Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Ann Oncol ; 28(2): 386-392, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28426102

ABSTRACT

Background: Comprehensive studies on neutropenia and infection-related complications in patients with acute lymphoblastic leukemia (ALL) are lacking. Patients and methods: We evaluated infection-related complications that were grade ≥3 on National Cancer Institute's Common Terminology Criteria for Adverse Events (version 3.0) and their risk factors in 409 children with newly diagnosed ALL throughout the treatment period. Results: Of the 2420 infection episodes, febrile neutropenia and clinically or microbiologically documented infection were seen in 1107 and 1313 episodes, respectively. Among documented infection episodes, upper respiratory tract was the most common site (n = 389), followed by ear (n = 151), bloodstream (n = 147), and gastrointestinal tract (n = 145) infections. These episodes were more common during intensified therapy phases such as remission induction and reinduction, but respiratory and ear infections, presumably viral in origin, also occurred during continuation phases. The 3-year cumulative incidence of infection-related death was low (1.0±0.9%, n = 4), including 2 from Bacillus cereus bacteremia. There was no fungal infection-related mortality. Age 1-9.9 years at diagnosis was associated with febrile neutropenia (P = 0.002) during induction and febrile neutropenia and documented infection (both P < 0.001) during later continuation. White race was associated with documented infection (P = 0.034) during induction. Compared with low-risk patients, standard- and high-risk patients received more intensive therapy during early continuation and had higher incidences of febrile neutropenia (P < 0.001) and documented infections (P = 0.043). Furthermore, poor neutrophil surge after dexamethasone pulses during continuation, which can reflect the poor bone marrow reserve, was associated with infections (P < 0.001). Conclusions: The incidence of infection-related death was low. However, young age, white race, intensive chemotherapy, and lack of neutrophil surge after dexamethasone treatment were associated with infection-related complications. Close monitoring for prompt administration of antibiotics and modification of chemotherapy should be considered in these patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy-Induced Febrile Neutropenia/mortality , Chemotherapy-Induced Febrile Neutropenia/therapy , Child , Child, Preschool , Dexamethasone/administration & dosage , Female , Humans , Infant , Leukocyte Count , Male , Neutrophils/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Respiratory Tract Infections/chemically induced , Respiratory Tract Infections/mortality , Retrospective Studies , Risk Factors , Treatment Outcome , Vincristine/administration & dosage
2.
Leukemia ; 31(2): 333-339, 2017 02.
Article in English | MEDLINE | ID: mdl-27560110

ABSTRACT

To determine the clinical significance of minimal residual disease (MRD) in patients with prognostically relevant subtypes of childhood acute lymphoblastic leukemia (ALL), we analyzed data from 488 patients treated in St Jude Total Therapy Study XV with treatment intensity based mainly on MRD levels measured during remission induction. MRD levels on day 19 predicted treatment outcome for patients with hyperdiploid >50 ALL, National Cancer Institute (NCI) standard-risk B-ALL or T-cell ALL, while MRD levels on day 46 were prognostic for patients with NCI standard-risk or high-risk B-ALL. Patients with t(12;21)/(ETV6-RUNX1) or hyperdiploidy >50 ALL had the best prognosis; those with a negative MRD on day 19 had a particularly low risk of relapse: 1.9% and 3.8%, respectively. Patients with NCI high-risk B-ALL or T-cell ALL had an inferior outcome; even with undetectable MRD on day 46, cumulative risk of relapse was 12.7% and 15.5%, respectively. Among patients with NCI standard-risk B-ALL, the outcome was intermediate overall but was poor if MRD was ⩾1% on day 19 or MRD was detectable at any level on day 46. Our results indicate that the clinical impact of MRD on treatment outcome in childhood ALL varies considerably according to leukemia subtype and time of measurement.


Subject(s)
Neoplasm, Residual/pathology , Neoplasm, Residual/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Recurrence , Remission Induction , Survival Analysis , Treatment Outcome
3.
Clin Pharmacol Ther ; 101(3): 373-381, 2017 03.
Article in English | MEDLINE | ID: mdl-27564568

ABSTRACT

We performed a genomewide association study (GWAS) of primary erythrocyte thiopurine S-methyltransferase (TPMT) activity in children with leukemia (n = 1,026). Adjusting for age and ancestry, TPMT was the only gene that reached genomewide significance (top hit rs1142345 or 719A>G; P = 8.6 × 10-61 ). Additional genetic variants (in addition to the three single-nucleotide polymorphisms [SNPs], rs1800462, rs1800460, and rs1142345, defining TPMT clinical genotype) did not significantly improve classification accuracy for TPMT phenotype. Clinical mercaptopurine tolerability in 839 patients was related to TPMT clinical genotype (P = 2.4 × 10-11 ). Using 177 lymphoblastoid cell lines (LCLs), there were 251 SNPs ranked higher than the top TPMT SNP (rs1142345; P = 6.8 × 10-5 ), revealing a limitation of LCLs for pharmacogenomic discovery. In a GWAS, TPMT activity in patients behaves as a monogenic trait, further bolstering the utility of TPMT genetic testing in the clinic.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Leukemia/drug therapy , Mercaptopurine/pharmacokinetics , Methyltransferases/genetics , Antimetabolites, Antineoplastic/administration & dosage , Child , Dose-Response Relationship, Drug , Female , Genome-Wide Association Study , Genotype , Humans , Male , Mercaptopurine/administration & dosage , Pharmacogenetics , Polymorphism, Single Nucleotide
4.
Leukemia ; 19(12): 2125-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16281077

ABSTRACT

Despite substantial progress in the management of childhood acute myeloid leukemia (AML), only about 50% of patients are cured by intensive chemotherapy. The long-term results of clinical trials may reveal principles that can guide the development of future therapy. From 1980 to 2000, 251 patients <15 years of age with newly diagnosed AML were enrolled on one of the five consecutive St Jude AML studies. The median age of the 128 boys and 123 girls was 6.2 years; 193 were white, 45 black, and 13 of other racial groups. With the exception of one protocol (AML-83), outcomes improved in general over the two decades. The estimated 5-year event-free survival (+/-s.e.) was 30.8+/-5.6% for AML-80; 11.1+/-4.3% for AML-83; 35.9+/-7.4% for AML-87; 43.5+/-6.2% for AML-91; and 45.0+/-11.1% for AML-97. Resistant or relapsed AML caused the great majority of treatment failures. Increasing the intensity of chemotherapy (AML-87) did not improve outcome, partially because of toxicity, nor did prolonging postremission therapy by adding sequential myeloablative (AML-80) or nonmyeloablative (AML-83) chemotherapy cycles. We conclude that subtype-specific therapies are needed to replace the 'one size fits all' strategy of the past two decades.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Protocols/standards , Leukemia, Myeloid/therapy , Acute Disease , Adolescent , Antineoplastic Combined Chemotherapy Protocols/toxicity , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Leukemia, Myeloid/mortality , Male , Remission Induction/methods , Survival Analysis , Treatment Failure , Treatment Outcome
5.
Leukemia ; 19(8): 1399-403, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15973454

ABSTRACT

To assess the prognosis of overt testicular disease at diagnosis of acute lymphoblastic leukemia, and any therapeutic role of irradiation for this involvement, we reviewed the data of 811 boys treated on St Jude studies Total X--XI (early period) and Total XII-XIV (recent period). In all, 19 boys (2.3%) had testicular disease at diagnosis. In the early period, patients with testicular leukemia had a poorer overall survival (OS) (P=0.003), event-free survival (EFS) (P=0.064), and higher cumulative incidence of relapse (P=0.041) than did other patients. During the recent period, patients with and without overt testicular leukemia did not differ in OS (P=0.257), EFS (P=0.102), or cumulative incidence of relapse (P=0.51). In a multivariate analysis, OS was lower for patients with testicular disease than for those without the involvement in the early period (P=0.047) but not in the recent one (P=0.75). Both patients who received irradiation for residual testicular disease at the end of induction subsequently died of leukemia. Of the other 17 patients who did not receive irradiation, only one developed testicular relapse in combination with bone marrow relapse. In conclusion, the prognostic impact of overt testicular disease has diminished. Irradiation appears to provide no survival advantage to this patient population.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/radiotherapy , Testicular Neoplasms/radiotherapy , Adolescent , Child , Child, Preschool , Humans , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Proportional Hazards Models , Radiotherapy, Adjuvant , Recurrence , Retrospective Studies , Survival Rate , Testicular Neoplasms/mortality , Testicular Neoplasms/therapy
6.
Clin Pharmacol Ther ; 99(6): 651-60, 2016 06.
Article in English | MEDLINE | ID: mdl-26663398

ABSTRACT

Using broad interrogation of clinically relevant drug absorption, distribution, metabolism, and excretion (ADME) genes on the DMET platform, we identified a genetic variant in SLCO1B1 (rs2291075; c.597C>T), encoding the transporter OATP1B1, associated with event-free (P = 0.006, hazard ratio = 1.74) and overall survival (P = 0.012, hazard ratio = 1.85) in children with de novo acute myeloid leukemia (AML). Lack of SLCO1B1 expression in leukemic blasts suggested the association might be due to an inherited rather than a somatic effect. rs2291075 was in strong linkage with known functional variants rs2306283 (c.388A>G) and rs4149056 (c.521T>C). Functional studies in vitro determined that four AML-directed chemotherapeutics (cytarabine, daunorubicin, etoposide, and mitoxantrone) are substrates for OATP1B1 and the mouse ortholog Oatp1b2. In vivo pharmacokinetic studies using Oatp1b2-deficient mice further confirmed our results. Collectively, these findings demonstrate an important role for OATP1B1 in the systemic pharmacokinetics of multiple drugs used in the treatment of AML and suggest that inherited variability in host transporter function influences the effectiveness of therapy.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Animals , Antineoplastic Agents/metabolism , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Cohort Studies , Cytarabine/pharmacokinetics , DNA/genetics , DNA/isolation & purification , Female , Genetic Linkage , Genetic Variation , Humans , Male , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Survival Analysis , Treatment Outcome
7.
Biochim Biophys Acta ; 677(2): 269-73, 1981 Oct 12.
Article in English | MEDLINE | ID: mdl-7197557

ABSTRACT

Methionine adenosyltransferase, one of the two major enzymes utilizing methionine, is regulated by the levels of methionine in the growth medium (Jacobsen, S.J., Hoffman, R.M. and Erbe, R.W. (1980) J. Natl. Cancer Inst. 65, 1237--1244, and Caboche, M. and Mulsant, P. (1978) Somatic Cell Genet. 4, 407--421). We report here that methionyl-tRNA synthetase, unlike methionine adenosyltransferase, behaves in a constitutive manner with respect to the concentration of methionine in the culture medium. This behavior is seen in Chinese hamster ovary cells and in normal diploid and SV40-transformed human fibroblasts. Although the kinetics of regulation of methionine adenosyltransferase and methionyl-tRNA synthetase by exogenous methionine are clearly different, the levels of the two enzymes in the human cell lines are similar.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Methionine Adenosyltransferase/metabolism , Methionine-tRNA Ligase/metabolism , Methionine/metabolism , Transferases/metabolism , Animals , Cell Line , Cricetinae , Enzyme Repression/drug effects , Female , Fibroblasts , Humans , Mesocricetus , Methionine/pharmacology , Ovary
8.
J Clin Oncol ; 18(7): 1525-32, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10735901

ABSTRACT

PURPOSE: Development of antibodies and hypersensitivity to asparaginase are common and may attenuate asparaginase effect. Our aim was to determine the relationship between antiasparaginase antibodies or hypersensitivity reactions and event-free survival (EFS). PATIENTS AND METHODS: One hundred fifty-four children with acute lymphoblastic leukemia received Escherichia coli asparaginase 10,000 IU/m(2) intramuscularly three times weekly for nine doses during multiagent induction and reinduction phases and for seven monthly doses during continuation treatment. Erwinia asparaginase was used in case of clinical hypersensitivity to E coli but not for subclinical development of antibodies. Plasma antiasparaginase antibody concentrations were measured on day 29 of induction in 152 patients. RESULTS: Antibodies were detectable in 54 patients (35.5%), of whom 30 (55.6%) exhibited hypersensitivity to asparaginase. Of the 98 patients who had no detectable antibodies, 18 (18.4%) had allergic reactions. Patients with antibodies were more likely to have a reaction than those without antibodies (P <.001). Among the 50 patients who experienced allergic reactions (including two for whom antibodies were not measured), 36 (72.0%) were subsequently given Erwinia asparaginase; seven (19.4%) reacted to this preparation. EFS did not differ among patients who did and did not have antibodies (P =.54), with 4-year EFS (+/- 1 SE) of 83% +/- 6% and 76% +/- 5%, respectively. Similarly, EFS did not differ among patients who did and did not develop allergic reactions (P =.68), with 4-year estimates of 82% +/- 6% and 78% +/- 5%, respectively. CONCLUSION: In this setting, in which most patients with allergy were switched to another preparation, there was no adverse prognostic impact of clinical or subclinical allergy to asparaginase.


Subject(s)
Antineoplastic Agents/immunology , Asparaginase/immunology , Drug Hypersensitivity , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Adolescent , Antibody Formation , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Asparaginase/pharmacology , Asparaginase/therapeutic use , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Injections, Intramuscular , Male , Prognosis , Treatment Outcome
9.
J Clin Oncol ; 17(1): 191-6, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10458233

ABSTRACT

PURPOSE: To determine the molecular characteristics, clinical features, and treatment outcomes of children with acute lymphoblastic leukemia (ALL) and the t(11;19)(q23,p13.3) translocation. PATIENTS AND METHODS: A retrospective analysis of leukemic cell karyotypes obtained from patients with new diagnoses of ALL who were treated at St. Jude Children's Research Hospital or by the Pediatric Oncology Group was performed to identify cases with the t(11;19)(q23;p13.3) translocation. Molecular analyses were performed on these cases to determine the status of the MLL gene and the presence of the MLL-ENL fusion transcript. RESULTS: Among 3,578 patients with ALL and successful cytogenetic analysis, we identified 35 patients with the t(11;19)(q23;p13.3) translocation: 13 infants and 11 older children had B-precursor leukemia, whereas 11 patients had a T-cell phenotype. Although all of the cases examined had MLL rearrangements and MLL-ENL fusion transcripts, outcome varied according to age and immunophenotype. Among B-precursor cases, only two of the 13 infants remain in complete remission, compared with six of the 11 older children. Most strikingly, no relapses have occurred among B-precursor patients 1 to 9 years of age or among T-cell patients. CONCLUSION: Although MLL gene rearrangements are generally associated with a dismal outcome in ALL, two distinct subsets with MLL-ENL fusions have an excellent prognosis. Our results suggest that patients with this genetic abnormality who have T-cell ALL or are 1 to 9 years of age should not be considered candidates for hematopoietic stem-cell transplantation during their first remission.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 19 , DNA-Binding Proteins/genetics , Gene Rearrangement , Neoplasm Proteins , Nuclear Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic , Adolescent , Child , Child, Preschool , Disease-Free Survival , Female , Histone-Lysine N-Methyltransferase , Humans , Immunophenotyping , Infant , Karyotyping , Male , Myeloid-Lymphoid Leukemia Protein , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Retrospective Studies
10.
J Clin Oncol ; 17(3): 818-24, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10071272

ABSTRACT

PURPOSE: Whether recent improvements in the treatment of childhood acute lymphoblastic leukemia (ALL) have nullified the adverse prognosis associated with male sex remains unclear. Therefore, we analyzed the survival experience and presenting clinical and laboratory features of boys and girls with newly diagnosed ALL who were treated at our institution over the past three decades. PATIENTS AND METHODS: One thousand one hundred fifty-one boys and 904 girls were treated in 13 consecutive Total Therapy studies between 1962 and 1994. An overview analysis was used to investigate the impact of sex on overall and event-free survival, both for the entire cohort and for subgroups defined by treatment era and blast-cell immunophenotype. Stratified analyses were performed to adjust for treatment protocol and known risk factors, and in the modern treatment era, for protocol, immunophenotype, and the DNA content of leukemic cells (ie, DNA index). The pharmacokinetics of methotrexate, teniposide, and cytarabine, as well as the thiopurine methyltransferase activity of erythrocytes, were compared between boys and girls treated on a single protocol. RESULTS: Compared with girls, boys were more likely to have T-cell ALL (20.9% v 10.7%, P < .001) and seemed less likely to have a favorable DNA index (17.8% v 25.1%, P = .072). There were no other statistically significant differences between the two sexes with respect to presenting features, including leukemic-cell genetic abnormalities, nor were there significant sex differences in the pharmacokinetics of methotrexate, teniposide, or cytarabine or in erythrocyte thiopurine methyltransferase activity. Girls clearly fared better than boys (P < .001) on protocols used during the early era of treatment (10-year event-free survival +/- 1 SE, 43.1%+/-2.1% v 31.5%+/-1.7%). Although prognosis improved for both sexes in the modern era, the difference in outcome between girls and boys persisted (P = .025) (10-year event-free survival, 73.4%+/-3.7% v 63.5%+/-4.0%). However, stratification of modern-era patients by protocol, immunophenotype, and DNA index mitigated statistical evidence of a sex difference in overall survival (P = .263) and event-free survival (P = .124). CONCLUSION: Although boys and girls alike have benefited from improvements in ALL therapy, these gains have not completely eliminated the sex difference in prognosis that has persisted since the early 1960s. The apparent difference in outcome is partially explained by differences between boys and girls in the distributions of ALL immunophenotype and DNA index.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Sex Factors , Adolescent , Child , Child, Preschool , Female , Humans , Immunophenotyping , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prognosis , Survival Analysis , Treatment Outcome
11.
J Clin Oncol ; 16(12): 3768-73, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9850020

ABSTRACT

PURPOSE: To reassess the clinical and biologic significance of myeloid-associated antigen expression in childhood acute lymphoblastic leukemia (ALL). PATIENTS AND METHODS: We prospectively studied 334 newly diagnosed cases of this disease, using a comprehensive panel of antibodies that represented five myeloid cluster groups (CD13, CD14, CD15, CD33, and CD65). Blast cells were tested for ETV6 and MLL rearrangement using Southern blot analysis. RESULTS: CD13 was expressed in 13.7% of cases, CD14 in 1%, CD15 in 6.6%, CD33 in 16%, and CD65 in 9.7%. Approximately one third of cases (31.4%) expressed one or more of these antigens (B-cell precursor, 31.9%; T-cell, 28.8%), while 10.5% expressed two or more (B-cell precursor, 11.3%; T-cell, 6.1%). Among the B-cell precursor leukemias, myeloid-associated antigen expression was significantly associated with a lack of hyperdiploidy and rearrangements of ETV6 or MLL gene. Most of the cases with MLL rearrangements (82%) expressed CD65, CD15, and CD33, either alone or in combination, whereas 48% of those with a rearranged ETV6 gene expressed CD13, CD33, or both. Myeloid-associated antigen expression did not correlate with event-free survival, whether the analysis was based on any of the five antigens in our panel or on the three more commonly tested antigens (CD13, CD33, and CD65). Importantly, this finding was not affected by exclusion of patients with ETV6 or MLL gene rearrangements. CONCLUSION: Even though blast cell expression of myeloid-associated antigen expression shows significant associations with specific genetic abnormalities, it lacks prognostic value in childhood ALL.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Bone Marrow Cells/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Adolescent , Blotting, Southern , Child , Child, Preschool , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Infant , Male , Predictive Value of Tests , Prospective Studies
12.
J Clin Oncol ; 15(3): 1150-7, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9060558

ABSTRACT

PURPOSE: TEL gene rearrangements due to the 12;21 chromosomal translocation are the most common molecular genetic abnormality in childhood acute lymphoblastic leukemia (ALL), occurring in approximately 25% of cases with a B-precursor immunophenotype. The limited number of clinically useful genetic markers in this leukemia subtype prompted us to assess TEL status as a predictor of treatment outcome. PATIENTS AND METHODS: We determined the status of the TEL gene (rearranged or germline) in 188 cases of B-precursor acute leukemia using Southern blot analysis and related the findings to event-free survival. All comparisons of outcome were stratified by treatment regimen, risk classification, age, and leukocyte count. RESULTS: Forty-eight patients (26%) had a rearranged TEL gene. At 5 years of follow-up, an estimated 91% +/- 5% (SE) of this group were event-free survivors, compared with only 65% +/- 5% of the group with germline TEL (stratified log-rank P = .011). For nonhyperdiploid patients, the odds ratio of an adverse event in the germline TEL group to that for the rearranged TEL group was 4.06 (95% confidence interval, 1.86 to 8.84). The relationship of TEL rearrangement to a favorable prognosis was independent of recognized good-risk features in B-precursor leukemia, including age, initial leukocyte count, and hyperdiploidy. CONCLUSION: Rearrangement of the TEL gene distinguishes a large subset of children with favorable-prognosis B-precursor leukemia who cannot be identified by standard prognostic features. It may be possible to treat these patients less aggressively without loss of therapeutic efficacy.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 21/genetics , Gene Rearrangement, B-Lymphocyte/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic/genetics , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Disease-Free Survival , Female , Genetic Markers , Humans , Infant , Male
13.
J Clin Oncol ; 18(18): 3273-9, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10986060

ABSTRACT

PURPOSE: To investigate the incidence of and risk factors for late sequelae of treatment in patients who survived for more than 10 years after the diagnosis of childhood acute myeloid leukemia (AML). PATIENTS AND METHODS: Of 77 survivors (median follow-up duration, 16. 7 years), 44 (group A) had received chemotherapy, 18 (group B) had received chemotherapy and cranial irradiation, and 15 (group C) had received chemotherapy, total-body irradiation, and allogeneic bone marrow transplantation. Late complications, tobacco use, and health insurance status were assessed. RESULTS: Growth abnormalities were found in 51% of survivors, neurocognitive abnormalities in 30%, transfusion-acquired hepatitis in 28%, endocrine abnormalities in 16%, cataracts in 12%, and cardiac abnormalities in 8%. Younger age at the time of diagnosis or initiation of radiation therapy, higher dose of radiation, and treatment in groups B and C were risk factors for the development of academic difficulties and greater decrease in height Z: score. In addition, treatment in group C was a risk factor for a greater decrease in weight Z: score and the development of growth-hormone deficiency, hypothyroidism, hypogonadism, infertility, and cataracts. The estimated cumulative risk of a second malignancy at 20 years after diagnosis was 1.8% (95% confidence interval, 0.3% to 11.8%). Twenty-two patients (29%) were smokers, and 11 (14%) had no medical insurance at the time of last follow-up. CONCLUSION: Late sequelae are common in long-term survivors of childhood AML. Our findings should be useful in defining areas for surveillance of and intervention for late sequelae and in assessing the risk of individual late effects on the basis of age and history of treatment.


Subject(s)
Leukemia, Myeloid/complications , Leukemia, Myeloid/therapy , Acute Disease , Adolescent , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Bone Marrow Transplantation/adverse effects , Child , Child, Preschool , Cognition Disorders/chemically induced , Cognition Disorders/etiology , Cranial Irradiation/adverse effects , Endocrine System Diseases/chemically induced , Endocrine System Diseases/etiology , Female , Fertility/drug effects , Fertility/radiation effects , Follow-Up Studies , Growth Disorders/chemically induced , Growth Disorders/etiology , Heart Diseases/chemically induced , Hepatitis B/etiology , Hepatitis C/etiology , Humans , Infant , Male , Neoplasm Recurrence, Local , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/etiology , Radiation Injuries/etiology , Risk Factors , Smoking , Time Factors , Whole-Body Irradiation/adverse effects
14.
Leukemia ; 10(1): 74-82, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8558942

ABSTRACT

The MLL gene, located at chromosome 11, band q23, is frequently disrupted by a variety of chromosomal rearrangements that occur in acute lymphoblastic leukemias and in a subset of de novo and secondary acute myeloid leukemias. In both scenarios, MLL rearrangements are associated with distinct clinical features and a poor prognosis. MLL encodes a large protein (MLL) that shares homology with the Drosophila trithorax protein. 11q23 translocations result in the generation of a series of acute leukemia-specific chimeric proteins that contain the N-terminus of MLL and are thought to be crucial to leukemogenesis. In this article, we review the structural features of the MLL fusion proteins as well as the clinical features and molecular diagnosis of acute leukemias containing MLL arrangements.


Subject(s)
Chromosomes, Human, Pair 11 , Gene Rearrangement , Leukemia/genetics , Proto-Oncogenes , Transcription Factors , Acute Disease , Chromosome Mapping , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Zinc Fingers/genetics
15.
Leukemia ; 13(1): 6-13, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10049061

ABSTRACT

The TEL and AML1 genes are common targets of chromosomal translocations in hematopoietic malignancies. The TEL-AML1 fusion gene, created by the t(12;21), is the most common genetic alteration in childhood acute lymphoblastic leukemia and is associated with a favorable outcome. This review summarizes the roles of the TEL and AML1 proteins in hematopoiesis, the potential transforming mechanisms of TEL fusion proteins, and the clinical significance of the TEL-AML1 fusion.


Subject(s)
Leukemia/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion , Translocation, Genetic , Adolescent , Artificial Gene Fusion , Child , Child, Preschool , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Core Binding Factor Alpha 2 Subunit , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogenes
16.
Leukemia ; 17(11): 2090-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14523477

ABSTRACT

To determine the clinical significance of central nervous system (CNS) involvement at the time of diagnosis of pediatric acute myeloid leukemia (AML), we analyzed clinical features and outcomes of 290 patients treated consecutively on four institutional trials (AML80, AML83, AML87, and AML91). CNS status was classified as CNS1 (no blast cells in CSF; n=205), CNS2 (<5 WBC/mul CSF with blast cells; n=37), or CNS3 (>/=5 WBC/mul CSF with blast cells, or signs of CNS involvement; n=48). Patients with CNS3 status were significantly younger than others (P=0.016) and significantly more likely to have the favorable cytogenetic features t(9;11), t(8;21), or inv(16) (P<0.001). The CNS3 group had a significantly greater probability (+/-s.e.) of 5-year event-free survival (43.7+/-7.0%) than did the CNS1 (27.8+/-3.2%, P=0.015) and CNS2 (24.3+/-7.5%, P=0.032) groups. However, after adjustment for favorable genetic features, there was no significant difference in EFS between the CNS3 and the combined CNS1+CNS2 groups (P=0.075). In all, 10 of 151 patients treated on AML80 and AML83, but none of 139 treated on AML87 and AML91, had primary CNS relapse. CNS involvement had no adverse prognostic significance, and patients with CNS2 status had similar outcome to CNS1 patients in this large group of pediatric patients with AML, treated at a single institution.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System Neoplasms/epidemiology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Acute Disease , Adolescent , Age Factors , Blast Crisis/pathology , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/radiotherapy , Child , Child, Preschool , Clinical Trials as Topic , Disease-Free Survival , Female , Humans , Leukemia, Myeloid/genetics , Male , Prognosis , Recurrence , Retrospective Studies , Time Factors
17.
Leukemia ; 11(7): 979-83, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9204978

ABSTRACT

Acute lymphoblastic leukemia (ALL) occurring in infants less than 1 year of age differs clinically and biologically from that observed in older children. Cytogenetically, 11q23 translocations are detected in approximately 50% of infant ALLs and fuse the 11q23 gene HRX with a variety of partner chromosomal loci. Overall, HRX rearrangements are detected molecularly in 70-80% of infant ALLs as compared to 5-7% of ALLs arising in older children. Two recently described molecular abnormalities in childhood ALL are ETV6 gene rearrangements and homozygous deletions of p16(INK4A) and/or p15(INK4B). Each of these abnormalities occurs in 15-20% of all childhood ALLs, and neither can be accurately identified by routine cytogenetic analyses. The incidence of these genetic abnormalities and their potential relationship to HRX gene status in infant ALL is unknown. Using Southern blot analyses, we determined ETV6 and p16(INK4A)/p15(INK4B) gene status in a cohort of infant ALLs. No ETV6 rearrangements or homozygous deletions (n=69) or homozygous p16(INK4A) and/or p15(INK4B) gene deletions (n=54) were detected in any of the infant ALLs. Therefore, ETV6 and p16(INK4A)/p15(INK4B) do not play a significant role in the pathogenesis of infant ALL, further emphasizing the distinctive biology of this subset of leukemias.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins , DNA-Binding Proteins/genetics , Gene Deletion , Gene Rearrangement , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogenes , Repressor Proteins , Transcription Factors/genetics , Tumor Suppressor Proteins , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , Histone-Lysine N-Methyltransferase , Humans , Infant , Myeloid-Lymphoid Leukemia Protein , Proto-Oncogene Proteins c-ets , ETS Translocation Variant 6 Protein
18.
Leukemia ; 10(9): 1456-8, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8751462

ABSTRACT

Cryptic t(12;21)(p12-13;q22) leading to TEL-AML1 fusion has recently been recognized as the most frequent genetic rearrangement in childhood acute lymphoblastic leukemia (ALL) in Western countries. More recently, we found a similar frequency of this abnormality in Chinese children with ALL in Taiwan. In this study, we assessed further the frequency of TEL-AML1 fusion as well as that of BCR-ABL in Chinese adults with ALL, using reverse transcriptase-polymerase chain reaction assays. Among the 81 cases with newly diagnosed B lineage ALL studied, none had the TEL-AML1 fusion whereas 30 had the BCR-ABL fusion. The lack of cases with the TEL-AML1 fusion together with the high frequency of BCR-ABL fusion could largely account for the poorer outcome of adult ALL as compared with childhood ALL.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , DNA-Binding Proteins/biosynthesis , Leukemia-Lymphoma, Adult T-Cell/metabolism , Neoplasm Proteins/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins , Repressor Proteins , Transcription Factors/biosynthesis , Translocation, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/genetics , Female , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Male , Middle Aged , Molecular Sequence Data , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-ets , Transcription Factors/genetics , ETS Translocation Variant 6 Protein
19.
Leukemia ; 12(5): 675-81, 1998 May.
Article in English | MEDLINE | ID: mdl-9593264

ABSTRACT

We investigated the level of minimal residual disease (MRD) in 26 children with B-lineage acute lymphoblastic leukemia (ALL) after intensive induction therapy. A quantitative semi-nested polymerase chain reaction (PCR) detecting the clone-specific rearranged immunoglobulin heavy chain genes was developed to improve sensitivity and specificity of amplification. In all patients, one leukemic cell could be detected in a background of 10(5) normal blood mononuclear cells. All patients investigated were in complete remission at the end of induction therapy as evaluated by morphologic criteria. Nineteen patients (73%) had no detectable residual leukemic cells using the sensitive semi-nested PCR. Seven patients (27%) were PCR positive. Three had a low level (<2 x 10(-5) leukemic cells per bone marrow cell), while four patients had a high level (>2 x 10(5)) of detectable residual leukemic cells. All patients with low or undetectable levels of residual leukemia remained in complete remission at a median of 63 months from diagnosis (range 40-80 months), while all four patients with a high level of residual leukemia subsequently relapsed at a median of 21 months from diagnosis (range 13-37 months). The patient groups with undetectable or low, and high level of MRD did not differ significantly in other clinical or genetic features with prognostic significance. We conclude that the level of MRD at the end of the intensive induction therapy period is predictive of outcome in childhood B lineage ALL. If confirmed by large prospective studies, the level of MRD might be useful in stratifying patients into high and low risk categories.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm, Residual/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Child , Child, Preschool , Female , Humans , Immunophenotyping , Infant , Male , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prognosis , Remission Induction , Retrospective Studies , Sensitivity and Specificity , Treatment Outcome
20.
Leukemia ; 10(6): 991-3, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8667657

ABSTRACT

Despite its rarity by routine karyotypic analysis, cryptic t(12;21)(p12-13;q22) translocation leading to TEL/AML1 fusion has been recognized as the most frequent genetic rearrangement in childhood acute lymphoblastic leukemia (ALL) in two recent studies, one from France and the other from the United States. To estimate the frequency of this abnormality in the Chinese population, we studied 41 children with ALL and 17 with acute myeloid leukemia (AML) in two medical centers in Taiwan, using the reverse transcriptase polymerase chain reaction (RT-PCR) assay. Results of this analysis demonstrated a 17% frequency of this translocation in the ALL population overall and 19% in patients with B-lineage ALL, similar to previous findings in Caucasian children. None of the patients with AML had TEL/AML1 fusion transcripts. In addition to its association with the B-lineage immunophenotype, TEL/AML1 was also correlated with a low presenting leukocyte count and favorable age (1-10 years). These findings, combined with earlier reports, indicate that TEL/AML1 fusion is the most frequent genetic abnormality in childhood ALL, regardless of race. Molecular diagnosis of t(12;21)-positive ALL may identify a subgroup of patients who do not require intensive treatment for cure.


Subject(s)
Burkitt Lymphoma/genetics , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins , Repressor Proteins , Transcription Factors/genetics , Translocation, Genetic , Adolescent , Base Sequence , Burkitt Lymphoma/epidemiology , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Gene Rearrangement , Humans , Incidence , Infant , Molecular Sequence Data , Polymerase Chain Reaction , Prognosis , Proto-Oncogene Proteins c-ets , Taiwan/epidemiology , ETS Translocation Variant 6 Protein
SELECTION OF CITATIONS
SEARCH DETAIL