Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Clin Oncol ; 27(26): 4392-7, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19652076

ABSTRACT

PURPOSE: To determine the efficacy and safety of clofarabine in pediatric patients with refractory or relapsed acute myeloid leukemia (AML). PATIENTS AND METHODS: A phase II, open-label, multicenter study was conducted with single-agent clofarabine in pediatric patients with refractory or relapsed AML. Clofarabine was administered intravenously over 2 hours at the pediatric maximum-tolerated dose (MTD) of 52 mg/m(2) daily for 5 consecutive days. Cycles were repeated every 2 to 6 weeks. Responses determined by an independent response review panel. RESULTS: The 42 patients treated on the study had a median age of 13 years (range, 2 to 22 years) and had received a median number of two (range, one to five) prior regimens. The response rate was 26% and included one complete response without platelet recovery and 10 partial responses. The median duration of response was 20 weeks (range, 2 to >or= 156 weeks). Six of 28 patients who were refractory to the immediately preceding therapy achieved response. Thirteen patients (31%), including seven responders, proceeded to hematopoietic stem-cell transplantation (HSCT) after treatment with clofarabine and survived between 24 to >or= 160 weeks. Five patients (12%) remain alive post-transplantation at >or= 63, >or= 71, >or= 86, >or= 114, and >or= 130 weeks. The most common grade 3 or greater adverse events without regard to causality were febrile neutropenia, catheter-related infection, epistaxis, hypotension, nausea, and fever. Transient elevation of liver enzymes and hypokalemia occurred frequently. Five patients died within 30 days of clofarabine administration secondary to progressive disease, and another five died as a result of an adverse event. CONCLUSION: Clofarabine is active in pediatric patients with multiply relapsed or refractory AML. Responses allowed several refractory patients to proceed to HSCT. The toxicity profile was expected in this patient population.


Subject(s)
Adenine Nucleotides/therapeutic use , Arabinonucleosides/therapeutic use , Leukemia, Myeloid/drug therapy , Acute Disease , Adenine Nucleotides/adverse effects , Adolescent , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Arabinonucleosides/adverse effects , Child , Child, Preschool , Clofarabine , Drug Resistance, Neoplasm , Female , Fever/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukemia, Myeloid/surgery , Male , Nausea/etiology , Neutropenia/etiology , Recurrence , Remission Induction , Treatment Outcome , Young Adult
3.
J Clin Oncol ; 26(16): 2767-78, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18509186

ABSTRACT

PURPOSE: Tumor lysis syndrome (TLS) has recently been subclassified into either laboratory TLS or clinical TLS, and a grading system has been established. Standardized guidelines, however, are needed to aid in the stratification of patients according to risk and to establish prophylaxis and treatment recommendations for patients at risk or with established TLS. METHODS: A panel of experts in pediatric and adult hematologic malignancies and TLS was assembled to develop recommendations and guidelines for TLS based on clinical evidence and standards of care. A review of relevant literature was also used. RESULTS: New guidelines are presented regarding the prevention and management of patients at risk of developing TLS. The best management of TLS is prevention. Prevention strategies include hydration and prophylactic rasburicase in high-risk patients, hydration plus allopurinol or rasburicase for intermediate-risk patients, and close monitoring for low-risk patients. Primary management of established TLS involves similar recommendations, with the addition of aggressive hydration and diuresis, plus allopurinol or rasburicase for hyperuricemia. Alkalinization is not recommended. Although guidelines for rasburicase use in adults are provided, this agent is currently only approved for use in pediatric patients in the United States. CONCLUSION: The potential severity of complications resulting from TLS requires measures for prevention in high-risk patients and prompts treatment in the event that symptoms arise. Recognition of risk factors, monitoring of at-risk patients, and appropriate interventions are the key to preventing or managing TLS. These guidelines should assist in the prevention of TLS and improve the management of patients with established TLS.


Subject(s)
Antineoplastic Agents/adverse effects , Evidence-Based Medicine/organization & administration , Renal Insufficiency/complications , Tumor Lysis Syndrome , Adult , Child, Preschool , Economics, Pharmaceutical , Evidence-Based Medicine/trends , Humans , Practice Guidelines as Topic , Renal Dialysis/economics , Renal Insufficiency/economics , Renal Insufficiency/therapy , Retrospective Studies , Risk Factors , Tumor Lysis Syndrome/etiology , Tumor Lysis Syndrome/prevention & control , Tumor Lysis Syndrome/therapy
4.
Am J Clin Pathol ; 129(5): 700-5, 2008 May.
Article in English | MEDLINE | ID: mdl-18426728

ABSTRACT

Normal precursor B cells or hematogones share morphologic and immunophenotypic similarities with lymphoblasts of precursor B-lymphoblastic leukemia. The numbers are often increased and difficult to distinguish in many patients following chemotherapy for precursor B-lymphoblastic leukemia. The purpose of this study was to establish a unique method for differentiating hematogones from lymphoblasts by evaluating the immunofluorescence pattern of nuclear terminal deoxynucleotidyl transferase (TdT) staining in 29 cases of TdT+ acute leukemia and 20 cases with increased numbers of hematogones. All 29 cases of TdT+ acute leukemia demonstrated a finely granular pattern of TdT immunofluorescence that was uniformly distributed in the nucleus, whereas all 20 cases with increased hematogones demonstrated a coarsely granular or speckled pattern of TdT immunofluorescence, which often intensely aligns the nuclear membrane. The nuclear pattern of immunofluorescence using antibodies to TdT is an effective method for distinguishing hematogones from leukemic blasts.


Subject(s)
B-Lymphocytes/metabolism , Cell Nucleus/metabolism , DNA Nucleotidylexotransferase/metabolism , Neoplastic Stem Cells/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Stem Cells/metabolism , Adolescent , Adult , Aged , B-Lymphocytes/pathology , Child , Child, Preschool , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Infant , Male , Middle Aged , Stem Cells/pathology
5.
Support Care Cancer ; 14(6): 528-32, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16775648

ABSTRACT

This review focuses on the value of several groups of agents for the prevention and treatment of mucositis. The review refers to alimentary mucositis as a generalized term that includes oral mucositis and gastrointestinal mucositis. This paper is part of the systematic review made by the mucositis study group which operates in the Multinational Association of Supportive Care in Cancer (MASCC)/International Society of Oral Oncology (ISOO). Several new guidelines are suggested in this review as an update to the primary systematic review that was published by the same group in 2004.


Subject(s)
Gastrointestinal Diseases/therapy , Mucositis/therapy , Neoplasms , Practice Guidelines as Topic , Stomatitis/therapy , Administration, Topical , Adult , Analgesics/therapeutic use , Anesthetics, Local/therapeutic use , Animals , Anti-Infective Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/adverse effects , Child , Clinical Trials as Topic , Curcumin/therapeutic use , Dietary Supplements/standards , Disease Models, Animal , Drug Evaluation , Evidence-Based Medicine , Gastrointestinal Diseases/etiology , Graft vs Host Disease/complications , Humans , Medical Oncology/methods , Mucositis/etiology , Neoplasms/complications , Neoplasms/therapy , Primary Prevention/methods , Radiotherapy/adverse effects , Stomatitis/etiology
6.
J Clin Oncol ; 24(19): 3150-6, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16717292

ABSTRACT

PURPOSE: To compare conventional sibling bone marrow transplantation (CBMT), BMT with alternative donor (ABMT), and chemotherapy (CT) for children with acute lymphoblastic leukemia (ALL) and an early first marrow relapse. PATIENTS AND METHODS: After informed consent, 214 patients with ALL and early marrow relapse began multiagent induction therapy. One hundred sixty-three patients with fewer than 25% marrow blasts and count recovery at the end of induction (second remission [CR2]) were allocated by donor availability. Fifty patients with sibling donors were allocated to CBMT. Seventy-two patients were randomly allocated between ABMT and CT while 41 patients refused allocation. RESULTS: Overall, 3-year event free survival from entry is 19% +/- 3%. Thirty-two of 50 CBMT patients (64%) and 19 of 37 ABMT patients (51%) underwent transplantation in CR2 with 3-year disease-free survival of 42% +/- 7% and 29% +/- 7%. The 3-year DFS is 29% +/- 7%, 21% +/- 7%, and 27% +/- 8% for patients allocated to CBMT, ABMT, and CT, respectively. Contrary to protocol, 12 of 35 patients allocated to CT underwent BMT in CR2. Of these, five patients died after BMT and 5 patients relapsed. CONCLUSION: More than one half of patients died, failed reinduction, or relapsed again before 3 months after CR2 (median time to BMT). Intent-to-treat pair-wise comparison of ABMT with CT, CT with CBMT, and CBMT with ABMT yields hazards of 1.2, 1.1, 0.8 with P values of .56, .80, and .36, respectively. Outcomes remain similar and poor for children with ALL and early marrow relapse. BMT is not a complete answer to the challenge of ALL and early marrow relapse.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Neoplasms/surgery , Bone Marrow Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Siblings , Survival Analysis , Transplantation, Homologous , Treatment Outcome
7.
J Clin Oncol ; 24(12): 1917-23, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16622268

ABSTRACT

PURPOSE: To evaluate the efficacy and safety of clofarabine, a novel deoxyadenosine analog, in pediatric patients with refractory or relapsed acute lymphoblastic leukemia (ALL). PATIENTS AND METHODS: In a phase II, open-label, multicenter study, 61 pediatric patients with refractory or relapsed ALL received clofarabine 52 mg/m2 intravenously over 2 hours daily for 5 days, every 2 to 6 weeks. The median age was 12 years (range, 1 to 20 years), and the median number of prior regimens was three (range, two to six regimens). RESULTS: The response rate was 30%, consisting of seven complete remissions (CR), five CRs without platelet recovery (CRp), and six partial remissions. Remissions were durable enough to allow patients to proceed to hematopoietic stem-cell transplantation (HSCT) after clofarabine. Median CR duration in patients who did not receive HSCT was 6 weeks, with four patients maintaining CR or CRp for 8 weeks or more (8+, 12, 37+, and 48 weeks) on clofarabine therapy alone. The most common adverse events of grade > or = 3 were febrile neutropenia, anorexia, hypotension, and nausea. CONCLUSION: Clofarabine is active as a single agent in pediatric patients with multiple relapsed or refractory ALL. The toxicity profile is as expected in this heavily pretreated patient population. Studies exploring rational combinations of clofarabine with other agents are ongoing in an effort to maximize clinical benefit.


Subject(s)
Arabinonucleosides/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adenine Nucleotides , Adolescent , Adult , Arabinonucleosides/administration & dosage , Arabinonucleosides/adverse effects , Child , Child, Preschool , Clofarabine , Female , Humans , Infant , Infusions, Intravenous , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Treatment Outcome
8.
Pediatr Blood Cancer ; 47(2): 141-6, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16425271

ABSTRACT

PURPOSE: Re-induction outcomes vary for children with acute lymphoblastic leukemia (ALL) and marrow relapse. We explored possible relationships among asparaginase (ASNase) activity levels, asparagine (ASN) depletion, anti-ASNase antibody titers, and response to re-induction therapy in children and adolescents with ALL and an 'early' first marrow relapse. PATIENTS AND METHODS: After appropriate informed consent, we enrolled children and adolescents 1-21 years old with ALL and first marrow relapse within 12 months of completion of primary therapy. Induction therapy included intramuscular pegylated ASNase on Days 2 and 16. We assessed ASNase activity, anti-ASNase antibody titers against native and pegylated (E. coli) ASNase, and amino acid levels of asparagine (ASN) and glutamine (GLN) on Days 0, 14, and 35 of re-induction. RESULTS: Ninety-three patients were at least partially assessable. Among 21 patients with M1 marrow status at Day 35, the median Day 14 ASN level was <1 microM. This is significantly lower than the median Day 14 ASN level of 4 microM in the group of patients with M3 marrow at Day 35. Neither Day 0 nor Day 35 antibody titers predicted ASNase enzymatic activity level on Day 14. Surprisingly, Day 14 ASNase activity did not predict serum ASN level on Day 14. However, Day 0 and Day 35 anti-native ASNase antibody titers, and Day 0 anti-PEG ASNase antibody titers correlated positively with Day 14 serum ASN levels as one might expect from neutralizing antibody. Day 35 anti-PEG ASNase antibody titers did not. CONCLUSIONS: Patients with greater ASN depletion were more likely to achieve second remission in the context of six-drug therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Asparaginase/pharmacology , Asparagine/drug effects , Polyethylene Glycols/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antibody Formation/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Asparaginase/immunology , Asparagine/metabolism , Child , Female , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Humans , Injections, Intramuscular , Male , Polyethylene Glycols/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Remission Induction , Statistics, Nonparametric
9.
J Clin Oncol ; 21(8): 1612-7, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12697887

ABSTRACT

PURPOSE: To investigate the effect of granulocyte colony-stimulating factor (G-CSF) on hematopoietic toxicities, supportive care requirements, time to complete intensive therapy, and event-free survival (EFS) and overall survival (OS) in children with high-risk acute lymphoblastic leukemia (HR-ALL). PATIENTS AND METHODS: A total of 287 children with HR-ALL were randomly assigned to intensive chemotherapy regimens (New York I [NY I] or NY II) as part of the Children's Cancer Group (CCG)-1901 protocol. The induction phases consisted of five drugs (vincristine, prednisone, l-asparaginase, daunorubicin, and cyclophosphamide). Initial consolidation comprised six-agent chemotherapy combined with 18 Gy of total-brain irradiation. Patients were randomly assigned to receive G-CSF (5 microg/kg/day) during either induction or initial consolidation. A crossover study analysis was done on the 259 patients who completed both phases of therapy. RESULTS: The mean time to neutrophil recovery (>/= 0.5 x 109/L) was reduced with G-CSF (16.7 v 19.1 days, P =.0003); however, patients who received G-CSF did not have significantly reduced episodes of febrile neutropenia (149 v 164, P =.41), positive blood cultures (57 v 61, P =.66), or serious infections (75 v 79, P =.62). Hospitalization (14.0 v 13.9 days, P =.87) and induction therapy completion times (NY I, 30.3 v 31.3 days, P =.11; NY II, 33.4 v 32.3 days, P =.40) were not significantly altered. There were no differences in 6-year EFS (P =.24) or OS (P =.54) between patients receiving or not receiving G-CSF on CCG-1901, NY I and NY II. CONCLUSION: Children with high-risk ALL do not appear to benefit from prophylactic G-CSF.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Neutropenia/prevention & control , Neutrophils/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Asparaginase/administration & dosage , Asparaginase/adverse effects , Child , Child, Preschool , Cross-Over Studies , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Daunorubicin/administration & dosage , Daunorubicin/adverse effects , Drug Administration Schedule , Female , Humans , Infant , Male , Neutropenia/chemically induced , Prednisone/administration & dosage , Prednisone/adverse effects , Remission Induction , Risk Assessment , Survival Analysis , Treatment Outcome , Vincristine/administration & dosage , Vincristine/adverse effects
11.
Clin Chim Acta ; 321(1-2): 49-53, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12031592

ABSTRACT

BACKGROUND: Mutations in the erythroid-specific 5-aminolevulinate-synthase gene (ALAS2) have been identified in many cases of X-linked sideroblastic anemia (XLSA). METHODS: A polymerase chain reaction-mediated restriction fragment length polymorphism (RFLP) assay was used. RESULTS: A G527T point mutation was identified. This resulted in a substitution of tyrosine for asparagine at residue 159 (D159Y). This mutation was also identified in the mother of the two probands. Mutations in all three individuals were confirmed by DNA sequencing analysis. CONCLUSIONS: We identified a missense mutation in exon 5 of the ALAS2 gene in two brothers of a consanguineous marriage, who were clinically pyridoxine-responsive.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/genetics , Exons/genetics , Genetic Linkage/genetics , Mutation, Missense/genetics , X Chromosome/genetics , Anemia, Sideroblastic/pathology , Base Sequence , Child, Preschool , Consanguinity , DNA Mutational Analysis , Erythrocytes/pathology , Female , Humans , Isoenzymes/genetics , Male , Molecular Sequence Data , Polymorphism, Restriction Fragment Length
SELECTION OF CITATIONS
SEARCH DETAIL
...