Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Clin Lymphoma Myeloma Leuk ; 14(5): 401-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24875590

ABSTRACT

INTRODUCTION/BACKGROUND: We evaluated the characteristics of a cohort of patients with myelodysplastic syndrome (MDS) related to therapy (t-MDS) to create a prognostic model. PATIENTS AND METHODS: We identified 281 patients with MDS who had received previous chemotherapy and/or radiotherapy for previous malignancy. Potential prognostic factors were determined using univariate and multivariate analyses. RESULTS: Multivariate Cox regression analysis identified 7 factors that independently predicted short survival in t-MDS: age ≥ 65 years (hazard ratio [HR], 1.63), Eastern Cooperative Oncology Group performance status 2-4 (HR, 1.86), poor cytogenetics (-7 and/or complex; HR, 2.47), World Health Organization MDS subtype (RARs or RAEB-1/2; HR, 1.92), hemoglobin (< 11 g/dL; HR, 2.24), platelets (< 50 × 10(9)/dL; HR, 2.01), and transfusion dependency (HR, 1.59). These risk factors were used to create a prognostic model that segregated patients into 3 groups with distinct median overall survival: good (0-2 risk factors; 34 months), intermediate (3-4 risk factors; 12 months), and poor (5-7 risk factors; 5 months) (P < .001) and 1-year leukemia-free survival (96%, 84%, and 72%, respectively, P = .003). This model also identified distinct survival groups according to t-MDS therapy. CONCLUSION: In summary, we devised a prognostic model specifically for patients with t-MDS that predicted overall survival and leukemia-free survival. This model might facilitate the development of risk-adapted therapeutic strategies.


Subject(s)
Antineoplastic Agents/adverse effects , Leukemia, Myeloid, Acute/etiology , Leukemia, Radiation-Induced/etiology , Models, Biological , Myelodysplastic Syndromes/etiology , Neoplasms, Second Primary/etiology , Radiotherapy/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Blood Transfusion/statistics & numerical data , Disease Progression , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hemoglobins/analysis , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/mortality , Leukemia, Radiation-Induced/pathology , Male , Middle Aged , Multivariate Analysis , Myelodysplastic Syndromes/chemically induced , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/mortality , Platelet Count , Prognosis , Proportional Hazards Models , Risk Factors , Young Adult
2.
Haematologica ; 98(1): 114-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22801963

ABSTRACT

We recently reported that clofarabine, high-dose cytarabine, and granulocyte colony-stimulating factor (GCLAC) produced a 46% complete remission rate in relapsed/refractory acute myeloid leukemia. GCLAC differs from FLAG by substitution of clofarabine for fludarabine, raising the question of the relative efficacy of these two regimens. We compared GCLAC given at the University of Washington Medical Center/Fred Hutchinson Cancer Research Center to fludarabine and cytarabine (FA) and FLAG given at MD Anderson Cancer Center. Independent multivariate analyses conducted at both institutions showed that after accounting for duration of first complete remission, salvage number, age, and cytogenetics, GCLAC was associated with a higher complete remission rate (odds ratio 9.57, P<0.0001) and longer survival (mortality hazard ratio 0.43, P=0.0002). Despite the retrospective nature of the analyses, GCLAC may be superior to FA/FLAG, particularly in patients with short duration of first complete remission or unfavorable cytogenetics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Salvage Therapy/methods , Adenine Nucleotides/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Arabinonucleosides/administration & dosage , Clofarabine , Cytarabine/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Leukemia, Myeloid, Acute/diagnosis , Middle Aged , Retrospective Studies , Survival Rate/trends , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Young Adult
3.
Cancer ; 118(12): 3116-22, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22370904

ABSTRACT

BACKGROUND: The long-term outcome of patients with chronic phase chronic myeloid leukemia treated with imatinib after failure of interferon alpha therapy has not been detailed. METHODS: In total, 368 patients were analyzed. Univariate and multivariate survival analyses were conducted using standard statistical methods. RESULTS: Overall, 247 patients (67%) achieved a complete cytogenetic response (CCyR). Of the 327 patients who were studied, 207 patients (63%) achieved a major molecular response (MMR), and 99 patients (30%) had undetectable breakpoint cluster region/c-abl oncogene (BCR-ABL) levels at some time during therapy. The estimated 10-year survival rate was 68%, the progression-free survival rate was 67%, and the event-free survival rate was 51%. In multivariate analysis, age ≥ 60 years, hemoglobin <10 g/dL, bone marrow basophils ≥ 5%, any peripheral blasts, and clonal evolution were independent adverse factors for survival. The estimated 7-year survival rate according to the presence of no factors (n = 154), 1 or 2 factors (n = 190), or ≥ 3 factors (n = 24) were 93%, 70%, and 25%, respectively (P < .01). Achieving an MMR, a CCyR, or a partial cytogenetic response at 12 months was associated with significantly better 10-year survival rate in a landmark analysis (10-year survival rate, 80%-90%) compared with achieving a minor cytogenetic response or a complete hematologic response (10-year survival rate, 55%-65%) or another response (10-year survival rate, 10%). In a landmark analysis that included imatinib response at 12 months, achieving a major cytogenetic response or better (hazard ratio, 0.12; P < .001) and achieving a complete hematologic response or a minor cytogenetic response (hazard ratio, 0.36; P = .003) were significant favorable prognostic factors. CONCLUSIONS: The current results indicated that the estimated 10-year survival rate of 68% for patients with chronic myeloid leukemia who receive imatinib after failure on interferon has improved.


Subject(s)
Interferon-alpha/therapeutic use , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/administration & dosage , Benzamides , Female , Follow-Up Studies , Fusion Proteins, bcr-abl/drug effects , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Leukemia, Myeloid, Chronic-Phase/mortality , Male , Middle Aged , Prognosis , Retreatment , Survival Rate , Treatment Failure , Treatment Outcome
4.
Cancer ; 118(18): 4462-70, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22252728

ABSTRACT

BACKGROUND: Although most patients with myelodysplastic syndrome (MDS) exhibit bone marrow hypercellularity, a subset of them present with a hypocellular bone marrow. Specific factors associated with poor prognosis have not been investigated in patients with hypocellular MDS. METHODS: The authors studied a cohort of 253 patients with hypocellular MDS diagnosed at The University of Texas MD Anderson Cancer Center between 1993 and 2007 and a cohort of 1725 patients with hyper-/normocellular MDS diagnosed during the same time period. RESULTS: Patients with hypocellular MDS presented more frequently with thrombocytopenia (P < .019), neutropenia (P < .001), low serum ß-2 microglobulin (P < .001), increased transfusion dependency (P < .001), and intermediate-2/high-risk disease (57% vs 42%, P = .02) compared with patients with hyper-/normocellular MDS. However, no difference in overall survival was observed between the 2 groups (P = .28). Multivariate analysis identified poor performance status (Eastern Cooperative Oncology Group ≥2), low hemoglobin (<10 g/dL), unfavorable cytogenetics (-7/7q or complex), increased bone marrow blasts (≥5%), and high serum lactate dehydrogenase (>600 IU/L) as adverse independent factors for survival. CONCLUSIONS: A new prognostic model based on these factors was built that segregated patients into 3 distinct risk categories independent of International Prognostic Scoring System (IPSS) score. This model is independent from the IPSS, further refines IPSS-based prognostication, and may be used to develop of risk-adapted therapeutic approaches for patients with hypocellular MDS.


Subject(s)
Bone Marrow Diseases/mortality , Myelodysplastic Syndromes/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Bone Marrow , Bone Marrow Diseases/blood , Bone Marrow Diseases/complications , Disease Progression , Disease-Free Survival , Female , Hemoglobins/analysis , Humans , L-Lactate Dehydrogenase/blood , Leukemia, Myeloid, Acute , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Neutropenia/complications , Prognosis , Retrospective Studies , Thrombocytopenia , Young Adult
5.
Haematologica ; 97(7): 1029-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22271898

ABSTRACT

BACKGROUND: Outcomes in chronic myeloid leukemia have improved with tyrosine kinase inhibitor treatment. However, little is known about outcomes of chronic myeloid leukemia in adolescent and young adult patients. DESIGN AND METHODS: We reviewed all 468 chronic myeloid leukemia patients treated at our institution with tyrosine kinase inhibitors as initial therapy: imatinib (n=281), nilotinib (n=98) or dasatinib (n=89). RESULTS: Median age was 47 years, median follow up 71 months and median treatment time with initial tyrosine kinase inhibitors 48 months. Adolescent and young adult was defined as aged 15-29 years. Sixty-one adolescent and young adult patients were identified. The only significant differences between adolescent and young adult and older patients were incidence of splenomegaly and distribution in Sokal risk groups. Only 3 adolescent and young adult patients have died. Rates of complete cytogenetic, major molecular and complete molecular response were significantly higher in older patients compared to adolescent and young adult patients, with a favorable trend in event-free survival for older patients. Transformation-free and overall survival were similar for the two groups. CONCLUSIONS: The unfavorable trend in outcome for adolescent and young adult patients with chronic myeloid leukemia is unexpected. Additional research in this population is required to better define outcomes, understand the cause of this difference, and to help make better treatment recommendations.


Subject(s)
Antineoplastic Agents/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Thiazoles/administration & dosage , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Benzamides , Dasatinib , Drug Administration Schedule , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Male , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Probability , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Survival Rate , Treatment Outcome
6.
J Clin Oncol ; 29(32): 4260-5, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21990394

ABSTRACT

PURPOSE: The response definitions proposed by the European LeukemiaNet (ELN) are defined on the basis of imatinib front-line therapy. It is unknown whether these definitions apply to patients treated with second-generation tyrosine kinase inhibitors (TKIs). PATIENTS AND METHODS: One hundred sixty-seven patients with newly diagnosed chronic myelogenous leukemia (CML) in chronic phase were treated with second-generation TKIs in phase II trials (nilotinib, 81; dasatinib, 86). Median follow-up was 33 months. Event-free survival (EFS) was measured from the start of treatment to the date of loss of complete hematologic response, loss of complete or major cytogenetic response, discontinuation of therapy for toxicity or lack of efficacy, progression to accelerated or blastic phases, or death at any time. RESULTS: Overall, 155 patients (93%) achieved complete cytogenetic response (CCyR), including 146 (87%) with major molecular response (MMR; complete in 46 patients [28%]). According to the ELN definitions, the rates of suboptimal response were 0%, 2%, 1%, and 12% at 3, 6, 12, and 18 months of therapy, respectively. There was no difference in EFS and CCyR duration between patients who achieved CCyR with and without MMR across all the landmark times of 3, 6, 12, and 18 months. CONCLUSION: The use of second-generation TKIs as initial therapy in CML induces high rates of CCyR at early time points. The ELN definitions of response proposed for imatinib therapy are not applicable in this setting. We propose that achievement of CCyR and partial cytogenetic response at 3 months should be considered optimal and suboptimal responses, respectively. The achievement of MMR offered no advantage over CCyR in defining long-term outcome in patients with newly diagnosed CML treated with second-generation TKIs.


Subject(s)
Leukemia, Myeloid, Chronic-Phase/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Humans , Leukemia, Myeloid, Chronic-Phase/genetics , Leukemia, Myeloid, Chronic-Phase/mortality , Middle Aged
7.
Br J Haematol ; 155(2): 182-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21848522

ABSTRACT

This phase I/II study was conducted to determine the maximum tolerated dose, toxicity, and efficacy of clofarabine in combination with high dose cytarabine and granulocyte colony-stimulating factor (G-CSF) priming (GCLAC), in the treatment of patients with relapsed or refractory acute myeloid leukaemia (AML). Dose escalation of clofarabine occurred without dose-limiting toxicity, so most patients were treated at the maximum dose, 25 mg/m(2) per day with cytarabine 2 g/m(2) per day, each for 5 d, and G-CSF 5 µg/kg, beginning the day before chemotherapy and continuing daily until neutrophil recovery. The complete remission (CR) rate among the 46 evaluable patients was 46% (95% confidence interval [CI] 31-61%) and the CR + CR but with a platelet count <100 × 10(9)/l rate was 61% (95% CI 45-75%). Multivariate analysis showed that responses to GCLAC were independent of age, cytogenetic risk category, and number of prior salvage regimens. GCLAC is highly active in relapsed and refractory AML and warrants prospective comparison to other regimens, as well as study in untreated patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Salvage Therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Cytarabine/administration & dosage , Cytarabine/adverse effects , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/adverse effects , Humans , Infections/etiology , Kaplan-Meier Estimate , Male , Maximum Tolerated Dose , Middle Aged , Proportional Hazards Models , Recurrence , Remission Induction , Risk , Young Adult
8.
Clin Lymphoma Myeloma Leuk ; 11(2): 237-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21575929

ABSTRACT

PURPOSE: To analyze the incidence and significance of thrombocytopenia in patients with myelodysplastic syndrome (MDS). PATIENTS AND METHODS: A total of 2517 patients with MDS referred to our institution since 1993 were analyzed, with a specific focus on the incidence and associations of thrombocytopenia. RESULTS: The median age of the study group was 66 years. The median survival was 13 months. Platelet counts <100 × 10(9)/L were noted in 65%, and platelets counts <30 × 10(9)/L in 26%. Each platelets count drop below the range of 200 × 10(9)/L has shown a larger magnitude change in terms of worsening effect on survival. Therefore, smaller ranges of platelet counts of <200 × 10(9)/L were studied. Platelet cutoffs of 30, 50, and 200 × 10(9)/L thus were identified to have significant associations with differences in survival. Significant thrombocytopenia was associated with poor performance, other cytopenias, adverse karyotype, and advanced MDS phases. Thrombocytopenia was associated with worse prognosis; it also was predicted for worse outcome within each of the International Prognostic Scoring System risk groups. CONCLUSION: Prognosis in MDS is directly associated with the severity of thrombocytopenia.


Subject(s)
Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/diagnosis , Thrombocytopenia/complications , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Incidence , Male , Middle Aged , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/mortality , Platelet Count , Prognosis , Survival Analysis , Thrombocytopenia/epidemiology , Young Adult
9.
J Clin Oncol ; 29(16): 2240-6, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21537048

ABSTRACT

PURPOSE: Patients with cancer often experience comorbidities that may affect their prognosis and outcome. The objective of this study was to determine the effect of comorbidities on the survival of patients with myelodysplastic syndrome (MDS). PATIENTS AND METHODS: We conducted a retrospective cohort study of 600 consecutive patients with MDS who presented to MD Anderson Cancer Center from January 2002 to December 2004. The Adult Comorbidity Evaluation-27 (ACE-27) scale was used to assess comorbidities. Data on demographics, International Prognostic Scoring System (IPSS), treatment, and outcome (leukemic transformation and survival) were collected. Kaplan-Meier methods and Cox regression were used to assess survival. A prognostic model incorporating baseline comorbidities with age and IPSS was developed to predict survival. RESULTS: Overall median survival was 18.6 months. According to the ACE-27 categories, median survival was 31.8, 16.8, 15.2, and 9.7 months for those with none, mild, moderate, and severe comorbidities, respectively (P < .001). Adjusted hazard ratios were 1.3, 1.6, and 2.3 for mild, moderate, and severe comorbidities, respectively, compared with no comorbidities (P < .001). A final prognostic model including age, IPSS, and comorbidity score predicted median survival of 43.0, 23.0, and 9.0 months for lower-, intermediate-, and high-risk groups, respectively (P < .001). CONCLUSION: Comorbidities have a significant impact on the survival of patients with MDS. Patients with severe comorbidity had a 50% decrease in survival, independent of age and IPSS risk group. A comprehensive assessment of the severity of comorbidities helps predict survival in patients with MDS.


Subject(s)
Myelodysplastic Syndromes/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Comorbidity , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Retrospective Studies , Young Adult
10.
Cancer ; 117(22): 5085-93, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21523765

ABSTRACT

BACKGROUND: Deletions of derivative chromosome 9 are a poor prognostic factor in patients with chronic myeloid leukemia (CML) treated with hydroxyurea, interferon, or stem cell transplantation. Imatinib may overcome the adverse prognostic impact of deletions of derivative chromosome 9. METHODS: A study was undertaken to investigate the prognostic impact of deletions of derivative chromosome 9 in 353 patients with CML receiving the second generation tyrosine kinase inhibitors (TKIs) nilotinib (n = 161) or dasatinib (n = 192). RESULTS: Deletion of derivative chromosome 9 status was determined in 245 (69%). Twenty-eight (11%) patients, 22 in chronic phase, 4 in accelerated phase, and 2 in blast phase, carried deletions of derivative chromosome 9, including 17 receiving nilotinib and 11 receiving dasatinib (P = .47). Overall survival (OS) at 24 months was similar between patients with or without deletions of derivative chromosome 9 (70% vs 71%, P = .76). For patients in chronic phase, no significant differences in overall major cytogenetic response (77% vs 82%, P = .57) or complete cytogenetic response (77% vs 81%, P = .71) rates were observed between patients with or without deletions of derivative chromosome 9. At 24 months, patients with CML in chronic phase without deletions of derivative chromosome 9 had improved event-free survival (EFS) (88% vs 66%, P = .07) and OS (96% vs 82%; P = .08) compared with those carrying deletions of derivative chromosome 9. However, multivariate analysis established second-line versus frontline second generation TKI therapy as the only adverse prognostic factor for EFS and increased bone marrow blast burden and older age as independent adverse prognostic factors for OS. CONCLUSIONS: Deletions of derivative chromosome 9 do not appear to be an independent risk factor for survival among patients with CML in chronic phase receiving second generation TKIs.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosomes, Human, Pair 9 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Dasatinib , Disease-Free Survival , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Male , Middle Aged , Prognosis , Protein-Tyrosine Kinases/antagonists & inhibitors , Sequence Deletion , Treatment Outcome
11.
Am J Hematol ; 86(7): 546-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21509801

ABSTRACT

Intracranial hemorrhage (ICH) is associated with great morbidity and mortality in patients with acute leukemia. We identified 118 patients with ICH from a total of 2,421 patient with leukemia who were treated at our institution between 2005-2009, and assessed the prognostic factors for mortality in the ICH cohort. Median age at time of ICH was 58 years, 49% were male, and 60% had acute myeloid leukemia. The relative incidence of ICH was highest in patients with chronic myeloid leukemia in blast crisis (12.1%). Mental status changes were the most common symptom which prompted work-up for ICH. Median survival for all patients after onset of ICH was 20 days. In multivariate analyses, four clinical characteristics were identified as independent adverse factors for mortality in patients with ICH: albumin <3.5 g/dL, lactate dehydrogenase (LDH) >835 U/L, age > 60 years, and relapsed disease status. Based on the number of risk factors, mortality after ICH was: 25% (0 risk factor), 47% (1), 78% (2), and >97% (3 or 4). In conclusion, patients with leukemia who develop ICH do not have universally poor outcomes, and a simple scoring system could serve to advise physicians and families of the prognosis and the potential benefit of aggressive treatment options.


Subject(s)
Intracranial Hemorrhages/mortality , Leukemia/mortality , Models, Biological , Acute Disease , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Incidence , Intracranial Hemorrhages/blood , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/therapy , L-Lactate Dehydrogenase/blood , Leukemia/blood , Leukemia/complications , Leukemia/therapy , Male , Middle Aged , Retrospective Studies , Risk Factors , Serum Albumin/analysis
12.
Haematologica ; 96(6): 918-21, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21357704

ABSTRACT

We investigated the impact of carrying more than one BCR-ABL1 mutation in 207 patients with chronic myeloid leukemia (102 chronic, 61 accelerated, and 44 blast phase) post-imatinib failure. Seven (8%) of 92 patients carrying mutations had more than one mutation: 4 (4%) in chronic phase, 2 (2%) in accelerated phase, and one (1 %) in blast phase. The cytogenetic response rate to second generation TKIs for patients with no, one, or more than one mutation was 88%, 69%, 50% (P=0.03) in chronic phase, 54%, 42%, 50% in accelerated phase (P=0.67), and 35%, 25%, 0% (P=0.63) in blast phase, respectively. No differences were observed in event free survival or overall survival in accelerated or blast phase according to their mutational status. However, the 4-year event free survival rates among patients in chronic phase with no, one, or more than one BCR-ABL1 mutation were 56%, 49%, and 0%, respectively (P=0.02), with overall survival rates of 91%, 69%, and 75%, respectively (P=0.13). In conclusion, patients with more than one BCR-ABL1 mutation fare worse than those with no or one mutation.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/pharmacology , Survival Analysis , Treatment Outcome , Young Adult
13.
Cancer ; 117(2): 327-35, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20845478

ABSTRACT

BACKGROUND: Imatinib is an effective tyrosine kinase inhibitor (TKI) for patients with chronic myeloid leukemia (CML) in chronic phase (CP). Although some patients may fail on therapy with imatinib, effective salvage therapy is available with second-generation TKIs. Current measurement of efficacy for each therapy is judged by its individual impact on overall survival and event-free survival (EFS). METHODS: In total, 586 patients with CML in CP who received imatinib were included in this analysis in 2 cohorts: imatinib as front-line therapy (n = 281) or after failure on interferon-α (IFN-α) (n = 305). By accounting for successful salvage treatment (ie, regain of complete cytogenetic response), the current EFS (CEFS) rate was calculated to obtain a more accurate impression of the outcome of patients with CML who received treatment with sequential TKIs. RESULTS: For patients who received imatinib after failing on IFN-α, the 7-year EFS rate was 61%, whereas the CEFS rate was 69%. The 7-year EFS rate for patients who received imatinib as initial therapy was 81% compared with a 7-year CEFS rate of 88%. CONCLUSIONS: CEFS provided a more accurate representation of the outcome of patients with CML in CP. These patients may frequently be treated successfully with subsequent TKIs after experiencing failure on the first TKI.


Subject(s)
Disease-Free Survival , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Benzamides , Female , Humans , Imatinib Mesylate , Interferon-alpha/therapeutic use , Leukemia, Myeloid, Chronic-Phase/mortality , Male , Middle Aged , Retreatment , Salvage Therapy
14.
Blood ; 116(26): 5818-23; quiz 6153, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20923968

ABSTRACT

Pretreatment characteristics and outcome of patients treated with induction regimens containing high-dose ara-C (HiDAC) at M. D. Anderson Cancer Center refractory to 1 cycle of induction were compared with similar patients achieving a complete response (CR). Among 1597 patients treated with HiDAC-based induction from 1995 to 2009, 285 were refractory to 1 cycle. Median age was 59 years (range, 18-85 years). Induction regimens included HiDAC with anthracyclines (n = 181; 64%) or HiDAC with nonanthracycline chemotherapy (n = 104; 36%). Refractory patients were older (median age, 59 vs 56 years; P < .001), more likely with unfavorable cytogenetics (P < .001) and antecedent hematologic disorder (P < .001), and had a higher presentation white blood cell count (P = .04), but not a higher incidence of FLT3 mutations (P = .85), than those achieving CR. Forty-three patients (22%) responded to salvage (35 CR and 8 CR without platelet recovery). With a median follow-up of 72 months (range, 27-118 months) in responders, 11 are alive. Nineteen patients (7%) were alive and in CR for at least 6 months, including 9 who underwent allogeneic stem cell transplantation. On multivariate analysis, severe thrombocytopenia, leukocytosis, increasing marrow blast percentage, unfavorable cytogenetics, and salvage not including allogeneic stem cell transplantation were associated with a worse survival. Alternative strategies are needed for these patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Anthracyclines/administration & dosage , Cytarabine/administration & dosage , Female , Humans , Male , Middle Aged , Remission Induction , Survival Rate , Treatment Outcome , Young Adult
15.
Blood ; 116(22): 4422-9, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20668231

ABSTRACT

Patients ≥ 70 years of age with acute myeloid leukemia (AML) have a poor prognosis. Recent studies suggested that intensive AML-type therapy is tolerated and may benefit most. We analyzed 446 patients ≥ 70 years of age with AML (≥ 20% blasts) treated with cytarabine-based intensive chemotherapy between 1990 and 2008 to identify risk groups for high induction (8-week) mortality. Excluding patients with favorable karyotypes, the overall complete response rate was 45%, 4-week mortality was 26%, and 8-week mortality was 36%. The median survival was 4.6 months, and the 1-year survival rate was 28%. Survival was similar among patients treated before 2000 and since 2000. A multivariate analysis of prognostic factors for 8-week mortality identified the following to be independently adverse: age ≥ 80 years, complex karyotypes, (≥ 3 abnormalities), poor performance (2-4 Eastern Cooperative Oncology Group), and elevated creatinine > 1.3 mg/dL. Patients with none (28%), 1 (40%), 2 (23%), or ≥ 3 factors (9%) had estimated 8-week mortality rates of 16%, 31%, 55%, and 71% respectively. The 8-week mortality model also predicted for differences in complete response and survival rates. In summary, the prognosis of most patients (72%) ≥ 70 years of age with AML is poor with intensive chemotherapy (8-week mortality ≥ 30%; median survival < 6 months).


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Age Factors , Aged , Aged, 80 and over , Humans , Prognosis , Risk Factors , Survival Rate
16.
Cancer ; 116(13): 3152-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20564631

ABSTRACT

BACKGROUND: Imatinib 400 mg daily is the standard treatment for patients with chronic myelogenous leukemia (CML). The safety and efficacy of imatinib in CML patients with pre-existing liver and/or renal dysfunction has not been analyzed. METHODS: The authors analyzed the outcome of 259 patients with early chronic phase CML treated with imatinib (starting dose 400 mg in 50, 800 mg in 209). Pre-existing liver and/or renal dysfunction was seen in 38 (15%) and 11 (4%) patients, respectively. RESULTS: Dose reductions were required in 91 (43%) of 210 patients with normal organ function, compared with 8 (73%) of 11 (P = .065) with renal dysfunction, and 19 (50%) of 38 (P = .271) with liver dysfunction. Grade 3-4 hematologic toxicities including anemia (29%, 10%, and 7% of patients with renal dysfunction, liver dysfunction, and normal organ function, respectively), neutropenia (57%, 30%, and 30%), and thrombocytopenia (43%, 30%, and 26%) were more frequent in patients with pre-existing renal dysfunction treated with high-dose imatinib. Grade 3-4 nonhematologic toxicities were observed at similar frequencies. Complete cytogenetic response rates, event-free survival, and overall survival were similar in all groups. CONCLUSIONS: Although patients with pre-existing liver and/or renal dysfunction might have a higher rate of hematologic toxicity and require more frequent dose reductions, most patients can be adequately managed, resulting in response rates and survival similar to those without pre-existing organ dysfunction.


Subject(s)
Antineoplastic Agents/therapeutic use , Kidney Diseases/complications , Leukemia, Myeloid, Chronic-Phase/complications , Leukemia, Myeloid, Chronic-Phase/drug therapy , Liver Diseases/complications , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Benzamides , Female , Humans , Imatinib Mesylate , Kidney Diseases/physiopathology , Leukemia, Myeloid, Chronic-Phase/mortality , Liver Diseases/physiopathology , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects
17.
Cancer ; 116(11): 2673-81, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20499401

ABSTRACT

BACKGROUND: Clonal evolution is frequently detected in patients developing resistance to imatinib. The outcome of patients with clonal evolution treated with second generation tyrosine kinase inhibitors is not known. METHODS: The authors analyzed the outcome of 177 CML patients after second tyrosine kinase inhibitor therapy. RESULTS: Ninety-five patients were in chronic phase, 30 had clonal evolution, 28 were in accelerated phase (AP), and 24 were in AP plus clonal evolution. Major cytogenetic response rates were 58%, 54%, 28%, and 13%; 2-year overall survival (OS) rates were 86%, 73%, 68%, and 33%; and 2-year event-free survival (EFS) rates were 69%, 67%, 31%, and 8%, respectively. The hematologic and cytogenetic response rates, OS, and EFS were no different between patients in chronic phase with clonal evolution and patients with chronic phase and no clonal evolution. However, clonal evolution had a significant adverse impact when associated with other features of AP. On multivariate analysis, clonal evolution had no independently significant effect on achieving major cytogenetic response on the second generation tyrosine kinase inhibitors. The factors predicting increasing major cytogenetic response to second generation tyrosine kinase inhibitors were prior achievement of major cytogenetic response with imatinib, higher hemoglobin levels, no splenomegaly, lower percentage of Philadelphia chromosome-positive metaphases, and no prior chemotherapy. CONCLUSIONS: Clonal evolution constitutes a heterogeneous entity with variable outcome with second generation tyrosine kinase inhibitors, with trisomy 8, chromosome 17, and complex abnormalities having the worst outcome, regardless of the number of metaphases involved. The molecular events behind these abnormalities and potential therapeutic approaches directed at them need to be defined.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Adult , Aged , Aged, 80 and over , Benzamides , Chromosome Aberrations , Dasatinib , Drug Resistance, Neoplasm , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Middle Aged , Piperazines/therapeutic use , Treatment Outcome
18.
J Clin Oncol ; 28(4): 605-13, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20038729

ABSTRACT

PURPOSE: The current classification systems of myelodysplastic syndromes (MDS), including the International Prognostic Scoring System (IPSS), do not fully reflect the molecular heterogeneity of the disease. Molecular characterization may predict clinical outcome and help stratify patients for targeted therapies. Epigenetic therapy using decitabine, a DNA hypomethylating agent, is clinically effective for the treatment of MDS. Therefore, we investigated the association between DNA methylation and clinical outcome in MDS. PATIENTS AND METHODS: We screened 24 patients with MDS for promoter CpG island methylation of 24 genes and identified aberrant hypermethylation at 10 genes. We then performed quantitative methylation analyses by bisulfite pyrosequencing of the identified genes in 317 patient samples from three independent studies and assessed relations between methylation and clinical outcome. RESULTS: In an initial training cohort of 89 patients with MDS, methylation frequencies of individual genes ranged from 7% to 70% and were highly concordant. Therefore, we defined a methylation z score based on all genes for each patient. We found that patients with higher levels of methylation, compared with patients with lower levels, had a shorter median overall survival (12.3 v 17.5 months, respectively; P = .04) and shorter median progression-free survival (6.4 v 14.9 months, respectively; P = .009). This methylation prognostic model was independent of age, sex, and IPSS group. Applied to two validation cohorts (228 patients), this model was confirmed as an independent prognostic predictor for survival. Although methylation at baseline did not correlate with clinical response to decitabine, we observed a significant correlation between reduced methylation over time and clinical responses. CONCLUSION: DNA methylation predicts overall and progression-free survival in MDS.


Subject(s)
CpG Islands , DNA Methylation , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Adult , Aged , Aged, 80 and over , Clinical Trials, Phase III as Topic , Cohort Studies , Disease-Free Survival , Female , Humans , Male , Middle Aged , Multicenter Studies as Topic , Myelodysplastic Syndromes/genetics , Neoplasm Staging , Polymerase Chain Reaction , Randomized Controlled Trials as Topic , Survival Rate , Treatment Outcome , Young Adult
19.
J Clin Oncol ; 28(3): 398-404, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20008620

ABSTRACT

PURPOSE: Dasatinib is effective therapy for chronic myeloid leukemia (CML) after imatinib failure. In this study, we investigate the efficacy of dasatinib as initial therapy for patients with CML in early chronic phase. PATIENTS AND METHODS: Patients with newly diagnosed CML in early chronic phase were randomly assigned to receive dasatinib 100 mg once daily or 50 mg twice daily as initial therapy. RESULTS: Among 50 patients observed for at least 3 months, 49 patients (98%) achieved a complete cytogenetic response (CCyR), and 41 patients (82%) achieved a major molecular response (MMR). Responses occurred rapidly, with 94% of patients achieving CCyR by 6 months. There was no difference in response rate by treatment arm. The projected event-free survival rate at 24 months is 88%, and all patients are alive after a median follow-up time of 24 months. Grade >or= 3 neutropenia and thrombocytopenia occurred in 21% and 10% of patients, respectively. Nonhematologic toxicity was usually grade 1 to 2. There was no significant difference in toxicity between the two arms, and the actual median dose at 12 months was 100 mg (range, 20 to 100 mg). CONCLUSION: Dasatinib is an effective agent for the initial management of CML in early chronic phase, producing high rates of CCyR and MMR.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Chronic-Phase/drug therapy , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , Adolescent , Adult , Aged , Dasatinib , Humans , Middle Aged , Young Adult
20.
J Clin Oncol ; 28(3): 392-7, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20008621

ABSTRACT

PURPOSE: Although most patients with chronic myeloid leukemia (CML) in chronic phase respond well to front-line therapy with imatinib, some patients do not achieve the desirable end point, and others may eventually lose response or are intolerant. PATIENTS AND METHODS: Patients with newly diagnosed CML in chronic phase were treated with nilotinib 400 mg twice daily on an empty stomach as initial therapy. RESULTS: Among 51 patients in chronic phase observed for at least 3 months, 50 (98%) achieved a complete cytogenetic remission (CCyR), and 39 (76%) achieved a major molecular response (MMR). Responses occurred rapidly, with 96% of patients achieving CCyR by 3 months and 98% achieving CCyR by 6 months. The projected event-free survival at 24 months is 90%, and all patients are alive after a median follow-up time of 17 months. Grade >or= 3 neutropenia occurred in 12% of patients, and thrombocytopenia in occurred 11%. Nonhematologic toxicity was usually grade 1 to 2 and manageable. The actual median dose at 12 months was 800 mg (range, 200 to 800 mg). CONCLUSION: Nilotinib is an effective option for the initial management of CML in early chronic phase, producing high rates of CCyR and MMR, with most patients reaching these responses early during their therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Chronic-Phase/drug therapy , Pyrimidines/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...