Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ann Oncol ; 25(9): 1691-1700, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24675021

RESUMO

Mast cell leukemia (MCL), the leukemic manifestation of systemic mastocytosis (SM), is characterized by leukemic expansion of immature mast cells (MCs) in the bone marrow (BM) and other internal organs; and a poor prognosis. In a subset of patients, circulating MCs are detectable. A major differential diagnosis to MCL is myelomastocytic leukemia (MML). Although criteria for both MCL and MML have been published, several questions remain concerning terminologies and subvariants. To discuss open issues, the EU/US-consensus group and the European Competence Network on Mastocytosis (ECNM) launched a series of meetings and workshops in 2011-2013. Resulting discussions and outcomes are provided in this article. The group recommends that MML be recognized as a distinct condition defined by mastocytic differentiation in advanced myeloid neoplasms without evidence of SM. The group also proposes that MCL be divided into acute MCL and chronic MCL, based on the presence or absence of C-Findings. In addition, a primary (de novo) form of MCL should be separated from secondary MCL that typically develops in the presence of a known antecedent MC neoplasm, usually aggressive SM (ASM) or MC sarcoma. For MCL, an imminent prephase is also proposed. This prephase represents ASM with rapid progression and 5%-19% MCs in BM smears, which is generally accepted to be of prognostic significance. We recommend that this condition be termed ASM in transformation to MCL (ASM-t). The refined classification of MCL fits within and extends the current WHO classification; and should improve prognostication and patient selection in practice as well as in clinical trials.


Assuntos
Leucemia de Mastócitos/classificação , Leucemia Mielomonocítica Aguda/classificação , Leucemia Mielomonocítica Crônica/classificação , Exame de Medula Óssea , Diagnóstico Diferencial , Progressão da Doença , Humanos , Leucemia de Mastócitos/diagnóstico , Leucemia Mielomonocítica Aguda/diagnóstico , Leucemia Mielomonocítica Crônica/diagnóstico , Mastócitos/patologia , Mastocitose/patologia
3.
Eur J Cancer Care (Engl) ; 22(5): 605-11, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23701251

RESUMO

Homoharringtonine is an alkaloid inhibitor of protein synthesis with activity in myeloid malignancies. We report a phase II pilot study of homoharringtonine in myelodysplastic syndrome (MDS). Induction consisted of homoharringtonine at 2.5 mg/m(2) via continuous infusion for 7 days. Maintenance was given every 4 weeks. Nine patients were enrolled: five with refractory anaemia with excess blasts, two with refractory anaemia with excess blasts in transformation, one each with refractory anaemia and chronic myelomonocytic leukaemia respectively. Median age was 70 years (55-84) and 6 (66%) were male. Per International Prognostic Scoring System (IPSS) two patients were intermediate-1, five intermediate-2 and two high-risk. Median chemotherapy courses were one (1-3). One patient (11%) responded with complete haematological and cytogenetic remission after one course. Eight patients did not respond (four had stable disease, two progressed to acute leukaemia and two died during induction - from aspergillus pneumonia and intracerebral haemorrhage respectively). Grade 3/4 myelosuppression seen in 56% (5/9). Serious non-haematological toxicities included one case of grade 4 left bundle branch heart block and one grade 3 nephrotoxicity. Median time between courses was 42 days (35-72 days). In conclusion homoharringtonine might have clinical activity in some patients with MDS.


Assuntos
Harringtoninas/administração & dosagem , Hematínicos/administração & dosagem , Síndromes Mielodisplásicas/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Esquema de Medicação , Feminino , Harringtoninas/efeitos adversos , Hematínicos/efeitos adversos , Mepesuccinato de Omacetaxina , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Resultado do Tratamento
4.
Ann Oncol ; 20(6): 1080-5, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19237479

RESUMO

BACKGROUND: Hepatosplenic T-cell lymphoma (HSTCL) is a rare peripheral T-cell lymphoma; treatment with standard anthracycline-containing chemotherapy regimens has been disappointing, and an optimal treatment strategy for this patient population has not yet been determined. METHODS: We identified 15 cases of pathologically confirmed HSTCL in the institution's database. Clinical characteristics and treatment results were reviewed. RESULTS: Complete responses (CRs) were achieved in 7 of 14 patients who received chemotherapy. Achievement of CR was followed by hematopoietic stem-cell transplantation in three patients. Median duration of CR was 8 months (range 2 to 32+ months) with four patients currently alive and in CR at 5, 8, 12, and 32 months, respectively. Median overall survival (OS) was 11 months (range 2 to 36+ months). Patients who achieved a CR had a median OS of 13 months, compared with 7.5 months in patients who did not achieve a CR. Risk factors associated with worse outcome included male gender, failure to achieve a CR, history of immunocompromise, and absence of a T-cell receptor gene rearrangement in the gamma chain. CONCLUSION: A better understanding of the pathophysiology of HSTCL and new therapeutic strategies are needed.


Assuntos
Linfoma de Células T Periférico/patologia , Linfoma de Células T Periférico/terapia , Adulto , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Linfoma de Células T Periférico/diagnóstico , Linfoma de Células T Periférico/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Análise de Sobrevida , Adulto Jovem
5.
Leukemia ; 21(7): 1395-404, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17495975

RESUMO

The FIP1-like-1 (FIP1L1)-platelet-derived growth factor receptor-alpha (FIP1L1-PDGFR-alpha) fusion kinase causes hypereosinophilic syndrome (HES) in a defined subset of patients. Imatinib mesylate is a potent inhibitor of ABL but also of PDGFR-alpha, and has been associated with durable hematologic responses in patients with HES. However, development of mutations in the tyrosine kinase domain may hamper the activity of tyrosine kinase inhibitors (TKIs), which suggests that novel agents are warranted to prevent or overcome resistance. We evaluated the efficacy of the novel TKI EXEL-0862 in FIP1L1-PDGFR-alpha-expressing cell lines and in cells from a patient with HES harboring the FIP1L1-PDGFR-alpha gene. EXEL-0862 inhibited the proliferation of EOL-1 and imatinib-resistant T674I FIP1L1-PDGFR-alpha-expressing cells and resulted in potent inhibition of the phosphorylation of PDGFR-alpha and downstream proteins STAT3 and Erk1/2, both in vitro and ex vivo. Moreover, EXEL-0862 induced apoptotic death in EOL-1 cells and imatinib-resistant T674I FIP1L1-PDGFR-alpha-expressing cells, and resulted in significant downregulation of the antiapoptotic protein Mcl-1 through a caspase-dependent mechanism. Our data establish EXEL-0862 as a solid candidate for the targeted treatment of patients with FIP1L1-PDGFR-alpha-positive HES.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Inibidores Enzimáticos/farmacologia , Síndrome Hipereosinofílica/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Fatores de Poliadenilação e Clivagem de mRNA , Benzamidas , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/uso terapêutico , Humanos , Síndrome Hipereosinofílica/patologia , Mesilato de Imatinib , Proteína de Sequência 1 de Leucemia de Células Mieloides , Piperazinas/farmacologia , Pirimidinas/farmacologia , Células Tumorais Cultivadas
6.
Leukemia ; 21(3): 480-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17215857

RESUMO

beta-2 Microglobulin (beta2M), a subunit of human leukocyte antigen-class I (HLA-I), is well established as a marker of prognosis in various solid tumors and hematologic malignancies. The prognostic role of intact free-circulating HLA-I (sHLA-I) is less well understood. We compared the clinical relevance of plasma levels of sHLA-I and beta2M in patients with acute myeloid leukemia (AML; n=209) or advanced myelodysplastic syndrome (MDS; n=98). sHLA-1 and beta2M levels were significantly higher in AML and MDS patients than in control subjects, but did not differ significantly between the two disease groups. In AML patients, multivariate analysis showed both sHLA-1 and beta2-M to be highly predictive of complete remission (CR), survival and duration of complete response (CRD). In MDS, the predictive value of the two markers differed substantially from one another: beta2M was associated with survival, CR and CRD, whereas sHLA-I was not. These findings not only establish the role of sHLA-I as a tumor marker in AML but also support that MDS is clinically and biologically distinct from AML. sHLA-I has been reported to be an immunomodulator inhibiting the cytotoxic effects of T-lymphocytes, which may offset its predictive value for disease aggressiveness in patients with MDS.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/sangue , Antígenos HLA/sangue , Leucemia Mieloide/sangue , Síndromes Mielodisplásicas/sangue , Proteínas de Neoplasias/sangue , Microglobulina beta-2/sangue , Doença Aguda , Adulto , Idoso , Idoso de 80 Anos ou mais , Citarabina/administração & dosagem , Progressão da Doença , Feminino , Humanos , Idarubicina/administração & dosagem , Estimativa de Kaplan-Meier , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/mortalidade , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/mortalidade , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Indução de Remissão , Solubilidade , Topotecan/administração & dosagem , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
7.
Leuk Res ; 31(5): 691-4, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17095087

RESUMO

In a study of 99 consecutive patients with "idiopathic" eosinophilia, clonal T-cells were demonstrated in blood, bone marrow, or other tissue samples of 14 patients including 6 who had an overt T-cell malignancy. The remaining eight patients (approximately 8%) with an "Occult" T-cell clone had predominantly cutaneous disease and FIP1L1-PDGFRA was absent in all six evaluable patients. Two patients were effectively treated with low-dose oral cyclophosphamide or methotrexate whereas Gleevec treatment was ineffective in another two patients. Two patients (25%) transformed into cutaneous T-cell lymphoma after 3-8 years of eosinophilic prodrome.


Assuntos
Eosinofilia/complicações , Leucemia Linfoide/complicações , Linfoma de Células T Periférico/complicações , Linfócitos T/imunologia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzamidas , Células Clonais , Ciclofosfamida/administração & dosagem , Eosinofilia/imunologia , Feminino , Humanos , Mesilato de Imatinib , Leucemia Linfoide/tratamento farmacológico , Leucemia Linfoide/imunologia , Linfoma de Células T Periférico/tratamento farmacológico , Linfoma de Células T Periférico/imunologia , Masculino , Metotrexato/administração & dosagem , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica/metabolismo , Piperazinas/administração & dosagem , Prevalência , Pirimidinas/administração & dosagem , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
8.
Leuk Res ; 31(11): 1503-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17397921

RESUMO

We recently developed a modified Dupriez prognostic scoring system (PSS) that effectively discriminated between high-, intermediate-, and low-risk young patients (age < or =60 years) with primary myelofibrosis (PMF) based on the respective presence of none, one, or two or more of the following parameters: hemoglobin <10 g/dL, leukocyte count <4 or >30 x 10(9)L(-1), and platelet count <100 x 10(9)L(-1). The current study (n=129; median age, 52 years; 69 males) reveals, on multivariable analysis, that an absolute monocyte count of > or =1 x 10(9)L(-1) carries an independent predictive value (p=0.02), for an inferior survival, in addition to that provided by hemoglobin level (p=0.002), platelet count (0.02), and leukocyte count (p=0.16). The inclusion of the monocyte count as a fourth risk factor enabled the construction of a new and improved Mayo PSS; median survival was 173, 61, and 26 months in the absence of all four (low-risk), three (intermediate-risk), or two or less (high-risk) adverse features, respectively (p<0.0001). The independent prognostic value of monocytosis was validated in a separate database of 97 patients with PMF from another institution.


Assuntos
Monócitos/patologia , Mielofibrose Primária/patologia , Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico
9.
Int J Immunopathol Pharmacol ; 20(2): 373-80, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17624250

RESUMO

In an effort to define the antigenic mechanism that contributes to beneficial therapeutic outcome in patients with polycythemia vera (PV), we screened a human testis cDNA library with serological cloning derived from sera of three PV patients who had undergone therapeutic-induced remission. As a result, we identified a novel antigen, MPD5, which belongs to the group of cryptic antigens with unconventional genomic intron/exon structure. Moreover, MPD5 elicited IgG antibody responses in a subset of PV patients who had benefited from a variety of therapies--including IFN-alpha, Hydroxyurea, Imatinib mesylate, Anagrelide, and phlebotomy--but not in untreated PV patients or healthy donors, suggesting that MPD5 is a PV-associated, therapy-related antigen. In the granulocytes of PV patients who are responsive to therapy, upregulated MPD5 expression may serve to enhance immune responses. These findings provide new insight into the mechanism underlying regulation of the self-antigen repertoire that elicits anti-tumor immune responses in patients with myeloproliferative diseases, indicating the potential of these self-antigens as targets of novel immunotherapy.


Assuntos
Antígenos de Neoplasias/imunologia , Policitemia Vera/imunologia , Antígenos de Neoplasias/sangue , Humanos , Masculino , Policitemia Vera/sangue
10.
Leukemia ; 20(10): 1767-73, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16855631

RESUMO

Mutations of the BCR-ABL kinase domain are a common mechanism of resistance to imatinib in chronic myeloid leukemia. We screened for mutations 171 patients failing imatinib therapy. Sixty-six mutations in 23 amino acids were identified in 62 (36%) patients not responding to imatinib. Phosphate-binding loop (P-loop) mutations were the most frequent (n=24; 36%). By multivariate analysis, factors associated with development of mutations were older age (P=0.026) prior interferon therapy (P=0.026), and accelerated phase or blast phase at time of imatinib failure (P=0.001). After a median follow-up of 38 months (range, 4-68 months) from the start of imatinib therapy, seven patients with non-P-loop and two with P-loop mutation died. By multivariate analysis, development of clonal evolution and higher percentage of peripheral blood basophils were associated with worse survival from the time of imatinib failure. Mutation status had no impact on survival. When survival was measured from the time therapy started, non-P-loop mutations together with duration of response and transformation at the time of failure to imatinib were associated with shorter survival. In conclusion, P-loop mutations were not associated with poor outcome, suggesting that the prognosis of patients who fail imatinib is multifactorial.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Basófilos/patologia , Benzamidas , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Pessoa de Meia-Idade , Análise Multivariada , Mutação Puntual , Prognóstico , Taxa de Sobrevida
11.
Leukemia ; 31(9): 1845-1854, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28529308

RESUMO

The molecular characterization of myeloproliferative neoplasms, including essential thrombocythemia (ET), has enabled deeper understanding of their pathogenesis. A driver lesion, namely, Janus kinase (JAK)2V617F, calreticulin (CALR) or myeloproliferative leukemia (MPL) gene mutation can be identified in the vast majority of patients. Each of these mutations is associated with distinct clinical features and may modulate the patients' clinical course, risk of complications, including vascular events, and survival. JAK2V617F appears to be a risk-modifying mutation and has been shown to increase the likelihood of thrombotic events in patients with ET across studies. As such, it has been included in prognostic models and its presence may influence treatment decisions. The association of CALR and MPL mutations with the incidence of vascular events has been less clear. Even more limited information is available on the contribution of additional non-driver lesions to the thrombotic risk. In this review we discuss the available evidence on the role of recurrent mutations in the risk of thrombotic complications in patients with ET and how these mutations weigh into modern prognostic scores.


Assuntos
Genômica , Trombocitemia Essencial/complicações , Trombose/etiologia , Humanos , Mutação , Prognóstico , Medição de Risco , Trombocitemia Essencial/genética , Trombose/genética
12.
Leukemia ; 31(2): 393-402, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27479177

RESUMO

NS-018 is a Janus-activated kinase 2 (JAK2)-selective inhibitor, targeting the JAK-signal transducer and activator of transcription (STAT) pathway that is deregulated in myelofibrosis. In this phase I, dose-escalation portion of a phase I/II study, patients with myelofibrosis received oral NS-018 in continuous 28-day cycles. The primary study objective was to evaluate safety, tolerability and clinically active dose of NS-018. Forty-eight patients were treated; 23 (48%) had previously received a JAK inhibitor (JAKi). The most common drug-related adverse events were thrombocytopenia (27%)/anemia (15%) for hematologic events, and dizziness (23%)/nausea (19%) for non-hematologic events. Once daily NS-018 at 300 mg was chosen as the phase II study dose based on improved tolerability compared with higher doses. A ⩾50% reduction in palpable spleen size was achieved in 56% of patients (47% of patients with prior JAKi treatment), and improvements were observed in myelofibrosis-associated symptoms. Bone marrow fibrosis grade (local assessment) improved from baseline in 11/30 evaluable patients (37%) after 3 cycles of NS-018. JAK2 allele burden was largely unchanged. Changes in cytokine/protein levels were noted after 4 weeks of treatment. NS-018 reached peak plasma concentration in 1-2 h and did not accumulate with multiple dosing. NS-018 will be assessed in patients with previous JAKi exposure in the phase II portion.


Assuntos
Antineoplásicos/uso terapêutico , Janus Quinase 2/antagonistas & inibidores , Terapia de Alvo Molecular , Mielofibrose Primária/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Antineoplásicos/farmacologia , Biomarcadores , Medula Óssea/patologia , Feminino , Seguimentos , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Fenótipo , Mielofibrose Primária/diagnóstico , Mielofibrose Primária/metabolismo , Mielofibrose Primária/terapia , Inibidores de Proteínas Quinases/farmacologia , Retratamento , Resultado do Tratamento
13.
Leukemia ; 31(9): 1951-1961, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28042144

RESUMO

The PROTAC (proteolysis-targeting chimera) ARV-825 recruits bromodomain and extraterminal (BET) proteins to the E3 ubiquitin ligase cereblon, leading to degradation of BET proteins, including BRD4. Although the BET-protein inhibitor (BETi) OTX015 caused accumulation of BRD4, treatment with equimolar concentrations of ARV-825 caused sustained and profound depletion (>90%) of BRD4 and induced significantly more apoptosis in cultured and patient-derived (PD) CD34+ post-MPN sAML cells, while relatively sparing the CD34+ normal hematopoietic progenitor cells. RNA-Seq, Reverse Phase Protein Array and mass cytometry 'CyTOF' analyses demonstrated that ARV-825 caused greater perturbations in messenger RNA (mRNA) and protein expressions than OTX015 in sAML cells. Specifically, compared with OTX015, ARV-825 treatment caused more robust and sustained depletion of c-Myc, CDK4/6, JAK2, p-STAT3/5, PIM1 and Bcl-xL, while increasing the levels of p21 and p27. Compared with OTX015, PROTAC ARV-771 treatment caused greater reduction in leukemia burden and further improved survival of NSG mice engrafted with luciferase-expressing HEL92.1.7 cells. Co-treatment with ARV-825 and JAK inhibitor ruxolitinib was synergistically lethal against established and PD CD34+ sAML cells. Notably, ARV-825 induced high levels of apoptosis in the in vitro generated ruxolitinib-persister or ruxolitinib-resistant sAML cells. These findings strongly support the in vivo testing of the BRD4-PROTAC based combinations against post-MPN sAML.


Assuntos
Azepinas , Leucemia Mieloide Aguda , Transtornos Mieloproliferativos , Proteínas Nucleares , Talidomida , Fatores de Transcrição , Animais , Humanos , Camundongos , Antígenos CD34 , Apoptose/efeitos dos fármacos , Azepinas/farmacologia , Azepinas/uso terapêutico , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Leucemia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Transtornos Mieloproliferativos/patologia , Nitrilas , Proteínas Nucleares/metabolismo , Proteólise , Pirazóis/farmacologia , Pirimidinas , Talidomida/análogos & derivados , Talidomida/farmacologia , Talidomida/uso terapêutico , Fatores de Transcrição/metabolismo , Carga Tumoral/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo
14.
Leukemia ; 31(2): 318-324, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27795561

RESUMO

Most clinical trials exclude patients with poor performance or comorbidities. To study whether patients with these characteristics can be treated within a clinical trial, we conducted a study for patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS) with poor performance, organ dysfunction or comorbidities. Primary endpoint was 60-day survival. Study included stopping rules for survival and response. Treatment consisted on a combination of azacitidine and vorinostat. Thirty patients (16 with MDS, 14 with AML) were enrolled. Median follow-up was 7.4 months (0.3-29). Sixty-day survival was 83%. No stopping rules were met. Main adverse events (AEs) were grades 1 and 2 gastrointestinal toxicities. In view of these results, we expanded the study and treated 79 additional patients: 27 with azacitidine (AZA) and 52 with azacitidine and vorinostat (AZA+V). Median follow-up was 22.7 months (12.6-47.5). Sixty-day survival rate was 79% (AZA=67%, AZA+V=85%, P=0.07). Median overall survival was 7.6 months (4.5-10.7). Median event-free survival was 4.5 months (3.5-5.6). Main AEs included grades 1 and 2 gastrointestinal toxicities. Our results suggest this subset of patients can be safely treated within clinical trials and derive clinical benefit. Relaxation of standard exclusion criteria may increase the pool of patients likely to benefit from therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores , Medula Óssea/patologia , Aberrações Cromossômicas , Comorbidade , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidade , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/mortalidade , Resultado do Tratamento
16.
Cancer Res ; 52(14): 3880-5, 1992 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-1617664

RESUMO

The capacity of recombinant human interleukin 2 (rH-IL2), alone or in combination with recombinant tumor necrosis factor (r-TNF alpha), to activate murine resident peritoneal macrophages to a tumoricidal state was examined. Resident peritoneal exudate cells from C57BL/6 mice were cultured for 18 h with activating agents and washed and the adherent cells (macrophages) were assessed for cytolytic activity against radiolabeled target tumor cells (EL4, P815). Under these conditions, rH-IL2 alone activated macrophages to a tumoricidal state in a concentration dependent fashion. Neither murine nor human r-TNF alpha alone had any activating effect but, when combined with rH-IL2, further stimulated rH-IL2-inducible responses. Using polymyxin B, it was shown that macrophage activation was not due to an inadvertent lipopolysaccharide contamination of the r-TNF alpha or rH-IL2 preparations. It was also unlikely that target cell lysis was a direct result of increased TNF alpha production by rH-IL2 stimulated macrophages since P815 is totally resistant to lysis by r-TNF alpha. Although the lytic effector function was mediated by adherent cells, nonadherent peritoneal exudate cells were required for activation to occur. Furthermore, antisera against murine gamma-interferon, when added to activation cultures, reduced the level of cytolytic activity which developed. These data suggest that rH-IL2-induced peritoneal macrophage activation requires stimulation of nonadherent cells and is dependent upon gamma-interferon mediated mechanisms.


Assuntos
Interleucina-2/farmacologia , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Neoplasias Experimentais/terapia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Sinergismo Farmacológico , Interferon gama/farmacologia , Linfoma de Células T/terapia , Sarcoma de Mastócitos/terapia , Cavidade Peritoneal/citologia , Polimixina B/farmacologia , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas
17.
Leukemia ; 30(2): 268-73, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26365212

RESUMO

Decitabine may open the chromatin structure of leukemia cells making them accessible to the calicheamicin epitope of gemtuzumab ozogamicin (GO). A total of 110 patients (median age 70 years; range 27-89 years) were treated with decitabine and GO in a trial designed on model-based futility to accommodate subject heterogeneity: group 1: relapsed/refractory acute myeloid leukemia (AML) with complete remission duration (CRD) <1 year (N=28, 25%); group 2: relapsed/refractory AML with CRD ⩾1 year (N=5, 5%); group 3: untreated AML unfit for intensive chemotherapy or untreated myelodysplastic syndrome (MDS) or untreated myelofibrosis (MF; N=57, 52%); and group 4: AML evolving from MDS or relapsed/refractory MDS or MF (N=20, 18%). Treatment consisted of decitabine 20 mg/m(2) daily for 5 days and GO 3 mg/m(2) on day 5. Post-induction therapy included five cycles of decitabine+GO followed by decitabine alone. Complete remission (CR)/CR with incomplete count recovery was achieved in 39 (35%) patients; group 1= 5/28 (17%), group 2=3/5 (60%), group 3=24/57 (42%) and group 4=7/20 (35%). The 8-week mortality in groups 3 and 4 was 16% and 10%, respectively. Common drug-related adverse events included nausea, mucositis and hemorrhage. Decitabine and GO improved the response rate but not overall survival compared with historical outcomes in untreated AML ⩾60 years.


Assuntos
Aminoglicosídeos/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azacitidina/análogos & derivados , Leucemia Mieloide Aguda/tratamento farmacológico , Síndromes Mielodisplásicas/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Aminoglicosídeos/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos , Azacitidina/administração & dosagem , Azacitidina/efeitos adversos , Decitabina , Feminino , Gemtuzumab , Humanos , Masculino , Pessoa de Meia-Idade , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/análise
18.
Leukemia ; 15(8): 1165-70, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11480557

RESUMO

Hepatocyte growth factor (HGF) is a potent angiogenic factor. The aim of our study was to evaluate plasma HGF levels and their prognostic significance in patients with newly diagnosed acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). The sandwich enzyme immunoassay technique was used to quantify HGF in stored samples obtained before treatment from patients with AML (59 patients) and MDS (42 patients) treated at The University of Texas MD Anderson Cancer Center. HGF levels were significantly higher in patients with AML or MDS than in healthy individuals (P < 0.0001). Higher HGF levels in both AML and MDS correlated significantly with white blood cell (P = 0.000001 for both groups) and monocyte counts (P = 0.0004 and 0.003, respectively), and with poor performance status (P = 0.03 and 0.001, respectively). Using Cox proportional hazard model and HGF levels as a continuous variable, plasma levels of HGF correlated with shorter survival of AML (P = 0.001), but not MDS (P = 0.34) patients. No significant correlation was observed between HGF levels and complete remission rate or duration. In the multivariate analysis HGF retained its significance as prognostic factor in AML (P = 0.02), along with age (P = 0.0005).


Assuntos
Fator de Crescimento de Hepatócito/sangue , Leucemia Mieloide/sangue , Síndromes Mielodisplásicas/sangue , Doença Aguda , Adulto , Biomarcadores Tumorais , Humanos , Leucemia Mieloide/mortalidade , Leucemia Mieloide/patologia , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/mortalidade , Síndromes Mielodisplásicas/patologia , Valor Preditivo dos Testes , Prognóstico , Análise de Sobrevida
19.
Leukemia ; 17(6): 1100-3, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12764375

RESUMO

Granulocytic sarcoma (GS) is an extramedullary tumor composed of immature myeloid cells. The objectives of this study were to describe the frequency, presenting characteristics, and survival in patients with nonleukemic GS by conducting a review of all untreated patients presenting to the MD Anderson Cancer Center between January 1990 and June 2002. In all, 21 patients with nonleukemic GS, 1520 patients with acute myeloid leukemia (AML), and 402 patients with high-risk myelodysplastic syndrome (MDS) were identified. GS occurred in 1.4% of patients with AML, and 1.1% of patients with AML or high-risk MDSs. The median patient age was 57 years (range, 7-81). Among 20 patients with available cytogenetics in tissue and/or bone marrow, six had chromosome 8 abnormalities. The median follow-up of surviving patients is 12 months (range, 7-75). In all, 20 patients were treated. Patients were treated with AML-type chemotherapy (n=16), chemotherapy and radiotherapy (n=3), or radiotherapy alone (n=1). A total of 13 patients (65%) achieved complete remission and one patient (5%) achieved partial remission. The median overall survival was 20 months (range, 1-75), median overall failure-free survival was 12 months (range, 1-75). The median survival of patients with chromosome 8 abnormalities was 12 months compared with 40 months of those without (P=0.17). Novel therapies for patients with GS are required.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Sarcoma Mieloide/tratamento farmacológico , Sarcoma Mieloide/radioterapia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Criança , Aberrações Cromossômicas , Cromossomos Humanos Par 8/genética , Terapia Combinada , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/mortalidade , Síndromes Mielodisplásicas/terapia , Indução de Remissão , Sarcoma Mieloide/genética , Sarcoma Mieloide/mortalidade , Taxa de Sobrevida , Resultado do Tratamento
20.
Clin Cancer Res ; 7(12): 3884-93, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11751479

RESUMO

STI571 is a 2-phenylalaminopyrimidine derivative that inhibits c-abl, Bcr-Abl, and platelet-derived growth factor receptor tyrosine kinases. Recently, inhibition of stem cell factor (SCF)-induced c-kit phosphorylation and cell proliferation by STI571 was reported in the human myeloid cell line MO7e. Because approximately 70% of acute myelogenous leukemia (AML) cases are c-kit positive, we evaluated in vitro effects of STI571 on c-kit-positive cell lines and primary AML blast cells. At concentrations >5 microM, the drug marginally inhibited SCF-independent proliferation of cell lines and most of AML blasts. Treatment of AML cells with cytarabine and STI571 showed synergistic effect at low concentrations. Western blotting analysis documented a distinct band of M(r) 145,000 specific for c-kit in cell lines and in AML samples. There was no correlation between the level of the c-kit expression evaluated by Western blotting and percentage of c-kit-positive blasts as measured by flow cytometry. Neither in cell lines nor in primary AML cells, c-kit autophosphorylation was detectable under standard growth conditions. SCF-induced phosphorylation of c-kit in MO7e cells was inhibited by STI571. In a c-kit-positive AML-4 cell line, as well as in AML samples, c-kit phosphorylation was not induced by SCF exposure, suggesting that in these cases, the receptor could not be functionally activated. In conclusion, with the exception of MO7e, SCF did not induce phosphorylation of c-kit, and cell proliferation was not modulated in the presence of STI571. We did not detect any SCF-independent c-kit phosphorylation in our experimental systems. Consequently, STI571 exerted only a limited inhibitory effect on the cell growth.


Assuntos
Leucemia Mieloide Aguda/fisiopatologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Benzamidas , Divisão Celular/efeitos dos fármacos , Citarabina/toxicidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Mesilato de Imatinib , Interleucina-3/farmacologia , Cinética , Leucemia Mieloide Aguda/sangue , Contagem de Leucócitos , Fosforilação , Proteínas Proto-Oncogênicas c-kit/metabolismo , Recidiva , Fator de Células-Tronco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA