Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nat Med ; 29(5): 1092-1102, 2023 05.
Article in English | MEDLINE | ID: mdl-37012551

ABSTRACT

Neuroblastomas harbor ALK aberrations clinically resistant to crizotinib yet sensitive pre-clinically to the third-generation ALK inhibitor lorlatinib. We conducted a first-in-child study evaluating lorlatinib with and without chemotherapy in children and adults with relapsed or refractory ALK-driven neuroblastoma. The trial is ongoing, and we report here on three cohorts that have met pre-specified primary endpoints: lorlatinib as a single agent in children (12 months to <18 years); lorlatinib as a single agent in adults (≥18 years); and lorlatinib in combination with topotecan/cyclophosphamide in children (<18 years). Primary endpoints were safety, pharmacokinetics and recommended phase 2 dose (RP2D). Secondary endpoints were response rate and 123I-metaiodobenzylguanidine (MIBG) response. Lorlatinib was evaluated at 45-115 mg/m2/dose in children and 100-150 mg in adults. Common adverse events (AEs) were hypertriglyceridemia (90%), hypercholesterolemia (79%) and weight gain (87%). Neurobehavioral AEs occurred mainly in adults and resolved with dose hold/reduction. The RP2D of lorlatinib with and without chemotherapy in children was 115 mg/m2. The single-agent adult RP2D was 150 mg. The single-agent response rate (complete/partial/minor) for <18 years was 30%; for ≥18 years, 67%; and for chemotherapy combination in <18 years, 63%; and 13 of 27 (48%) responders achieved MIBG complete responses, supporting lorlatinib's rapid translation into active phase 3 trials for patients with newly diagnosed high-risk, ALK-driven neuroblastoma. ClinicalTrials.gov registration: NCT03107988 .


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neuroblastoma , Adult , Humans , 3-Iodobenzylguanidine/therapeutic use , Aminopyridines/therapeutic use , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Lactams, Macrocyclic/adverse effects , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Protein Kinase Inhibitors/therapeutic use , Child , Infant , Child, Preschool , Adolescent
2.
Int J Radiat Oncol Biol Phys ; 115(5): 1115-1128, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36526235

ABSTRACT

PURPOSE: 131I-metaiodobenzylguanidine (131I-MIBG) has demonstrated efficacy as a single agent in neuroblastoma. Recent trials have focused on 131I-MIBG combination strategies, though little is known about the effect of putative radiosensitizers on biological markers of radiation exposure. METHODS AND MATERIALS: NANT2011-01 evaluated 131I-MIBG therapy alone (arm A) or in combination with vincristine/irinotecan (arm B) or vorinostat (arm C) for patients with relapsed or refractory neuroblastoma. Blood samples were collected before and after 131I-MIBG infusion to determine levels of radiation-associated biomarkers (transcript and protein). The association of biomarker with treatment arm, clinical response, and treatment toxicity was analyzed. RESULTS: The cohort included 99 patients who had at least 1 biomarker available for analysis. Significant modulation in most biomarkers between baseline, 72, and 96 hours following 131I-MIBG was observed. Patients in arm C had the lowest degree of modulation in FLT3 ligand protein. Lower baseline BCL2 transcript levels were associated with higher overall response. Patients with greater increases in FLT3 ligand at 96 hours after 131I-MIBG therapy were significantly more likely to have grade 4 thrombocytopenia. Peripheral blood gene expression of the BCL2 family of apoptotic markers (BCL2L1 and BAX transcripts) was significantly associated with grade 4 hematologic toxicity. RNA sequencing demonstrated little overlap in the top modulated peripheral blood transcripts between randomized arms. CONCLUSIONS: Peripheral blood biomarkers relevant to radiation exposure demonstrate significant modulation after 131I-MIBG and concomitant radiation sensitizers affect extent of modulation. Biomarkers related to hematopoietic damage and apoptosis were associated with hematologic toxicity.


Subject(s)
Neuroblastoma , Radiation-Sensitizing Agents , Humans , 3-Iodobenzylguanidine/adverse effects , Radiation-Sensitizing Agents/adverse effects , Neuroblastoma/drug therapy , Neuroblastoma/radiotherapy , Biomarkers , Proto-Oncogene Proteins c-bcl-2
3.
J Clin Oncol ; 41(3): 508-516, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36206505

ABSTRACT

PURPOSE: Although chemoimmunotherapy is widely used for treatment of children with relapsed high-risk neuroblastoma (HRNB), little is known about timing, duration, and evolution of response after irinotecan/temozolomide/dinutuximab/granulocyte-macrophage colony-stimulating factor (I/T/DIN/GM-CSF) therapy. PATIENTS AND METHODS: Patients eligible for this retrospective study were age < 30 years at diagnosis of HRNB and received ≥ 1 cycle of I/T/DIN/GM-CSF for relapsed or progressive disease. Patients with primary refractory disease who progressed through induction were excluded. Responses were evaluated using the International Neuroblastoma Response Criteria. RESULTS: One hundred forty-six patients were included. Tumors were MYCN-amplified in 50 of 134 (37%). Seventy-one patients (49%) had an objective response to I/T/DIN/GM-CSF (objective response; 29% complete response, 14% partial response [PR], 5% minor response [MR], 21% stable disease [SD], and 30% progressive disease). Of patients with SD or better at first post-I/T/DIN/GM-CSF disease evaluation, 22% had an improved response per International Neuroblastoma Response Criteria on subsequent evaluation (13% of patients with initial SD, 33% with MR, and 41% with PR). Patients received a median of 4.5 (range, 1-31) cycles. The median progression-free survival (PFS) was 13.1 months, and the 1-year PFS and 2-year PFS were 50% and 28%, respectively. The median duration of response was 15.9 months; the median PFS off all anticancer therapy was 10.4 months after discontinuation of I/T/DIN/GM-CSF. CONCLUSION: Approximately half of patients receiving I/T/DIN/GM-CSF for relapsed HRNB had objective responses. Patients with initial SD were unlikely to have an objective response, but > 1 of 3 patients with MR/PR on first evaluation ultimately had complete response. I/T/DIN/GM-CSF was associated with extended PFS in responders both during and after discontinuation of treatment. This study establishes a new comparator for response and survival in patients with relapsed HRNB.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Neuroblastoma , Child , Humans , Adult , Progression-Free Survival , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Irinotecan/therapeutic use , Temozolomide/therapeutic use , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neuroblastoma/pathology
4.
J Clin Oncol ; 40(33): 3839-3847, 2022 11 20.
Article in English | MEDLINE | ID: mdl-35820112

ABSTRACT

PURPOSE: Metastatic retinoblastoma has a poor prognosis when treated with conventional chemotherapy and radiation therapy (RT). Intensified therapy may improve the outcome. METHODS: A prospective, international trial enrolled patients with extraocular retinoblastoma. Patients with stage II or III (locoregional) retinoblastoma received four cycles of chemotherapy, followed by involved field RT (45 Gy). Patients with stage IVa or IVb (metastatic or trilateral) retinoblastoma also received four cycles of chemotherapy and those with ≥ partial response then received one cycle of high-dose carboplatin, thiotepa, and etoposide with autologous hematopoietic stem-cell support. Patients with stage IVa or IVb with residual tumor postchemotherapy received RT. The proportion of patients who achieved event-free survival would be reported and compared with historical controls separately for each of the three groups of patients. RESULTS: Fifty-seven eligible patients were included in the analyses. Event-free survival at 1 year was 88.1% (90% CI, 66.6 to 96.2) for stage II-III, 82.6% (90% CI, 61.0 to 92.9) for stage IVa, and 28.3% (90% CI, 12.7 to 46.2) for stage IVb/trilateral. Toxicity was significant as expected and included two therapy-related deaths. CONCLUSION: Intensive multimodality therapy is highly effective for patients with regional extraocular retinoblastoma and stage IVa metastatic retinoblastoma. Although the study met its aim for stage IVb, more effective therapy is still required for patients with CNS involvement (ClinicalTrials.gov identifier: NCT00554788).


Subject(s)
Retinal Neoplasms , Retinoblastoma , Child , Humans , Combined Modality Therapy/adverse effects , Prospective Studies , Retinal Neoplasms/therapy , Retinal Neoplasms/pathology , Retinoblastoma/therapy , Retinoblastoma/pathology
5.
JCO Clin Cancer Inform ; 5: 1181-1188, 2021 12.
Article in English | MEDLINE | ID: mdl-34882497

ABSTRACT

PURPOSE: Metaiodobenzylguanidine (MIBG) scans are a radionucleotide imaging modality that undergo Curie scoring to semiquantitatively assess neuroblastoma burden, which can be used as a marker of therapy response. We hypothesized that a convolutional neural network (CNN) could be developed that uses diagnostic MIBG scans to predict response to induction chemotherapy. METHODS: We analyzed MIBG scans housed in the International Neuroblastoma Risk Group Data Commons from patients enrolled in the Children's Oncology Group high-risk neuroblastoma study ANBL12P1. The primary outcome was response to upfront chemotherapy, defined as a Curie score ≤ 2 after four cycles of induction chemotherapy. We derived and validated a CNN using two-dimensional whole-body MIBG scans from diagnosis and evaluated model performance using area under the receiver operating characteristic curve (AUC). We also developed a clinical classification model to predict response on the basis of age, stage, and MYCN amplification. RESULTS: Among 103 patients with high-risk neuroblastoma included in the final cohort, 67 (65%) were responders. Performance in predicting response to upfront chemotherapy was equivalent using the CNN and the clinical model. Class-activation heatmaps verified that the CNN used areas of disease within the MIBG scans to make predictions. Furthermore, integrating predictions using a geometric mean approach improved detection of responders to upfront chemotherapy (geometric mean AUC 0.73 v CNN AUC 0.63, P < .05; v clinical model AUC 0.65, P < .05). CONCLUSION: We demonstrate feasibility in using machine learning of diagnostic MIBG scans to predict response to induction chemotherapy for patients with high-risk neuroblastoma. We highlight improvements when clinical risk factors are also integrated, laying the foundation for using a multimodal approach to guiding treatment decisions for patients with high-risk neuroblastoma.


Subject(s)
3-Iodobenzylguanidine , Neuroblastoma , 3-Iodobenzylguanidine/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Neuroblastoma/diagnostic imaging , Neuroblastoma/drug therapy , Radionuclide Imaging , Research Report
6.
J Nucl Med Technol ; 48(1): 81-84, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31604895

ABSTRACT

131I-metaiodobenzylguanidine (131I-MIBG) is a theranostic agent useful for treatment of neuroendocrine malignancies. In this case, a child with a Curie score of 21 was administered 17.871 GBq (483 mCi) of 131I-MIBG. The elimination half-life progressively increased from 23 h to 77 h during the 11 d that the patient was hospitalized for radiation isolation. Six weeks after the posttherapy scan, a survey with an ion-chamber device yielded readings of 0.3 µSv/h (0.03 mR/h) on contact with spinal regions that had shown increased uptake on the scan. A planar image obtained using the 131I setting and a high-energy collimator did not demonstrate any focal uptake. 123I-MIBG was administered, and the 24-h scan was of diagnostic quality, without degradation from the remaining 131I-MIBG. Additional study is needed on whether the Curie score affects elimination of 131I-MIBG and on whether the period of hospitalized radiation isolation needs to be extended.


Subject(s)
Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Neuroendocrine Tumors/radiotherapy , Radiopharmaceuticals/pharmacokinetics , Whole Body Imaging/methods , Child , Female , Humans , Iodine Radioisotopes/urine , Radionuclide Imaging/methods , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/urine
7.
Clin Cancer Res ; 23(18): 5374-5383, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28559462

ABSTRACT

Purpose: We determined whether quantifying neuroblastoma-associated mRNAs (NB-mRNAs) in bone marrow and blood improves assessment of disease and prediction of disease progression in patients with relapsed/refractory neuroblastoma.Experimental Design: mRNA for CHGA, DCX, DDC, PHOX2B, and TH was quantified in bone marrow and blood from 101 patients concurrently with clinical disease evaluations. Correlation between NB-mRNA (delta cycle threshold, ΔCt, for the geometric mean of genes from the TaqMan Low Density Array NB5 assay) and morphologically defined tumor cell percentage in bone marrow, 123I-meta-iodobenzylguanidine (MIBG) Curie score, and CT/MRI-defined tumor longest diameter was determined. Time-dependent covariate Cox regression was used to analyze the relationship between ΔCt and progression-free survival (PFS).Results: NB-mRNA was detectable in 83% of bone marrow (185/223) and 63% (89/142) of blood specimens, and their ΔCt values were correlated (Spearman r = 0.67, P < 0.0001), although bone marrow Ct was 7.9 ± 0.5 Ct stronger than blood Ct When bone marrow morphology, MIBG, or CT/MRI were positive, NB-mRNA was detected in 99% (99/100), 88% (100/113), and 81% (82/101) of bone marrow samples. When all three were negative, NB-mRNA was detected in 55% (11/20) of bone marrow samples. Bone marrow NB-mRNA correlated with bone marrow morphology or MIBG positivity (P < 0.0001 and P = 0.007). Bone marrow and blood ΔCt values correlated with PFS (P < 0.001; P = 0.001) even when bone marrow was morphologically negative (P = 0.001; P = 0.014). Multivariate analysis showed that bone marrow and blood ΔCt values were associated with PFS independently of clinical disease and MYCN gene status (P < 0.001; P = 0.055).Conclusions: This five-gene NB5 assay for NB-mRNA improves definition of disease status and correlates independently with PFS in relapsed/refractory neuroblastoma. Clin Cancer Res; 23(18); 5374-83. ©2017 AACR.


Subject(s)
Biomarkers, Tumor , Bone Marrow/metabolism , Bone Marrow/pathology , Gene Expression , Neuroblastoma/diagnosis , Neuroblastoma/genetics , Biopsy , Child , Child, Preschool , Drug Resistance, Neoplasm , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Neuroblastoma/mortality , Neuroblastoma/therapy , Prognosis , Recurrence , Survival Analysis , Tomography, X-Ray Computed
8.
J Pediatr Oncol Nurs ; 34(3): 160-172, 2017.
Article in English | MEDLINE | ID: mdl-28061552

ABSTRACT

Neuroblastoma, an embryonic cancer of the sympathetic nervous system, is the most common extracranial solid tumor in childhood. Dinutuximab (formerly called ch14.18), a monoclonal antibody targeting the disialoganglioside GD2, has been shown to significantly improve survival rates in patients with high-risk neuroblastoma. However, the safe and effective use of dinutuximab therapy in these high-risk patients requires medical expertise in patient selection, treatment administration, and the monitoring and management of adverse events. Findings of the randomized phase III study (ANBL0032) led to the approval of dinutuximab for the treatment of children with high-risk neuroblastoma. Multi-institutional nursing approaches to implementing standard protocols ensure the effective management of high-risk neuroblastoma patients receiving dinutuximab immunotherapy. Understanding and implementing recommendations for the management of the clinically important and most common adverse events are essential to ensuring patient continuation of therapy and improving patient outcomes.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Maintenance Chemotherapy/nursing , Neuroblastoma/drug therapy , Neuroblastoma/nursing , Nurse's Role , Child , Child, Preschool , Clinical Trials, Phase III as Topic , Humans
9.
PLoS Genet ; 12(12): e1006501, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27997549

ABSTRACT

Neuroblastoma is characterized by a relative paucity of recurrent somatic mutations at diagnosis. However, recent studies have shown that the mutational burden increases at relapse, likely as a result of clonal evolution of mutation-carrying cells during primary treatment. To inform the development of personalized therapies, we sought to further define the frequency of potentially actionable mutations in neuroblastoma, both at diagnosis and after chemotherapy. We performed a retrospective study to determine mutation frequency, the only inclusion criterion being availability of cancer gene panel sequencing data from Foundation Medicine. We analyzed 151 neuroblastoma tumor samples: 44 obtained at diagnosis, 42 at second look surgery or biopsy for stable disease after chemotherapy, and 59 at relapse (6 were obtained at unknown time points). Nine patients had multiple tumor biopsies. ALK was the most commonly mutated gene in this cohort, and we observed a higher frequency of suspected oncogenic ALK mutations in relapsed disease than at diagnosis. Patients with relapsed disease had, on average, a greater number of mutations reported to be recurrent in cancer, and a greater number of mutations in genes that are potentially targetable with available therapeutics. We also observed an enrichment of reported recurrent RAS/MAPK pathway mutations in tumors obtained after chemotherapy. Our data support recent evidence suggesting that neuroblastomas undergo substantial mutational evolution during therapy, and that relapsed disease is more likely to be driven by a targetable oncogenic pathway, highlighting that it is critical to base treatment decisions on the molecular profile of the tumor at the time of treatment. However, it will be necessary to conduct prospective clinical trials that match sequencing results to targeted therapeutic intervention to determine if cancer genomic profiling improves patient outcomes.


Subject(s)
Clonal Evolution/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adolescent , Adult , Aged , Anaplastic Lymphoma Kinase , Child , Child, Preschool , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , MAP Kinase Signaling System/genetics , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Neuroblastoma/surgery , Retrospective Studies , ras Proteins/genetics
10.
J Nucl Med Technol ; 42(2): 109-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24799606

ABSTRACT

UNLABELLED: Syringe pumps are commonly used to administer therapeutic (131)I-metaiodobenzylguanidine. Here we describe our recent experience with a peristaltic infusion pump system in a pediatric setting. This method can easily accommodate infusions from several vials simultaneously and is adaptable to various types of peristaltic pump. METHODS: Simple off-the-shelf components are used to vent the vial: a charcoal filter, a 0.22-µm syringe filter, and a 2.54-cm (1-in) needle. The vial is connected to the primary infusion set using a male/male extension line and a 19-gauge × 8.89-cm (3.5-in) aspirating needle. With aseptic technique, the extension line is attached to the Y connector closest to the primary intravenous line leading from the saline reservoir to the infusion pump. An A-clamp is attached to the primary intravenous line, immediately before the entrance to the pump. Gravity is allowed to clear the air from the extension set and the aspirating needle. After all the air has been purged, the aspirating needle is inserted into the therapy vial using aseptic technique. The pump is programmed with the desired infusion rate and volume to be infused. RESULTS: Twenty-one consecutive infusions have been performed to date using this method. Most of the infusions involved the use of 1 vial. On 7 occasions, 2 or 3 vials connected in series were used to successfully administer the therapy. Overestimation of the volume in the vials or of the total infusion time required can cause air to be pulled into the lines. To prevent this, the volume in the vials is equalized to 30 mL, facilitating calculation of the infusion time. If the infusion is observed over the last 2 or 3 mL and the pump stops when the air-fluid mark is about halfway up the extension set, air will be kept out of the primary infusion set. CONCLUSION: This method for infusing one or more vials of therapeutic radiopharmaceuticals is robust and easy to use. During infusion, the radiopharmaceutical remains in a shielded vial. Multiple vials can be connected in series to infuse the entire dose simultaneously.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , Administration, Intravenous/instrumentation , Infusion Pumps , Asepsis , Infusion Pumps/microbiology
11.
Pediatr Blood Cancer ; 60(5): 879-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23024113

ABSTRACT

BACKGROUND: (131) I-metaiodobenzylguanidine (MIBG) produces a 37% response rate in relapsed/refractory neuroblastoma, and could be used to improve remission status prior to myeloablative chemotherapy with autologous stem cell transplant (ASCT). The purpose of our report was to evaluate safety and response with MIBG therapy followed by myeloablative busulfan and melphalan (BuMel) with ASCT in patients with refractory neuroblastoma. METHODS: Retrospective chart review was done on patients treated with MIBG (18 mCi/kg) on Day 1 and ASCT on day 14. Six to eight weeks after MIBG, patients without progressive disease received IV busulfan on days -6 to -2 (target Css 700-900), melphalan (140 mg/m2 IV) on day -1, and ASCT on Day 0. Response and toxicity were evaluated after MIBG and again after myeloablative therapy. RESULTS: Eight patients completed MIBG/ASCT followed by BuMel/ASCT. MIBG was well tolerated, with grade 3 or 4 non-hematologic toxicity limited to one patient with sepsis. Grade 3 mucositis occurred in six patients after BuMel/ASCT. One patient developed sinusoidal obstructive syndrome (SOS) and died 50 days post-ASCT following myeloablative conditioning. All patients engrafted neutrophils (median 16.5 days) and platelets (median 32 days) after BuMel, excluding the patient with SOS. After all therapy, there were three complete, two partial, and one minor response in seven evaluable patients. CONCLUSIONS: MIBG at doses up to 18 mCi/kg can be safely administered 6 weeks prior to a BuMel consolidative regimen for refractory neuroblastoma. Preceding MIBG did not impair engraftment following BuMel. This regimen is being further evaluated in a Children's Oncology Group (COG) trial.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/therapy , Stem Cell Transplantation , 3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/adverse effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Busulfan/administration & dosage , Busulfan/adverse effects , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Neuroblastoma/drug therapy , Neuroblastoma/radiotherapy , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/therapeutic use , Retrospective Studies , Transplantation, Autologous
12.
Pediatr Blood Cancer ; 59(5): 902-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22744917

ABSTRACT

BACKGROUND: The Pediatric Oncology Group performed a pilot study to assess the feasibility of tandem high dose chemotherapy (HDC) with stem cell rescue (HDC/SCR). We report here the results of this single arm trial of induction chemotherapy, local control measures (surgery and local radiation), and tandem HDC/SCR. PROCEDURE: Patients with high risk neuroblastoma (NBL) underwent five cycles of induction chemotherapy and resection of primary tumors. Peripheral blood stem cells (PBSC) were collected after Course 3 without exvivo manipulation. Myeloablative chemotherapy was performed in rapid sequence after induction chemotherapy and surgery. The ability of patients to complete both cycles of HDC/SCR was a primary endpoint. Transplant-related toxicity, progression-free survival (PFS) and overall survival (OS) were recorded. RESULTS: A total of 33 patients were enrolled. Twenty-two patients completed at least one HDC/SCR procedure and 17 patients completed both. Only one patient had insufficient stem cells collected for both transplants. There was one transplant-related death; engraftment was rapid and toxicity was as expected. The PFS of the 33 patients treated on this study is 24.2% ± 7.5% and OS is 36.4% ± 8.4% at 5 years. For patients who received at least one transplant PFS is 36.4% ± 11.0% and OS is 45.5% ± 11.2% at 5 years. CONCLUSIONS: The treatment of high risk NBL with tandem HDC/SCR is feasible in terms of transplant-related mortality and the ability to collect adequate PBSC for two transplants. The outcomes from this intensified treatment have been used to design a Children's Oncology Group Phase III study testing the efficacy of tandem HDC/SCR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neuroblastoma/mortality , Neuroblastoma/therapy , Peripheral Blood Stem Cell Transplantation , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Disease-Free Survival , Humans , Infant , Male , Survival Rate , Transplantation, Autologous
13.
J Pediatr Hematol Oncol ; 32(5): e167-72, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20606544

ABSTRACT

To longitudinally assess serum concentrations of rituximab, it was administered intravenously to 25 children with opsoclonus-myoclonus syndrome at 375 mg/m2 on each of 4 consecutive weeks with (Group I and II) or without (Group III) conventional immunotherapy. Serum rituximab levels, drawn before and after each infusion and at later intervals, were analyzed by enzyme-linked immunosorbent assay. Rituximab concentration increased stepwise with each infusion, dropping by the next infusion, thereby forming 4 discrete peaks (Cmax) and troughs (Cmin). It then fell precipitously to trace levels at 4 months. However, Cmax and Cmin curves differed significantly between groups. Compared with the youngest children (Group I), the oldest (Group III) had a 34% lower rituximab concentration at the fourth infusion, 45% less IgM depletion 1 month later, and received 20% less rituximab when the dose was recalculated as mg/kg. Serum IgM and rituximab levels were negatively correlated. Peak rituximab concentration did not correlate with adrenocorticotropic hormone dose. These results indicate that the degree of serum IgM depletion is a useful indicator for rituximab dose equivalency in children of different ages. They also suggest that pediatric rituximab dosing should be based on body weight, not surface area. (ClinicalTrials.gov NCT00244361).


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Immunotherapy , Opsoclonus-Myoclonus Syndrome/metabolism , Adolescent , Adrenocorticotropic Hormone/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/blood , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin M/blood , Immunoglobulins, Intravenous/administration & dosage , Infant , Male , Opsoclonus-Myoclonus Syndrome/immunology , Prospective Studies , Rituximab , Treatment Outcome
14.
Crit Rev Oncol Hematol ; 41(1): 29-40, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11796230

ABSTRACT

The telomere-telomerase hypothesis is the science of cellular aging (senescence) and cancer. The ends of chromosomes, telomeres, count the number of divisions a cell can undergo before entering permanent growth arrest. As divisions are being counted, events occur on the cellular and molecular level, which may either delay or hasten this arrest. As humans age, a particular concern is the accumulation of events that lead to the progression of cancer. Telomerase is a mechanism that most normal cells do not possess, but almost all cancer cells acquire, to overcome their mortality and extend their lifespan. This review aims to provide a comprehensive understanding of the role of telomerase in cancer development, progression, diagnosis, and in the future, treatment. The ultimate goal of telomerase research is to use our understanding to develop anti-telomerase therapies, an almost universal tumor target.


Subject(s)
Aging/physiology , Neoplasms/enzymology , Telomerase/physiology , Telomere/physiology , Animals , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...