Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Int J Lab Hematol ; 36(4): 459-64, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24400650

RESUMO

INTRODUCTION: Atypical chronic lymphocytic leukemia (aCLL) is a morphologic variant found in approximately 25% of patients with chronic lymphocytic leukemia (CLL). Although aCLL has a more aggressive course compared to typical CLL (tCLL), it is not usually reported. This retrospective study used digital microscopy to morphologically classify CLL patients as aCLL or tCLL, and determined the prevalence of prognostic markers in each group. METHODS: CellaVision AB (Lund, Sweden) was used to evaluate lymphocyte morphology on archived blood films of 97 CLL patients, and results of their prognostic marker analysis at diagnosis were obtained. The unpaired t-test, Chi-square, or Fisher's Exact test were used for statistical analysis. RESULTS: 27% of CLL cases were morphologically classified as aCLL. The aCLL group had a higher prevalence of trisomy 12, unmutated IgVH, and CD38 expression (markers associated with poor prognosis), and a lower prevalence of 13q14 deletions compared to tCLL; this was statistically significant. CONCLUSION: Using digital imaging to identify aCLL is feasible, economical, and may provide clinically relevant prognostic information at diagnosis and during periodic monitoring. Further study of a larger number of patients is needed to assess the clinical utility of reporting aCLL morphology.


Assuntos
Linfócitos B/patologia , Deleção Cromossômica , Cromossomos Humanos Par 13 , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Trissomia , ADP-Ribosil Ciclase 1/genética , Cromossomos Humanos Par 12 , Feminino , Expressão Gênica , Humanos , Processamento de Imagem Assistida por Computador , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia Linfocítica Crônica de Células B/diagnóstico , Masculino , Microscopia/métodos
3.
Curr Top Microbiol Immunol ; 313: 205-19, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17217045

RESUMO

Acute promyelocytic leukemia (APL) is a rare subtype of acute myeloid leukemia (AML) for which a number of targeted therapies have been developed. The "targets" have included both genotypic and phenotypic features of the disease. The application of monoclonal antibodies (MAbs) to this disease to date have been limited to a relatively small number of studies where this therapy has been used to supplement effective approaches to the disease. The preliminary results have been promising, and further development of this modality as an effective adjunct to existing treatment regimens will most certainly occur in the near future.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Aminoglicosídeos/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Gemtuzumab , Células HL-60 , Humanos , Camundongos , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Resultado do Tratamento
4.
Leukemia ; 20(2): 212-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16357841

RESUMO

Epigenetic mechanisms underlying tumorigenesis have recently received much attention as potential therapeutic targets of human cancer. We designed a pilot study to target DNA methylation and histone deacetylation through the sequential administration of 5-azacytidine followed by sodium phenylbutyrate (PB) in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Ten evaluable patients (eight AML, two MDS) were treated with seven consecutive daily subcutaneous injections of 5-azacytidine at 75 mg/m2 followed by 5 days of sodium PB given intravenously at a dose of 200 mg/kg. Five patients (50%) were able to achieve a beneficial clinical response (partial remission or stable disease). One patient with MDS proceeded to allogeneic stem cell transplantation and is alive without evidence of disease 39 months later. The combination regimen was well tolerated with common toxicities of injection site skin reaction (90% of the patients) from 5-azacytidine, and somnolence/fatigue from the sodium PB infusion (80% of the patients). Correlative laboratory studies demonstrated the consistent reacetylation of histone H4, although no relationship with the clinical response could be demonstrated. Results from this pilot study demonstrate that a combination approach targeting different mechanisms of transcriptional modulation is clinically feasible with acceptable toxicity and measurable biologic and clinical outcomes.


Assuntos
Antineoplásicos/uso terapêutico , Azacitidina/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Fenilbutiratos/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Acetilação/efeitos dos fármacos , Doença Aguda , Adulto , Idoso , Antimetabólitos Antineoplásicos/uso terapêutico , Metilação de DNA , Esquema de Medicação , Quimioterapia Combinada , Feminino , Histonas/efeitos dos fármacos , Histonas/metabolismo , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/genética , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/genética , Projetos Piloto , Resultado do Tratamento
5.
Cancer Genet Cytogenet ; 127(1): 71-3, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11408069

RESUMO

A case of acute myelocytic leukemia with a translocation (5;18)(q35;q21) is reported. Cytogenetic abnormalities of the long arm of chromosome 5 have long been known to affect hematopoiesis. Although translocations between 5q and other chromosomes have been associated with malignancy, this is the first reported case of a t(5;18) resulting in acute myeloid leukemia. Possible molecular mechanisms underlying the pathogenesis of the disease are discussed.


Assuntos
Cromossomos Humanos Par 18/genética , Cromossomos Humanos Par 5/genética , Leucemia Mieloide Aguda/genética , Translocação Genética , Adulto , Medula Óssea/patologia , Bandeamento Cromossômico , Feminino , Humanos , Imunofenotipagem
6.
J Clin Oncol ; 19(7): 1985-92, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11283131

RESUMO

PURPOSE: Flavopiridol is the first cyclin-dependent kinase inhibitor to enter clinical trials. Activity in gastric cancer xenografts and in a patient with gastric cancer on the phase I trial led to this phase II study of flavopiridol in patients with metastatic gastric cancer. PATIENTS AND METHODS: Sixteen patients were entered onto the study, and 14 were assessable for response. Flavopiridol was administered initially at a dose of 50 mg/m(2)/d by continuous infusion for 72 hours every 2 weeks. Assessment of plasma pharmacokinetics was performed in all patients. Peripheral mononuclear cells were collected throughout the 72-hour infusion for determinants of apoptosis. RESULTS: There were no major objective responses (exact confidence interval 0% to 23%). One patient achieved a minor response in his liver metastases, though the primary progressed. Other patients exhibited histologic and radiographic evidence of tumor necrosis. Common toxicities included fatigue in 93% of patients (grade 3 or 4 in 27%) and diarrhea in 73% of patients (grade 3 or 4 in 20%). Five patients (33%) developed venous thromboses at the central catheter tip. The studies performed on peripheral mononuclear cells indicated no induction of apoptosis. CONCLUSION: Flavopiridol administered as a single agent for 72 hours every 14 days is inactive in the treatment of gastric cancer. The drug also induced an unexpected higher incidence of vascular thrombosis and fatigue than was anticipated from the phase I trials. Future development of flavopiridol will depend on other doses and schedules in combination with chemotherapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Quinases Ciclina-Dependentes/antagonistas & inibidores , Flavonoides/uso terapêutico , Piperidinas/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Adenocarcinoma/patologia , Adulto , Idoso , Antineoplásicos/farmacologia , Fadiga/induzido quimicamente , Feminino , Flavonoides/farmacologia , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Piperidinas/farmacologia , Neoplasias Gástricas/patologia , Trombose Venosa/etiologia
7.
Blood ; 97(7): 1929-36, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11264154

RESUMO

Patient-tumor-specific oligonucleotides were generated for the detection of minimal residual disease (MRD) in a highly specific and sensitive clonotypic polymerase chain reaction (cPCR). The clone-specific region of highest diversity, CDR-III, was PCR amplified and sequenced. Nested CDR-III clonotypic primers were used in a semi-nested cPCR with a sensitivity of at least 1 in 10(5) cells. Patients with protocol-eligible Rai intermediate or high-risk chronic lymphocytic leukemia (CLL) received induction with fludarabine 25 mg/m(2) per day for 5 days every 4 weeks for 6 cycles, followed by consolidative high-dose cyclophosphamide (1.5, 2.25, or 3g/m(2)). cPCR was performed on peripheral blood and bone marrow mononuclear cells. All 5 patients achieving a clinical partial remission (PR) studied by cPCR were positive. Five patients achieved nodular PR (nPR) (residual nodules or suspicious lymphocytic infiltrates in a bone marrow biopsy as the sole suggestion of residual disease). Five of 5 patients with nPR were cPCR positive. In contrast, flow cytometry for CD5-CD19 dual staining and kappa--lambda clonal excess detected MRD in only 3 of the same 5 nPR patients, all of whom were cPCR positive, and immunohistochemistry detected MRD in only 1 of 4 assessable patients. Three of 7 CR patients evaluable by cPCR had MRD. Only 1 CR patient had MRD by flow cytometry; that patient was also cPCR positive. These data support the conclusions that nodular PR in CLL represents MRD and that clonotypic PCR detects MRD in CLL more frequently than flow cytometry or immunohistochemistry. (Blood. 2001;97:1929-1936)


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Linfocítica Crônica de Células B/patologia , Reação em Cadeia da Polimerase/métodos , Vidarabina/análogos & derivados , Antígenos CD/análise , Biomarcadores Tumorais , Medula Óssea/química , Medula Óssea/patologia , Células Clonais/química , Células Clonais/patologia , Ciclofosfamida/administração & dosagem , Citometria de Fluxo , Rearranjo Gênico de Cadeia Pesada de Linfócito B , Genes de Imunoglobulinas , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias kappa de Imunoglobulina/análise , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/análise , Cadeias lambda de Imunoglobulina/genética , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/genética , Infiltração Leucêmica , Linfonodos/química , Linfonodos/patologia , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/genética , Neoplasia Residual , Indução de Remissão , Sensibilidade e Especificidade , Resultado do Tratamento , Vidarabina/administração & dosagem
8.
Expert Opin Investig Drugs ; 9(6): 1197-205, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11060736

RESUMO

Although the standard approach to myeloid leukaemias remains chemotherapy, the agents currently available rarely result in cure. Recent advances in understanding the biology of these disorders have lead to the development of targeted treatment strategies. In acute promyelocytic leukaemia (APL), all-trans retinoic acid (ATRA), sodium phenylbutyrate and arsenic trioxide are agents which either induce differentiation or apoptosis and have been used to successfully achieve remission. The tyrosine kinase inhibitor, STI-571, antisense oligonucleotides, and bcr-abl vaccines are strategies which focus on the oncogenic events in chronic myelogenous leukaemia (CML). Two anti-CD33 monoclonal antibody conjugates, Y90-HuM195 and CMA-676, have been used in acute myelogenous leukaemia (AML) and have shown some efficacy. Although the preliminary results with these targeted therapies are promising, further studies are needed to establish them as effective, less toxic alternatives to the current standard of care.


Assuntos
Antineoplásicos/uso terapêutico , Leucemia Mieloide/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Humanos , Leucemia Mieloide/patologia
9.
Leukemia ; 14(9): 1577-82, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10995003

RESUMO

Fludarabine is the most active agent in the treatment of chronic lymphocytic leukemia (CLL). Despite this activity only a minority of patients treated with fludarabine achieve a complete response. We evaluated a new treatment program of sequential therapy with fludarabine followed by high-dose cyclophosphamide in previously untreated patients with CLL. This report details the results in 25 patients with previously untreated CLL. Patients received fludarabine (25 mg/m2/day x 5 days every 4 weeks for six cycles) as induction followed by consolidation with high-dose cyclophosphamide at one of three dose levels 1.5 g/m2, 2.25 g/m2, or 3 g/m2 administered every 2 weeks for three doses. High-dose cyclophosphamide was given with G-CSF support (5 microg/kg/day days 3-12). Complete response (CR) required a normal physical examination, normal CBC, a normal bone marrow evaluation including no residual lymphoid nodules on biopsy. A nodular response was defined as a complete response with the exception of an occasional residual nodule seen on bone marrow biopsy. Flow cytometric analysis for CD5:CD19 dual staining and kappa/lambda clonal excess was performed in all patients as a sensitive measure of minimal residual disease (MRD). Selected patients had patient/tumor-specific oligonucleotides generated that were subsequently used in a polymerase chain reaction as an extremely sensitive measure of MRD. There were no treatment-related deaths and no patient encountered unacceptable toxicity. After completion of this sequential regimen 76% (95% confidence interval: 59-93%) of patients had a major response: eight (32%) achieved a CR, four (16%) a nodular response, seven (28%) a PR, and six patients (24%) failed. Four patients withdrew from study during induction with fludarabine and did not receive at least one cycle of cyclophosphamide. Of the 21 patients who received consolidation with cyclophosphamide 10 (48%) had an improved quality of response when compared to that achieved with fludarabine. Two patients (8%) had no disease detectable by flow cytometry ('flow cytometric' CR) after six cycles of fludarabine. This improved to nine patients (36%) after high-dose cyclophosphamide. Following consolidation with high-dose cyclophosphamide three patients (12%) tested negative by PCR. All of these patients had morphologic evidence of residual disease after six cycles of fludarabine. Consolidation with high-dose cyclophosphamide increased the fraction of patients achieving a nodular response or CR three-fold (16% to 48%). This appears to be clinically relevant because with a median follow-up of 52 (range 34-78) months the projected 6-year survival for patients achieving a CR or NR is 91% compared to 41% for all others (P = 0.012). We conclude that sequential therapy with fludarabine followed by high-dose cyclophosphamide in previously untreated patients with CLL is safe and can improve the quality of response in a large proportion of patients compared to therapy with fludarabine alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclofosfamida/uso terapêutico , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Vidarabina/análogos & derivados , Vidarabina/uso terapêutico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/mortalidade , Masculino , Pessoa de Meia-Idade , Indução de Remissão , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/efeitos adversos
11.
N Engl J Med ; 339(19): 1341-8, 1998 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-9801394

RESUMO

BACKGROUND: Two reports from China have suggested that arsenic trioxide can induce complete remissions in patients with acute promyelocytic leukemia (APL). We evaluated this drug in patients with APL in an attempt to elucidate its mechanism of action. METHODS: Twelve patients with APL who had relapsed after extensive prior therapy were treated with arsenic trioxide at doses ranging from 0.06 to 0.2 mg per kilogram of body weight per day until visible leukemic cells were eliminated from the bone marrow. Bone marrow mononuclear cells were serially monitored by flow cytometry for immunophenotype, fluorescence in situ hybridization, reverse-transcription-polymerase-chain-reaction (RT-PCR) assay for PML-RAR-alpha fusion transcripts, and Western blot analysis for expression of the apoptosis-associated proteins caspases 1, 2, and 3. RESULTS: Of the 12 patients studied, 11 achieved complete remission after treatment that lasted from 12 to 39 days (range of cumulative doses, 160 to 495 mg). Adverse effects were relatively mild and included rash, lightheadedness, fatigue, and musculoskeletal pain. Cells that expressed both CD11b and CD33 (antigens characteristic of mature and immature cells, respectively), and which were found by fluorescence in situ hybridization to carry the t(15;17) translocation, increased progressively in number during treatment and persisted in the early phase of complete remission. Eight of 11 patients who initially tested positive for the PML-RAR-alpha fusion transcript by the RT-PCR assay later tested negative; 3 other patients, who persistently tested positive, relapsed early. Arsenic trioxide induced the expression of the proenzymes of caspase 2 and caspase 3 and activation of both caspase 1 and caspase 3. CONCLUSIONS: Low doses of arsenic trioxide can induce complete remissions in patients with APL who have relapsed. The clinical response is associated with incomplete cytodifferentiation and the induction of apoptosis with caspase activation in leukemic cells.


Assuntos
Antineoplásicos/uso terapêutico , Arsenicais/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Óxidos/uso terapêutico , Adolescente , Adulto , Idoso , Antígenos CD/análise , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Apoptose , Trióxido de Arsênio , Arsenicais/administração & dosagem , Arsenicais/efeitos adversos , Células da Medula Óssea/imunologia , Caspases , Diferenciação Celular , Criança , Humanos , Imunofenotipagem , Leucemia Promielocítica Aguda/imunologia , Leucemia Promielocítica Aguda/patologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Pessoa de Meia-Idade , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/efeitos dos fármacos , Proteínas de Fusão Oncogênica/análise , Proteínas de Fusão Oncogênica/efeitos dos fármacos , Óxidos/administração & dosagem , Óxidos/efeitos adversos , Recidiva , Indução de Remissão/métodos
12.
Semin Hematol ; 35(3): 253-60, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9685171

RESUMO

Suppression of normal hematopoiesis is a frequent complication of cancer or its treatment. The basis for dose-intensified cancer therapy in the 1990s was the discovery that hematopoietic growth factors and peripheral blood progenitor cell infusions can ameliorate some of its associated hematologic toxicities. Both granulocyte colony-stimulating factor and granulocyte macrophage colony-stimulating factor accelerate neutrophil recovery after chemotherapy and can mobilize peripheral blood progenitor cells for use in autologous or allogeneic transplantation. Unfortunately, the duration and severity of chemotherapy-induced thrombocytopenia is unaffected by the use of these myeloid growth factors. During the last 5 years, the activities of a variety of potential platelet or megakaryocyte-stimulating factors have been determined in clinical trials. The results of these studies are described.


Assuntos
Interleucina-11/uso terapêutico , Interleucina-3/uso terapêutico , Interleucina-6/uso terapêutico , Fator de Células-Tronco/uso terapêutico , Trombocitopenia/tratamento farmacológico , Humanos , Proteínas Recombinantes/uso terapêutico
13.
Leukemia ; 12(6): 865-8, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9639412

RESUMO

Relapsed or refractory adult acute lymphoblastic leukemia (ALL) carries a grave prognosis. The most promising strategy for curing these patients is through re-induction chemotherapy followed by successful allogeneic transplant. We studied a new high-dose induction regimen in order to improve the outcome for these patients. Eighteen adult patients with relapsed/refractory ALL were treated on a phase I study of high-dose cytarabine combined with a single escalating dose of idarubicin. Five patients had primary refractory disease and 13 were treated in refractory relapse. Nine patients (50%) had Ph+ ALL. The induction regimen was cytarabine 3 g/m2/day intravenously days 1-5 and idarubicin as a single intravenous dose on day 3. G-CSF 5 microg/kg subcutaneously every 12 h was started on day 7. The initial idarubicin dose was 20 mg/m2 with dose escalations of 10 mg m2. Cohorts of three patients were treated at each idarubicin dose level. Unacceptable toxicity was encountered at 50 mg/m2 with one death from infection and one death from cardiotoxicity in a patient with significant prior anthracycline exposure. There were no instances of grade 4 non-hematologic toxicity encountered at idarubicin doses of 20 mg/m2, 30 mg/m2, or 40 mg/m2. The data suggest a dose-response relationship for increasing doses of idarubicin with 0/3 complete responses (CR) at 20 mg/m2, 1/3 CR at 30 mg/m2, and 7/12 (58%) CR at idarubicin doses > or = 40 mg/m2. We conclude that concomitant administration of cytarabine 3 g/m2/day x 5 and high-dose idarubicin at 40 mg/m2 as a single dose on day 3 can be administered safely to patients with refractory and relapsed ALL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Feminino , Humanos , Idarubicina/administração & dosagem , Idarubicina/efeitos adversos , Masculino , Pessoa de Meia-Idade
14.
Clin Cancer Res ; 4(2): 311-6, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9516916

RESUMO

The objective of this study was to identify factors associated with poor mobilization of peripheral blood progenitor cells (PBPCs) or delayed platelet engraftment after high-dose therapy and autologous stem cell transplantation in patients with lymphoma. Fifty-eight patients with Hodgkin's disease or non-Hodgkin's lymphoma underwent PBPC transplantation as the "best available therapy" at Memorial Sloan-Kettering Cancer Center (New York, NY) between 1993 and 1995. PBPCs were mobilized with either granulocyte colony-stimulating factor (G-CSF) alone (n = 19) or G-CSF following combination chemotherapy (n = 39). Forty-eight of these patients underwent a PBPC transplant, receiving a conditioning regimen containing cyclophosphamide, etoposide, and either total body irradiation, total lymphoid irradiation, or carmustine. A median number of 4.6 x 10(6) CD34+ cells/kg were obtained with a median of three leukapheresis procedures. Mobilization of PBPCs using chemotherapy plus G-CSF was superior to G-CSF alone (6.7 x 10(6) versus 1.5 x 10(6) CD34+ cells/kg; P = 0.0002). Poorer mobilization of progenitor cells was observed in patients who had previously received stem cell-toxic chemotherapy, including (a) nitrogen mustard, procarbazine, melphalan, carmustine or > 7.5 g of cytarabine chemotherapy premobilization (2.0 x 10(6) versus 6.0 x 10(6) CD34+ cells/kg; P = 0.005), or (b) > or = 11 cycles of any previous chemotherapy (2.6 x 10(6) versus 6.7 x 10(6) CD34+ cells/kg; P = 0.02). Platelet recovery to > 20,000/microliter was delayed in patients who received < 2.0 x 10(6) CD34+ cells (median, 13 versus 22 days; P = 0.06). Patients who received > or = 11 cycles of chemotherapy prior to PBPC mobilization tended to have delayed platelet recovery to > 20,000/microliter and to require more platelet transfusions than less extensively pretreated patients (median, 13.5 versus 23.5 days; P = 0.15; median number of platelet transfusion episodes, 13 versus 9; P = 0.17). These data suggest that current strategies to mobilize PBPCs may be suboptimal in patients who have received either stem cell-toxic chemotherapy or > or = 11 cycles of chemotherapy prior to PBPC mobilization. Alternative approaches, such as ex vivo expansion or the use of other growth factors in addition to G-CSF, may improve mobilization of progenitor cells for PBPC transplantation.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Doença de Hodgkin/terapia , Linfoma não Hodgkin/terapia , Análise de Variância , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Transfusão de Eritrócitos , Feminino , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Doença de Hodgkin/sangue , Doença de Hodgkin/tratamento farmacológico , Humanos , Tempo de Internação , Leucaférese , Linfoma não Hodgkin/sangue , Linfoma não Hodgkin/tratamento farmacológico , Masculino , Transfusão de Plaquetas , Resultado do Tratamento
15.
Clin Cancer Res ; 3(9): 1467-72, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9815832

RESUMO

Flavopiridol (L86-8275) is a synthetic flavone currently undergoing Phase I clinical trials. It is active against a series of human cancer cell lines and has been shown to inhibit a broad range of protein kinases, including cyclin-dependent kinases and protein kinase C (PKC). Previous studies have shown that the PKC-specific inhibitor safingol significantly enhances the induction of apoptosis by mitomycin-C (MMC) in gastric cancer cells. Because flavopiridol can potentially inhibit PKC, we elected to determine the extent to which flavopiridol would promote MMC-induced apoptosis in both gastric and breast cancer cells. For these studies, MKN-74 gastric cancer cells and MDA-MB-468 breast cancer cells were exposed to either no drug, 1 microgram/ml MMC alone, 300 nM flavopiridol alone, or a combination of chemotherapy with flavopiridol for 24 h. Sequence specificity was also examined by first exposing cells to MMC for 24 h followed by flavopiridol for 24 h or to the same drugs in the reverse order. Apoptosis was measured by quantitative fluorescence microscopy of nuclear chromatin condensation in cells stained with the dye, bisbenzimide trihydrochloride. Exposure of MKN-74 cells to flavopiridol alone induced apoptosis in 12 +/- 1% of the cells, and exposure to MMC alone induced apoptosis in 10 +/- 1%. However, the combination of flavopiridol and MMC increased the induction of apoptosis to 55 +/- 3% of the cells (P < 0.005 for the drug combination versus flavopiridol alone). Pretreatment with the PKC activator 3-phorbol 12-myristate 13-acetate only partially reversed this effect (43 +/- 1%; P < 0.025). In MDA-MB-468 cells, flavopiridol alone induced apoptosis in 17 +/- 1% of the cells, and MMC alone induced apoptosis in 10 +/- 1% of the cells. The combination of flavopiridol and MMC increased the percentage of MDA-MB-468 cells undergoing apoptosis to 58 +/- 4% (P < 0.005 for the drug combination versus flavopiridol alone). Sequential treatment with MMC followed by flavopiridol induced apoptosis in 63 +/- 2% of the MKN-74 cells (P < 0.05 versus the concomitant drug combination) and in 76 +/- 2% of the MDA-MB-468 cells (P < 0.025 versus the concomitant drug combination), whereas flavopiridol followed by MMC did not increase the induction of apoptosis in either cell line. As determined by the terminal deoxynucleotidyl transferase labeling of the 3' ends of DNA fragments produced in apoptotic cells, the induction of apoptosis with the combination of flavopiridol and MMC occurred to MKN-74 cells in all phases of the cell cycle (i.e., G0-G1, S, and G2-M). These results indicate that flavopiridol potentiates the cytotoxic effect of the chemotherapeutic agent MMC by promoting drug-induced apoptosis in tumor cells. Sequencing studies suggest that MMC followed by flavopiridol or simultaneous treatment is superior to flavopiridol followed by MMC. The enhancement of MMC-induced apoptosis by flavopiridol may be partially PKC dependent and is not associated with one specific region of the cell cycle.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Flavonoides/farmacologia , Mitomicina/farmacologia , Piperidinas/farmacologia , Neoplasias Gástricas/patologia , Ciclo Celular , Fragmentação do DNA , Esquema de Medicação , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Humanos , Estimulação Química , Células Tumorais Cultivadas/efeitos dos fármacos
17.
J Clin Oncol ; 14(9): 2480-5, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8823326

RESUMO

PURPOSE: To evaluate the efficacy and safety of a new induction regimen for adult acute lymphoblastic leukemia (ALL) that does not contain vincristine or corticosteroids. PATIENTS AND METHODS: Thirty-seven adult patients with newly diagnosed ALL and lymphoblastic lymphoma were treated with a dose-intense induction regimen. This regimen was designed to increase the fraction of patients achieving an early complete remission (CR) in an attempt to increase long-term disease-free survival. The induction regimen was cytarabine (Ara-C) 3 g/m2/d for 5 days and mitoxantrone 80 mg/m2 as a single dose on day 3. Granulocyte colony-stimulating factor (G-CSF) 200 micrograms/ m2/d beginning on day 7 was used to promote early myeloid recovery. RESULTS: There were 31 CRs (84%). Median time to CR was 34 days, median hospital stay was 28 days, and the median number of days with a neutrophil count less than 500/microL was 18. There were three patients with resistant disease who experienced treatment failure and three early deaths from sepsis. Four patients with Philadelphia chromosome-positive (Ph+) ALL achieved hematologic and cytogenetic CRs. CONCLUSION: This dose-intense induction regimen produced a high incidence of CRs with acceptable toxicity without the use of vincristine or corticosteroids. Comparisons with our prior vincristine/prednisone-based induction regimen (the L-20 protocol) suggest that patients treated on the current study were more likely to achieve a CR and that they achieved this remission earlier than patients treated with a traditional four-drug (vincristine, prednisone, doxorubicin, and cyclophosphamide) induction regimen.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Intervalo Livre de Doença , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Mitoxantrona/administração & dosagem , Mitoxantrona/efeitos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Prednisona/administração & dosagem , Prednisona/efeitos adversos , Indução de Remissão , Vincristina/administração & dosagem , Vincristina/efeitos adversos
18.
Am J Hematol ; 51(2): 117-21, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8579051

RESUMO

5-azacytidine (5-AZA) and carboplatin (CBDCA) are two agents which have demonstrated antileukemic activity in a number of phase I-II trials. Their mechanisms of action and pharmacology related to cell resistance suggested suitability for combination therapy. The aim of this pilot was to evaluate the effects of this combination in the treatment of patients with relapsed/refractory acute leukemia. A total of 21 patients was enrolled. 5-azacytidine, at doses ranging from 50-150 mg/m2/day, was administered as a 2-hr infusion for 5 consecutive days. On day 3, patients began a 5-day course of CBDCA given as a 24-hr continuous intravenous infusion of 250 mg/m2/day. There were no complete remissions with this regimen. Although there were three partial responses, these were generally of short duration. Nonhematologic toxicities were mild. No correlation was seen between response and serum platinum levels. These results demonstrate that the 5-AZA/CBDCA combination is ineffective therapy for heavily pretreated patients with acute leukemia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia/tratamento farmacológico , Adulto , Idoso , Azacitidina/administração & dosagem , Azacitidina/efeitos adversos , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Recidiva
19.
Leukemia ; 10(1): 32-9, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8558934

RESUMO

Given the high treatment-related mortality in elderly patients with acute myelogenous leukemia (AML), we undertook a study using granulocyte colony-stimulating factor (G-CSF) following chemotherapy in an effort to ameliorate toxicity. Patients ( > 60 years) received induction with idarubicin 12 mg/m2/day x 3 and cytosine arabinoside (Ara-C) 200 mg/m2/day x 5. A second course of chemotherapy consisting of mitoxantrone 12mg/m2/day x 3, etoposide (VP-16) 150 mg/m2/day x 3 and Ara-C 200 mg/m2/day x 4 was given approximately 1 month after achieving a complete remission (CR) or immediately if patients failed the first induction. Twenty-four hours following completion of the chemotherapy, G-CSF (10 micrograms/kg/day continuous i.v. infusion) was started. A historical control group of 28 patients treated without G-CSF was used for comparison. Twenty-six patients were evaluable for response. Following induction, the recovery of neutrophils to greater than 500/microliters and 1000/microliters was more raped in the responders who received G-CSF compared to historical controls (median 13 vs 17 days, P = 0.008; 14 vs 19 days, P = 0.005). The toxic death rate of 8% in the study group was significantly lower than the 32% mortality observed in the historical controls (P = 0.04). There was no difference in supportive care requirements or infectious complications. The complete remission (CR) rate was 58% in the entire study group with 71% of de novo AML patients achieving CR. Disease-free survival and overall survival were comparable between the study and historical control groups. These results indicate that G-CSF benefits elderly patients after intensive chemotherapy for AML by decreasing the duration of neutropenia. The reduced neutropenic period may have contributed to the small number of early toxic deaths.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Citarabina/administração & dosagem , Citarabina/efeitos adversos , Intervalo Livre de Doença , Etoposídeo/administração & dosagem , Etoposídeo/efeitos adversos , Feminino , Humanos , Idarubicina/administração & dosagem , Idarubicina/efeitos adversos , Leucemia Mieloide Aguda/mortalidade , Masculino , Mitoxantrona/administração & dosagem , Mitoxantrona/efeitos adversos , Neutropenia/induzido quimicamente , Neutropenia/prevenção & controle , Projetos Piloto , Indução de Remissão , Taxa de Sobrevida
20.
J Natl Cancer Inst ; 87(18): 1394-9, 1995 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-7658500

RESUMO

BACKGROUND: Protein kinase C (PKC) is a family of enzymes that function in processes relevant to carcinogenesis, tumor cell metastasis, and apoptosis. Safingol, an optical isomer (the L-threo enantiomer) of dihydrosphingosine, is a specific inhibitor of PKC and might represent a novel agent for anticancer therapy. Preclinical animal studies show that safingol alone has a minimal effect on tumor cell growth, but combining this compound with conventional chemotherapy agents dramatically potentiates their antitumor effects. It has been suggested that many chemotherapeutic agents exert their antitumor effects by inducing apoptosis. PURPOSE: We wanted to determine the extent to which safingol, alone or in combination with a standard chemotherapeutic agent (mitomycin C [MMC]), would promote apoptosis in gastric cancer cells in vitro. Furthermore, we investigated whether the induction of apoptosis in the treated cells was affected by their p53 tumor suppressor status or their drug-resistance status. METHODS: SK-GT-5 (p53-deficient and MMC-resistant) and MKN-74 (p53 wild-type and MMC-sensitive) gastric cancer cells were exposed to either no drug, safingol (50 microM) alone, MMC (5 micrograms/mL) alone, or a combination of safingol (50 microM) and MMC (5 micrograms/mL). In some experiments, cells were exposed simultaneously to safingol and the PKC activator, 3-phorbol 12-myristate 13-acetate (PMA), prior to treatment with MMC. Apoptosis was measured by two methods: 1) quantitative fluorescence microscopy of nuclear chromatin condensation in cells stained with the dye, bisbenzamide trihydrochloride (Hoechst-33258), and 2) terminal deoxynucleotidyl transferase (TdT) labeling of the 3'-OH ends of DNA fragments produced in apoptotic cells. RESULTS: As determined by quantitative fluorescence microscopy, exposure of SK-GT-5 cells to safingol alone induced apoptosis in 2% +/- 1% (mean +/- SD) of the cells, and MMC alone increased that level to 18% +/- 1%. However, the combination of safingol and MMC induced apoptosis in 39% +/- 1% of the cells (P < .001, for the drug combination versus MMC alone). With MKN-74 cells, safingol alone induced apoptosis in 8% +/- 3% of the cells, whereas MMC alone induced apoptosis in 40% +/- 4% of treated cells and the combination of safingol and MMC induced apoptosis in 83% +/- 4% of the cells. Similar results were obtained with the TdT assay. Simultaneous exposure of cells to safingol and PMA abrogated the safingol-mediated enhancement of MMC-induced apoptosis. CONCLUSIONS: The PKC inhibitor safingol enhances the cytotoxic effect of the chemotherapeutic agent MMC in gastric cancer cells by promoting drug-induced apoptosis. The induction of apoptosis occurs regardless of the p53 status or the drug-resistance status of the cells.


Assuntos
Mitomicina/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Esfingosina/análogos & derivados , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/fisiopatologia , Apoptose/efeitos dos fármacos , Resistência a Medicamentos , Sinergismo Farmacológico , Humanos , Esfingosina/farmacologia , Estereoisomerismo , Neoplasias Gástricas/química , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA