Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Jpn J Clin Oncol ; 53(7): 595-603, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37017320

ABSTRACT

BACKGROUND: In a Phase 3 international clinical trial (VIALE-C), venetoclax plus low-dose cytarabine improved the response rate and overall survival versus placebo plus low-dose cytarabine in patients with newly diagnosed acute myeloid leukemia who were ineligible for intensive chemotherapy. After the enrollment period of VIALE-C ended, we conducted an expanded access study to provide preapproval access to venetoclax in combination with low-dose cytarabine in Japan. METHODS: Previously, untreated patients with acute myeloid leukemia who were ineligible for intensive chemotherapy were enrolled according to the VIALE-C criteria. Patients received venetoclax (600 mg, Days 1-28, 4-day ramp-up in Cycle 1) in 28-day cycles and low-dose cytarabine (20 mg/m2, Days 1-10). All patients took tumor lysis syndrome prophylactic agents and hydration. Safety endpoints were assessed. RESULTS: Fourteen patients were enrolled in this study. The median age was 77.5 years (range = 61-84), with 78.6% over 75 years old. The most common grade ≥ 3 treatment-emergent adverse event was neutropenia (57.1%). Febrile neutropenia was the most frequent serious adverse event (21.4%). One patient developed treatment-related acute kidney injury, leading to discontinuation of treatment. Two patients died because of cardiac failure and disease progression that were judged not related to study treatment. No patients developed tumor lysis syndrome. CONCLUSIONS: The safety outcomes were similar to those in VIALE-C without new safety signals and were well managed with standard medical care. In clinical practice, more patients with severe background disease are expected, in comparison with in VIALE-C, suggesting that it is important to carefully manage and prevent adverse events.


Subject(s)
Leukemia, Myeloid, Acute , Tumor Lysis Syndrome , Aged , Aged, 80 and over , Humans , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytarabine/therapeutic use , Japan , Leukemia, Myeloid, Acute/drug therapy , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/prevention & control , Tumor Lysis Syndrome/drug therapy
2.
Acta Haematol ; 134(1): 25-31, 2015.
Article in English | MEDLINE | ID: mdl-25871926

ABSTRACT

AIMS: To explore the biomarker for predicting the occurrence of adverse events in myeloma patients treated by intravenous bortezomib, we measured proteasome activity in peripheral blood mononuclear cells. METHODS: Samples were obtained from 34 bortezomib-naïve patients. Proteasome activity was measured at pre- and postchemotherapy phase by using a synthetic substrate. RESULTS: Bortezomib injection resulted in a dramatic decrease in proteasome activity, reaching 32.4 ± 18.79% (mean ± SD) of the pretreatment level at 1 h, but it generally recovered at the end of the first course. In total, 6 patients manifested with severe bortezomib-induced peripheral neuropathy (sBIPN) in the second-third course. There was a nonsignificant trend for these patients to have lower levels of the relative proteasome activity at the end of the first course than those without sBIPN (median: 74.03 vs. 103.2%, p = 0.052). Moreover, in all of them, proteasome activity did not recover to the pretreatment level, whereas no patients with complete recovery manifested with sBIPN. Analysis with Fisher's exact test demonstrated that incomplete recovery of proteasome activity is a significant risk factor for sBIPN (p = 0.014). CONCLUSION: Patients with incomplete recovery of proteasome activity are at high risk for developing sBIPN, and the susceptible patients can be indicated by monitoring proteasome activity.


Subject(s)
Antineoplastic Agents/adverse effects , Boronic Acids/adverse effects , Leukocytes, Mononuclear , Multiple Myeloma , Neoplasm Proteins/metabolism , Peripheral Nervous System Diseases , Proteasome Endopeptidase Complex/metabolism , Pyrazines/adverse effects , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Boronic Acids/administration & dosage , Bortezomib , Disease Susceptibility , Female , Follow-Up Studies , Humans , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/enzymology , Peripheral Nervous System Diseases/pathology , Pyrazines/administration & dosage
3.
PLoS One ; 10(3): e0119442, 2015.
Article in English | MEDLINE | ID: mdl-25790231

ABSTRACT

Erythropoietin (Epo) and its receptor (EpoR) are required for the regulation of erythropoiesis. Epo binds to the EpoR homodimer on the surface of erythroid progenitors and erythroblasts, and positions the intracellular domains of the homodimer to be in close proximity with each other. This conformational change is sufficient for the initiation of Epo-EpoR signal transduction. Here, we established a system of chemically regulated erythropoiesis in transgenic mice expressing a modified EpoR intracellular domain (amino acids 247-406) in which dimerization is induced using a specific compound (chemical inducer of dimerization, CID). Erythropoiesis is reversibly induced by oral administration of the CID to the transgenic mice. Because transgene expression is limited to hematopoietic cells by the Gata1 gene regulatory region, the effect of the CID is limited to erythropoiesis without adverse effects. Additionally, we show that the 160 amino acid sequence is the minimal essential domain of EpoR for intracellular signaling of chemically inducible erythropoiesis in vivo. We propose that the CID-dependent dimerization system combined with the EpoR intracellular domain and the Gata1 gene regulatory region generates a novel peroral strategy for the treatment of anemia.


Subject(s)
Anemia/genetics , Erythropoiesis/genetics , Erythropoietin/genetics , GATA1 Transcription Factor/genetics , Receptors, Erythropoietin/genetics , Amino Acid Sequence/genetics , Anemia/drug therapy , Animals , Erythropoiesis/drug effects , Erythropoietin/biosynthesis , Gene Expression Regulation, Developmental/drug effects , Mice , Mice, Transgenic , Protein Multimerization/drug effects , Protein Structure, Tertiary , Receptors, Erythropoietin/biosynthesis , Signal Transduction/drug effects , Signal Transduction/genetics , Tacrolimus/administration & dosage , Tacrolimus/analogs & derivatives
4.
Gan To Kagaku Ryoho ; 41(7): 811-6, 2014 Jul.
Article in Japanese | MEDLINE | ID: mdl-25131865

ABSTRACT

Abiraterone acetate(AA)has been approved in more than 80 countries for the treatment of patients with metastatic castration-resistant prostate cancer(mCRPC). In July 2013, a marketing approval application for AA was submitted to the Japanese Ministry of Health, Labour, and Welfare. AA is a selective inhibitor of CYP17A1, a crucial enzyme for androgen biosynthesis. AA exerts its anti-tumor activity by directly inhibiting androgen production at all three sources, i. e., the testes, adrenal glands, and tumor itself. Data from international phase III studies and phase I and II studies in Japan have indicated that AA improves the overall survival and quality of life(QoL)of patients with mCRPC. Herein, we have summarized the development of AA and the results of important international and local clinical trials in Japan. In addition, the effect of food on AA bioavailability, concomitant steroid use, and liver function test abnormalities have been discussed regarding the appropriate use of AA.


Subject(s)
Androstadienes/therapeutic use , Androstenols/therapeutic use , Clinical Trials as Topic , Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Abiraterone Acetate , Androstadienes/adverse effects , Androstenes , Androstenols/adverse effects , Chemical and Drug Induced Liver Injury , Enzyme Inhibitors/adverse effects , Humans , Male , Prostatic Neoplasms, Castration-Resistant/enzymology
5.
Int J Hematol ; 99(6): 726-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24706190

ABSTRACT

Several clinical trials have demonstrated the effectiveness of bortezomib in combination with various anti-myeloma agents; however, no definitive information is available regarding drugs best suited for use in combination with bortezomib. Using isobologram analysis, we investigated the combined effects of bortezomib with four key anti-myeloma drugs (melphalan, cyclophosphamide, doxorubicin and lenalidomide), which represent components of major bortezomib-based regimens with corticosteroids, in three myeloma cell lines (U266, RPMI8226 and KMS-12BM) under various conditions. Melphalan showed the best performance with bortezomib under all culture conditions tested (liquid culture, on fibronectin-coated plates, and co-culture with bone marrow stromal cells), whereas cyclophosphamide was antagonistic with bortezomib especially in the presence of stromal cells. Doxorubicin showed additive effects under stroma-free conditions and in contact with fibronectin, but was rather antagonistic in the presence of stromal cells. In contrast, lenalidomide exerted the most favorable effect with bortezomib in contact with stromal cells. Consistent with these results, caspase-3 was activated more strongly by melphalan than by other agents in combination with bortezomib. Moreover, bortezomib-induced up-regulation of CHOP was readily enhanced by lenalidomide in contact with stromal cells. The present findings may provide fundamental information for the selection of bortezomib-based regimens for myeloma patients.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Fibronectins/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Pyrazines/pharmacology , Bone Marrow/metabolism , Bone Marrow/pathology , Bortezomib , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Drug Combinations , Drug Resistance, Neoplasm , Drug Synergism , Humans , Tumor Microenvironment
6.
Cancer Sci ; 105(2): 195-201, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24329927

ABSTRACT

Because of the potentially high mortality rate (6.5%) associated with bortezomib-induced lung disease (BILD) in Japanese patients with relapsed or refractory multiple myeloma, we evaluated the incidence, mortality and clinical features of BILD in a Japanese population. This study was conducted under the Risk Minimization Action Plan (RMAP), which was collaboratively developed by the pharmaceutical industry and public health authority. The RMAP consisted of an intensive dissemination of risk information and a recommended countermeasure to health-care professionals. All patients treated with bortezomib were consecutively registered in the study within 1 year and monitored for emerging BILD. Of the 1010 patients registered, 45 (4.5%) developed BILD, 5 (0.50%) of whom had fatal cases. The median time to BILD onset from the first bortezomib dose was 14.5 days, and most of the patients responded well to corticosteroid therapy. A retrospective review by the Lung Injury Medical Expert Panel revealed that the types with capillary leak syndrome and hypoxia without infiltrative shadows were uniquely and frequently observed in patients with BILD compared with those with conditions associated with other molecular-targeted anticancer drugs. The incidence rate of BILD in Japan remains high compared with that reported in other countries, but the incidence and mortality rates are lower than expected before the introduction of bortezomib in Japan. This study describes the radiographic pattern and clinical characterization of BILD in the Japanese population. The RMAP seemed clinically effective in minimizing the BILD risk among our Japanese population.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Boronic Acids/adverse effects , Boronic Acids/therapeutic use , Lung Diseases/chemically induced , Multiple Myeloma/drug therapy , Pyrazines/adverse effects , Pyrazines/therapeutic use , Adult , Aged , Aged, 80 and over , Bortezomib , Female , Humans , Incidence , Japan/epidemiology , Lung Diseases/mortality , Lung Diseases/pathology , Male , Middle Aged , Retrospective Studies , Risk
7.
Genes Cells ; 18(10): 886-98, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23890289

ABSTRACT

GATA1 is a transcription factor essential for erythropoiesis and megakaryopoiesis. It has been found that Gata1 gene knockdown heterozygous female (Gata1(G1.05/+)) mice spontaneously develop erythroblastic leukemias. In this study, we have generated a novel Gata1 knockdown erythroblastic cell line, designated GAK14, from the leukemia cells in the Gata1(G1.05/+) mice. Although GAK14 cells maintain immature phenotype on OP9 stromal cells in the presence of erythropoietin and stem cell factor, the cells produce Gr-1-, Mac1-, B220-, CD3e- or CD49b-positive hematopoietic cells when co-cultured with DAS104-8 feeder cells. However, GAK14 cells did not produce erythroid and megakaryocytic lineages, perhaps due to the absence of GATA1. Indeed, GAK14 cells became capable of differentiating into mature erythroid cells when complemented with full-length GATA1 and co-cultured with fetal liver-derived FLS5 stromal cells. This differentiation potential was impaired when GATA1 lacking the N-terminal domain was complemented. The N-terminal domain is known to contribute to the pathogenesis of transient abnormal myelopoiesis and acute megakaryoblastic leukemia related to Down syndrome. These results thus showed that GAK14 cells will serve as a powerful tool for dissecting domain function of GATA1 and that the GATA1 N-terminal domain is essential for the erythroid differentiation of GAK14 cells.


Subject(s)
Amino Acids/genetics , Cell Line, Tumor , Erythroid Precursor Cells/physiology , Erythropoiesis , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/metabolism , Leukemia, Erythroblastic, Acute , Animals , Cell Lineage , Coculture Techniques , Erythroid Precursor Cells/metabolism , Female , GATA1 Transcription Factor/genetics , Gene Expression , Gene Knockdown Techniques , Megakaryocyte Progenitor Cells/physiology , Mice , Myelopoiesis , Protein Structure, Tertiary
8.
Cancer Sci ; 104(7): 912-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23574271

ABSTRACT

This phase I/II study was conducted to evaluate the safety and efficacy of bortezomib-melphalan-prednisolone in Japanese patients with previously untreated multiple myeloma who are ineligible for hematopoietic stem cell transplantation. One hundred and one patients were enrolled, and 99 patients received up to nine 6-week cycles of bortezomib (0.7/1.0/1.3 mg/m²) on days 1, 4, 8, 11, 22, 25, 29, and 32 in cycles 1-4 and on days 1, 8, 22, and 29 in cycles 5-9, with melphalan (9 mg/m²) and prednisolone (60 mg/m²) on days 1-4 of each cycle. The recommended dose was determined in the phase I portion, and the overall response rate and safety of bortezomib-melphalan-prednisolone at the recommended dose were assessed in the phase II portion. The recommended dose of bortezomib was determined to be 1.3 mg/m². Grade 3 or higher non-hematological adverse events included diarrhea (12%) and peripheral neuropathy (10%); grade 4 hematological adverse events included lymphopenia (41%), neutropenia (30%), and thrombocytopenia (22%). Eleven patients had lung injury associated with bortezomib; two had grade 3 disease, and the other nine had grade 1 or 2 disease. Of the 86 patients treated with 1.3-mg/m² bortezomib in phases I and II, the median number of treatment cycles was 4.5, and the overall response rate was 70% (95% confidence interval: 59-79%). Bortezomib-melphalan-prednisolone with 1.3-mg/m² bortezomib was considered to be tolerable and effective in Japanese patients with previously untreated multiple myeloma. However, further investigation is needed to refine the administration schedule.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , Combined Modality Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/surgery , Prednisolone/administration & dosage , Prednisolone/adverse effects , Pyrazines/administration & dosage , Pyrazines/adverse effects , Treatment Outcome
9.
Mol Cell Biol ; 33(8): 1687-95, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23428869

ABSTRACT

The human ß-globin locus is comprised of embryonic, fetal, and adult globin genes, each of which is expressed at distinct stages of pre- and postnatal development. Functional defects in globin proteins or expression results in mild to severe anemia, such as in sickle-cell disease or ß-thalassemia, but the clinical symptoms of both disorders are ameliorated by persistent expression of the fetal globin genes. Recent genome-wide association studies (GWAS) identified the intergenic region between the HBS1L and MYB loci as a candidate modifier of fetal hemoglobin expression in adults. However, it remains to be clarified whether the enhancer activity within the HBS1L-MYB regulatory domain contributes to the production of fetal hemoglobin in adults. Here we report a new mouse model of hereditary persistence of fetal hemoglobin (HPFH) in which a transgene was randomly inserted into the orthologous murine Hbs1l-Myb locus. This mutant mouse exhibited typically elevated expression of embryonic globins and hematopoietic parameters similar to those observed in human HPFH. These results support the contention that mutation of the HBS1L-MYB genomic domain is responsible for elevated expression of the fetal globin genes, and this model serves as an important means for the analysis of networks that regulate fetal globin gene expression.


Subject(s)
Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Proto-Oncogene Proteins c-myb/genetics , beta-Globins/genetics , Animals , Cell Line , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , Mice, Transgenic , RNA Interference , RNA, Small Interfering
10.
Cancer Sci ; 103(10): 1839-47, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22816487

ABSTRACT

The management of myelodysplastic syndrome (MDS) remains challenging. We performed a phase I/II study to evaluate the safety and efficacy of decitabine in patients with MDS in Japan. Patients with MDS with red cell transfusion dependence or 5-30% blasts in marrow and with an International Prognostic Scoring System score of intermediate-1 or higher were eligible. Patients received intravenous decitabine at 15 or 20 mg/m(2) daily for 5 days every 4 weeks. A total of 37 patients were enrolled. Three patients received 15 mg/m(2) and experienced no dose limiting toxicity during the first cycle. Thirty-four patients received 20 mg/m(2) . Grade 3 or greater non-hematologic toxicities included cerebral infarction (n = 1), subdural hematoma (n = 1), elevated blood glucose (n = 1), and pulmonary hypertension (n = 1). At 20 mg/m(2) , complete response, partial response, and hematologic improvement were observed in 7 (20.6%), 2 (5.9%), and 7 (20.6%) patients, respectively. Complete cytogenetic response was observed in 30% of evaluable 20 patients. The median number of cycles to clinical response was 4 (range 4-8), and duration of remission was 474+ days (range 294-598+). The 2-year rate of acute myeloid leukemia-free survival was 52%. Correlative studies revealed hypomethylation in multiple genes in peripheral blood cells after treatment. Hypomethylation was generally more profound in CD15 + peripheral blood cells, which reflects myeloid cells, than in peripheral blood mononuclear cells. In summary, decitabine was safe and demonstrated efficacy in Japanese patients with high-risk MDS. This trial was registered at ClinicalTrials.gov (NCT00796003).


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Azacitidine/analogs & derivatives , Myelodysplastic Syndromes/drug therapy , Aged , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/pharmacokinetics , Decitabine , Dose-Response Relationship, Drug , Female , Humans , Japan , Male , Middle Aged , Treatment Outcome
11.
Int J Hematol ; 93(5): 618-623, 2011 May.
Article in English | MEDLINE | ID: mdl-21523339

ABSTRACT

The correlation between imatinib (IM) trough plasma concentration (Cmin) and clinical response was assessed in patients with chronic-phase chronic myeloid leukemia. The Cmin correlated with neither the achievement of complete cytogenetic response (977 vs. 993 ng/ml, P = 0.48) nor a major molecular response (1,044 vs. 818 ng/ml, P = 0.17). Although this was significantly higher in patients with complete molecular response (CMR) than in those without (1,430 vs. 859 ng/ml, P = 0.04), the difference was not significant in the sub-population treated with a standard dose of IM (400 mg/day). Finally, multivariate analysis showed that the IM standard dose, but not Cmin, was predictive of the achievement of CMR. We thus suggest that, in practical clinics at least, adherence to the standard dose is the most important factor for the achievement of CMR.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myeloid, Chronic-Phase/blood , Leukemia, Myeloid, Chronic-Phase/drug therapy , Piperazines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacokinetics , Aged , Benzamides , Cross-Sectional Studies , Cytogenetic Analysis , Dose-Response Relationship, Drug , Drug Dosage Calculations , Female , Fusion Proteins, bcr-abl/blood , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Japan , Leukemia, Myeloid, Chronic-Phase/pathology , Male , Middle Aged , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Protein-Tyrosine Kinases/blood , Protein-Tyrosine Kinases/genetics , Pyrimidines/administration & dosage , Remission Induction/methods , Retrospective Studies , Treatment Outcome
12.
Nat Commun ; 2: 181, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21304510

ABSTRACT

Post-translational protein modifications are systems designed to expand restricted genomic information through functional conversion of target molecules. Ubiquitin-like post-translational modifiers regulate numerous cellular events through their covalent linkages to target protein(s) by an enzymatic cascade analogous to ubiquitylation consisting of E1 (activating), E2 (conjugating) and E3 (ligating) enzymes. In this study, we report the essential role of Uba5, a specific activating enzyme for the ubiquitin-like modifier, Ufm1, in erythroid development. Mice lacking Uba5 exhibited severe anaemia, followed by death in utero. Although Uba5 was dispensable for the production of erythropoietin, its genetic loss led to impaired development of megakaryocyte and erythroid progenitors from common myeloid progenitors. Intriguingly, transgenic expression of Uba5 in the erythroid lineage rescued the Uba5-deficient embryos from anaemia and prolonged their survival, demonstrating the importance of Uba5 in cell-autonomous erythroid differentiation. Our results suggest that one of the ubiquitin-like protein modification systems, the Ufm1 system, is involved in the regulation of haematopoiesis.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells , Hematopoiesis/physiology , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Animals , Cell Line , Colony-Forming Units Assay , DNA Primers/genetics , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Activating Enzymes/genetics
13.
Rinsho Ketsueki ; 52(12): 1859-69, 2011 Dec.
Article in Japanese | MEDLINE | ID: mdl-22241153

ABSTRACT

Bortezomib (Velcade(®)), a proteasome inhibitor, was launched for the treatment of relapsed or refractory multiple myeloma in Japan in December 2006. Prior to approval in Japan, high incidence (15.2%) and mortality (6.5%) of bortezomib therapy-related lung disorders were reported with private import treatment. Therefore the Velcade Lung Disorder Panel (the Panel) was established and the cases have been reviewed. A total of 3,556 patients, including 823 post-marketing surveillance (PMS) patients, have received bortezomib since April 25, 2009. The incidence of lung disorders associated with bortezomib therapy was 2.33% (3.77% in case of PMS). The panel reviewed the detailed information of 70 cases and classified the CT and X-ray images as follows: (1)Interstitial pneumonia; diffuse alveolar damage pattern, hypersensitivity pneumonia pattern and others (2)Vascular hyperpermiability; (non-cardiogenic) pulmonary edema pattern and capillary leak syndrome-like pattern (3)Hypoxia. These post-marketing data showed that the incidence of lung disorders in Japan was lower than expected based on private import data.


Subject(s)
Antineoplastic Agents/adverse effects , Boronic Acids/adverse effects , Lung Diseases/chemically induced , Pyrazines/adverse effects , Adult , Aged , Aged, 80 and over , Bortezomib , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Product Surveillance, Postmarketing
14.
Rinsho Ketsueki ; 51(2): 127-31, 2010 Feb.
Article in Japanese | MEDLINE | ID: mdl-20379104

ABSTRACT

We report a patient with refractory idiopathic thrombotic thrombocytopenic purpura (TTP) who was successfully treated with rituximab. A 50-year-old woman was referred to our hospital with progressive psychoneurotic symptoms, hemolytic anemia and thrombocytopenia. The diagnosis of TTP was confirmed by the absence of ADAMTS13 activity with the presence of circulating ADAMTS13 inhibitor. High-dose steroid therapy and plasma exchange were performed. Despite 21 sessions of plasma exchange, however, there was no remarkable improvement. We then administered rituximab. Fifteen days after the first infusion of rituximab, she achieved complete remission and ADAMTS13 activity increased up to 14%. The patient has remained in remission for more than 9 months.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Plasma Exchange , Purpura, Thrombotic Thrombocytopenic/therapy , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/blood , ADAMTS13 Protein , Antibodies, Monoclonal, Murine-Derived , Biomarkers/blood , Drug Administration Schedule , Female , Humans , Middle Aged , Purpura, Thrombotic Thrombocytopenic/diagnosis , Remission Induction , Rituximab , Treatment Outcome
15.
BMJ Case Rep ; 20092009.
Article in English | MEDLINE | ID: mdl-21686374

ABSTRACT

Intravascular large B cell lymphoma (IVLBCL) presents various neurological symptoms, and the prognosis frequently deteriorates with a delay in diagnosis. In addition, for the diagnosis of IVLBCL, invasive biopsies are generally performed in main organs, such as the brain. We report a case of IVLBCL in which an early diagnosis was enabled by skin biopsy. The patient in this case had cauda equine syndrome and had developed multiple brain infarctions. She received six cycles of R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone) treatment and is currently in complete remission. At the macroscopic level, her lesions resembled senile angioma, commonly observed in normal elderly persons. Eruptions of this type are not currently recognised as IVLBCL lesions and might easily be overlooked. In cases in which IVLBCL could be suspected, an active search and biopsy of skin lesions, including an eruption of this type, are useful for early diagnosis and treatment.

16.
Blood ; 111(8): 4375-85, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18258797

ABSTRACT

The transcription factor Gata1 is expressed in several hematopoietic lineages and plays essential roles in normal hematopoietic development during embryonic stages. The lethality of Gata1-null embryos has precluded determination of its role in adult erythropoiesis. Here we have examined the effects of Gata1 loss in adult erythropoiesis using conditional Gata1 knockout mice expressing either interferon- or tamoxifen-inducible Cre recombinase (Mx-Cre and Tx-Cre, respectively). Mx-Cre-mediated Gata1 recombination, although incomplete, resulted in maturation arrest of Gata1-null erythroid cells at the proerythroblast stage, thrombocytopenia, and excessive proliferation of megakaryocytes in the spleen. Tx-Cre-mediated Gata1 recombination resulted in depletion of the erythroid compartment in bone marrow and spleen. Formation of the early and late erythroid progenitors in bone marrow was significantly reduced in the absence of Gata1. Furthermore, on treatment with a hemolytic agent, these mice failed to activate a stress erythropoietic response, despite the rising erythropoietin levels. These results indicate that, in addition to the requirement of Gata1 in adult megakaryopoiesis, Gata1 is necessary for steady-state erythropoiesis and for erythroid expansion in response to anemia. Thus, ablation of Gata1 in adult mice results in a condition resembling aplastic crisis in human.


Subject(s)
Anemia, Aplastic/genetics , Erythropoiesis , GATA1 Transcription Factor/deficiency , Gene Deletion , Animals , Animals, Newborn , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Count , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Erythroid Cells/drug effects , Erythroid Cells/pathology , Erythropoiesis/drug effects , Integrases/metabolism , Interferons/pharmacology , Megakaryocytes/drug effects , Megakaryocytes/pathology , Mice , Models, Biological , Spleen/drug effects , Spleen/pathology , Tamoxifen/pharmacology , Thrombocytopenia/pathology
17.
Eur J Haematol ; 80(5): 444-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18221386

ABSTRACT

Acute promyelocytic leukemia (APL) is characterized by chromosomal rearrangements of 17q21, leading to fusion of the gene-encoding retinoic acid receptor alpha (RARA) with a number of alternative partner genes. Signal transducer and activator of transcription 5 beta (STAT5B) is one of the alternative partners. We report a rare case of APL with STAT5B-RARA fusion transcript and the normal chromosome 17 on G-banding. Administration of all trans-retinoic acid improved disseminated intravascular coagulation without decrease of the leukemia cells in his peripheral blood and bone marrow. The molecular mechanism of fusion between STAT5B and RARA by chromosomal rearrangement is discussed based on the data from genome database. Clinical characteristics of APL with STAT5B-RARA are also discussed.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/genetics , STAT5 Transcription Factor/analysis , STAT5 Transcription Factor/genetics , Transcription, Genetic/genetics , Adult , Aged , Amino Acid Sequence , Base Sequence , Humans , Karyotyping , Male , Middle Aged , Molecular Sequence Data , Oncogene Proteins, Fusion/chemistry , STAT5 Transcription Factor/chemistry
19.
Mol Cell Biol ; 26(21): 7953-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16940183

ABSTRACT

The nuclear proto-oncogene c-myb plays crucial roles in the growth, survival, and differentiation of hematopoietic cells. We established three lines of erythropoietin receptor-transgenic mice and found that one of them exhibited anemia, thrombocythemia, and splenomegaly. These abnormalities were independent of the function of the transgenic erythropoietin receptor and were observed exclusively in mice harboring the transgene homozygously, suggesting transgenic disruption of a certain gene. The transgene was inserted 77 kb upstream of the c-myb gene, and c-Myb expression was markedly decreased in megakaryocyte/erythrocyte lineage-restricted progenitors (MEPs) of the homozygous mutant mice. In the bone marrows and spleens of the mutant mice, numbers of megakaryocytes were increased and numbers of erythroid progenitors were decreased. These abnormalities were reproducible in vitro in a coculture assay of MEPs with OP9 cells but eliminated by the retroviral expression of c-Myb in MEPs. The erythroid/megakaryocytic abnormalities were reconstituted in mice in vivo by transplantation of mutant mouse bone marrow cells. These results demonstrate that the transgene insertion into the c-myb gene far upstream regulatory region affects the gene expression at the stage of MEPs, leading to an imbalance between erythroid and megakaryocytic cells, and suggest that c-Myb is an essential regulator of the erythroid-megakaryocytic lineage bifurcation.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Erythroid Cells/metabolism , Genes, myb , Megakaryocytes/physiology , Regulatory Sequences, Nucleic Acid , Transgenes , Animals , Blood Platelets/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Erythroid Cells/chemistry , Genetic Complementation Test , Hematopoiesis/physiology , Megakaryocytes/cytology , Mice , Mice, Transgenic , Mutagenesis, Insertional , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Retroviridae/genetics , Retroviridae/metabolism , Spleen/cytology , Spleen/pathology , Spleen/physiology , Stem Cells/cytology , Stem Cells/metabolism
20.
Int J Hematol ; 83(3): 247-51, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16720556

ABSTRACT

Results of recent studies with animal models suggest that expression of MLL fusion proteins promotes acute leukemogenesis. However, the most potent MLL fusion proteins are not sufficient for the development of acute myeloid leukemia (AML). The clinical data on the pathogenesis of this type of leukemia are limited. We analyzed the case of a patient with therapy-related AML with MLL rearrangement. The patient initially developed AML with t(8;21). Although the patient achieved complete remission with chemotherapy, an abnormal karyotype, inv(11)(q21q23), was detected. After 6-year persistence of a clone with the inversion 11 karyotype in the bone marrow, secondary AML developed. Results of fluorescence in situ hybridization analysis combined with magnet-activated cell sorting analysis showed that MLL rearrangement was detected in CD34+ and CD13+ fractions but not in a CD3+ fraction of the bone marrow. There were 2 important clinical findings. One was that MLL rearrangement was not sufficient for the development of leukemia. The other was that MLL rearrangement targets specific lineages.


Subject(s)
Chromosome Inversion/genetics , Chromosomes, Human, Pair 11/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasms, Second Primary/genetics , Antigens, CD34/biosynthesis , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD13 Antigens/biosynthesis , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology
SELECTION OF CITATIONS
SEARCH DETAIL