continuous infusion.


Asunto(s)
Antineoplásicos Alquilantes/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Ifosfamida/administración & dosificación , Sarcoma/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Adolescente , Adulto , Anciano , Antineoplásicos Alquilantes/efectos adversos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Quimioterapia Combinada , Femenino , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Ifosfamida/efectos adversos , Masculino , Mesna/uso terapéutico , Persona de Mediana Edad , Proyectos Piloto , Resultado del Tratamiento
18.
Ann Intern Med ; 126(9): 673-81, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9139552

RESUMEN

BACKGROUND: Thrombocytopenia is frequently encountered in patients with cancer. It is associated with an increased risk for clinically important bleeding episodes, which increases the demand for platelet transfusion. OBJECTIVE: To assess hematopoietic response to and clinical tolerance of recombinant human thrombopoietin, a recently cloned novel cytokine. DESIGN: Phase I and II clinical cohort study. SETTING: The University of Texas M.D. Anderson Cancer Center, Houston, Texas. PATIENTS: 12 patients with sarcoma who had high risk for severe chemotherapy-induced thrombocytopenia. INTERVENTION: A single intravenous dose of thrombopoietin (0.3 to 2.4 micrograms/kg of body weight) 3 weeks before chemotherapy. MEASUREMENTS: Peripheral blood and bone marrow evaluation before and after thrombopoietin administration. RESULTS: A single dose of thrombopoietin was associated with an increase in platelet counts (mean increase from baseline, 61% to 213%; P = 0.002) in a dose-related manner. This increase began by day 4 in most patients and peaked on a median of day 12. This sustained response was associated with a prolonged serum thrombopoietin half life (20 to 30 hours). The platelets appeared morphologically normal and showed normal aggregation in response to various agonists. Platelet response was accompanied by a dose-related increase in bone marrow megakaryocytes (as much as 4-fold); the expansion of the bone marrow progenitors of myeloid, erythroid, multipotential, and megakaryocytic lineages; and the marked mobilization of progenitors (maximum, 5.7-fold to 10-fold) of multiple cell lineages in the peripheral blood. Treatment was well tolerated, and no serious adverse events occurred. CONCLUSIONS: Thrombopoietin, administered as a single dose, is a potent stimulus for prolonged platelet production in humans. It merits further evaluation for the prevention and treatment of thrombocytopenia.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , Neoplasias/sangre , Trombocitopenia/prevención & control , Trombopoyetina/administración & dosificación , Anticuerpos/sangre , Antineoplásicos/efectos adversos , Plaquetas/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Humanos , Inyecciones Intravenosas , Megacariocitos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Recuento de Plaquetas/efectos de los fármacos , Ploidias , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Trombocitopenia/inducido químicamente , Trombopoyetina/farmacocinética
20.
J Clin Oncol ; 15(3): 1218-34, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9060566

RESUMEN

PURPOSE: To study the impact of Procrit (epoetin alfa; Amgen Inc, Thousand Oaks, CA) on quality of life, transfusion requirements, and hemoglobin in anemic cancer patients receiving chemotherapy. PATIENTS AND METHODS: More than 500 community-based oncologists enrolled 2,342 patients with malignancies undergoing cytotoxic chemotherapy in an open-label study. Patients were treated with epoetin alfa 150 U/kg three times weekly, which could be doubled if the therapuetic response was judged inadequate. Total treatment was up to 4 months. RESULTS: Of the 2,342 patients enrolled, data were available for 2,030 patients. Of the 2,030, 1,047 patients completed all 4 months of epoetin alfa therapy. Epoetin alfa was associated with significant increases in mean self-rated scores for energy level, activity level, and overall quality of life; these improvements correlated with the magnitude of the hemoglobin increase and were independent of tumor response. In addition, epoetin alfa was associated with a significant increase in mean hemoglobin and with a significant decrease in the proportion of patients requiring transfusions (baseline to final value, P < .001). Epoetin alfa was well tolerated. CONCLUSION: Epoetin alfa is effective in improving the functional status and quality of life in anemic cancer patients receiving chemotherapy, as well as increasing hemoglobin level and decreasing transfusion requirements. Improvement in functional status can be attributed to an increase in hemoglobin level, demonstrating that quality of life in this group of patients can be improved by aggressively treating anemia. Further studies will be required to define the optimal doses and schedules for epoetin alfa.


Asunto(s)
Anemia/tratamiento farmacológico , Transfusión Sanguínea/estadística & datos numéricos , Eritropoyetina/uso terapéutico , Hematínicos/uso terapéutico , Hemoglobinas/efectos de los fármacos , Neoplasias/sangre , Calidad de Vida , Anemia/sangre , Anemia/inducido químicamente , Epoetina alfa , Femenino , Hemoglobinas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Recombinantes , Diálisis Renal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...