Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Clin Oncol ; 41(17): 3149-3159, 2023 06 10.
Article in English | MEDLINE | ID: mdl-37098238

ABSTRACT

PURPOSE: In 2006, Children's Oncology Group (COG) reclassified subgroups of toddlers diagnosed with neuroblastoma from high-risk to intermediate-risk, when the age cutoff for high-risk assignment was raised from 365 days (12 months) to 547 days (18 months). The primary aim of this retrospective study was to determine if excellent outcome was maintained after assigned reduction of therapy. PATIENTS AND METHODS: Children <3 years old at diagnosis, enrolled on a COG biology study from 1990 to 2018, were eligible (n = 9,189). Assigned therapy was reduced for two cohorts of interest on the basis of the age cutoff change: 365-546 days old with International Neuroblastoma Staging System (INSS) stage 4, MYCN not amplified (MYCN-NA), favorable International Neuroblastoma Pathology Classification (INPC), hyperdiploid tumors (12-18mo/Stage4/FavBiology), and 365-546 days old with INSS stage 3, MYCN-NA, and unfavorable INPC tumors (12-18mo/Stage3/MYCN-NA/Unfav). Log-rank tests compared event-free survival (EFS) and overall survival (OS) curves. RESULTS: For 12-18mo/Stage4/FavBiology, 5-year EFS/OS (± SE) before (≤2006; n = 40) versus after (>2006; n = 55) assigned reduction in therapy was similar: 89% ± 5.1%/89% ± 5.1% versus 87% ± 4.6%/94% ± 3.2% (P = .7; P = .4, respectively). For 12-18mo/Stage3/MYCN-NA/Unfav, the 5-year EFS and OS were both 100%, before (n = 6) and after (n = 4) 2006. The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as high-risk ≤2006 had an EFS/OS of 91% ± 4.4%/91% ± 4.5% versus 38% ± 1.3%/43% ± 1.3% for all other high-risk patients <3 years old (P < .0001; P < .0001, respectively). The 12-18mo/Stage4/FavBiology plus 12-18mo/Stage3/MYCN-NA/Unfav classified as intermediate-risk >2006 had an EFS/OS of 88% ± 4.3%/95% ± 2.9% versus 88% ± 0.9%/95% ± 0.6% for all other intermediate-risk patients <3 years old (P = .87; P = .85, respectively). CONCLUSION: Excellent outcome was maintained among subsets of toddlers with neuroblastoma assigned to reduced treatment after reclassification of risk group from high to intermediate on the basis of new age cutoffs. Importantly, as documented in prior trials, intermediate-risk therapy is not associated with the degree of acute toxicity and late effects commonly observed with high-risk regimens.


Subject(s)
Neuroblastoma , Humans , Infant , Child, Preschool , Prognosis , N-Myc Proto-Oncogene Protein/genetics , Retrospective Studies , Disease-Free Survival , Neoplasm Staging , Neuroblastoma/pathology , Risk Assessment
2.
Pediatr Blood Cancer ; 66(7): e27736, 2019 07.
Article in English | MEDLINE | ID: mdl-30968542

ABSTRACT

PURPOSE: Neuroblastoma is the most common extracranial solid pediatric malignancy, with poor outcomes in high-risk disease. Standard treatment approaches employ an increasing array of aggressive multimodal therapies, of which local control with surgery and radiotherapy remains a backbone; however, the benefit of broad regional nodal irradiation remains controversial. We analyzed centrally reviewed radiation therapy data from patients enrolled on COG A3973 to evaluate the impact of primary site irradiation and the extent of regional nodal coverage stratified by extent of surgical resection. METHODS: Three hundred thirty high-risk neuroblastoma patients with centrally reviewed radiotherapy plans were analyzed. Outcome was evaluated by the extent of nodal irradiation. For the 171 patients who also underwent surgery (centrally reviewed), outcome was likewise analyzed according to the extent of resection. Overall survival (OS), event-free survival (EFS), and cumulative incidence of local progression (CILP) were examined by Kaplan-Meier, log-rank test (EFS, OS), and Grey test (CILP). RESULTS: The five-year CILP, EFS, and OS for all 330 patients receiving radiotherapy on A3973 were 8.5% ± 1.5%, 47.2% ± 3.0%, and 59.7% ± 3.0%, respectively. There were no significant differences in outcomes based on the extent of lymph node irradiation regardless of the degree of surgical resection (< 90% or ≥90%). CONCLUSION: Although local control remains a significant component of treatment of high-risk neuroblastoma, our results suggest there is no benefit of extensive lymph node irradiation, irrespective of the extent of surgical resection preceding stem cell transplant.


Subject(s)
Lymph Nodes , Neuroblastoma , Disease-Free Survival , Female , Humans , Infant , Infant, Newborn , Male , Neuroblastoma/mortality , Neuroblastoma/radiotherapy , Survival Rate
3.
Pediatr Blood Cancer ; 62(4): 629-36, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25704135

ABSTRACT

BACKGROUND: Juvenile myelomonocytic leukemia (JMML) is not durably responsive to chemotherapy, and approximately 50% of patients relapse after hematopoietic stem cell transplant (HSCT). Here we report the activity and acute toxicity of the farnesyl transferase inhibitor tipifarnib, the response rate to 13-cis retinoic acid (CRA) in combination with cytoreductive chemotherapy, and survival following HSCT in children with JMML. PROCEDURE: Eighty-five patients with newly diagnosed JMML were enrolled on AAML0122 between 2001 and 2006. Forty-seven consented to receive tipifarnib in a phase II window before proceeding to a phase III trial of CRA in combination with fludarabine and cytarabine followed by HSCT and maintenance CRA. Thirty-eight patients enrolled only in the phase III trial. RESULTS: Overall response rate was 51% after tipifarnib and 68% after fludarabine/cytarabine/CRA. Tipifarnib did not increase pre-transplant toxicities. Forty-six percent of the 44 patients who received protocol compliant HSCT relapsed. Five-year overall survival was 55 ± 11% and event-free survival was 41 ± 11%, with no significant difference between patients who did or did not receive tipifarnib. CONCLUSIONS: Administration of tipifarnib in the window setting followed by HSCT in patients with newly diagnosed JMML was safe and yielded a 51% initial response rate as a single agent, but failed to reduce relapse rates or improve long-term overall survival.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Leukemia, Myelomonocytic, Juvenile/drug therapy , Quinolones/administration & dosage , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Cytarabine/administration & dosage , Disease-Free Survival , Enzyme Inhibitors/administration & dosage , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Isotretinoin/administration & dosage , Leukemia, Myelomonocytic, Juvenile/enzymology , Leukemia, Myelomonocytic, Juvenile/mortality , Leukemia, Myelomonocytic, Juvenile/pathology , Male , Middle Aged , Survival Rate , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
5.
J Clin Oncol ; 30(15): 1842-8, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22529259

ABSTRACT

PURPOSE: The primary objective of Children's Oncology Group study P9641 was to demonstrate that surgery alone would achieve 3-year overall survival (OS) ≥ 95% for patients with asymptomatic International Neuroblastoma Staging System stages 2a and 2b neuroblastoma (NBL). Secondary objectives focused on other low-risk patients with NBL and on those who required chemotherapy according to protocol-defined criteria. PATIENTS AND METHODS: Patients underwent maximally safe resection of tumor. Chemotherapy was reserved for patients with, or at risk for, symptomatic disease, with less than 50% tumor resection at diagnosis, or with unresectable progressive disease after surgery alone. RESULTS: For all 915 eligible patients, 5-year event-free survival (EFS) and OS were 89% ± 1% and 97% ± 1%, respectively. For patients with asymptomatic stage 2a or 2b disease, 5-year EFS and OS were 87% ± 2% and 96% ± 1%, respectively. Among patients with stage 2b disease, EFS and OS were significantly lower for those with unfavorable histology or diploid tumors, and OS was significantly lower for those ≥ 18 months old. For patients with stage 1 and 4s NBL, 5-year OS rates were 99% ± 1% and 91% ± 1%, respectively. Patients who required chemotherapy at diagnosis achieved 5-year OS of 98% ± 1%. Of all patients observed after surgery, 11.1% experienced recurrence or progression of disease. CONCLUSION: Excellent survival rates can be achieved in asymptomatic low-risk patients with stages 2a and 2b NBL after surgery alone. Immediate use of chemotherapy may be restricted to a minority of patients with low-risk NBL. Patients with stage 2b disease who are older or have diploid or unfavorable histology tumors fare less well. Future studies will seek to refine risk classification.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/drug therapy , Neuroblastoma/surgery , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Child , Child, Preschool , Disease Progression , Disease-Free Survival , Female , Humans , Infant , Kaplan-Meier Estimate , Male , Neoplasm Staging , Neuroblastoma/mortality , Neuroblastoma/pathology , Patient Selection , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , United States , Young Adult
6.
N Engl J Med ; 363(14): 1313-23, 2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20879880

ABSTRACT

BACKGROUND: The survival rate among patients with intermediate-risk neuroblastoma who receive dose-intensive chemotherapy is excellent, but the survival rate among patients who receive reduced doses of chemotherapy for shorter periods of time is not known. METHODS: We conducted a prospective, phase 3, nonrandomized trial to determine whether a 3-year estimated overall survival of more than 90% could be maintained with reductions in the duration of therapy and drug doses, using a tumor biology-based therapy assignment. Eligible patients had newly diagnosed, intermediate-risk neuroblastoma without MYCN amplification; these patients included infants (<365 days of age) who had stage 3 or 4 disease, children (≥365 days of age) who had stage 3 tumors with favorable histopathological features, and infants who had stage 4S disease with a diploid DNA index or unfavorable histopathological features. Patients who had disease with favorable histopathological features and hyperdiploidy were assigned to four cycles of chemotherapy, and those with an incomplete response or either unfavorable feature were assigned to eight cycles. RESULTS: Between 1997 and 2005, a total of 479 eligible patients were enrolled in this trial (270 patients with stage 3 disease, 178 with stage 4 disease, and 31 with stage 4S disease). A total of 323 patients had tumors with favorable biologic features, and 141 had tumors with unfavorable biologic features. Ploidy, but not histopathological features, was significantly predictive of the outcome. Severe adverse events without disease progression occurred in 10 patients (2.1%), including secondary leukemia (in 3 patients), death from infection (in 3 patients), and death at surgery (in 4 patients). The 3-year estimate (±SE) of overall survival for the entire group was 96±1%, with an overall survival rate of 98±1% among patients who had tumors with favorable biologic features and 93±2% among patients who had tumors with unfavorable biologic features. CONCLUSIONS: A very high rate of survival among patients with intermediate-risk neuroblastoma was achieved with a biologically based treatment assignment involving a substantially reduced duration of chemotherapy and reduced doses of chemotherapeutic agents as compared with the regimens used in earlier trials. These data provide support for further reduction in chemotherapy with more refined risk stratification. (Funded by the National Cancer Institute; ClinicalTrials.gov number, NCT00003093.)


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neuroblastoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Humans , Infant , Intention to Treat Analysis , Neoplasm Staging , Neuroblastoma/mortality , Neuroblastoma/pathology , Prospective Studies , Survival Analysis , Treatment Outcome
7.
J Clin Oncol ; 28(24): 3808-15, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20660830

ABSTRACT

PURPOSE: Single-agent topotecan (TOPO) and combination topotecan and cyclophosphamide (TOPO/CTX) were compared in a phase II randomized trial in relapsed/refractory neuroblastoma. Because responders often underwent further therapies, novel statistical methods were required to compare the long-term outcome of the two treatments. PATIENTS AND METHODS: Children with refractory/recurrent neuroblastoma (only one prior aggressive chemotherapy regimen) were randomly assigned to daily 5-day topotecan (2 mg/m(2)) or combination topotecan (0.75 mg/m(2)) and cyclophosphamide (250 mg/m(2)). A randomized two-stage group sequential design enrolled 119 eligible patients. Toxicity and response were estimated. Long-term outcome of protocol therapy was assessed using novel methods-causal inference-which allowed adjustment for the confounding effect of off-study therapies. RESULTS: Seven more responses were observed for TOPO/CTX (complete response [CR] plus partial response [PR], 18 [32%] of 57) than TOPO (CR+PR, 11 [19%] of 59;P = .081); toxicity was similar. At 3 years, progression-free survival (PFS) and overall survival (OS) were 4% +/- 2% and 15% +/- 4%, respectively. PFS was significantly better for TOPO/CTX (P = .029); there was no difference in OS. Older age at diagnosis and lack of MYCN amplification predicted increased OS (P < .05). Adjusting for randomized treatment effect and subsequent autologous stem-cell transplantation, there was no difference between TOPO and TOPO/CTX in terms of the proportion alive at 2 years. CONCLUSION: TOPO/CTX was superior to TOPO in terms of PFS, but there was no OS difference. After adjustment for subsequent therapies, no difference was detected in the proportion alive at 2 years. Causal inference methods for assessing long-term outcomes of phase II therapies after subsequent treatment can elucidate effects of initial therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclophosphamide/administration & dosage , Models, Statistical , Neuroblastoma/drug therapy , Topotecan/administration & dosage , Adolescent , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Causality , Child , Child, Preschool , Confounding Factors, Epidemiologic , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Infant , Kaplan-Meier Estimate , Male , Prognosis , Recurrence , Risk Factors , Treatment Outcome , Young Adult
8.
Pediatr Blood Cancer ; 54(1): 103-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19774634

ABSTRACT

INTRODUCTION: Approximately 80% of children currently survive 5 years following diagnosis of their cancer. Studies based on limited data have implicated certain cancer therapies in the development of ocular sequelae in these survivors. PROCEDURE: The Childhood Cancer Survivor Study (CCSS) is a retrospective cohort study investigating health outcomes of 5+ year survivors diagnosed and treated between 1970 and 1986 compared to a sibling cohort. The baseline questionnaire included questions about the first occurrence of six ocular conditions. Relative risks (RR) and 95% confidence intervals (CI) were calculated from responses of 14,362 survivors and 3,901 siblings. RESULTS: Five or more years from the diagnosis, survivors were at increased risk of cataracts (RR: 10.8; 95% CI: 6.2-18.9), glaucoma (RR: 2.5; 95% CI: 1.1-5.7), legal blindness (RR: 2.6; 95% CI: 1.7-4.0), double vision (RR: 4.1; 95% CI: 2.7-6.1), and dry eyes (RR: 1.9; 95% CI: 1.6-2.4), when compared to siblings. Dose of radiation to the eye was significantly associated with risk of cataracts, legal blindness, double vision, and dry eyes, in a dose-dependent manner. Risk of cataracts were also associated with radiation 3,000+ cGy to the posterior fossa (RR: 8.4; 95% CI: 5.0-14.3), temporal lobe (RR: 9.4; 95% CI: 5.6-15.6), and exposure to prednisone (RR: 2.3; 95% CI: 1.6-3.4). CONCLUSIONS: Childhood cancer survivors are at risk of developing late occurring ocular complications, with exposure to glucocorticoids and cranial radiation being important determinants of increased risk. Long-term follow-up is needed to evaluate potential progression of ocular deficits and impact on quality of life.


Subject(s)
Eye Diseases/etiology , Neoplasms/complications , Adolescent , Adult , Child , Child, Preschool , Eye/drug effects , Eye Diseases/diagnosis , Female , Glucocorticoids/adverse effects , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neoplasms/mortality , Neoplasms/therapy , Neoplasms, Radiation-Induced/etiology , Prognosis , Radiation Injuries/diagnosis , Radiation Injuries/etiology , Retrospective Studies , Surveys and Questionnaires , Survival Rate , Survivors , Young Adult
9.
Leuk Res ; 33(5): 671-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19010541

ABSTRACT

The biological hallmark of juvenile myelomonocytic leukemia (JMML) is selective GM-CSF hypersensitivity. We hypothesized that PTEN protein deficiency might lead to insufficient negative growth signals to counter the hyperactive Ras signaling and therefore aid in the acceleration of the malignant transformation of JMML. In screening 34 JMML patients we found: (1) decreased PTEN protein in 67% of patients; (2) significantly lower PTEN mRNA levels in patients compared to controls (p<0.01); (3) a hypermethylated PTEN promoter in 77% of patients; and (4) constitutive-hyperactive Akt and MAPK in 55% and 73% of patients, respectively. These findings suggest that PTEN deficiency is very common in JMML and is in part due to hypermethylation of the PTEN gene promoter.


Subject(s)
Leukemia, Myelomonocytic, Juvenile/genetics , PTEN Phosphohydrolase/genetics , Base Sequence , Blotting, Western , DNA Methylation , DNA Primers , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Promoter Regions, Genetic , Protein Kinases/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , Transcription, Genetic
10.
Pediatr Blood Cancer ; 51(6): 747-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18704922

ABSTRACT

BACKGROUND: From 1993 to 1995, the Pediatric Oncology Group (POG) enrolled patients with high-risk neuroblastoma on three sequential, conjoined studies: a phase II induction window (9340), followed by intensive multiagent induction chemotherapy (9341), and subsequent myeloablative therapy with autologous stem cell rescue (9342). We report here the outcomes of patients treated on these studies. PATIENTS AND METHODS: Patients were between 1 and 21 years old with high-risk neuroblastoma. Phase II window therapy consisted of two courses of either paclitaxel, topotecan, or cyclophosphamide with topotecan. Induction therapy consisted of at least five cycles of intensive chemotherapy, followed by myeloablative therapy with purged autologous stem cell reinfusion. Patient responses, treatment toxicities, and overall and event-free survival rates were calculated. RESULTS: Eighty-four percent of patients responded to induction chemotherapy, with 39% achieving complete response. Toxicities were primarily hematologic. The 7-year EFS and OS rates for all eligible patients on POG 9341 were 23 +/- 4% and 28 +/- 4%, respectively. The 7-year EFS and OS rates for patients treated on POG 9342 were 27 +/- 6% and 29 +/- 6%, respectively. CONCLUSIONS: These studies were the first attempt by POG to use autologous stem cell transplantation for neuroblastoma treatment in a cooperative group setting. Toxicities and outcomes were comparable to contemporary cooperative group studies. The phase II induction window had no detectable effect on outcomes. New strategies are needed to improve survival for this devastating disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Neuroblastoma/therapy , Adolescent , Adult , Carboplatin/administration & dosage , Child , Child, Preschool , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Disease-Free Survival , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Female , Humans , Ifosfamide/administration & dosage , Infant , Male , Neuroblastoma/drug therapy , Paclitaxel/administration & dosage , Survival Rate , Topotecan/administration & dosage , Transplantation Conditioning , Treatment Outcome , Vincristine/administration & dosage , Young Adult
11.
J Clin Oncol ; 26(6): 913-8, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-18281664

ABSTRACT

PURPOSE: MYCN amplification is rarely detected in patients with favorable-stage neuroblastoma (NB). To determine the clinical significance of MYCN amplification in children with favorable-stage NB, we performed a retrospective review of data from the Pediatric Oncology Group (POG) biology study 9047. PATIENTS AND METHODS: MYCN status, tumor cell ploidy, treatment, and outcome of patients with stage A, B, or Ds NB, enrolled on POG 9047 between 1990 and 1999 were analyzed. Event-free survival (EFS) and overall (OS) survival rates were analyzed using the Kaplan-Meier method. RESULTS: Of the 1,667 patients enrolled on POG 9047, 643 had favorable-stage disease. Of these, follow-up data were available on 568 (34%) with stage A, B, or Ds disease and normal MYCN copy number, and 32 (1.9%) patients with MYCN-amplified, stage A, B, or Ds tumors. Within the cohort lacking MYCN amplification, the 7-year EFS and OS rates (+/- SE) were 91% +/- 1% and 96% +/- 1%, respectively. Patients with MYCN amplification had significantly worse EFS and OS (50% +/- 9% and 59% +/- 9%, respectively, P < .0001). Within the cohort of children with MYCN amplification, the 7-year EFS and OS rates were 80% +/- 10% and 87% +/- 9%, respectively for patients with hyperdiploid tumors and 25% +/- 11% and 38% +/- 12% for patients with diploid/hypodiploid NBs (P = .0063 and P = .0074, respectively). CONCLUSION: Tumor cell ploidy may be a clinically useful factor for prognostication and treatment stratification in children with MYCN-amplified, favorable-stage NB tumors.


Subject(s)
Gene Amplification , Neuroblastoma/genetics , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Ploidies , Adolescent , Child , Child, Preschool , Diploidy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , N-Myc Proto-Oncogene Protein , Neoplasm Staging , Neuroblastoma/therapy , Predictive Value of Tests , Prognosis , Retrospective Studies , Salvage Therapy , Treatment Outcome , United States
12.
Blood ; 111(3): 1124-7, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18000165

ABSTRACT

Juvenile myelomonocytic leukemia is an aggressive and frequently lethal myeloproliferative disorder of childhood. Somatic mutations in NRAS, KRAS, or PTPN11 occur in 60% of cases. Monitoring disease status is difficult because of the lack of characteristic leukemic blasts at diagnosis. We designed a fluorescently based, allele-specific polymerase chain reaction assay called TaqMAMA to detect the most common RAS or PTPN11 mutations. We analyzed peripheral blood and/or bone marrow of 25 patients for levels of mutant alleles over time. Analysis of pre-hematopoietic stem-cell transplantation, samples revealed a broad distribution of the quantity of the mutant alleles. After hematopoietic stem-cell transplantation, the level of the mutant allele rose rapidly in patients who relapsed and correlated well with falling donor chimerism. Simultaneously analyzed peripheral blood and bone marrow samples demonstrate that blood can be monitored for residual disease. Importantly, these assays provide a sensitive strategy to evaluate molecular responses to new therapeutic strategies.


Subject(s)
Alleles , Leukemia, Myelomonocytic, Juvenile/diagnosis , Leukemia, Myelomonocytic, Juvenile/genetics , Polymerase Chain Reaction/methods , Bone Marrow/metabolism , Child , Chimerism , Female , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myelomonocytic, Juvenile/surgery , Male , Recurrence , Sensitivity and Specificity
13.
Pediatr Blood Cancer ; 49(7 Suppl): 1060-5, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17943963

ABSTRACT

Neuroblastoma is the most common malignancy in infants and 40% of neuroblastomas are diagnosed in the first year of life. While generally neuroblastoma behaves less aggressively in this age group, tumors that have adverse biologic characteristics do not differ in their behavior from counterparts in older children. Clinical and biologic behavior of neuroblastoma in children up to 460 days of age is similar to that in children less than 1 year of age. Thus the categorization of children up to 18 months of age into risk category is critically dependent on biologic characterization and assignment to appropriate treatment intensity categories.


Subject(s)
Biomedical Research/trends , Neuroblastoma/therapy , Humans , Infant , Infant, Newborn , Neoplasm Staging , Neuroblastoma/pathology , Prognosis , Risk Factors
14.
J Clin Oncol ; 24(18): 2879-84, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16782927

ABSTRACT

PURPOSE: The INT-0098 Intergroup Liver Tumor Study demonstrated no statistically significant differences in event-free and overall survival between patients randomized to treatment with either cisplatin + fluorouracil + vincristine (C5V) or cisplatin + doxorubicin. Results from this and other therapeutic trials suggested that cisplatin was the most active agent against hepatoblastoma. To increase the platinum dose-intensity, a novel regimen was developed alternating carboplatin and cisplatin (CC) every 2 weeks. The P9645 study was designed to compare the risk of treatment failure for patients with stage III/IV hepatoblastoma randomized to either C5V or CC. METHODS: C5V was given according to INT-0098 and CC consisted of carboplatin at 700 mg/m2 on day 0 (560 mg/m2 after two cycles) followed by cisplatin 100 mg/m2 on day 14. Granulocyte colony-stimulating factor was used after each CC cycle. All patients received four to six cycles of chemotherapy. RESULTS: From the time the study was opened until the time that random assignment was halted, 56 patients received CC and 53 patients received C5V. The 1-year event-free survival was 37% for patients receiving CC and 57% for those receiving C5V (P = .017). Patients randomly assigned to CC required more blood product support. As a result of a semiannual review by the Children's Oncology Group Data and Safety Monitoring Committee, random assignment was discontinued after 3 years of enrollment because the projected improvement in long-term outcome associated with CC was statistically excluded as a possible outcome of this trial. CONCLUSION: Intensification of therapy by alternating platinum analogs increased the risk of adverse outcome in children with unresectable or metastatic hepatoblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Cisplatin/administration & dosage , Hepatoblastoma/drug therapy , Liver Neoplasms/drug therapy , Adolescent , Child , Child, Preschool , Female , Fluorouracil/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Infant , Male , Vincristine/administration & dosage
15.
J Clin Oncol ; 24(16): 2544-8, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16735707

ABSTRACT

PURPOSE: To investigate prognostic factors for pediatric extragonadal malignant germ cell tumors (PEMGCT). MATERIALS AND METHODS: Between 1990 and 1996, patients with stage I through IV PEMGCT were eligible for a trial of cisplatin dose intensity. We retrospectively investigated prognostic factors for PEMGCT, including age, stage, primary site, treatment, and elevated alfa fetoprotein by univariate and multivariate analysis. RESULTS: The 165 patients had a median age of 1.9 years (range, 3 days to 18.5 years); 109 were female; and 99 had alfa fetoprotein > or = 10,000. There were 30 stage I/II, 61 stage III, and 74 stage IV tumors; primary sites included 88 sacrococcygeal, 39 thoracic, and 38 others. The 5-year overall survival (OS) and event-free survival (EFS) rates with standard deviations were 83.4% +/- 3.7% and 79.0% +/- 4.1%, respectively. Univariate analysis identified age > or = 12 years as a highly significant prognostic factor for EFS (5-year EFS, 48.9% +/- 15.6% v 84.1% +/- 3.9%; P < .0001) and for OS (5-year OS, 53.7% +/- 14.9% v 88.5% +/- 3.4%; P < .0001), whereas treatment was of borderline significance (P = .0777). Multivariate Cox proportional hazards regression identified only age > or = 12 years as a significant prognostic factor for EFS (P = .0002). In multivariate Cox regression for OS, the combination of age and primary site was highly significant (P < .0001). Patients > or = 12 years of age with thoracic tumors had six times the risk of death compared with patients younger than 12 years with other primaries. CONCLUSION: Age is the most predictive factor of EFS in PEMGCT. There is a significant interaction between age and primary site, suggesting that patients > or = 12 years of age with thoracic tumors are a biologically distinct group.


Subject(s)
Germinoma/diagnosis , Adolescent , Age Factors , Analysis of Variance , Biomarkers, Tumor/blood , Child , Disease-Free Survival , Female , Germinoma/blood , Germinoma/pathology , Germinoma/therapy , Humans , Male , Neoplasm Staging , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Survival Analysis , alpha-Fetoproteins/metabolism
16.
J Clin Oncol ; 23(34): 8819-27, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16314642

ABSTRACT

PURPOSE: The goal of Pediatric Oncology Group 9243 was to improve outcomes for children with intermediate-risk neuroblastoma (NB). PATIENTS AND METHODS: Patients were assigned to treatments on the basis of age, tumor MYCN status, and tumor cell ploidy. Children in the less intensive arm A received cyclophosphamide/doxorubicin and surgery. Patients not in complete remission postoperatively were treated with cisplatin/etoposide, cyclophosphamide/doxorubicin, and additional surgery. Patients with less favorable features were assigned to arm B, which consisted of carboplatin, etoposide, ifosfamide, and surgery. Survival rates were determined using an intent-to-treat approach. RESULTS: For arm-A patients, the 6-year event-free survival (EFS) was 86% with an SE of 3%. For arm-B patients, the 6-year EFS was 46% with an SE of 7%. MYCN status was the only statistically significant prognostic variable. Among patients whose tumors were MYCN nonamplified, a trend toward improved EFS was seen in children with hyperdiploid versus diploid tumors. However, many of these children responded well to salvage therapy, and overall survival rates did not differ on the basis of ploidy. Six-year EFS rates for arm B were patients with MYCN nonamplified, hyperdiploid tumors, 86% with an SE of 3%; patients with MYCN nonamplified, diploid tumors, 74% with an SE of 10%; patients with MYCN-amplified, hyperdiploid tumors, 46% with an SE of 15%; and patients with MYCN-amplified, diploid tumors, 22% with an SE of 10%. CONCLUSION: Outcomes for patients with MYCN-nonamplified, hyperdiploid tumors were excellent. Therapy reductions for these patients merit study. A trend toward less favorable outcomes for patients with MYCN-nonamplified, diploid tumors was observed; more children may need to be evaluated before therapy is reduced for this subgroup. For patients with MYCN-amplified tumors, new strategies are needed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/drug therapy , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Ploidies , Abdominal Neoplasms/drug therapy , Abdominal Neoplasms/genetics , Abdominal Neoplasms/pathology , Adolescent , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/administration & dosage , Carboplatin/adverse effects , Child , Child, Preschool , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Follow-Up Studies , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Ifosfamide/administration & dosage , Ifosfamide/adverse effects , Infant , Male , N-Myc Proto-Oncogene Protein , Neoplasm Staging , Neuroblastoma/genetics , Neuroblastoma/pathology , Neutropenia/chemically induced , Prognosis , Risk Factors , Treatment Outcome
17.
J Clin Oncol ; 23(27): 6499-507, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16170159

ABSTRACT

PURPOSE: To determine the influence of race/ethnicity on outcomes in the Childhood Cancer Survivor Study (CCSS). PATIENTS AND METHODS: Of CCSS adult survivors in the United States, 443 (4.9%) were black, 503 (5.6%) were Hispanic and 7,821 (86.6%) were white. Mean age at interview, 26.9 years (range, 18 to 48 years); mean follow-up, 17.2 years (range, 8.7 to 28.4 years). Late mortality, second malignancy (SMN) rates, health care utilization, and health status and behaviors were assessed for blacks and Hispanics and compared with white survivors. RESULTS: Late mortality rate (6.5%) and 15-year cumulative incidence of SMN (3.5%) were similar across racial/ethnic groups. Minority survivors were more likely to have lower socioeconomic status (SES); final models were adjusted for income, education, and health insurance. Although overall health status was similar, black survivors were less likely to report adverse mental health (females: odds ratio [OR], 0.6; 95% CI, 0.4 to 0.9; males: OR, 0.5; 95% CI, 0.3 to 0.8). Differences in health care utilization and behaviors noted: Hispanic survivors were more likely to report a cancer center visit (females: OR, 1.5; 95% CI, 1.1 to 2.0; males: OR, 1.7; 95% CI, 1.2 to 2.3); black females were more likely (OR, 1.6; 95% CI, 1.1 to 2.4), and Hispanic females less likely to have a recent Pap smear (OR, 0.7; 95% CI, 0.5 to 1.0); black and Hispanic survivors were less likely to report smoking; black survivors were less likely to report problem drinking. CONCLUSION: Adjusted for SES, adverse outcomes in CCSS were not associated with minority status. Importantly, black survivors reported less risky behaviors and better preventive practices. Hispanic survivors had equitable access to cancer related care.


Subject(s)
Attitude to Health/ethnology , Ethnicity/statistics & numerical data , Neoplasms/ethnology , Neoplasms/mortality , Adolescent , Adult , Age Distribution , Black People/statistics & numerical data , Child , Child, Preschool , Cohort Studies , Confidence Intervals , Female , Hispanic or Latino/statistics & numerical data , Humans , Infant , Male , Middle Aged , Minority Groups/statistics & numerical data , Neoplasms/diagnosis , Neoplasms/therapy , Odds Ratio , Patient Compliance , Retrospective Studies , Risk Factors , Sex Distribution , Socioeconomic Factors , Survival Rate , Survivors , United States , White People/statistics & numerical data
18.
J Clin Oncol ; 23(27): 6466-73, 2005 Sep 20.
Article in English | MEDLINE | ID: mdl-16116152

ABSTRACT

PURPOSE: To determine predictive strength of tumor cell ploidy and MYCN gene amplification on survival of children older than 12 months with disseminated neuroblastoma (NB). PATIENTS AND METHODS: Of 648 children with stage D NB enrolled onto the Pediatric Oncology Group NB Biology Study 9047 (1990-2000), 560 children were assessable for ploidy and MYCN amplification. Treatment of patients older than 12 months varied; most receiving high-dose chemotherapy with stem-cell rescue. Infants received standard chemotherapy, depending on MYCN status and ploidy. RESULTS: Among stage D MYCN-amplified patients, 4-year event-free survival (EFS) +/- SE had no prognostic significance for tumor cell ploidy for patients either younger than 12 months or > or = 12 months old. However, among stage D nonamplified-MYCN patients, 4-year EFS for those with tumor hyperdiploidy (DNA index [DI] > 1) was clearly superior to those with diploidy (DI < or = 1): younger than 12 months, 83.7% +/- 4.4% (n = 87) versus 46.2% +/- 13.8% (n = 13; P = .0003); and for 12- to 24-month-old children, 72.7% +/- 10.2% (n = 22) versus 26.7% +/- 13.2% (n = 16; P = .0092). Further analysis suggested better prognoses in the 12- to 18-month-old subgroup with hyperdiploid tumors (4-year EFS, 92.9% +/- 7.2%) compared with the 19- to 24-month-old subgroup (4-year EFS, 37.5% +/- 21.0%; P = .0037). In children older than 24 months, outcome was dire (< 20% long-term survival), regardless of ploidy or MYCN status. CONCLUSION: Children 12 to 18 months old with metastatic NB had favorable outcomes with high-dose therapy if their tumors were hyperdiploid and lacked MYCN amplification. This subgroup may respond well to contemporary chemotherapy, and could be spared intensive myeloablative therapy with stem-cell rescue.


Subject(s)
Neoplasm Invasiveness/pathology , Neuroblastoma/genetics , Neuroblastoma/mortality , Proto-Oncogene Proteins c-myc/metabolism , Age Factors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child, Preschool , Diploidy , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genes, myc/genetics , Genetic Markers/genetics , Humans , Infant , Male , Neoplasm Staging , Neuroblastoma/pathology , Neuroblastoma/therapy , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Risk Assessment , Stem Cell Transplantation , Survival Analysis
19.
Cancer Lett ; 228(1-2): 257-66, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16024170

ABSTRACT

Recent evidence suggests that the cut-off for age utilized in neuroblastoma risk groups should be increased from the 365-day cut-off currently in use. Separate cooperative group analyses were performed by German and Italian groups and two analyses by the Children's Oncology Group (North America, Australia, New Zealand, Switzerland, Netherlands). In general, the results are in agreement regarding the prognostic contribution of age. There is strong evidence to support an increase in the age cut-off to a value in the range of 15-18 months based on the results from the German analysis and two COG analyses. However, Italian results in INSS stage 4 patients show that outcome in patients 12-17 months is not better than that of older patients. Further analyses are warrented.


Subject(s)
Age Factors , Neuroblastoma/epidemiology , Child , Germany/epidemiology , Humans , Italy/epidemiology , Medical Oncology , Risk Factors
20.
Blood ; 106(6): 2183-5, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-15928039

ABSTRACT

Germ line PTPN11 mutations cause 50% of cases of Noonan syndrome (NS). Somatic mutations in PTPN11 occur in 35% of patients with de novo, nonsyndromic juvenile myelomonocytic leukemia (JMML). Myeloproliferative disorders (MPDs), either transient or more fulminant forms, can also occur in infants with NS (NS/MPD). We identified PTPN11 mutations in blood or bone marrow specimens from 77 newly reported patients with JMML (n = 69) or NS/MPD (n = 8). Together with previous reports, we compared the spectrum of PTPN11 mutations in 3 groups: (1) patients with JMML (n = 107); (2) patients with NS/MPD (n = 19); and (3) patients with NS (n = 243). Glu76 was the most commonly affected residue in JMML (n = 45), with the Glu76Lys alteration (n = 29) being most frequent. Eight of 19 patients with NS/MPD carried the Thr73Ile substitution. These data suggest that there is a genotype/phenotype correlation in the spectrum of PTPN11 mutations found in patients with JMML, NS/MPD, and NS. This supports the need to characterize the spectrum of hematologic abnormalities in individuals with NS and to better define the impact of the PTPN11 lesion on the disease course in patients with NS/MPD and JMML.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Myeloproliferative Disorders/genetics , Noonan Syndrome/genetics , Protein Tyrosine Phosphatases/genetics , Child , DNA Mutational Analysis , Humans , Mutation, Missense , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11
SELECTION OF CITATIONS
SEARCH DETAIL
...