Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Pharmacol Ther ; 114(4): 845-852, 2023 10.
Article in English | MEDLINE | ID: mdl-37422689

ABSTRACT

Oral azacitidine (oral-AZA) maintenance is approved for adults with acute myeloid leukemia (AML) in remission post-intensive chemotherapy, not proceeding to hematopoietic stem cell transplantation. This study aimed to develop a population pharmacokinetic (PopPK) model to characterize oral-AZA concentration-time profiles in patients with AML, myelodysplastic syndrome, or chronic myelomonocytic leukemia. PopPK-estimated exposure parameters were used to evaluate exposure-response relationships in the phase III QUAZAR AML-001 study. The PopPK dataset comprised 286 patients with 1,933 evaluable oral-AZA concentration records. The final PopPK model was a one-compartment model with first-order absorption incorporating an absorption lag time and first-order elimination. Regression analyses identified two oral-AZA exposure parameters (area under the plasma concentration-time curve at steady state (AUCss ); maximum plasma concentration (Cmax )) as statistically significant predictors for relapse-free survival (hazard ratio (HR) = 0.521, P < 0.001; HR = 0.630, P = 0.013; respectively), and AUCss as a significant predictor for overall survival (HR = 0.673, P = 0.042). The probability of grade ≥ 3 neutropenia was significantly increased with increases in AUCss (odds ratio (OR) = 5.71, 95% confidence interval (CI) = 2.73-12.62, P < 0.001), cumulative AUC through cycles 1 to 6 (OR = 2.71, 95% CI = 1.76-4.44, P < 0.001), and Cmax at steady-state (OR = 2.38, 95% CI = 1.23-4.76, P = 0.012). A decreasing trend was identified between AUCss and relapse-related schedule extensions, vs. an increasing trend between AUCss and event-related dose reductions. As the majority (56.8%) of patients required no dose modifications, and the proportions requiring schedule extension (19.4%) or dose reduction (22.9%) were almost equal, oral-AZA 300 mg once daily for 14 days is the optimal dosing schedule balancing survival benefit and safety risk.


Subject(s)
Leukemia, Myeloid, Acute , Leukemia, Myelomonocytic, Chronic , Myelodysplastic Syndromes , Adult , Humans , Antimetabolites , Antimetabolites, Antineoplastic , Azacitidine/adverse effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukemia, Myelomonocytic, Chronic/drug therapy , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/chemically induced , Clinical Trials, Phase III as Topic
2.
Wien Med Wochenschr ; 173(1-2): 34-40, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36282401

ABSTRACT

The impact of treatment on the outcome of chronic myelomonocytic leukemia (CMML) patients over a longer period of time and the potential role of predictive factors are not well defined. In a retrospective observational study, we analyzed 168 CMML patients regarding treatment options and survival during the past three decades. The proportion of patients treated with hydroxyurea (HU), intensive chemotherapy, and azacitidine (AZA) was 65/19/0% before 2000, 51/25/32% from 2000-2010, and 36/12/53% after 2010, respectively. Median overall survival (OS) increased from 10 months before 2000 to 23 months thereafter (p = 0.021). AZA-treated patients but not patients treated with other treatment options had improved survival as compared to CMML patients without AZA therapy (19 vs. 25 months, p = 0.041). When looking at subgroups, the following patient cohorts had a significant survival benefit in association with AZA therapy: patients with Hb > 10 g/dL, patients with monocytosis > 10 G/L, and patients with mutations in RASopathy genes.


Subject(s)
Leukemia, Myelomonocytic, Chronic , Humans , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/chemically induced , Antimetabolites, Antineoplastic/adverse effects , Azacitidine/therapeutic use , Azacitidine/adverse effects , Retrospective Studies , Hydroxyurea/therapeutic use
3.
BMC Cancer ; 22(1): 569, 2022 May 21.
Article in English | MEDLINE | ID: mdl-35597904

ABSTRACT

BACKGROUND: This report summarizes three phase I studies evaluating volasertib, a polo-like kinase inhibitor, plus azacitidine in adults with myelodysplastic syndromes (MDS), chronic myelomonocytic leukemia, or acute myeloid leukemia. METHODS: Patients received intravenous volasertib in 28-day cycles (dose-escalation schedules). In Part 1 of 1230.33 (Study 1; NCT01957644), patients received 250-350 mg volasertib on day (D)1 and D15; in Part 2, patients received different schedules [A, D1: 170 mg/m2; B, D7: 170 mg/m2; C, D1 and D7: 110 mg/m2]. In 1230.35 (Study 2; NCT02201329), patients received 200-300 mg volasertib on D1 and D15. In 1230.43 (Study 3; NCT02721875), patients received 110 mg/m2 volasertib on D1 and D8. All patients in Studies 1 and 2, and approximately half of the patients in Study 3, were scheduled to receive subcutaneous azacitidine 75 mg/m2 on D1-7. RESULTS: Overall, 22 patients were treated (17 with MDS; 12 previously untreated). Across Studies 1 and 2 (n = 21), the most common drug-related adverse events were hematological (thrombocytopenia [n = 11]; neutropenia [n = 8]). All dose-limiting toxicities were grade 4 thrombocytopenia. The only treated patient in Study 3 experienced 18 adverse events following volasertib monotherapy. Studies 1 and 2 showed preliminary activity (objective response rates: 25 and 40%). CONCLUSIONS: The safety of volasertib with azacitidine in patients with MDS was consistent with other volasertib studies. All studies were terminated prematurely following the discontinuation of volasertib for non-clinical reasons by Boehringer Ingelheim; however, safety information on volasertib plus azacitidine are of interest for future studies in other diseases.


Subject(s)
Leukemia, Myeloid, Acute , Leukemia, Myelomonocytic, Chronic , Myelodysplastic Syndromes , Thrombocytopenia , Adult , Azacitidine/therapeutic use , Clinical Trials, Phase I as Topic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukemia, Myelomonocytic, Chronic/drug therapy , Myelodysplastic Syndromes/chemically induced , Myelodysplastic Syndromes/drug therapy , Pteridines , Thrombocytopenia/chemically induced
5.
Blood ; 122(16): 2807-11; quiz 2920, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-23896412

ABSTRACT

We sought to describe the clinical features and outcomes of therapy-related chronic myelomonocytic leukemia (t-CMML) and compare with those of de novo CMML. We identified 358 CMML patients, of whom 39 (11%) had t-CMML. Although the groups had similar demographic, hematologic, and molecular alteration profiles, the proportion of patients with intermediate or high CMML-specific cytogenetic risk in the t-CMML was significantly higher than that in the de novo CMML (P = .011). The median latency to develop t-CMML was 6 years. The median overall and leukemia-free survival duration of the t-CMML were shorter than those of the de novo CMML; however, t-CMML itself was not prognostic after adjusting for the effects of other covariates including cytogenetics. These results suggest that compared with de novo CMML, t-CMML is associated with more high-risk cytogenetics that manifest as poor outcomes. We propose that t-CMML be recognized as one of the therapy-related myeloid neoplasms.


Subject(s)
Antineoplastic Agents/adverse effects , Leukemia, Myelomonocytic, Chronic/pathology , Neoplasms, Radiation-Induced/diagnosis , Radiotherapy/adverse effects , Adult , Aged , Aged, 80 and over , Cytogenetics , Female , Humans , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukemia, Myelomonocytic, Chronic/therapy , Male , Middle Aged , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/radiotherapy , Prognosis , Time Factors , Treatment Outcome , Young Adult
6.
N Engl J Med ; 367(24): 2316-21, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23134356

ABSTRACT

Vemurafenib, a selective RAF inhibitor, extends survival among patients with BRAF V600E-mutant melanoma. Vemurafenib inhibits ERK signaling in BRAF V600E-mutant cells but activates ERK signaling in BRAF wild-type cells. This paradoxical activation of ERK signaling is the mechanistic basis for the development of RAS-mutant squamous-cell skin cancers in patients treated with RAF inhibitors. We report the accelerated growth of a previously unsuspected RAS-mutant leukemia in a patient with melanoma who was receiving vemurafenib. Exposure to vemurafenib induced hyperactivation of ERK signaling and proliferation of the leukemic cell population, an effect that was reversed on drug withdrawal.


Subject(s)
Genes, ras , Indoles/adverse effects , Leukemia, Myelomonocytic, Chronic/genetics , Melanoma/genetics , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/adverse effects , Aged , Cell Proliferation/drug effects , Disease Progression , Humans , Indoles/therapeutic use , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukocyte Count , Male , Melanoma/drug therapy , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use , Vemurafenib
7.
Int J Hematol ; 89(5): 699-703, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19430863

ABSTRACT

Therapy related chronic myelomonocytic leukemia (t-CMML) is rare. We report a 23-year-old female who developed acute fulminant hepatic failure after drug overdose. She underwent ABO incompatible orthotopic liver transplant. She received cyclophosphamide along with other immunosuppressants. Seven years later, she was diagnosed with t-CMML-2 with 45XX,-7 karyotype. She received 4 cycles of azacitidine and proceeded with allogeneic bone marrow transplant. This is the first a case of t-CMML reported in a liver transplant recipient. In this article, we also summarize all reported cases of t-CMML, and we review therapy related MDS in recipients of solid organ transplant.


Subject(s)
Leukemia, Myelomonocytic, Chronic/chemically induced , Liver Transplantation/adverse effects , Azacitidine/therapeutic use , Bone Marrow Transplantation , Cyclophosphamide/adverse effects , Female , Humans , Immunosuppressive Agents/adverse effects , Leukemia, Myelomonocytic, Chronic/therapy , Liver Failure, Acute/surgery , Liver Transplantation/methods , Young Adult
9.
J Clin Pharm Ther ; 31(4): 401-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16882113

ABSTRACT

Chronic myelomonocytic leukaemia (CMML) is a preleukaemic condition with myeloproliferative features, and classified as a part of myelodysplastic syndrome (MDS). Other than alkylating agents and topoisomerase II inhibitors, there is less evidence that chemotherapeutic drugs are associated with therapy-related CMML, acute leukaemia or MDS. We present a patient who developed CMML within 2 years of platinum-based chemotherapy for a metastatic non-small cell lung cancer. He received a cumulative dose of 240 mg/m(2) of cisplatin, and 1123 mg/m(2) of carboplatin before developing CMML. The cytogenetic study revealed trisomy 8. This is the first reported case that links platinum-based therapy with development of CMML with trisomy 8. Although the relationship between platinum therapy and the development of CMML is difficult to assess due to combinational nature of therapy in most cases, physicians should consider the possibility of CMML in patients with symptoms or signs suggestive of haematologic malignancy after platinum therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/adverse effects , Leukemia, Myelomonocytic, Chronic/chemically induced , Lung Neoplasms/drug therapy , Etoposide/administration & dosage , Fatal Outcome , Humans , Leukemia, Myelomonocytic, Chronic/physiopathology , Male , Middle Aged
11.
Genes Chromosomes Cancer ; 20(1): 60-3, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9290955

ABSTRACT

CBP, which is located on 16p13 and encodes a transcriptional adaptor/coactivator protein, has been shown to fuse by the t(8;16)(p11;p13) translocation to MOZ on 8p11 in acute myeloid leukemia. We found a t(11;16)(q23;p13) in a child with therapy-related chronic myelomonocytic leukemia. Subsequent reverse transcriptase-polymerase chain reaction and direct sequencing analyses revealed the MLL-CBP fusion transcript in CMML cells. Because 11q23 translocations involving MLL and t(8;16) involving MOZ and CBP have been reported in therapy-related leukemias, both the MLL and CBP genes may be targets for topoisomerase II inhibitors. Accordingly, we believe that most t(11;16)-associated leukemias may develop in patients who have been treated with cytotoxic chemotherapy for primary malignant diseases.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 16/genetics , DNA-Binding Proteins/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Neoplasms, Second Primary/genetics , Nuclear Proteins/genetics , Proto-Oncogenes , Recombination, Genetic , Trans-Activators , Transcription Factors/genetics , Translocation, Genetic , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blotting, Southern , CREB-Binding Protein , Child , Chromosome Banding , DNA Primers , DNA, Neoplasm/analysis , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myelomonocytic, Chronic/chemically induced , Male , Myeloid-Lymphoid Leukemia Protein , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA-Directed DNA Polymerase , Remission Induction , Sequence Analysis, DNA
12.
Acta Haematol ; 96(4): 251-4, 1996.
Article in English | MEDLINE | ID: mdl-8922495

ABSTRACT

A 19-year-old male with chronic myelomonocytic leukaemia (CMML) with bone marrow eosinophilia terminating in myeloblastic transformation is described. Before the appearance of CMML, he received a total dose of 2,894 mg of nimustine for the treatment of a pontine glioma over 12 years. Peripheral blood count showed leucocytosis with mature neutrophils and monocytes. Bone marrow smears showed myeloid hyperplasia with increased blasts and abnormal eosinophils and dysplastic features in the myeloid and megakaryocytic cells. A karyotype analysis of the bone marrow cells indicated 46, XY, der(11)t(1;11)(q21;q14). No bcr/abl rearrangement was observed in the cells. He was treated with hydroxyurea to control the leucocyte count, but myeloblastic transformation developed.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow/pathology , Brain Neoplasms/drug therapy , Eosinophilia/complications , Eosinophilia/genetics , Glioma/drug therapy , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukemia, Myelomonocytic, Chronic/complications , Nimustine/adverse effects , Brain Neoplasms/radiotherapy , Child , Chromosome Aberrations , Fatal Outcome , Glioma/radiotherapy , Humans , Hydroxyurea/pharmacology , Karyotyping , Leukocyte Count/drug effects , Lymphocyte Activation , Male
13.
Int J Hematol ; 60(4): 263-5, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7894029

ABSTRACT

A 35-year-old male presented with chronic myelomonocytic leukemia (CMMoL) after 6.5 years of alkylating agent therapy for IgG-kappa type multiple myeloma. The total dose of melphalan was 0.648 g. CMMoL was stable with weekly injection of alpha-interferon for one year. Thereafter, monocytosis and thrombocytopenia aggravated, and the patient died of disseminating intravascular coagulation. Prolonged drug therapy can induce CMMoL, as well as other myelodysplastic syndromes.


Subject(s)
Leukemia, Myelomonocytic, Chronic/chemically induced , Melphalan/adverse effects , Multiple Myeloma/drug therapy , Neoplasms, Second Primary/chemically induced , Adult , Disseminated Intravascular Coagulation/chemically induced , Humans , Male , Multiple Myeloma/pathology , Myelodysplastic Syndromes/chemically induced
14.
Am J Clin Pathol ; 100(3): 270-5, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8379535

ABSTRACT

Three cases of secondary (therapy-related) hematologic malignant conditions were identified among 95 children as old as 18 years of age; the cases were diagnosed between 1984 and 1990 and consisted of acute lymphoblastic leukemia, acute myeloid leukemia (AML), and myelodysplastic syndrome (MDSs). They constituted 10% of all new cases of AML and MDS seen at the University Hospitals of Cleveland during this time and were not related to congenital factors. The primary malignant conditions were malignant thoracopulmonary tumor (Askin tumor), neuroblastoma, and Burkitt's lymphoma. The secondary hematologic disorders all showed a prominent monocytic component: acute monocytic leukemia, MDSs evolving to acute myelomonocytic leukemia, and chronic myelomonocytic leukemia. The mean interval between treatment for the primary malignant condition and the onset of secondary disease was 36 months. All had received cyclophosphamide and an epipodophyllotoxin for the primary tumor; two were treated with radiation therapy. Cytogenetic abnormalities included del(5), del(13), t(1;6), and t(9;11)(p22[symbol:see text]3). The survival time after the onset of secondary disease was short.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Leukemia, Myelomonocytic, Acute/chemically induced , Leukemia, Myelomonocytic, Chronic/chemically induced , Leukemia, Promyelocytic, Acute/chemically induced , Neoplasms, Second Primary/chemically induced , Adolescent , Antigens, CD/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Chromosome Aberrations/chemically induced , Chromosome Disorders , Combined Modality Therapy , Female , Humans , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/immunology , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/immunology , Male , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/immunology
15.
J Med ; 22(3): 157-61, 1991.
Article in English | MEDLINE | ID: mdl-1770323

ABSTRACT

From 1980 to 1987, three cases of chronic myelomonocytic leukemia (CMML) were encountered among 68 cases of multiple myeloma who survived more than three years from the diagnosis. The incidence (4.8%) of secondary myelodysplastic syndrome (MDS) is almost identical to previous reports, but case reports of chronic myelomonocytic leukemia were rare. In Japan, there are few reports of multiple myeloma patients who later developed secondary MDS or acute myelogenous leukemia (AML). In our cases, none of the 31 patients treated with cyclophosphamide developed secondary MDS, while three of 37 patients treated with melphalan developed CMML. This difference is not statistically significant.


Subject(s)
Leukemia, Myelomonocytic, Chronic/chemically induced , Melphalan/adverse effects , Multiple Myeloma/complications , Aged , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Myelodysplastic Syndromes/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...