Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Cell ; 171(5): 1042-1056.e10, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29056344

ABSTRACT

We present an extensive assessment of mutation burden through sequencing analysis of >81,000 tumors from pediatric and adult patients, including tumors with hypermutation caused by chemotherapy, carcinogens, or germline alterations. Hypermutation was detected in tumor types not previously associated with high mutation burden. Replication repair deficiency was a major contributing factor. We uncovered new driver mutations in the replication-repair-associated DNA polymerases and a distinct impact of microsatellite instability and replication repair deficiency on the scale of mutation load. Unbiased clustering, based on mutational context, revealed clinically relevant subgroups regardless of the tumors' tissue of origin, highlighting similarities in evolutionary dynamics leading to hypermutation. Mutagens, such as UV light, were implicated in unexpected cancers, including sarcomas and lung tumors. The order of mutational signatures identified previous treatment and germline replication repair deficiency, which improved management of patients and families. These data will inform tumor classification, genetic testing, and clinical trial design.


Subject(s)
Neoplasms/genetics , Adult , Child , Cluster Analysis , DNA Polymerase II/genetics , DNA Polymerase III/genetics , DNA Replication , Humans , Mutation , Neoplasms/classification , Neoplasms/pathology , Neoplasms/therapy , Poly-ADP-Ribose Binding Proteins/genetics
2.
Pediatr Blood Cancer ; 71(7): e31051, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38706187

ABSTRACT

It is not clear whether trial access disparities exist in the Children's Oncology Group (COG). Here, we leverage a cohort of children with high-risk neuroblastoma (HR-NBL) enrolled on the COG ANBL00B1 neuroblastoma biology study to examine subsequent enrollment to upfront COG therapeutic trials by race, ethnicity, and proxied poverty status. Among 1917 children with HR-NBL enrolled on ANBL00B1, 696 (36.3%) subsequently enrolled on an upfront therapeutic trial with no difference by race, ethnicity, or proxied poverty status. In neuroblastoma, trial access disparities are not comparable to adult oncology, and efforts to advance equity should prioritize other mechanisms of survival disparities.


Subject(s)
Neuroblastoma , Poverty , Humans , Neuroblastoma/therapy , Neuroblastoma/ethnology , Male , Female , Child , Child, Preschool , Infant , Ethnicity/statistics & numerical data , Clinical Trials as Topic/statistics & numerical data , Healthcare Disparities , Adolescent , Follow-Up Studies
3.
Pediatr Blood Cancer ; 70(6): e30286, 2023 06.
Article in English | MEDLINE | ID: mdl-36975166

ABSTRACT

PURPOSE: To determine whether extent of surgical resection of the primary tumor correlates with survival in patients with International Neuroblastoma Staging System (INSS) stage 4, high-risk neuroblastoma. METHODS: Data were extracted for patients with newly diagnosed INSS stage 4, high-risk neuroblastoma between 2001 and 2019 from the national Cancer in Young People in Canada (CYPC) database. Complete resection was defined as gross total resection of primary tumor based on operative reports. Primary endpoints were 3 and 5-year event-free (EFS) and overall survival (OS). Survival analyses were completed using log-rank test and Cox proportional hazards regression including covariates of age, sex, decade of treatment (2001-2009 vs. 2010-2019), immunotherapy, and tandem stem cell transplant (SCT). RESULTS: One-hundred and forty patients with complete surgical data were included. On univariate analysis, 3-year EFS and OS for patients that had complete versus incomplete resection was 71% (95% CI 57-80%) vs. 48% (36-60%) and 86% (75-93%) vs. 64% (51-74%), p = .008 and p = .002, respectively. 5-year EFS and OS for patients with complete resection also demonstrated significantly improved survival. On Cox Proportional Hazards models adjusted for age, immunotherapy, tandem SCT, and surgical resection, only complete resection was associated with statistically significant improved 3 year EFS and OS, HR = 0.48 (0.29-0.81; p = .006) and HR = 0.42 (0.24-0.73; p = .002). CONCLUSIONS: In a large Canadian INSS stage 4 high-risk neuroblastoma cohort, complete surgical resection was associated with increased EFS and OS. Within the constraints of a retrospective study, these results suggest that the ability to achieve primary tumor complete resection in patients with metastatic high-risk disease is associated with improved survival.


Subject(s)
Neuroblastoma , Humans , Infant , Adolescent , Retrospective Studies , Neoplasm Staging , Canada , Survival Analysis , Neuroblastoma/pathology , Disease-Free Survival
4.
Pediatr Blood Cancer ; 70 Suppl 6: e30572, 2023 09.
Article in English | MEDLINE | ID: mdl-37458162

ABSTRACT

Neuroblastoma is the most common extra-cranial solid tumor in children and is known for its clinical heterogeneity. A greater understanding of the biology of this disease has led to both improved risk stratification and new approaches to therapy. Outcomes for children with low and intermediate risk disease are excellent overall, and efforts to decrease therapy for such patients have been largely successful. Although survival has improved over time for patients with high-risk disease and treatments evaluated in the relapse setting are now being moved into earlier phases of treatment, much work remains to improve survival and decrease therapy-related toxicities. Studies of highly annotated biobanked samples continue to lead to important insights regarding neuroblastoma biology. Such studies, along with correlative biology studies incorporated into therapeutic trials, are expected to continue to provide insights that lead to new and more effective therapies. A focus on translational science is accompanied by an emphasis on new agent development, optimized risk stratification, and international collaboration to address questions relevant to molecularly defined subsets of patients. In addition, the COG Neuroblastoma Committee is committed to addressing the patient/family experience, mitigating late effects of therapy, and studying social determinants of health in patients with neuroblastoma.


Subject(s)
Neuroblastoma , Child , Humans , Neuroblastoma/pathology , Medical Oncology , Disease Progression
5.
Microvasc Res ; 143: 104397, 2022 09.
Article in English | MEDLINE | ID: mdl-35671835

ABSTRACT

Mammalian target of rapamycin (mTOR) inhibitors are clinically effective at treating some complex lymphatic malformations (LM). The mTOR inhibitor rapamycin blocks the phosphoinositide 3-kinase (PI3K) pathway, which is commonly mutated in this condition. Although rapamycin is effective at controlling symptoms of LM, treatment courses are long, not all LMs respond to treatment, and many patients relapse after treatment has stopped. Concurrent rat sarcoma virus (RAS) pathway abnormalities have been identified in LM, which may limit the effectiveness of rapamycin. Protein tyrosine phosphatase-2 (SHP2) controls the RAS pathway upstream, and SHP2 inhibitors are being investigated for treatment of various tumors. The objective of this study was to determine the impact of SHP2 inhibition in combination with rapamycin on LM growth in vitro. Using primary patient cells isolated from a surgically resected LM, we found that combination treatment with rapamycin and the SHP2 inhibitor SHP099 caused a synergistic reduction in cell growth, migration and lymphangiogenesis. These results suggest that combination treatment targeting the PI3K and RAS signaling pathways may result in effective treatment of LMs of the head and neck.


Subject(s)
Endothelial Cells , Phosphatidylinositol 3-Kinases , Phosphatidylinositol 3-Kinases/pharmacology , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
6.
Environ Health ; 21(1): 30, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35255910

ABSTRACT

BACKGROUND: The aetiology of neuroblastic tumours is likely to involve both genetic and environmental factors. A number of possible environmental risk factors have been suggested, including infection. If an irregular temporal pattern in incidence is found, this might suggest that a transient agent, such as an infection, is implicated. Previous work has found evidence for temporal clustering in children and young adults living in northern England. METHODS: We examined data from a second population-based registry from Ontario, Canada to determine whether there was evidence of temporal clustering of neuroblastic tumours. Cases diagnosed in children and young adults aged 0-19 years between 1985 and 2016 were extracted from the population-based Pediatric Oncology Group of Ontario Networked Information System (POGONIS). A modified version of the Potthoff-Whittinghill method was used to test for temporal clustering. Estimates of extra-Poisson variation (EPV) and standard errors (SE) were obtained. RESULTS: Eight hundred seventy-six cases of neuroblastic tumours were diagnosed during the study period. Overall, no evidence of temporal clustering was found between fortnights, between months or between quarters within years. However, significant EPV was found between years within the full study period (EPV = 1.05, SE = 0.25; P = 0.005). CONCLUSIONS: The findings are consistent with the possibility that a transient agent, such as an infection that is characterised by 'peaks and troughs' in its occurrence, might be implicated in the aetiology of neuroblastic tumours. However, this pattern may also reflect a long-term increase in the numbers of cases, rather than peaks and troughs.


Subject(s)
Neoplasms , Child , Cluster Analysis , England/epidemiology , Humans , Incidence , Infant , Neoplasms/epidemiology , Ontario/epidemiology , Young Adult
7.
Lancet Oncol ; 22(12): 1764-1776, 2021 12.
Article in English | MEDLINE | ID: mdl-34780709

ABSTRACT

BACKGROUND: Several paediatric malignancies, including anaplastic large cell lymphoma (ALCL), inflammatory myofibroblastic tumour (IMT), neuroblastoma, and rhabdomyosarcoma, harbour activation of anaplastic lymphoma kinase (ALK) through different mechanisms. Here, we report the safety, pharmacokinetics, and efficacy of ceritinib in paediatric patients with ALK-positive malignancies. METHODS: This multicentre, open-label, phase 1 trial was done at 23 academic hospitals in ten countries. Children (aged ≥12 months to <18 years) diagnosed with locally advanced or metastatic ALK-positive malignancies that had progressed despite standard therapy, or for which no effective standard therapy were available, were eligible. ALK-positive malignancies were defined as those with ALK rearrangement, amplification, point mutation, or in the case of rhabdomyosarcoma, expression in the absence of any genetic alteration. Eligible patients had evaluable or measurable disease as defined by either Response Evaluation Criteria in Solid Tumours, version 1.1 for patients with non-haematological malignancies, International Neuroblastoma Response Criteria scan for patients with neuroblastoma, or International Working Group criteria for patients with lymphoma. Other eligibility criteria were Karnofsky performance status score of at least 60% for patients older than 12 years or Lansky score of at least 50% for patients aged 12 years or younger. This study included a dose-escalation part, followed by a dose-expansion part, in which all patients received treatment at the recommended dose for expansion (RDE) established in the dose-escalation part. Both parts of the study were done in fasted and fed states. In the dose-escalation part, patients were treated with once-daily ceritinib orally, with dose adjusted for body-surface area, rounded to the nearest multiple of the 50 mg dose strength. The starting dose in the fasted state was 300 mg/m2 daily and for the fed state was 320 mg/m2 daily. The primary objective of this study was to establish the maximum tolerated dose (ie, RDE) of ceritinib in the fasted and fed states. The RDE was established on the basis of the incidence of dose-limiting toxicities in patients who completed a minimum of 21 days of treatment with safety assessments and at least 75% drug exposure, or who discontinued treatment earlier because of dose-limiting toxicity. Overall response rate (defined as the proportion of patients with a best overall response of complete response or partial response) was a secondary endpoint. Activity and safety analyses were done in all patients who received at least one dose of ceritinib. This trial is registered with ClinicalTrials.gov (NCT01742286) and is completed. FINDINGS: Between Aug 28, 2013, and Oct 17, 2017, 83 children with ALK-positive malignancies were enrolled to the dose-escalation (n=40) and dose-expansion (n=43) groups. The RDE of ceritinib was established as 510 mg/m2 (fasted) and 500 mg/m2 (fed). 55 patients (30 with neuroblastoma, ten with IMT, eight with ALCL, and seven with other tumour types) were treated with ceritinib at the RDE (13 patients at 510 mg/m2 fasted and 42 patients at 500 mg/m2 fed). The median follow-up was 33·3 months (IQR 24·8-39·3) for patients with neuroblastoma, 33·2 months (27·9-35·9) for those with IMT, 34·0 months (21·9-46·4) for those with ALCL, and 27·5 months (22·4-36·9) for patients with other tumour types. An overall response was recorded in six (20%; 95% CI 8-39) of 30 patients with neuroblastoma, seven (70%; 33-93) of ten patients with IMT, six (75%; 35-97) of eight patients with ALCL, and one (14%; <1-58) of seven patients with other tumours. The safety profile of ceritinib was consistent with that observed in adult patients. All patients had at least one adverse event. Grade 3 or 4 adverse events occurred in 67 (81%) of 83 patients and were mostly increases in aminotransferases (alanine aminotransferase increase in 38 [46%] patients and aspartate aminotransferase increase in 27 [33%] patients). At least one serious adverse event was reported in 40 (48%) of 83 patients and 31 (37%) of 83 patients had at least one grade 3 or 4 serious adverse event. 14 (17%) deaths occurred during the study, of which 12 were on-treatment deaths and two were after 30 days of the last dose. Of the 12 on-treatment deaths, ten were due to disease progression (neuroblastoma), one due to sepsis, and one due to intractable hypotension. INTERPRETATION: Ceritinib 500 mg/m2 once daily with food is the recommended dose for paediatric patients with ALK-positive malignancies. Ceritinib showed promising preliminary antitumour activity in patients with ALK-positive refractory or recurrent IMT or ALCL, and in a subset of patients with relapsed or refractory neuroblastoma, with a manageable safety profile. Our data support the notion that ALK inhibitors should be considered in therapeutic strategies for paediatric patients with malignancies with genetic ALK alterations. FUNDING: Novartis Pharmaceutical Corporation.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Neoplasms/drug therapy , Pyrimidines/therapeutic use , Sulfones/therapeutic use , Adolescent , Anaplastic Lymphoma Kinase/genetics , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Male , Maximum Tolerated Dose , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Survival Rate
8.
Pediatr Blood Cancer ; 68(12): e29289, 2021 12.
Article in English | MEDLINE | ID: mdl-34411405

ABSTRACT

BACKGROUND: Meta-iodobenzylguanidine(MIBG) scans are used to detect neuroblastoma metastatic lesions at diagnosis and during posttreatment surveillance. MIBG positivity following induction chemotherapy correlates with poor outcome; however, there are reports of patients with progression-free survival despite MIBG positivity at the end of therapy. The factors distinguishing these survivors from patients who progress or relapse are unclear. FDG-positron-emission tomography (PET) scans can also detect metastatic lesions at diagnosis; however, their role in posttherapy surveillance is less well studied. METHODS: We performed a retrospective analysis of International Neuroblastoma Staging System (INSS) stage 4 patients to identify those with residual MIBG-avid metastatic lesions on end-of-therapy scans without prior progression. Data collected included age, disease sites, histopathology, biomarkers, treatment, imaging studies, and response. RESULTS: Eleven of 265 patients met inclusion criteria. At diagnosis three of 11 patients were classified as intermediate and eight of 11 high risk; nine of 11 had documented marrow involvement. Histologic classification was favorable for four of 10 and MYCN amplification was detected in zero of 11 cases. The median time with persistent MIBG positivity following treatment was 1.5 years. Seven patients had at least one PET scan with low or background activity. Biopsies of three of three MIBG-avid residual lesions showed differentiation. All patients remain alive with no disease progression at a median of 4.0 years since end of therapy. CONCLUSION: Persistently MIBG-avid metastatic lesions in subsets of patients following completion of therapy may not represent active disease that will progress. Further studies are needed to determine whether MYCN status or other biomarkers, and/or PET scans, may help identify patients with residual inactive MIBG lesions who require no further therapy.


Subject(s)
Neoplasms, Second Primary , Neuroblastoma , 3-Iodobenzylguanidine , Guanidine/therapeutic use , Humans , N-Myc Proto-Oncogene Protein , Neoplasm Recurrence, Local , Neuroblastoma/drug therapy , Neuroblastoma/therapy , Positron-Emission Tomography/methods , Retrospective Studies
9.
Pediatr Blood Cancer ; 68(3): e28794, 2021 03.
Article in English | MEDLINE | ID: mdl-33205902

ABSTRACT

BACKGROUND: Long-term outcome remains poor for children with high-risk neuroblastoma (five-year overall survival [OS] ∼50%). Our objectives were to (a) identify prognostic biomarkers and apply them in a nomogram to identify the subgroup of ultra-high-risk patients at highest risk of disease progression/death, for whom novel frontline therapy is urgently needed; and (b) validate the nomogram in an independent cohort. METHODS: A total of 1820 high-risk patients (≥18 months old with metastatic neuroblastoma), diagnosed 1998-2015, from the International Neuroblastoma Risk Groups (INRG) Data Commons were analyzed in a retrospective cohort study. Using multivariable Cox regression of OS from diagnosis, a nomogram was created from prognostic biomarkers to predict three-year OS. External validation was performed using the SIOPEN HR-NBL1 trial cohort (n = 521), evidenced by receiver operating characteristic curves. RESULTS: The nomogram, including MYCN status (P < 0.0001), lactate dehydrogenase (LDH) (P = 0.0007), and presence of bone marrow metastases (P = 0.004), had robust performance and was validated. Applying the nomogram at diagnosis (a) gives prognosis of an individual patient and (b) identifies patients predicted to have poor outcome (three-year OS was 30% ± 5% for patients with a nomogram score of > 82 points; 58% ± 1% for those ≤82 points). Median follow-up time was 5.5 years (range, 0-14.1). CONCLUSIONS: In high-risk neuroblastoma, a novel, publicly available nomogram using prognostic biomarkers (MYCN status, LDH, presence of bone marrow metastases; https://neuroblastoma.shinyapps.io/High-Risk-Neuroblastoma-Nomogram/) has the flexibility to apply a clinically suitable and context-specific cutoff to identify patients at highest risk of death. This will facilitate testing urgently needed new frontline treatment options to improve outcome for these children.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Bone Marrow Neoplasms/mortality , L-Lactate Dehydrogenase/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/mortality , Nomograms , Age Factors , Bone Marrow Neoplasms/drug therapy , Bone Marrow Neoplasms/metabolism , Bone Marrow Neoplasms/secondary , Child, Preschool , Female , Follow-Up Studies , Gene Amplification , Humans , Male , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Prognosis , Retrospective Studies , Survival Rate
10.
J Pediatr Hematol Oncol ; 42(1): 1-7, 2020 01.
Article in English | MEDLINE | ID: mdl-31599856

ABSTRACT

Although intensive multimodal treatment has improved outcomes for patients with high-risk neuroblastoma, the specific role of primary tumor resection remains controversial. Many studies have been designed to determine whether the extent of surgical resection impacts survival; however, these reports have demonstrated conflicting results. There is also ongoing debate regarding the timing of primary tumor resection, with subtle differences in the approach between the large pediatric oncology cooperative consortia. Most of the published literature to date has been approached from a surgical viewpoint. Although most evidence supports surgery as part of the local control approach for high-risk neuroblastoma, recommendations for timing and extent of surgical resection are not consistent. This review summarizes our current understanding from the perspectives of both the pediatric oncologist and pediatric surgeons and discusses how the objectives of neuroblastoma primary surgical resection are different from that of other malignancies. Furthermore, this commentary will address how retrospective surgical outcome data may be interpreted in the setting of modern era high-risk neuroblastoma treatment.


Subject(s)
Neuroblastoma/mortality , Neuroblastoma/surgery , Disease-Free Survival , Humans , Neoplasm Staging , Retrospective Studies
12.
Pediatr Blood Cancer ; 66(8): e27776, 2019 08.
Article in English | MEDLINE | ID: mdl-31033188

ABSTRACT

BACKGROUND: Treatment of opsoclonus-myoclonus syndrome (OMS) has historically involved corticosteroids and intravenous immunoglobulin (IVIG) for a duration of 6-12 months or longer. This study evaluated whether a brief upfront immunomodulatory therapy protocol with rituximab reduces the duration of OMS therapy without adversely affecting OMS outcomes. PROCEDURE: Retrospective chart review was performed for consecutive children diagnosed with OMS from 2006 to 2019 at The Hospital for Sick Children (Toronto, Canada). Children treated within 3 months of diagnosis with a treatment protocol involving pulse methylprednisolone (3-5 days, followed by an oral steroid taper), IVIG and/or plasma exchange, and rituximab (protocol group, n = 7) were compared to a historical group treated primarily with prednisone and IVIG (n = 8). RESULTS: The duration of corticosteroid treatment was shorter in the protocol (median 4.5 [range 3-12] months) compared to that in the historical group (median 21.5 [range 6-70] months, P = .005), and subjects in the protocol group received fewer cycles of IVIG (median 1 [range 0-7] cycle vs 7 [range 1-70] cycles, P = .01). The proportion of children with OMS relapse was similar between the protocol and historic groups (2/6 vs 5/8, P = .59). OMS symptom rating scales at 12-month follow-up were similar in the protocol group (median 2.5, range 0-3) compared to that in the historical group (median 1, range 0-7; P = .66). CONCLUSIONS: An upfront immunomodulatory therapy protocol with rituximab permits reduction in the duration of corticosteroid and IVIG therapy without a detrimental effect on OMS outcomes. Future studies with longer follow-up will have to determine whether neurocognitive and psychosocial outcomes are improved by this approach.


Subject(s)
Immunomodulation , Immunotherapy/methods , Neoplasm Recurrence, Local/therapy , Opsoclonus-Myoclonus Syndrome/therapy , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Male , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Opsoclonus-Myoclonus Syndrome/immunology , Opsoclonus-Myoclonus Syndrome/pathology , Prognosis , Retrospective Studies
13.
Pediatr Blood Cancer ; 66(4): e27556, 2019 04.
Article in English | MEDLINE | ID: mdl-30479064

ABSTRACT

Given the biological and clinical heterogeneity of neuroblastoma, risk stratification is vital to determining appropriate treatment. Historically, most patients with high-risk neuroblastoma (HR-NBL) have been treated uniformly without further stratification. Attempts have been made to identify factors that can be used to risk stratify these patients and to characterize an "ultra-high-risk" (UHR) subpopulation with particularly poor outcome. However, among published data, there is a lack of consensus in the definition of the UHR population and heterogeneity in the endpoints and statistical methods used. This review summarizes our current understanding of stratification of HR-NBL and discusses the complex issues in defining UHR neuroblastoma.


Subject(s)
Neuroblastoma/diagnosis , Neuroblastoma/mortality , Disease-Free Survival , Humans , Neuroblastoma/therapy , Risk Assessment , Risk Factors , Survival Rate
14.
J Pediatr Hematol Oncol ; 41(3): 222-227, 2019 04.
Article in English | MEDLINE | ID: mdl-30334904

ABSTRACT

Alternative radiolabeled, targeted agents are being investigated for children with relapsed neuroblastoma (NB) who do not respond to I-metaiodobenzylguanidine (MIBG) therapy. (DOTA-Tyr)-octreotate targets somatostatin receptors (SSTRs), particularly SSTR2, which are expressed on NB cells. We investigated SSTR2 expression in NB tumors (36 high-risk [HR]; 33 non-HR patients) and correlated SSTR2 levels with clinical features, norepinephrine transporter (NET) expression, and MIBG avidity. SSTR2 and NET immunohistochemistry scores (0 to 3) were calculated on biopsies using digital image analysis based on staining intensity and distribution. Clinical data were correlated with SSTR2 expression. Median SSTR2 score for 69 patients was 1.31 (0.26 to 2.55). Non-HR NB was associated with a higher SSTR2 score (P=0.032). The SSTR2 expression did not correlate with age, International Neuroblastoma Staging System (INSS) stage, MYCN amplification and histology. Higher SSTR2 scores were observed in MIBG-avid versus MIBG-nonavid NB. SSTR2 score was not significantly associated with NET score (r=-0.062, P=0.62). Twenty-six patients who relapsed or progressed had a median SSTR2 score of 1.33 (0.26 to 2.55). Patients with NB including relapsed or progressive disease showed SSTR2 expression at diagnosis, suggesting they could be candidates for radiolabeled-DOTA-conjugated peptide imaging or therapy.


Subject(s)
Neuroblastoma/chemistry , Receptors, Somatostatin/metabolism , 3-Iodobenzylguanidine/analysis , Disease Progression , Female , Humans , Immunohistochemistry , Male , Molecular Imaging/methods , Molecular Targeted Therapy/methods , Neuroblastoma/diagnostic imaging , Neuroblastoma/radiotherapy , Norepinephrine Plasma Membrane Transport Proteins/analysis , Prevalence , Radiopharmaceuticals/therapeutic use , Receptors, Somatostatin/analysis , Recurrence
15.
J Pediatr Hematol Oncol ; 41(5): 388-391, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31094905

ABSTRACT

Congenital neuroblastoma with placental involvement is exceptionally rare, but mortality is high. Detailed examination of placenta including MYCN amplification and segmental chromosomal aberrations should be performed in all suspected cases, as it is noninvasive and readily available. Maternal dissemination has not been reported. In this manuscript, we describe an infant with placental diagnosis of MYCN nonamplified congenital neuroblastoma. This is the first report of a recurrence of congenital 4S neuroblastoma following resolution in which MYCN amplification is only detected in the recurrence. Germline sequencing using a large comprehensive cancer panel did not reveal variants in candidate cancer predisposition genes.


Subject(s)
N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Adult , Chromosome Aberrations , Female , Gene Amplification , Humans , Infant , Neuroblastoma/congenital , Neuroblastoma/pathology , Placenta Diseases , Pregnancy , Recurrence
16.
Pediatr Blood Cancer ; 65(5): e26964, 2018 05.
Article in English | MEDLINE | ID: mdl-29369484

ABSTRACT

BACKGROUND: Ganglioneuromas (GNs) usually demonstrate favorable histological and clinical features. Surgery is often performed due to clinical symptoms and/or theoretical concerns that GN may transform into neuroblastoma (NB); however, several studies have identified significant GN-surgical morbidities. OBJECTIVES: We compared the natural history, biological and clinical features of GN and ganglioneuroblastoma-intermixed (GNB-I) managed by surgery or observation to inform management and surveillance. PROCEDURES: This retrospective study includes patients (n = 67) with histological diagnosis of GN (50/67) and GNB-I (17/67) at the Hospital for Sick Children between 1990 and 2014. Clinical, pathological features, tumor dimensions, and management were recorded. RESULTS: Median age and maximal tumor diameter were 6 years (1.3-17.8) and 6.3 cm (1.4-16.9), respectively. Of the 67 patients, 46 (69%) had upfront surgery and 21 (31%) were observed. Of the 21 observed patients 4 later underwent resection. There were post-operative complications in 15 of the 50 (30%) surgical patients. The presence of imaging-defined risk factors correlated with complications (P = 0.005). Observed patients were older (median 8.4 vs. 5.3 years) and diagnosed more recently. Median growth was 0.3 cm/year and 6 of 21 had progressive disease (PD). At median follow-up of 2.2 years (0.2-14.3), all patients were alive and for those with evaluable imaging there were 27 complete and 10 partial responses, 19 stable and 6 PD. Pathology classification changed at resection for three cases, but no GN was reclassified to NB. CONCLUSIONS: GN and GNB-I have a slow growth rate and resection can be associated with significant morbidity. Watch and wait approaches should be considered for some GN and GNB-I.


Subject(s)
Ganglioneuroblastoma/pathology , Ganglioneuroblastoma/surgery , Ganglioneuroma/pathology , Ganglioneuroma/surgery , Adolescent , Adult , Child , Child, Preschool , Disease Management , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Prognosis , Retrospective Studies , Young Adult
17.
Pediatr Blood Cancer ; 65(8): e27110, 2018 08.
Article in English | MEDLINE | ID: mdl-29693780

ABSTRACT

Opsoclonus, myoclonus, ataxia syndrome (OMA) is a severe neurologic disorder often associated with neuroblastoma. It is challenging to treat and can have long-term neurologic sequelae. Current recommended therapies include intravenous immunoglobulin, corticosteroids, rituximab, and chemotherapy (cyclophosphamide). We present two cases who were refractory to conventional therapy and underwent autologous stem cell transplantation (ASCT). One patient had complete resolution of symptoms following ASCT and the other patient had minimal change in symptoms with this therapy. These findings support consideration of ASCT as a therapeutic option for patients with refractory OMA after failure of known effective therapies.


Subject(s)
Opsoclonus-Myoclonus Syndrome/therapy , Stem Cell Transplantation , Female , Humans , Infant , Male , Transplantation, Autologous
18.
Pediatr Blood Cancer ; 65(12): e27390, 2018 12.
Article in English | MEDLINE | ID: mdl-30117275

ABSTRACT

BACKGROUND: Neuroblastoma is the most common pediatric extracranial solid tumor. Germline pathogenic variants in ALK and PHOX2B, as well as other cancer predisposition genes, are increasingly implicated in the pathogenesis of neuroblastic tumors. A challenge for clinicians is the identification of children with neuroblastoma who require genetics evaluation for underlying cancer predisposition syndromes (CPS). PROCEDURE: We developed a decisional algorithm (MIPOGG) to identify which patients with neuroblastic tumors have an increased likelihood of an underlying CPS. This algorithm, comprising 11 Yes/No questions, evaluates features in the tumor, personal and family history that are suggestive of an underlying CPS. We assessed the algorithm's performance in a retrospective cohort. RESULTS: Two hundred and nine of 278 consecutive patients with neuroblastic tumors at The Hospital for Sick Children (2007-2016) had sufficient clinical data for retrospective application of the decisional algorithm. Fifty-one of 209 patients had been referred to genetics for CPS evaluation; 6/51 had a genetic or clinical confirmation of a CPS. The algorithm correctly identified all six children (Beckwith-Wiedemann (n = 2), Fanconi anemia, RB1, PHOX2B, chromosome duplication involving ALK) as requiring a genetic evaluation by using clinical features present at diagnosis. The level of agreement between the algorithm and physicians was 83.9%, with 15 more patients identified by the algorithm than by physicians as requiring a genetics referral. CONCLUSIONS: This decisional algorithm appropriately detected all patients who, following genetic evaluation, were confirmed to have a CPS and may improve the detection of CPS in patients with neuroblastic tumors compared with current practice.


Subject(s)
Algorithms , Decision Support Systems, Clinical , Genetic Predisposition to Disease , Genetic Testing/methods , Neuroblastoma/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Referral and Consultation , Retrospective Studies
19.
Pediatr Blood Cancer ; 65(7): e27023, 2018 07.
Article in English | MEDLINE | ID: mdl-29603591

ABSTRACT

BACKGROUND: Vorinostat combined with retinoids produces additive antitumor effects in preclinical studies of neuroblastoma. Higher systemic exposures of vorinostat than achieved in pediatric phase I trials with continuous daily dosing are necessary for in vivo increased histone acetylation and cytotoxic activity. We conducted a phase I trial in children with relapsed/refractory neuroblastoma to determine the maximum tolerated dose (MTD) of vorinostat on an interrupted schedule, escalating beyond the previously identified pediatric MTD. METHODS: Isotretinoin (cis-13-retinoic acid) 80 mg/m2 /dose was administered by mouth twice daily on days 1-14 in combination with escalating doses of daily vorinostat up to 430 mg/m2 /dose (days 1-4; 8-11) in each 28-day cycle using the standard 3 + 3 design. Vorinostat pharmacokinetic testing and histone acetylation assays were performed. RESULTS: Twenty-nine patients with refractory or relapsed neuroblastoma were enrolled and 28 were evaluable for dose escalation decisions. Median number of cycles completed was two (range 1-15); 11 patients received four or more cycles. Three patients experienced cycle 1 dose-limiting toxicities. A total of 18 patients experienced grade 3/4 toxicities related to study therapy. The maximum intended dose of vorinostat (430 mg/m2 /day, days 1-4; 8-11) was tolerable and led to increased histone acetylation in surrogate tissues when compared to lower doses of vorinostat (P = 0.009). No objective responses were seen. CONCLUSIONS: Increased dose vorinostat (430 mg/m2 /day) on an interrupted schedule is tolerable in combination with isotretinoin. This dose led to increased vorinostat exposures and demonstrated increased histone acetylation. Prolonged stable disease in patients with minimal residual disease warrants further investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/drug effects , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Salvage Therapy , Adolescent , Adult , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Isotretinoin/administration & dosage , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Prognosis , Survival Rate , Vorinostat/administration & dosage , Young Adult
20.
Genes Chromosomes Cancer ; 56(1): 28-41, 2017 01.
Article in English | MEDLINE | ID: mdl-27465929

ABSTRACT

Amplification of the MYCN gene in neuroblastoma is associated with a poor prognosis and is considered to remain unchanged in post-treatment specimens and metastases. While heterogeneity of MYCN copy number in tumor cells has been reported, serial samples have only been studied in a limited way, and the biologic relevance of this finding is not well understood. We used in situ hybridization on paraffin sections of 102 specimens from 30 patients with MYCN-amplified neuroblastoma to determine MYCN copy number in the primary tumor, pre- and post-treatment, and in metastatic samples. Nineteen cases (63%) showed diffuse MYCN amplification in all samples tested. Nine cases (30%) showed a reduction in MYCN copy number: five cases with diffuse amplification subsequently showed focal amplification, one case with diffuse MYCN amplification showed MYCN gain after treatment, and three focally amplified cases were non-amplified in later specimens. In two cases (7%), focal amplification became diffuse in subsequent samples. Histology was not predictive of the temporal or spatial pattern of MYCN amplification for a particular tumor. If extent of amplification (focal vs. diffuse) is not considered, 26/30 (87%) of cases were consistently MYCN-amplified. However, our data suggest that MYCN status can be heterogeneous between tumor sites, during tumor progression or following treatment, challenging the notion that MYCN copy number does not change for a particular neuroblastoma. Assessing the biologic significance of MYCN heterogeneity will require larger studies of clinically annotated tumor samples, and will depend on interpreting heterogeneity in MYCN status in combination with other genetic changes. © 2016 Wiley Periodicals, Inc.


Subject(s)
Biomarkers, Tumor/genetics , DNA, Neoplasm/genetics , Gene Amplification , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Child , Child, Preschool , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Infant , Male , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Neuroblastoma/diagnosis , Neuroblastoma/therapy , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL