Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
2.
Leukemia ; 16(6): 1121-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040442

ABSTRACT

Significant predictors of treatment outcome are poorly defined for patients with T-lineage acute lymphoblastic leukemia (T-ALL). A high WBC at diagnosis, which has traditionally been a predictor of poor response in T-ALL, has considerably weakened prognostic significance in the face of modern, more intensive chemotherapy. To test the hypothesis that bone marrow stroma-supported leukemic cell recovery might identify children at high risk for relapse, we measured the ex vivo recovery of T-ALL lymphoblasts from 29 newly diagnosed patients using a stromal cell co-culture assay. In all cases the T-ALL lymphoblasts showed an increase in recovery of T-ALL cells (RTC), ranging from 4 to 343%, in comparison to samples maintained without stroma. Since we were blinded to patient outcome in this case-control study, we then correlated patient outcome with RTC. The RTC for 18 patients in complete continuous remission (CCR) for greater than 4 years was stochastically larger than for the 11 patients who eventually relapsed (P = 0.011, by the two-sided Wilcoxon test). Furthermore, 100% of patients with an RTC of more than 26% had a CCR greater than 4 years while 78% of the patients with an RTC of less than 25% relapsed within 4 years. This is the first report to show that higher lymphoblast recovery may predict a more favorable outcome for children with T-ALL. A prospective study is needed to test whether stroma-supported leukemic cell recovery might serve as a basis for assigning risk-adjusted therapy.


Subject(s)
Bone Marrow Cells/cytology , Coculture Techniques/methods , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Neoplasm Recurrence, Local/diagnosis , Stromal Cells/physiology , Adolescent , Adult , Case-Control Studies , Cell Line , Cell Lineage , Cell Survival , Child , Child, Preschool , Humans , Infant , Leukemia-Lymphoma, Adult T-Cell/diagnosis , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukocyte Count , Treatment Outcome , Tumor Cells, Cultured
3.
Br J Haematol ; 113(3): 746-56, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11380466

ABSTRACT

The downstream effects of p15 and p16 gene deletions and loss of transcripts on dihydrofolate reductase (DHFR) were examined in 63 B-precursor (BP) acute lymphoblastic leukaemia (ALL) samples. p15 and/or p16 gene deletions were seen in 6% and 8%, respectively, of BP-ALL samples; however, losses of p15 and/or p16 transcripts were seen in 26 out of 63 (41%) samples. Loss of p15 transcripts (36.5%) exceeded that for p16 (17.5%). For the 26 BP-ALLs that lacked p15 and/or p16 transcripts, only six (23%) exhibited low levels of DHFR by flow cytometry assay with Pt430, a fluorescent anti-folate. Conversely, 18 out of 37 (49%) BP-ALL samples with intact p15 and/or p16 genes and transcripts showed low levels of DHFR (P = 0.04). In p15- and p16-null K562 cells transfected with a tetracycline-inducible p15 cDNA construct, induction of p15 transcripts and protein was accompanied by decreased growth rates, decreased S-phase fraction, decreased retinoblastoma protein phosphorylation, and markedly reduced levels of DHFR transcripts and protein. Collectively, our results suggest that losses of p15 and/or p16 gene expression result in elevated levels of DHFR in BP-ALL in children. However, additional downstream factors undoubtedly also contribute to elevated levels of this enzyme target.


Subject(s)
Burkitt Lymphoma/genetics , Cell Cycle Proteins , Gene Deletion , Genes, p16 , Tetrahydrofolate Dehydrogenase/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins , Adolescent , Blotting, Southern , Burkitt Lymphoma/enzymology , Case-Control Studies , Cell Cycle , Child , Child, Preschool , Confidence Intervals , Cyclin-Dependent Kinase Inhibitor p15 , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Female , Flow Cytometry , Gene Expression/drug effects , Humans , Infant , K562 Cells , Logistic Models , Male , Odds Ratio , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
Biol Blood Marrow Transplant ; 7(1): 49-57, 2001.
Article in English | MEDLINE | ID: mdl-11215699

ABSTRACT

The use of high-dose therapy (HDT) and autologous hematopoietic cell transplantation (AHCT) for children and adolescents with primary refractory and relapsed Hodgkin's disease is increasing. The purpose of this retrospective analysis was to: (1) evaluate the outcome of HDT and AHCT in pediatric patients with Hodgkin's disease, and (2) identify factors that predispose patients to the development of transplantation-related complications. We describe the experiences of 34 pediatric patients from a single institution with primary refractory or relapsed Hodgkin's disease. HDT regimens consisted of cyclophosphamide and etoposide combined with either carmustine, chloroethylcyclohexylnitrosurea, or fractionated total body irradiation. Kaplan-Meier survival predicts that 67% (95% confidence interval [CI] 47%-87%) of patients will be alive and disease-free at 5 years. Nine patients had disease recurrence, of whom 5 relapsed after 1 year (1.5-6.3 years). Five patients succumbed to treatment-related toxicities, of whom 4 died of pulmonary failure. Fifteen patients (44%) developed post-AHCT idiopathic diffuse lung injury syndrome: acute alveolitis (n = 2); diffuse alveolar hemorrhage (n = 2); acute respiratory distress syndrome (n = 2); delayed interstitial pneumonitis (n = 8); and bronchiolitis obliterans (n = 1). The following factors did not predict for the development of a diffuse lung injury syndrome in univariate analysis: prior treatment with bleomycin, pre-HDT pulmonary function tests, and prior thoracic irradiation. Of the patients in our cohort, 44% had a history of atopy (allergic rhinitis and/or asthma). Multivariate logistic analysis revealed that a preexisting history of atopy was highly predictive of idiopathic pulmonary complications (P = .0001, odds ratio = 21, CI 3.6-125). Our experience shows that HDT followed by AHCT results in durable remissions in two thirds of pediatric patients with refractory and relapsed Hodgkin's disease, and a history of atopy is associated with post-AHCT pulmonary complications.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Adolescent , Child , Child, Preschool , Combined Modality Therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hodgkin Disease/complications , Hodgkin Disease/pathology , Humans , Hypersensitivity, Immediate , Predictive Value of Tests , Prognosis , Recurrence , Respiratory Distress Syndrome/etiology , Retrospective Studies , Transplantation, Autologous
5.
Blood ; 96(7): 2543-9, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11001909

ABSTRACT

To further define the cytogenetic differences between B-cell lineage (B-lineage) acute lymphoblastic leukemia (ALL) and T-cell lineage ALL (T-ALL) and to determine the prognostic value of cytogenetics in childhood T-ALL, the blast cell karyotypes of 343 cases of pediatric T-ALL, the largest series reported to date, were evaluated. Cytogenetics were performed in a single central laboratory, and the children were treated using a single Pediatric Oncology Group protocol. Clear differences between the karyotypic characteristics of B-lineage ALL and T-ALL were confirmed. This study suggests that there may be survival differences associated with some T-ALL blast cell karyotypes. Better survival is associated with only normal karyotypes and with t(10;14) (translocation of chromosomes 10 and 14); worse survival is associated with the presence of any derivative chromosome. Two new recurring chromosome aberrations previously not reported in T-ALL were found: del(1)(p22) and t(8;12)(q13;p13). Ten aberrations found in this series, which were reported only once previously in T-ALL, can now be considered recurring abnormalities in T-ALL. All 12 of these new recurring aberrations are targets for discovery and characterization of new genes that are important in T-cell development and leukemogenesis.


Subject(s)
Chromosome Aberrations , Karyotyping , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Adolescent , Adult , Aneuploidy , Burkitt Lymphoma/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 14 , Female , Humans , Infant , Leukemia-Lymphoma, Adult T-Cell/mortality , Male , Prognosis , Survival Rate , Translocation, Genetic
6.
Biol Blood Marrow Transplant ; 6(3): 254-61, 2000.
Article in English | MEDLINE | ID: mdl-10871150

ABSTRACT

We have previously demonstrated a decrease in the incidence of acute graft-versus-host disease (GVHD) with the addition of methotrexate (MTX) to cyclosporine (CSP) and prednisone (PSE) chemotherapy in patients with leukemia. We have now completed a prospective randomized trial comparing the 3-drug regimen (CSP/MTX/PSE, including 3 doses of MTX) to the standard 2-drug regimen (CSP/MTX, including 4 doses of MTX) to investigate the benefit of PSE used up front for the prevention of acute and chronic GVHD. In the trial, 193 patients were randomized and 186 were included in the final analysis. All patients received a bone marrow graft from a fully histocompatible sibling donor. The preparatory regimen consisted of fractionated total-body irradiation (fTBI) and etoposide in all but 13 patients, who received fTBI and cyclophosphamide. The patients were randomized to receive either CSP/MTX/PSE or CSP/MTX. The 2 groups were well balanced with respect to diagnosis, disease stage, age, donor-recipient sex, and parity. In an intent-to-treat analysis, the incidence of acute GVHD was 18% (95% confidence interval [CI] 12-28) for the CSP/MTX/PSE group compared with 20% (CI 10-26) for the CSP/,MTX group (P = .60), with a median follow up of 2.2 years. Overall survival was 65% for those receiving CSP/MTX/PSE and 72% for those receiving CSP/MTX (P = .10); the relapse rate was 15% for the CSP/MTX/PSE group and 12% for the CSP/MTX group (P = .83). The incidence of chronic GVHD was similar (46% versus 52%; P = .38), with a follow-up of 0.7 to 6.0 years. Of interest, 21 patients went off study due to GVHD (5 in the CSP/MTX/PSE group and 16 in the CSP/MITX group [P = .02]), and 11 patients went off study because of alveolar hemorrhage (3 in the CSP/MTX/PSE group and 8 in the CSP/MTX group [P = .22]). The addition of PSE did not result in a higher incidence of infectious complications, bacterial (66% versus 58%), viral (77% versus 66%), or fungal (20% versus 20%), in those receiving CSP/MTX/PSE versus CSP/MTX, respectively. These data suggest that the addition of PSE was associated with a somewhat lower incidence of early posttransplantation complications but did not have a positive impact on the incidence of acute or chronic GVHD or event-free or overall survival.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Bone Marrow Transplantation , Cyclosporine/administration & dosage , Graft vs Host Disease/prevention & control , Immunosuppressive Agents/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia/therapy , Methotrexate/administration & dosage , Prednisone/administration & dosage , Acute Disease , Adolescent , Adult , Bone Marrow Transplantation/adverse effects , Child , Child, Preschool , Double-Blind Method , Graft vs Host Disease/etiology , Humans , Infant , Middle Aged , Prospective Studies , Survival Analysis , Transplantation, Homologous , Treatment Outcome
7.
Leukemia ; 14(5): 792-5, 2000 May.
Article in English | MEDLINE | ID: mdl-10803508

ABSTRACT

The rapidity of response to induction therapy is emerging as an important prognostic factor in children and adolescents with acute lymphoblastic leukemia (ALL). We studied the relationship between rapidity of reduction in peripheral blood blast count and treatment outcome in children with T cell ALL (T-ALL). Initial systemic chemotherapy included prednisone, vincristine, doxorubicin and cyclophosphamide. A Cox analysis evaluated the correlation between the length of time that the peripheral blood absolute blast count (ABC) remained above 1000/mm3 following the start of treatment and event-free survival (EFS). Data were available for 281 patients. Patients for whom the ABC remained >1000/mm3 for 3 or more days following administration of intensive therapy had an estimated 5-year EFS of 34.2% (s.e. = 7.2) vs 58.3% (3.5) for those whose ABC was <1000/mm3 within 0-2 days, with a hazard ratio (HR) of failure of 2.03 (95% CI = 1.35-3.06, P < 0.001) for the slower responding patients. Pre-treatment of some type (usually with prednisone) occurred in 128 patients (average duration 1.7 days). When this was accounted for, patients with an ABC >1000/mm3 for 5 or more days following the start of treatment of any kind had a HR for failure of 2.27 (95% CI = 1.38-3.72, P < 0.001) compared to those responding within 0-4 days. Inclusion of other clinical and biological factors in a multivariate analysis did not alter the prognostic importance of slower blast clearance. Pediatric patients with T-ALL who have a circulating blast count >1000/mm3 at diagnosis and a relatively slower response to initial treatment are at increased risk of treatment failure. Rapidity of response may therefore be a clinically useful prognostic factor for patients with T-ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/therapeutic use , Blast Crisis/blood , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Adolescent , Blast Crisis/pathology , Child , Confidence Intervals , Cyclophosphamide/administration & dosage , Disease-Free Survival , Doxorubicin/administration & dosage , Humans , Leukemia-Lymphoma, Adult T-Cell/blood , Leukemia-Lymphoma, Adult T-Cell/mortality , Multivariate Analysis , Predictive Value of Tests , Prednisone/administration & dosage , Prognosis , Regression Analysis , Remission Induction , Vincristine/administration & dosage
8.
Leukemia ; 14(3): 369-73, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720128

ABSTRACT

Contemporary chemotherapy has significantly improved event-free survival among patients with T cell-lineage acute lymphoblastic leukemia (T-ALL). Unlike B-precursor ALL, most investigators are still using cranial radiation (CRT) and are hesitant to rely solely on intrathecal therapy for T-ALL. In this study we assessed the effects of CRT upon event-free survival and central nervous system (CNS) relapses in a cohort of children with high risk features of T cell leukemia. In a series of six consecutive studies (1987-1995) patients were non-randomly assigned their CNS prophylaxis per individual protocol. These protocols were based on POG 8704 which relied on rotating drug combinations (cytarabine/cyclophosphamide, teniposide/Ara-C, and vincristine/doxorubicin/6-MP/prednisone) postinduction. Modifications such as high-dose cytarabine, intermediate-dose methotrexate, and the addition of G-CSF, were designed to give higher CNS drug levels (decreasing the need for CRT), to eliminate epidophyllotoxin (decreasing the risk of secondary leukemia), and to reduce therapy-related neutropenia (pilot studies POG 9086, 9295, 9296, 9297, 9398). All patients included in this analysis qualified for POG high risk criteria, WBC >50000/mm3 and/or CNS leukemia. Patients without CNS involvement received 16 doses of age-adjusted triple intra-thecal therapy (TIT = hydrocortisone, MTX, and cytarabine) whereas patients with CNS disease received three more doses of TIT during induction and consolidation. Patients who received CRT were treated with 2400 cGy (POG 8704) or 1800 cGy (POG 9086 and 9295). CNS therapy included CRT in 144 patients while the remaining 78 patients received no radiation by original protocol design. There were 155 males and 57 females with a median age of 8.2 years. The median WBC for the CRT+ and CRT- patients were 186000/mm3 and 200000/mm3, respectively. CNS involvement at diagnosis was seen in 16% of the CRT+ and 23% of the CRT- groups. The complete continuous remission rate (CCR) was not significantly different for the irradiated vs. non-irradiated groups (P = 0.46). The 3-year event-free survival was 65% (s.e. 6%) and 63% (s.e. 4%) for the non-irradiated vs. the radiated group. However, the 3-year CNS relapse rate was significantly higher amongst patients who did not receive CRT; 18% (s.e. 5%) vs. 7% (s.e. 3%) in the irradiated group (P = 0.012). Our analysis in a non-randomized setting, suggests that CRT did not significantly correlate with event-free survival but omitting it had an adverse effect on the CNS involvement at the time of relapse.


Subject(s)
Cranial Irradiation , Leukemia-Lymphoma, Adult T-Cell/radiotherapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System/pathology , Child , Child, Preschool , Cohort Studies , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Disease-Free Survival , Female , Humans , Infant , Injections, Spinal , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemic Infiltration/prevention & control , Male , Methotrexate/administration & dosage , Podophyllotoxin/administration & dosage , Prognosis , Remission Induction , Risk , Teniposide/administration & dosage , Treatment Outcome
9.
Leukemia ; 14(2): 232-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10673738

ABSTRACT

Epipodophyllotoxin-associated secondary myeloid leukemia is a devastating complication of acute lymphoblastic leukemia (ALL) therapy. The risk factors for treatment-related myeloid leukemia remain incompletely defined. Genetic deficiencies in glutathione S-transferase (GST) activities have been linked to higher frequencies of a number of human malignancies. Our objective was to determine whether the null genotype for GSTM1, GSTT1, or both, was more frequent in children with ALL who developed treatment-related myeloid malignancies as compared to those who did not. A PCR technique was used to assay for the null genotype for GSTM1 and GSTT1 in 302 children with ALL, 57 of whom also subsequently developed treatment-related acute myeloid leukemia or myelodysplastic syndrome. Among children with ALL who did not develop treatment-related myeloid malignancies, the frequencies of GSTM1 and GSTT1 wild-type, GSTM1 null-GSTT1 wild-type, GSTM1 wild-type-GSTT1 null, and GSTM1 and GSTT1 null genotypes were 40%, 42%, 9% and 9%, respectively. The corresponding frequencies for patients who developed acute myeloid malignancies were 42%, 32%, 11% and 16%, respectively (P = 0.26). A statistically significant increase in the frequency of the GST null genotype was observed in male patients who developed myeloid malignancies as compared to male ALL control patients (P = 0.036), but was not observed in female patients (P = 0.51). Moreover, a logistic regression analysis of possible predictors for myeloid malignancies, controlling for gender and race, did not reveal an association of GSTM1 or GSTT1 null genotypes (P = 0.62 and 0.11, respectively) with treatment-related malignancies. Our data suggest that GSTM1 and GSTT1 null genotypes may not predispose to epipodophyllotoxin-associated myeloid malignancies.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Glutathione Transferase/genetics , Leukemia, Myeloid, Acute/chemically induced , Neoplasms, Second Primary/chemically induced , Podophyllotoxin/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Child , Child, Preschool , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Female , Genotype , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/ethnology , Leukemia, Myeloid, Acute/genetics , Male , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/metabolism , Neoplasms, Second Primary/enzymology , Neoplasms, Second Primary/ethnology , Neoplasms, Second Primary/genetics , Podophyllotoxin/therapeutic use , Polymerase Chain Reaction/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Risk Factors , United States
10.
Med Pediatr Oncol ; 34(2): 92-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10657867

ABSTRACT

BACKGROUND: Growth factor use has been shown to ameliorate chemotherapy-induced neutropenia, leading to shorter hospital stays and lower use of parenteral antibiotics, two costly areas of cancer treatment. Prior reports on pediatric patients have shown evidence of cost savings in some studies, but no such evidence in others. In this study a retrospective analysis compared the costs of inpatient supportive care for pediatric patients with T-cell leukemia and advanced lymphoblastic lymphoma enrolled in a Pediatric Oncology Group trial. PROCEDURE: Patients 1-22 years of age were randomized to receive either granulocyte colony-stimulating factor (G-CSF; n = 45) or no G-CSF (n = 43) following induction and two cycles of maintenance therapy. There were no significant differences in neutropenia-related outcomes during the induction phase. During maintenance therapy, G-CSF patients had significantly fewer days to an ANC >500 cells/microl and a trend towards fewer days of hospitalization. Data on resource utilization were tabulated from case report forms. Costs were imputed from national data on hospitalization costs, average wholesale prices of pharmaceuticals, and patient billing information from a single institution. RESULTS: Total median costs of supportive care were $34,190 for patients receiving G-CSF and $28,653 for patients not receiving G-CSF (P > 0. 05 for the cost difference). Sensitivity analyses demonstrated that the total cost difference was not statistically significant, even in scenarios that included reasonable variations in estimates of the range of the length of stay, antibiotic regimen, and dosage and cost of G-CSF. CONCLUSIONS: In the setting of pediatric leukemia, the cost of growth factor may offset potential savings from shorter hospital stays or lower antibiotic use, a finding consistent with that from the Children's Cancer Study Group.


Subject(s)
Granulocyte Colony-Stimulating Factor/economics , Granulocyte Colony-Stimulating Factor/therapeutic use , Leukemia, T-Cell/drug therapy , Neutropenia/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Adult , Antineoplastic Agents/adverse effects , Child , Child, Preschool , Costs and Cost Analysis , Female , Filgrastim , Humans , Infant , Length of Stay , Male , Neutropenia/chemically induced , Prospective Studies , Recombinant Proteins , Retrospective Studies
11.
Leukemia ; 13(12): 2053-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10602428

ABSTRACT

Between 1984 and 1997, 23 consecutive patients with Philadelphia chromosome-positive acute lymphoblastic leukemia in first complete remission were treated with allogeneic bone marrow transplants from HLA-matched siblings. All patients but one were conditioned with fractionated total body irradiation (1320 cGy) and high-dose etoposide (60 mg/kg). One patient received high-dose cyclophosphamide instead of etoposide, and another patient received both drugs. Nine patients died following BMT, two from relapsed leukemia, and seven from transplant-related causes. The 3-year probabilities of disease-free survival and relapse are 65% and 12%, respectively. For patients transplanted after 1992, these probabilities are 81% (48-95%, 95% confidence interval) and 11% (2-50%), respectively. The relatively low relapse rate in this group of patients compared to published reports may reflect the enhanced anti-leukemic activity of etoposide in combination with FTBI compared to other conditioning regimens. The enhancement in overall survival for patients transplanted after 1992 may reflect improvements in supportive care, in particular, the prophylaxis of serious fungal and viral infections.


Subject(s)
Bone Marrow Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation, Homologous , Adult , Child , Disease-Free Survival , Female , Follow-Up Studies , Fusion Proteins, bcr-abl/genetics , Humans , Male , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Recurrence
12.
Leukemia ; 13(11): 1696-707, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10557041

ABSTRACT

T cell acute lymphocytic leukemia (T-ALL) and B-precursor ALL differ significantly in the clinical characteristics of the patients at presentation and in laboratory-defined characteristics of the leukemic cells. We assessed for pediatric patients with T-ALL the relative importance of prognostic factors previously demonstrated to predict outcome in B-precursor ALL. Presenting clinical and laboratory features were correlated with outcome for 441 children 12 months to 21 years of age with previously untreated T-ALL, registered on the Pediatric Oncology Group (POG) T3 protocol between 1986 and 1992. These T-ALL prognostic factor analyses were then compared to similar analyses for 1993 patients with B-precursor ALL enrolled during the same time period on the POG ALinC 14 protocol. Quantitative interaction between phenotype and each prognostic factor was studied to determine the relative importance of the prognostic factor for each of the two major immunophenotypes. We also analyzed the importance of maturational stage as a T-ALL prognostic factor, using a modified Ludwig definition of maturational stage. We conclude that several of the clinical and laboratory prognostic factors, which are used reliably for B-precursor ALL, are much less predictive in T-ALL (ie age, WBC, consensus risk group, hyperdiploidy, presence of trans- locations and CALLA expression). There was no significant difference between the phenotypes in the prognostic importance of race or gender. Our data demonstrate a significant difference in outcome among the three maturational stages of T-cell ALL, with the intermediate group faring best. Using traditional risk group criteria to stratify patients with T-ALL for therapy may not be appropriate.


Subject(s)
Burkitt Lymphoma/diagnosis , Membrane Glycoproteins , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Adolescent , Adult , Age Factors , Antigens, CD/analysis , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Burkitt Lymphoma/therapy , CD24 Antigen , Central Nervous System/pathology , Child , Child, Preschool , Humans , Immunophenotyping , Infant , Leukocyte Count , Ploidies , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/classification , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prognosis , Racial Groups , Risk Factors , Sex Factors , Survival Rate , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Translocation, Genetic/genetics , Treatment Outcome
13.
Biol Blood Marrow Transplant ; 5(5): 285-91, 1999.
Article in English | MEDLINE | ID: mdl-10534058

ABSTRACT

Graft-vs.-host disease (GVHD) is a major predictor of outcome following allogeneic bone marrow transplantation (BMT). For patients alive at day 100 after BMT, the presence or absence of chronic GVHD is one of the most important determinants of survival and quality of life. We wished to determine the effects on chronic GVHD of two regimens used for the prophylaxis of acute GVHD: cyclosporine, methotrexate, and prednisone (CSA/MTX/PSE) and cyclosporine and prednisone (CSA/PSE). One hundred forty-nine evaluable patients were entered into the acute GVHD study. As of 31 March 1997, 63 months after the last patient underwent BMT, the median survival time was 4.5 years (range 0.09-9.9). The incidence of chronic GVHD was independent of the prophylactic regimen (55 vs. 54%), and extensive chronic GVHD occurred in 25 and 24% of patients receiving CSA/MTX/PSE and CSA/PSE, respectively. Of note, the median Karnofsky performance status of both groups was 100% (range 70-100%), reflecting the low incidence of extensive chronic GVHD. Survival rates free of chronic GVHD were 52 vs. 42% (p = 0.29) for patients receiving CSA/MTX/PSE vs. CSA/PSE. The incidence of relapse was also similar in both groups of patients. These data suggest that the combinations of CSA/MTX/PSE and CSA/PSE result in comparable chronic GVHD-free survival without an increase in leukemic relapse.


Subject(s)
Cyclosporine/therapeutic use , Graft vs Host Disease/prevention & control , Methotrexate/therapeutic use , Prednisone/therapeutic use , Acute Disease , Adolescent , Adult , Anti-Inflammatory Agents/therapeutic use , Bone Marrow Transplantation , Cause of Death , Child , Child, Preschool , Chronic Disease , Communicable Diseases/etiology , Disease-Free Survival , Female , Graft vs Host Disease/mortality , Humans , Immunosuppressive Agents/therapeutic use , Infant , Leukemia/complications , Leukemia/drug therapy , Leukemia/therapy , Longitudinal Studies , Male , Middle Aged , Recurrence , Survival Rate , Transplantation, Homologous
14.
Bone Marrow Transplant ; 23(9): 929-32, 1999 May.
Article in English | MEDLINE | ID: mdl-10338049

ABSTRACT

Intravenous immunoglobulin has been used after bone marrow transplants to prevent infections and acute graft-versus-host disease. However, the minimum dose required for protection is unknown. This may have significant economic implications. A multicenter randomized clinical trial compared the impact of two intravenous immunoglobulin doses on systemic infections and acute graft-versus-host disease in transplant recipients. Either 250 mg/kg or 500 mg/kg was given weekly from day -8 to day +111. Multivariate analysis was used to assess the effect of dose and other risk factors on event-free survival, systemic infection, and acute graft-versus-host disease. The two-dose cohorts had similar event-free survival and infection frequencies. The higher dose was associated with less acute graft-versus-host disease (P = 0.03).


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/prevention & control , Immunoglobulins, Intravenous/administration & dosage , Virus Diseases/prevention & control , Disease-Free Survival , Graft vs Host Disease/etiology , Humans , Incidence , Transplantation, Homologous
15.
Cancer Res ; 59(7): 1492-7, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10197619

ABSTRACT

Methylthioadenosine phosphorylase (MTAP) is an important enzyme for the salvage of adenine and methionine and is deficient in a variety of cancers including T-cell acute lymphocytic leukemia (T-ALL). Previously, we reported that the MTAP gene was deleted in over 30% of T-ALL patients at both diagnosis and relapse. We now report that MTAP-primary T-ALL cells are more sensitive to the toxicity of L-alanosine, an inhibitor of de novo AMP synthesis, than are MTAP+ primary T-ALL cells. As measured by [3H]thymidine incorporation, DNA synthesis in all seven MTAP-primary T-ALL cells was inhibited by L-alanosine with a mean IC50 of 4.8+/-5.3 ILM (range, 0.3-11.3 microM). On the other hand, the IC50 for 60% (12 of 20) of MTAP+ primary T-ALL was 19+/-18 microM (range, 1.7-67 microM; P = 0.02), whereas the remaining 40% (8 of 20) had an IC50 of >80 microM4. Furthermore, normal lymphocytes and MTAP+ primary T-ALL cells were rescued from L-alanosine toxicity by the MTAP substrate 5'-deoxyadenosine, but MTAP-T-ALL cells were not. These results indicate that normal cells, which are intrinsically MTAP+, would be protected from L.-alanosine toxicity, whereas MTAP-tumor cells would be killed. Thus, our results support the use of L-alanosine alone or in combination with a salvage agent as a MTAP-selective therapy and therefore lay the foundation for the initiation of clinical trials for the treatment of T-ALL and other MTAP-deficient malignancies with L-alanosine.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Purine-Nucleoside Phosphorylase/metabolism , Adenosine Monophosphate/biosynthesis , Alanine/analogs & derivatives , Alanine/therapeutic use , Humans , Leukemia-Lymphoma, Adult T-Cell/metabolism , Polymerase Chain Reaction , Purine-Nucleoside Phosphorylase/deficiency , Purine-Nucleoside Phosphorylase/genetics , Thymidine/metabolism
16.
Leukemia ; 13(3): 335-42, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10086723

ABSTRACT

This study was designed to test the hypothesis that high-dose asparaginase consolidation therapy improves survival in pediatric patients with T cell acute lymphoblastic leukemia and advanced stage lymphoblastic lymphoma. Five hundred and fifty-two patients (357 patients with T cell acute lymphoblastic leukemia (ALL) and 195 patients with advanced stage lymphoblastic lymphoma) were enrolled in POG study 8704 (T-3). Treatment included rotating combinations of high-dose myelosuppressive chemotherapy agents proven to be effective in T cell ALL in other POG group-wide or local institutional protocols (including vincristine, doxorubicin, cyclophosphamide, prednisone, asparaginase, teniposide, cytarabine and mercaptopurine). After achieving a complete remission (CR), patients were randomized to receive or not receive high-dose intensive asparaginase consolidation (25,000 IU/m2) given weekly for 20 weeks by intramuscular injection. Intrathecal chemotherapy (methotrexate, hydrocortisone and cytarabine) was given to prevent CNS disease, and CNS irradiation was used only for patients with leukemia and an initial WBC of >50,000/microl or patients with active CNS disease at diagnosis. CR was achieved in 96% of patients. The high-dose asparaginase regimen was significantly superior to the control regimen for both the leukemia and lymphoma subgroups. Four-year continuous complete remission rate (CCR) for the leukemia patients was 68% (s.e. 4%) with asparaginase as compared to 55% (s.e. 4%) without. For the lymphoma patients, 4-year CCR was 78% (s.e. 5%) with asparaginase and 64% (s.e. 6%) in the controls. The overall one-sided logrank test had a P value <0.001 favoring asparaginase, while corresponding values were P = 0.002 for ALL and P = 0.048 lymphoblastic lymphoma. Toxicities were tolerable, but there were 18 failures due to secondary malignancies (16 with non-lymphocytic leukemia or myelodysplasia). Neither WBC at diagnosis (leukemia patients) nor lymphoma stage were major prognostic factors. We conclude that when added to a backbone of effective rotating agents, repeated doses of asparaginase during early treatment improve the outcome for patients with T cell leukemia and advanced stage lymphoblastic lymphoma.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/drug therapy , Lymphoma, T-Cell/drug therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Asparaginase/therapeutic use , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Male , Remission Induction
17.
Int J Cancer Suppl ; 12: 52-7, 1999.
Article in English | MEDLINE | ID: mdl-10679871

ABSTRACT

This descriptive study of health-related quality of life of children with cancer compared children/adolescents', parents' and teachers' ratings for somatization, depression and anxiety to determine if there were significant correlations among respondent scores. In addition, the percentage of agreement among respondents and significant differences based on age, gender, use of cranial radiation and treatment status were measured. Forty-three children/adolescents with cancer, currently receiving therapy for at least 1 year or who had completed therapy for no more than 3 years (excluding children who had received bone marrow transplants or who had brain tumors), were recruited, with a parent and teacher, from 3 university medical centers. The Behavioral Assessment System for Children questionnaires for children/adolescents, parents and teachers were used. Parents reported a higher level of depression for the children/adolescents with cancer than did the teachers or the children/adolescents themselves. Parents reported a higher level of anxiety for the children/adolescents than did the teachers. High positive correlations were found among scores from parents and teachers and among scores from parents and children/adolescents for the anxiety and depression but not somatization subscales. Children/adolescents and teachers had high, positively correlated scores only for the depression subscale. High, positive correlations were found between somatization, anxiety and depression within each group of respondents. A significant percentage of agreement between all respondents on ratings for at-risk status was obtained only for the depression subscale. Age was the only variable found to have an influence on scores and only for the anxiety subscale.


Subject(s)
Anxiety/etiology , Depression/etiology , Health Status , Neoplasms/psychology , Quality of Life , Somatoform Disorders/etiology , Adolescent , Child , Female , Humans , Male
19.
J Clin Oncol ; 16(2): 522-6, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9469336

ABSTRACT

PURPOSE: Contemporary chemotherapy has significantly improved the event-free survival (EFS) among patients with T-cell disease. However, myelosuppression has been a significant adverse effect of this approach. In this study, we assessed the impact of r-metHu granulocyte colony-stimulating factor (G-CSF) on the period of neutropenia, number of days of hospitalization, and delays in subsequent administration of chemotherapy in a cohort of patients with T-cell leukemia (T-ALL) or advanced stage lymphoblastic lymphoma (ASLL). PATIENTS AND METHODS: This open-label, randomized trial incorporated r-metHuG-CSF into the induction and two consecutive continuation-therapy cycles of our intensive program for T-cell malignancies. In the induction phase, r-metHuG-CSF was given after two different combinations of chemotherapy, one of which included vincristine, prednisone, cyclophosphamide, and adriamycin and the other a continuous infusion of high-dose ara-C and L-asparaginase. In the two continuation-therapy cycles, r-metHuG-CSF was given following the combination of vincristine, adriamycin, prednisone, and 6-mercaptopurine (MP) and after continuous infusion of high-dose cytarabine (ara-C). RESULTS: Fifty-six patients with T-ALL and 33 with ASLL were enrolled onto study from April 1994 to December 1995. Our data show no significant difference in number of days of absolute neutrophil count (ANC) less than 500/microL, hospitalizations, or delays in therapy in the induction phase. However, in the continuation-therapy phase the number of days of ANC less than 500/microL was significantly shorter (P = .017) on the G-CSF-arm without significantly affecting the number of days of hospitalizations or delays in therapy. CONCLUSION: r-metHuG-CSF did not significantly affect the period of neutropenia, hospitalization, or delays in therapy in the induction phase, whereas in the two cycles of continuation therapy, it significantly shortened the period of neutropenia.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Leukemia-Lymphoma, Adult T-Cell/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Female , Filgrastim , Humans , Infant , Male , Pilot Projects , Recombinant Proteins
20.
Blood ; 90(2): 578-89, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9226157

ABSTRACT

The relationships between dihydrofolate reductase (DHFR) levels or methotrexate membrane transport and acute lymphoblastic leukemia (ALL) immunophenotype were evaluated on 51 T-cell and 44 B-precursor ALL specimens from 90 pediatric ALL patients at diagnosis and relapse, using a fluorescent methotrexate analog (PT430) and flow cytometry assay (Matherly et al, Blood 85:500, 1995). For T-cell ALL, 35 of 45 (78%) of newly diagnosed patients' specimens exhibited elevated DHFR relative to DHFR levels in ALL blasts from methotrexate-responsive patients. For 30 of 45 diagnostic T-ALL specimens, DHFR expression was heterogeneous, with up to 3 separate subpopulations covering a 44-fold range; the DHFR-overproducing fractions comprised 10% to 88% of the total blasts. Elevated DHFR was less common in B-precursor ALL at diagnosis, being detected in only 17 of 36 specimens (47%); 11 of these samples exhibited DHFR heterogeneity. Median maximal DHFR levels were fourfold higher in T-cell than B-precursor ALL at diagnosis. Within a particular phenotypic group, there were no correlations between DHFR levels and patient prognostic features, including age, sex, chromosomal abnormalities, white blood cell counts (WBCs), and percentage of S-phase. However, for B-precursor ALL, there was a notably higher fraction of African-American than white patients with elevated DHFR. For patients (both phenotypes) with low WBCs (<50,000/ microL), event-free survival times were significantly shorter for those expressing DHFR above a threshold level than for patients expressing DHFR below this level (P < .016); this relationship was not seen for patients with high WBCs (>50,000/microL). Elevated DHFR was detected in 11 of 14 relapse specimens (5 of 6 T-cell and 6 of 8 B-precursor). Two of five paired relapse specimens (both T-cell) from patients treated with methotrexate exhibited increased DHFR levels over those at diagnosis (2.5- to 5-fold); the fraction of DHFR-overproducing blasts was also increased in 4 of 5 paired relapse specimens (2 B-precursor and 2 T-cell). In contrast to the variations in DHFR, highly impaired methotrexate transport was detected in only 6 of 95 ALL specimens, including both diagnosis and relapse. There was no correlation between phenotype and impaired transport. These data provide further rationale for the use of mechanistically based prognostic factors to complement known biologic or disease-based prognostic indicators in the design of ALL therapy including methotrexate, particularly with patients presenting with low WBCs. The finding of a markedly increased frequency of DHFR overexpression in T-cell over B-precursor ALL suggests that this difference is associated with the poorer prognosis of patients with T-cell ALL treated with standard-dose antimetabolite therapy and implies that higher-dose methotrexate (> or = 1 g/m2) during consolidation therapy may be useful in the treatment of this disease.


Subject(s)
ATP-Binding Cassette Transporters , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Burkitt Lymphoma/enzymology , Chromosome Aberrations , Chromosome Disorders , Neoplasm Proteins , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Tetrahydrofolate Dehydrogenase/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adolescent , Blast Crisis , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Carrier Proteins/analysis , Child , Child, Preschool , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Leukocyte Count , Male , Methotrexate/metabolism , Methotrexate/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Recurrence , Tetrahydrofolate Dehydrogenase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...