Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Blood Adv ; 6(2): 521-527, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34710216

ABSTRACT

Secondary myelodysplastic syndromes and acute myeloid leukemia (sMDS/AML) are rare in children and adolescents and have a dismal prognosis. The mainstay therapy is hematopoietic cell transplantation (HCT), but there has been no innovation in cytoreductive regimens. CP X-351, a fixed 5:1 molar ratio of liposomal cytarabine to daunorubicin, has shown favorable safety and efficacy in elderly individuals with secondary AML and children with relapsed de novo AML. We report the outcomes of 7 young patients (6 with newly diagnosed sMDS/AML and 1 with primary MDS/AML) uniformly treated with CP X-351. Five patients had previously received chemotherapy for osteosarcoma, Ewing sarcoma, neuroblastoma, or T-cell acute lymphoblastic leukemia; 1 had predisposing genomic instability disorder (Cornelia de Lange syndrome) and 1 had MDS-related AML and multiorgan failure. The median age at diagnosis of myeloid malignancy was 17 years (range, 13-23 years). Patients received 1 to 3 cycles of CP X-351 (cytarabine 100 mg/m2 plus daunorubicin 44 mg/m2) on days 1, 3, and 5, resulting in complete morphologic remission without overt toxicity or treatment-related mortality. This approach allowed for adding an FLT3 inhibitor as individualized therapy in 1 patient. Six patients were alive and leukemia-free at 0.5 to 3.3 years after HCT. One patient died as a result of disease progression before HCT. To summarize, CP X-351 is an effective and well-tolerated regimen for cytoreduction in pediatric sMDS/AML that warrants prospective studies.


Subject(s)
Leukemia, Myeloid, Acute , Neoplasms, Second Primary , Adolescent , Aged , Child , Cytarabine/therapeutic use , Daunorubicin/therapeutic use , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Neoplasms, Second Primary/drug therapy , Prospective Studies
3.
Pediatr Blood Cancer ; 60(7): E38-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23417921

ABSTRACT

Dendritic cell leukemia (DCL) or hematodermic tumor is an uncommon subtype of acute leukemia. In contrast to adult cases, children tend to have a less aggressive course. The diagnosis of DCL should be considered when its characteristic morphologic features are present and leukemic cells co-express CD4 and CD56. Cases of DCL among pediatric patients have been reported to respond to therapeutic regimens for acute lymphoblastic leukemia, but details regarding the specifics of therapy are lacking.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dendritic Cells/pathology , Leukemia/drug therapy , Leukemia/physiopathology , Child , Child, Preschool , Female , Humans , Male
4.
Drug Metab Dispos ; 41(4): 923-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23298861

ABSTRACT

The subarachnoid space, where cerebrospinal fluid (CSF) flows over the brain and spinal cord, is lined on one side by arachnoid barrier (AB) cells that form part of the blood-CSF barrier. However, despite the fact that drugs are administered into the CSF and CSF drug concentrations are used as a surrogate for brain drug concentration following systemic drug administration, the tight-junctioned AB cells have never been examined for whether they express drug transporters that would influence CSF and central nervous system drug disposition. Hence, we characterized drug transporter expression and function in AB cells. Immunohistochemical analysis showed P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in mouse AB cells but not other meningeal tissue. The Gene Expression Nervous System Atlas (GENSAT) database and the Allen Mouse Brain Atlas confirmed these observations. Microarray analysis of mouse and human arachnoidal tissue revealed expression of many drug transporters and some drug-metabolizing enzymes. Immortalized mouse AB cells express functional P-gp on the apical (dura-facing) membrane and BCRP on both apical and basal (CSF-facing) membranes. Thus, like blood-brain barrier cells and choroid plexus cells, AB cells highly express drug transport proteins and likely contribute to the blood-CSF drug permeation barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Arachnoid/cytology , Blood-Brain Barrier/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport/genetics , Brain/metabolism , Cell Line , Gene Expression , Haplorhini , Humans , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Spinal Cord/metabolism
5.
J Clin Oncol ; 30(29): 3625-32, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22965955

ABSTRACT

PURPOSE: In acute myeloid leukemia (AML), initial treatment response by morphologic analysis of bone marrow predicts long-term outcome. Response can now be assessed by minimal residual disease (MRD) monitoring with flow cytometry or polymerase chain reaction (PCR). We determined the relation among the results of these approaches and their prognostic value. PATIENTS AND METHODS: In the multicenter AML02 study, follow-up bone marrow samples from 203 children and adolescents with newly diagnosed AML were examined by flow cytometry (n = 1,514), morphology (n = 1,382), and PCR amplification of fusion transcripts (n = 508). Results were correlated with treatment outcome. RESULTS: Among 1,215 samples with less than 5% leukemic myeloblasts by morphology, 100 (8.2%) were MRD positive (≥ 0.1%) by flow cytometry, whereas 96 (57.5%) of the 167 samples with ≥ 5% blasts were MRD negative. Virtually all (308 of 311; 99.0%) MRD-negative samples by PCR were also MRD negative by flow cytometry. However, only 19 (9.6%) of the 197 PCR-positive samples were flow cytometry positive, with analyses of AML1-ETO and CBFß-MYH11 accounting for most discrepancies, whereas eight of 13 MLL-positive samples had detectable MRD by flow cytometry. MRD by flow cytometry after induction 1 or 2 predicted lower event-free survival and higher relapse rate (P < .001) and was an independent prognostic factor in a multivariable analysis; prediction was not improved by morphologic information or molecular findings. CONCLUSION: In childhood AML, morphologic assessment of treatment response has limited value if MRD is measured by flow cytometry. MLL fusion transcripts can provide prognostic information in some patients, whereas monitoring of AML1-ETO and CBFß-MYH11 transcripts is largely uninformative.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Neoplasm, Residual/diagnosis , Neoplasm, Residual/mortality , Adolescent , Age Factors , Analysis of Variance , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Cohort Studies , Disease-Free Survival , Female , Flow Cytometry/methods , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Male , Monte Carlo Method , Neoplasm, Residual/drug therapy , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Severity of Illness Index , Sex Factors , Survival Analysis
6.
Invest New Drugs ; 30(4): 1660-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21796439

ABSTRACT

BACKGROUND: Gefitinib potently inhibits neuroblastoma proliferation in vitro, and the gefitinib/irinotecan combination shows greater than additive activity against neuroblastoma xenografts. This Phase II pilot study estimated the rate of response to two courses of intravenous irinotecan plus oral gefitinib in children with untreated high-risk neuroblastoma. METHODS: Two courses of irinotecan [15 mg/m(2)/day (daily ×5)×2] were combined with 12 daily doses of gefitinib (112.5 mg/m(2)/day). Response was assessed after 6 weeks. A response rate >55% was sought. RESULTS: Of the 23 children enrolled, 19 were evaluable for response. Median age at diagnosis was 3.1 years (range, 18 days-12.7 years). Most patients were older than 24 months (n = 20; 87%), male (n = 18; 78%), white (n = 16; 70%), had INSS 4 disease (n = 19; 83%), and had adrenal primary tumors (n = 18; 78%); nine patients (39%) had amplified tumor MYCN. The toxicity of gefitinib/irinotecan was mild and reversible (nausea, 5/20; diarrhea, 8/20; vomiting, 7/20). Five patients had partial responses; 9 others had a 23%-60% decrease in primary tumor volume and/or improved MIBG scans or decreased bone or bone marrow tumor burden. Median (range) systemic irinotecan exposure (AUC) was 283 ng/ml*hr (range, 163-890 ng/ml*hr) and 28 ng/ml*hr (3.6-297 ng/ml*hr) for the active metabolite, SN-38. No relation was observed between response and tumor expression of EGFR, MRP2-4, ABCG2, and Pgp. CONCLUSIONS: Although the gefitinib/irinotecan combination was very tolerable and induced responses, it was not sufficiently active to warrant further investigation. Initial investigational studies of this type can preclude the necessity for larger, longer, and costlier trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Neuroblastoma/diagnosis , Neuroblastoma/drug therapy , Quinazolines/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Camptothecin/therapeutic use , Child , Child, Preschool , Female , Gefitinib , Homovanillic Acid/urine , Humans , Immunohistochemistry , Infant , Infant, Newborn , Irinotecan , L-Lactate Dehydrogenase/blood , Male , Neoplasm Proteins/metabolism , Neuroblastoma/blood , Neuroblastoma/urine , Pilot Projects , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Risk Factors , Treatment Outcome , Tumor Burden/drug effects , Vanilmandelic Acid/urine
7.
J Clin Oncol ; 29(24): 3293-300, 2011 Aug 20.
Article in English | MEDLINE | ID: mdl-21768474

ABSTRACT

PURPOSE: To assess the toxicity, pharmacokinetics, and pharmacodynamics of multikinase inhibitor sorafenib in combination with clofarabine and cytarabine in children with relapsed/refractory leukemia. PATIENTS AND METHODS: Twelve patients with acute leukemia (11 with acute myeloid leukemia [AML]) received sorafenib on days 1 to 7 and then concurrently with cytarabine (1 g/m(2)) and clofarabine (stratum one: 40 mg/m(2), n = 10; stratum two [recent transplantation or fungal infection]: 20 mg/m(2), n = 2) on days 8 to 12. Sorafenib was continued until day 28 if tolerated. Two sorafenib dose levels (200 mg/m(2) and 150 mg/m(2) twice daily) were planned. Sorafenib pharmacokinetic and pharmacodynamic studies were performed on days 7 and 8. RESULTS: At sorafenib 200 mg/m(2), two of four patients in stratum one and one of two patients in stratum two had grade 3 hand-foot skin reaction and/or rash as dose-limiting toxicities (DLTs). No DLTs were observed in six patients in stratum one at sorafenib 150 mg/m(2). Sorafenib inhibited the phosphorylation of AKT, S6 ribosomal protein, and 4E-BP1 in leukemia cells. The rate of sorafenib conversion to its metabolite sorafenib N-oxide was high (mean, 33%; range, 17% to 69%). In vitro, the N-oxide potently inhibited FLT3-internal tandem duplication (ITD; binding constant, 70 nmol/L) and the viability of five AML cell lines. On day 8, sorafenib decreased blast percentages in 10 of 12 patients (median, 66%; range, 9% to 95%). After combination chemotherapy, six patients (three FLT3-ITD and three FLT3 wild-type AML) achieved complete remission, two (both FLT3-ITD AML) had complete remission with incomplete blood count recovery, and one (FLT3 wild-type AML) had partial remission. CONCLUSION: Sorafenib in combination with clofarabine and cytarabine is tolerable and shows activity in relapsed/refractory pediatric AML.


Subject(s)
Adenine Nucleotides/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arabinonucleosides/administration & dosage , Benzenesulfonates/administration & dosage , Cytarabine/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Adenine Nucleotides/pharmacokinetics , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arabinonucleosides/pharmacokinetics , Child , Clofarabine , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Leukemia/drug therapy , Male , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Kinase Inhibitors/pharmacokinetics , Recurrence , Sorafenib
9.
J Clin Oncol ; 29(4): 386-91, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21172890

ABSTRACT

PURPOSE: The prognosis for older adolescents and young adults with acute lymphoblastic leukemia (ALL) has been historically much worse than that for younger patients. We reviewed the outcome of older adolescents (age 15 to 18 years) treated in four consecutive Total Therapy studies to determine if recent improved treatment extended to this high-risk group. PATIENTS AND METHODS: Between 1991 and 2007, 963 pediatric patients, including 89 older adolescents, were enrolled on Total Therapy studies XIIIA, XIIIB, XIV, and XV. In the first three studies, treatment selection was based on presenting clinical features and leukemic cell genetics. In study XV, the level of residual disease was used to guide treatment, which featured intensive methotrexate, glucocorticoid, vincristine, and asparaginase, as well as early triple intrathecal therapy for higher-risk ALL. RESULTS: The 89 older adolescents were significantly more likely to have T-cell ALL, the t(4;11)(MLL-AF4), and detectable minimal residual disease during or at the end of remission induction; they were less likely to have the t(12;21)(ETV6-RUNX1) compared with younger patients. In the first three studies, the 44 older adolescents had significantly poorer event-free survival and overall survival than the 403 younger patients. This gap in prognosis was abolished in study XV: event-free survival rates at 5 years were 86.4% ± 5.2% (standard error) for the 45 older adolescents and 87.4% ± 1.7% for the 453 younger patients; overall survival rates were 87.9% ± 5.1% versus 94.1% ± 1.2%, respectively. CONCLUSION: Most older adolescents with ALL can be cured with risk-adjusted intensive chemotherapy without stem-cell transplantation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Age Factors , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chi-Square Distribution , Child , Child, Preschool , Disease-Free Survival , Female , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cell Transplantation , Humans , Infant , Kaplan-Meier Estimate , Male , Neoplasm, Residual , Patient Selection , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Risk Assessment , Risk Factors , Survival Rate , Time Factors , Transplantation, Homologous , Treatment Outcome
10.
J AAPOS ; 14(5): 453-4, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20863726

ABSTRACT

Mixed-phenotype acute leukemia is a rare condition with no previously reported intraocular involvement. We present clinical, radiologic, and cytologic findings of leukemic intraocular relapse in a 23-month-old girl, with lineage switch presenting as conjunctivitis after allogeneic bone marrow transplantation. A diagnostic approach using fine needle aspiration is described.


Subject(s)
Hematopoietic Stem Cell Transplantation , Iris Neoplasms/genetics , Iris Neoplasms/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Acute Disease , Fatal Outcome , Female , Humans , Infant , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Phenotype , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Recurrence , Translocation, Genetic
11.
Lancet Oncol ; 11(6): 543-52, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20451454

ABSTRACT

BACKGROUND: We sought to improve outcome in patients with childhood acute myeloid leukaemia (AML) by applying risk-directed therapy that was based on genetic abnormalities of the leukaemic cells and measurements of minimal residual disease (MRD) done by flow cytometry during treatment. METHODS: From Oct 13, 2002, to June 19, 2008, 232 patients with de-novo AML (n=206), therapy-related or myelodysplasia-related AML (n=12), or mixed-lineage leukaemia (n=14) were enrolled at eight centres. 230 patients were assigned by block, non-blinded randomisation, stratified by cytogenetic or morphological subtype, to high-dose (18 g/m(2), n=113) or low-dose (2 g/m(2), n=117) cytarabine given with daunorubicin and etoposide (ADE; induction 1). The primary aim of the study was to compare the incidence of MRD positivity of the high-dose group and the low-dose group at day 22 of induction 1. Induction 2 consisted of ADE with or without gemtuzumab ozogamicin (GO anti-CD33 monoclonal antibody); consolidation therapy included three additional courses of chemotherapy or haematopoietic stem-cell transplantation (HSCT). Levels of MRD were used to allocate GO and to determine the timing of induction 2. Both MRD and genetic abnormalities at diagnosis were used to determine the final risk classification. Low-risk patients (n=68) received five courses of chemotherapy, whereas high-risk patients (n=79), and standard-risk patients (n=69) with matched sibling donors, were eligible for HSCT (done for 48 high-risk and eight standard-risk patients). All 230 randomised patients were analysed for the primary endpoint. Other analyses were limited to the 216 patients with AML, excluding those with mixed-lineage leukaemia. This trial is closed to accrual and is registered with ClinicalTrials.gov, number NCT00136084. FINDINGS: Complete remission was achieved in 80% (173 of 216 patients) after induction 1 and 94% (203 of 216) after induction 2. Induction failures included two deaths from toxic effects and ten cases of resistant leukaemia. The introduction of high-dose versus low-dose cytarabine did not significantly lower the rate of MRD-positivity after induction 1 (34%vs 42%, p=0.17). The 6-month cumulative incidence of grade 3 or higher infection was 79.3% (SE 4.0) for patients in the high-dose group and 75.5% (4.2) for the low-dose group. 3-year event-free survival and overall survival were 63.0% (SE 4.1) and 71.1% (3.8), respectively. 80% (155 of 193) of patients achieved MRD of less than 0.1% after induction 2, and the cumulative incidence of relapse for this group was 17% (SE 3). MRD of 1% or higher after induction 1 was the only significant independent adverse prognostic factor for both event-free (hazard ratio 2.41, 95% CI 1.36-4.26; p=0.003) and overall survival (2.11, 1.09-4.11; p=0.028). INTERPRETATION: Our findings suggest that the use of targeted chemotherapy and HSCT, in the context of a comprehensive risk-stratification strategy based on genetic features and MRD findings, can improve outcome in patients with childhood AML. FUNDING: National Institutes of Health and American Lebanese Syrian Associated Charities (ALSAC).


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Adolescent , Aminoglycosides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Cytarabine/administration & dosage , Cytogenetic Analysis , Daunorubicin/administration & dosage , Disease-Free Survival , Etoposide/administration & dosage , Female , Flow Cytometry , Gemtuzumab , Humans , Infant , Infant, Newborn , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Neoplasm, Residual , Remission Induction , Survival Rate , Young Adult
12.
Eur J Haematol ; 85(1): 51-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20374273

ABSTRACT

The rate and pattern of iron deposition and accumulation are important determinants of liver damage in chronically transfused patients. To investigate iron distribution patterns at various tissue iron concentrations, effects of chelation on hepatic iron compartmentalization, and differences between patients with sickle cell disease (SCD) and thalassemia major (TM), we prospectively investigated hepatic histologic and biochemical findings in 44 patients with iron overload (35 SCD and 9 TM). The median hepatic iron content (HIC) in patients with TM and SCD was similar at 12.9 and 10.3 mg Fe/g dry weight, respectively (P = 0.73), but patients with SCD had significantly less hepatic fibrosis and inflammation (P < 0.05), less hepatic injury, and significantly less blood exposure. Patients with SCD had predominantly sinusoidal iron deposition, but hepatocyte iron deposition was observed even at low HIC. Chelated patients had more hepatocyte and portal tract iron than non-chelated ones, but similar sinusoidal iron deposition. These data suggest that iron deposition in patients with SCD generally follows the traditional pattern of transfusional iron overload; however, parenchymal hepatocyte deposition also occurs early and chelation removes iron preferentially from the reticuloendothelium. Pathophysiological and genetic differences affecting iron deposition and accumulation in SCD and TM warrants further investigation.


Subject(s)
Anemia, Sickle Cell/complications , Anemia, Sickle Cell/metabolism , Iron Overload/etiology , Iron Overload/metabolism , Iron/metabolism , Liver/metabolism , beta-Thalassemia/complications , beta-Thalassemia/metabolism , Adolescent , Adult , Anemia, Sickle Cell/therapy , Biopsy , Chelation Therapy , Child , Female , Humans , Iron Chelating Agents/therapeutic use , Iron Overload/pathology , Liver/pathology , Magnetic Resonance Imaging , Male , Prospective Studies , Tissue Distribution , Transfusion Reaction , Young Adult , beta-Thalassemia/therapy
13.
Surg Pathol Clin ; 3(4): 1091-125, 2010 Dec.
Article in English | MEDLINE | ID: mdl-26839300

ABSTRACT

The evaluation of pediatric bone marrow poses specific challenges when compared with the general adult population. These challenges stem in part from the higher likelihood of congenital disorders with hematopoietic manifestations, some of which may give rise to hematologic malignancies. Familiarity with the spectrum of disorders seen in the pediatric age group allows for an appropriate and focused differential diagnosis. This review addresses the diagnostic workup of pediatric bone marrow samples, as directed by the peripheral blood and bone marrow findings in the context of the patient's clinical history. Recommendations for the appropriate use of ancillary studies in various scenarios are provided.

14.
Hematol Oncol Clin North Am ; 23(4): 655-74, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19577163

ABSTRACT

Acute lymphoblastic leukemia and lymphoblastic lymphoma constitute a family of genetically heterogeneous lymphoid neoplasms derived from B- and T-lymphoid progenitors. Diagnosis is based on morphologic, immunophenotypic, and genetic features that allow differentiation from normal progenitors and other hematopoietic and nonhematopoietic neoplasms. Current intensive chemotherapy regimens have accomplished overall cure rates of 85% to 90% in children and 40% to 50% in adults, with outcomes depending on the genetic subtype of disease and clinical features at presentation. Therapy is optimized using minimal residual disease studies that employ flow cytometric and molecular methodologies, and are important determinants of prognosis. Genetic analyses currently underway are likely to provide insight into biology, mechanisms of relapse, pharmacogenetics, and new potential therapeutic targets, which should aid in further improvement of outcome in this disease.


Subject(s)
Immunophenotyping/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Diagnosis, Differential , Flow Cytometry , Gene Rearrangement , Humans , Immunoglobulins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Antigen, T-Cell/genetics
15.
Blood ; 114(10): 2087-96, 2009 Sep 03.
Article in English | MEDLINE | ID: mdl-19542301

ABSTRACT

RNA-binding motif protein 15 (RBM15) is involved in the RBM15-megakaryoblastic leukemia 1 fusion in acute megakaryoblastic leukemia. Although Rbm15 has been reported to be required for B-cell differentiation and to inhibit myeloid and megakaryocytic expansion, it is not clear what the normal functions of Rbm15 are in the regulation of hematopoietic stem cell (HSC) and megakaryocyte development. In this study, we report that Rbm15 may function in part through regulation of expression of the proto-oncogene c-Myc. Similar to c-Myc knockout (c-Myc-KO) mice, long-term (LT) HSCs are significantly increased in Rbm15-KO mice due to an apparent LT-HSC to short-term HSC differentiation defect associated with abnormal HSC-niche interactions caused by increased N-cadherin and beta(1) integrin expression on mutant HSCs. Both serial transplantation and competitive reconstitution capabilities of Rbm15-KO LT-HSCs are greatly compromised. Rbm15-KO and c-Myc-KO mice also share related abnormalities in megakaryocyte development, with mutant progenitors producing increased, abnormally small low-ploidy megakaryocytes. Consistent with a possible functional interplay between Rbm15 and c-Myc, the megakaryocyte increase in Rbm15-KO mice could be partially reversed by ectopic c-Myc. Thus, Rbm15 appears to be required for normal HSC-niche interactions, for the ability of HSCs to contribute normally to adult hematopoiesis, and for normal megakaryocyte development; these effects of Rbm15 on hematopoiesis may be mediated at least in part by c-Myc.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/metabolism , Adult Stem Cells/cytology , Animals , Cell Communication/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Mice , Mice, Knockout , Ploidies , Proto-Oncogene Proteins c-myc/genetics , RNA-Binding Proteins/genetics , Transplantation, Homologous
16.
N Engl J Med ; 360(26): 2730-41, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19553647

ABSTRACT

BACKGROUND: Prophylactic cranial irradiation has been a standard treatment in children with acute lymphoblastic leukemia (ALL) who are at high risk for central nervous system (CNS) relapse. METHODS: We conducted a clinical trial to test whether prophylactic cranial irradiation could be omitted from treatment in all children with newly diagnosed ALL. A total of 498 patients who could be evaluated were enrolled. Treatment intensity was based on presenting features and the level of minimal residual disease after remission-induction treatment. The duration of continuous complete remission in the 71 patients who previously would have received prophylactic cranial irradiation was compared with that of 56 historical controls who received it. RESULTS: The 5-year event-free and overall survival probabilities for all 498 patients were 85.6% (95% confidence interval [CI], 79.9 to 91.3) and 93.5% (95% CI, 89.8 to 97.2), respectively. The 5-year cumulative risk of isolated CNS relapse was 2.7% (95% CI, 1.1 to 4.3), and that of any CNS relapse (including isolated relapse and combined relapse) was 3.9% (95% CI, 1.9 to 5.9). The 71 patients had significantly longer continuous complete remission than the 56 historical controls (P=0.04). All 11 patients with isolated CNS relapse remained in second remission for 0.4 to 5.5 years. CNS leukemia (CNS-3 status) or a traumatic lumbar puncture with blast cells at diagnosis and a high level of minimal residual disease (> or = 1%) after 6 weeks of remission induction were significantly associated with poorer event-free survival. Risk factors for CNS relapse included the genetic abnormality t(1;19)(TCF3-PBX1), any CNS involvement at diagnosis, and T-cell immunophenotype. Common adverse effects included allergic reactions to asparaginase, osteonecrosis, thrombosis, and disseminated fungal infection. CONCLUSIONS: With effective risk-adjusted chemotherapy, prophylactic cranial irradiation can be safely omitted from the treatment of childhood ALL. (ClinicalTrials.gov number, NCT00137111.)


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asparaginase/administration & dosage , Asparaginase/adverse effects , Central Nervous System Neoplasms/drug therapy , Child , Child, Preschool , Combined Modality Therapy , Cranial Irradiation , Cyclophosphamide/administration & dosage , Daunorubicin/administration & dosage , Dexamethasone/administration & dosage , Hematopoietic Stem Cell Transplantation , Humans , Infant , Mercaptopurine/administration & dosage , Methotrexate/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Remission Induction/methods , Risk Factors , Secondary Prevention , Survival Analysis , Treatment Outcome , Vincristine/administration & dosage
17.
Blood ; 113(20): 4853-5, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19264677

ABSTRACT

R2* magnetic resonance imaging (R2*-MRI) can quantify hepatic iron content (HIC) by noninvasive means but is not fully investigated. Patients with iron overload completed 1.5T R2*-MRI examination and liver biopsy within 30 days. Forty-three patients (sickle cell anemia, n = 32; beta-thalassemia major, n = 6; and bone marrow failure, n = 5) were analyzed: median age, 14 years, median transfusion duration, 15 months, average (+/-SD) serum ferritin 2718 plus or minus 1994 ng/mL, and average HIC 10.9 plus or minus 6.8 mg Fe/g dry weight liver. Regions of interest were drawn and analyzed by 3 independent reviewers with excellent agreement of their measurements (intraclass correlation coefficient = 0.98). Ferritin and R2*-MRI were weakly but significantly associated (range of correlation coefficients among the 3 reviewers, 0.41-0.48; all P < .01). R2*-MRI was strongly associated with HIC for all 3 reviewers (correlation coefficients, 0.96-0.98; all P < .001). This high correlation confirms prior reports, calibrates R2*-MRI measurements, and suggests its clinical utility for predicting HIC using R2*-MRI. This study was registered at www.clinicaltrials.gov as #NCT00675038.


Subject(s)
Iron Overload/diagnostic imaging , Liver/diagnostic imaging , Magnetic Resonance Imaging/methods , Adolescent , Adult , Biopsy , Calibration , Child , Female , Ferritins/analysis , Ferritins/blood , Humans , Iron Overload/pathology , Liver/pathology , Magnetic Resonance Imaging/standards , Male , Radiography , Single-Blind Method , Young Adult
18.
Lancet Oncol ; 10(2): 147-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19147408

ABSTRACT

BACKGROUND: About a fifth of children with acute T-lymphoblastic leukaemia (T-ALL) succumb to the disease, suggesting an unrecognised biological heterogeneity that might contribute to drug resistance. We postulated that T-ALL originating from early T-cell precursors (ETPs), a recently defined subset of thymocytes that retain stem-cell-like features, would respond poorly to lymphoid-cell-directed therapy. We studied leukaemic cells, collected at diagnosis, to identify cases with ETP features and determine their clinical outcome. METHODS: Leukaemic cells from 239 patients with T-ALL enrolled at St Jude Children's Research Hospital (n=139) and in the Italian national study Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) ALL-2000 (n=100) were assessed by gene-expression profiling, flow cytometry, and single nucleotide polymorphism array analysis. Probabilities of survival and treatment failure were calculated for subgroups considered to have ETP-ALL or typical T-ALL. FINDINGS: 30 patients (12.6%) had leukaemic lymphoblasts with an ETP-related gene-expression signature or its associated distinctive immunophenotype (CD1a(-), CD8(-), CD5(weak) with stem-cell or myeloid markers). Cases of ETP-ALL showed increased genomic instability, in terms of number and size of gene lesions, compared with those with typical T-ALL. Patients with this form of leukaemia had high risk of remission failure or haematological relapse (72% [95% CI 40-100] at 10 years vs 10% [4-16] at 10 years for patients with typical T-ALL treated at St Jude Children's Research Hospital; and 57% [25-89] at 2 years vs 14% [6-22] at 2 years for patients treated in the AIEOP trial). INTERPRETATION: ETP-ALL is a distinct, previously unrecognised, pathobiological entity that confers a poor prognosis with use of standard intensive chemotherapy. Its early recognition, by use of the gene expression and immunophenotypic criteria outlined here, is essential for the development of an effective clinical management strategy. FUNDING: US National Cancer Institute, Cariplo Foundation, Citta della Speranza Foundation, Italian Association for Cancer Research (AIRC), Italian Ministry for University and Research, and American Lebanese Syrian Associated Charities (ALSAC).


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Child , Child, Preschool , Female , Flow Cytometry , Humans , Immunophenotyping , Infant , Kaplan-Meier Estimate , Male , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Risk Factors
19.
Blood ; 113(21): 5083-9, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19131545

ABSTRACT

To characterize the biology and optimal therapy of acute mixed-lineage leukemia in children, we reviewed the pathologic and clinical features, including response to therapy, of 35 patients with mixed-lineage leukemia. The majority of cases (91%) had blasts cells that simultaneously expressed either T-lineage plus myeloid markers (T/myeloid, n = 20) or B-lineage plus myeloid markers (B/myeloid, n = 12). Overall survival rates for the B/myeloid and T/myeloid subgroups were not significantly different from each other or from the rate for acute myeloid leukemia (AML) but were inferior to the outcome in children with acute lymphoblastic leukemia (ALL). Patients who failed to achieve complete remission with AML-directed therapy could often be induced with a regimen of prednisone, vincristine, and L-asparaginase. Analysis of gene-expression patterns identified a subset of biphenotypic leukemias that did not cluster with T-cell ALL, B-progenitor ALL, or AML. We propose that treatment for biphenotypic leukemia begin with one course of AML-type induction therapy, with a provision for a switch to lymphoid-type induction therapy with a glucocorticoid, vincristine, and L-asparaginase if the patient responds poorly. We also suggest that hematopoietic stem cell transplantation is often not required for cure of these patients.


Subject(s)
Leukemia, Biphenotypic, Acute/pathology , Leukemia, Biphenotypic, Acute/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/pathology , Child , Gene Expression Profiling , Humans , Immunophenotyping , Leukemia, Biphenotypic, Acute/mortality , Leukemia, Myeloid, Acute , Myeloid Cells/pathology , Practice Guidelines as Topic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Retrospective Studies , Survival Rate , T-Lymphocytes/pathology , Treatment Outcome
20.
Am J Surg Pathol ; 32(9): 1421-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18670345

ABSTRACT

Primary cutaneous anaplastic large-cell lymphoma (ALCL) ordinarily is distinguished from systemic ALCL by clinical presentation, absence of anaplastic lymphoma kinase (ALK) expression, and immunophenotype (CLA+, EMA/MUC1-). We present an exceptional case of an elderly man with primary cutaneous ALCL and no systemic disease for a 13-year period. Recurrent skin tumors in this patient were characterized by anaplastic, often multinucleated, cells infiltrating the lymphatics and associated with pseudoepitheliomatous hyperplasia. Cutaneous lymphocyte antigen was absent and EMA/MUC1, typical of systemic ALCL, was strongly expressed by the tumor cells. Remarkably, the tumor cells expressed a cytoplasmic-only variant of ALK protein, as reported in 3 previous cases of primary cutaneous ALCL. Fluorescence in situ hybridization revealed lack of rearrangements of the chromosome 2 ALK gene locus usually involved by translocation t(2;5) or other chromosomal rearrangements that generate nucleophosmin-ALK or the variant ALK fusions that occur in systemic ALCL. Nonetheless, the cytoplasmic ALK protein in the patient's tumor cells was shown to be phosphorylated/activated, suggesting a novel mechanism of ALK activation. Primary cutaneous ALCL of this novel subtype should be distinguished from systemic ALCL to ensure proper clinical management.


Subject(s)
Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Large-Cell, Anaplastic/pathology , Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Age of Onset , Aged , Anaplastic Lymphoma Kinase , Antigens, Differentiation, T-Lymphocyte , Antigens, Neoplasm/biosynthesis , Enzyme Activation/physiology , Humans , In Situ Hybridization, Fluorescence , Lymphoma, Large-Cell, Anaplastic/genetics , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Mucin-1/biosynthesis , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Phenotype , Phosphorylation , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL