Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cancer ; 129(7): 989-991, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36704992

ABSTRACT

Children with cancer receive many medications outside the hospital administered by their caregivers. The study by Walsh et al. shows the number and types of medication errors in these patients. The study includes data from three different centers. Importantly, the study shows the types of errors that cause harm. The authors describe how the harmful errors can be prevented. We suggest ways these results can be used to identify which patients and families will benefit from additional attention. Providing more help at clinic and in the home may help prevent harmful medication errors in children with cancer.


Subject(s)
Medication Errors , Neoplasms , Humans , Child , Medication Errors/prevention & control , Neoplasms/drug therapy , Hospitals
2.
Auris Nasus Larynx ; 50(4): 576-585, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36496304

ABSTRACT

OBJECTIVE: Head and neck cancers represent critical challenges due to the restricted anatomical space in children and the proximity of critical neurovascular structures which can compromise complete tumor resection. Applications of Indocyanine green (ICG) near infrared (NIR) fluorescent image-guided surgery (FGS) have recently expanded into the pediatric population, emphasizing its relevance for tumor delineation and evaluation of tissue perfusion. The objectives of the present study are twofold. First, we aim to assess the role of ICG NIR imagery in identifying neoplastic disease and second, to evaluate its role as an adjunct to identify metastatic deposits in cervical lymph node in children, adolescents, and young adults with head and neck cancers. METHODS: Eight patients with head and neck malignancies underwent ICG NIR FGS, between January 2019 and November 2021. ICG NIR findings were compared with preoperative cross-sectional imaging and results of operative tissue pathology analyses. RESULTS: All primary tumors were identified on preoperative imaging and intraoperatively with ICG NIR; however, for one case, extension of tumor was revealed with ICG NIR and confirmed by histopathological examination but was not otherwise visible on preoperative imaging or with naked eye visual and tactile assessment. ICG NIR assisted the decision process in a difficult case for which curative resection, without significant functional morbidity and potential mortality, was unrealistic. Although ICG NIR evaluation of the surgical bed did not display residual tumor, margins were found positive in two cases. ICG NIR evaluation for local metastases changed the surgical strategy in one patient by prompting conversion to bilateral neck dissections. The sensitivity of preoperative multimodality imaging to identify cervical levels of invasion was 75% with a specificity of 70%, a PPV of 33%, a NPV of 78% and an accuracy of 72%. The ICG NIR sensitivity was 83%, its specificity was 88% with a PPV of 91%, a NPV of 80% and an accuracy of 86%. The combination of preoperative multimodality imaging with ICG NIR findings led to a sensitivity of 83%, specificity of 88% and accuracy of 86%. CONCLUSION: This case series provides a proof of concept of the feasibility of ICG NIR, as an adjunct in tumor and local metastases identification in young patients with head and neck tumors. It revealed to be feasible and safe for intra-operative tumor identification, thus guiding and facilitating resection. However, it showed some limitations in precise tumor margin assessment. The combination of preoperative multimodality imaging with ICG NIR findings improved local metastases localization.


Subject(s)
Head and Neck Neoplasms , Indocyanine Green , Child , Humans , Young Adult , Adolescent , Spectroscopy, Near-Infrared/methods , Coloring Agents , Lymph Nodes , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/surgery
4.
Cancer ; 128(3): 606-614, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34643950

ABSTRACT

BACKGROUND: Data on primary hypothyroidism and its long-term impact on the health, cognition, and quality of life (QOL) of childhood cancer survivors are limited. This study examined the prevalence of and risk factors for primary hypothyroidism and its associations with physical, neurocognitive, and psychosocial outcomes. METHODS: This was a retrospective study with a cross-sectional health outcome analysis of an established cohort comprising 2965 survivors of childhood cancer (52.8% male; median current age, 30.9 years, median time since cancer diagnosis, 22.3 years). Multivariable logistic regression estimated odds ratios (ORs) and 95% confidence intervals (CIs) for associations between primary hypothyroidism and cancer-related risk factors, cardiovascular disease risk factors, frailty, neurocognitive and QOL outcomes, social attainment, and subsequent thyroid carcinoma. Associations between serum free thyroxine and thyrotropin levels at assessment and health outcomes were explored. RESULTS: The prevalence of primary hypothyroidism was 14.7% (95% CI, 13.5%-16.0%). It was more likely in females (OR, 1.06; 95% CI, 1.03-1.08), was less likely in non-Whites (OR, 0.96; 95% CI, 0.93-0.99), was associated with thyroid radiotherapy (higher risk at higher doses), and was more common if cancer was diagnosed at an age ≥ 15.0 years versus an age < 5 years (OR, 1.05; 95% CI, 1.01-1.09). Primary hypothyroidism was associated with frailty (OR, 1.54; 95% CI, 1.05-2.26), dyslipidemia (OR, 1.52; 95% CI, 1.14-2.04), impaired physical QOL (OR, 1.66; 95% CI, 1.12-2.48), and having health care insurance (OR, 1.51; 95% CI, 1.07-2.12). CONCLUSIONS: Primary hypothyroidism is common in survivors and is associated with unfavorable physical health and QOL outcomes. The impact of thyroid hormone replacement practices on these outcomes should be investigated further.


Subject(s)
Cancer Survivors , Hypothyroidism , Leukemia, Myeloid, Acute , Adolescent , Adult , Cancer Survivors/psychology , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Hypothyroidism/epidemiology , Leukemia, Myeloid, Acute/complications , Male , Prevalence , Quality of Life , Retrospective Studies , Risk Factors
5.
J Clin Oncol ; 40(4): 335-344, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34871104

ABSTRACT

PURPOSE: We evaluated whether combining a humanized antidisialoganglioside monoclonal antibody (hu14.18K322A) throughout therapy improves early response and outcomes in children with newly diagnosed high-risk neuroblastoma. PATIENTS AND METHODS: We conducted a prospective, single-arm, three-stage, phase II clinical trial. Six cycles of induction chemotherapy were coadministered with hu14.18K322A, granulocyte-macrophage colony-stimulating factor (GM-CSF), and low-dose interleukin-2 (IL-2). The consolidation regimen included busulfan and melphalan. When available, an additional cycle of parent-derived natural killer cells with hu14.18K322A was administered during consolidation (n = 31). Radiation therapy was administered at the end of consolidation. Postconsolidation treatment included hu14.18K322A, GM-CSF, IL-2, and isotretinoin. Early response was assessed after the first two cycles of induction therapy. End-of-induction response, event-free survival (EFS), and overall survival (OS) were evaluated. RESULTS: Sixty-four patients received hu14.18K322A with induction chemotherapy. This regimen was well tolerated, with continuous infusion narcotics. Partial responses (PRs) or better after the first two chemoimmunotherapy cycles occurred in 42 of 63 evaluable patients (66.7%; 95% CI, 55.0 to 78.3). Primary tumor volume decreased by a median of 75% (range, 100% [complete disappearance]-5% growth). Median peak hu14.18K322A serum levels in cycle one correlated with early response to therapy (P = .0154, one-sided t-test). Sixty of 62 patients (97%) had an end-of-induction partial response or better. No patients experienced progressive disease during induction. The 3-year EFS was 73.7% (95% CI, 60.0 to 83.4), and the OS was 86.0% (95% CI, 73.8 to 92.8), respectively. CONCLUSION: Adding hu14.18K322A to induction chemotherapy improved early objective responses, significantly reduced tumor volumes in most patients, improved end-of-induction response rates, and yielded an encouraging 3-year EFS. These results, if validated in a larger study, may be practice changing.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Interleukin-2/therapeutic use , Neuroblastoma/drug therapy , Adolescent , Age Factors , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Child, Preschool , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Induction Chemotherapy , Infant , Interleukin-2/adverse effects , Male , Neuroblastoma/immunology , Neuroblastoma/mortality , Neuroblastoma/pathology , Progression-Free Survival , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Tumor Burden/drug effects
6.
J Pediatr Hematol Oncol ; 43(5): e692-e696, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33181583

ABSTRACT

Although outcomes for patients with high-risk neuroblastoma improved after the addition of a chimeric anti-GD2 monoclonal antibody (dinutuximab) as treatment for minimal residual disease, nearly half of these patients die of disease. Recent studies demonstrated efficacy of the combination of chemotherapy with anti-GD2 mAb in patients with relapsed or newly diagnosed disease. This retrospective case series describes 6 patients treated at St Jude Children's Research Hospital with an induction regimen containing dinutuximab and chemotherapy, followed by consolidation and postconsolidation therapy. The treatment was well tolerated with expected toxicities. All patients completed induction therapy and demonstrated a clinical response. Further studies are warranted.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cytokines/therapeutic use , Neuroblastoma/drug therapy , Child, Preschool , Female , Humans , Induction Chemotherapy , Infant , Male , Retrospective Studies
7.
Eur J Cancer ; 137: 204-213, 2020 09.
Article in English | MEDLINE | ID: mdl-32795876

ABSTRACT

BACKGROUND: Talazoparib combined with irinotecan and temozolomide demonstrated efficacy in a murine Ewing sarcoma model. Based on these data, we conducted a phase I trial of talazoparib and irinotecan with/without temozolomide in paediatric patients with recurrent/refractory solid malignancies. PATIENTS AND METHODS: Cohorts of 3-6 patients with recurrent/refractory solid malignancies received escalating doses of oral talazoparib and intravenous irinotecan (arm A) and oral talazoparib, oral temozolomide and intravenous irinotecan (arm B) in a 3 + 3 design. Talazoparib was administered on days 1-6, and intravenous irinotecan and oral temozolomide were administered on days 2-6, of a 21-day course. Serum for talazoparib and irinotecan pharmacokinetics was obtained during course 1. UGT1A1 polymorphism and Schlafen family member 11 (SLFN11) immunohistochemical staining were performed. RESULTS: Forty-one patients (20 males; median age, 14.6 years; 24 with recurrent disease) were evaluable for dose escalation. Twenty-nine and 12 patients were treated on arm A and arm B, respectively, for a total of 208 courses. The most common diagnosis was Ewing sarcoma (53%). The most common ≥grade III haematologic toxicities in arms A and B included neutropenia (78% and 31%, respectively) and thrombocytopenia (42% and 31%, respectively). In arms A and B, febrile neutropenia (24% and 14%, respectively) and diarrhoea (21% and 7%, respectively) were the most common ≥grade III non-hematologic toxicities. Six patients (Ewing sarcoma [5 patients] and synovial sarcoma [1 patient]) had a response (1 with a complete response, 5 with a partial response). The objective response rates were 10.3% (arm A) and 25% (arm B). Pharmacokinetic testing demonstrated no evidence of drug-drug interaction between talazoparib and irinotecan. UGT1A1 was not related to response. SLFN11 positivity was associated with best response to therapy. CONCLUSIONS: The combination of talazoparib and irinotecan with/without temozolomide is feasible and active in Ewing sarcoma, and further investigation is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Irinotecan/therapeutic use , Neoplasms/drug therapy , Phthalazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Temozolomide/therapeutic use , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Child , Child, Preschool , Female , Humans , Irinotecan/pharmacology , Male , Neoplasms/pathology , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Temozolomide/pharmacology , Young Adult
8.
Pediatr Qual Saf ; 5(2): e269, 2020.
Article in English | MEDLINE | ID: mdl-32426635

ABSTRACT

INTRODUCTION: Handoffs and transitions of care are common weak points in healthcare provider communication as patients move between sites. With no consistent pattern of communication between St. Jude Children's Research Hospital (St. Jude) and its affiliated clinics, the Affiliate Program Office at St. Jude developed and implemented a standardized communication tool to facilitate patient transitions between different healthcare sites. METHODS: Each team of providers created flow diagrams to define the current state of communication when patients were transitioning between remote sites. Fishbone diagrams identified the common barriers to effective communication as a lack of consistent communication and ownership. We developed a communication tool to address these barriers, which was disseminated by secure email. We measured the percent usage of the completed hand-off tool before a patient transitioned, staff experience, and the number of errors. RESULTS: The time to send or receive the communication bundle was <10 minutes. Within 3 months of implementing the SMART bundle at 3 pilot sites, the bundle was used completely in 6 of 8 patient transitions and was associated with somewhat improved staff satisfaction. We identified no adverse events related to the communication bundle. CONCLUSIONS: In this small pilot study, we accomplished closed-loop communication between geographically remote healthcare sites by using an electronically transmitted standardized communication bundle.

9.
Pediatr Blood Cancer ; 67(9): e28330, 2020 09.
Article in English | MEDLINE | ID: mdl-32452122

ABSTRACT

Infantile fibrosarcoma (IFS) is a rare pediatric cancer that typically presents early in life. Surgical resection is commonly curative; however, resection is sometimes not possible requiring additional multimodal treatment. IFS commonly harbors a fusion in one of the neurotrophic receptor tyrosine kinase (NTRK) genes. Larotrectinib, a highly selective inhibitor of tropomyosin receptor kinase (TRK), has been shown to be well tolerated and effective in children as young as 1-month old. We report a case of IFS in a newborn treated with larotrectinib. The patient experienced a rapid clinical and radiographic response demonstrating the potential to treat newborns with larotrectinib.


Subject(s)
Fibrosarcoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Humans , Infant, Newborn , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Oncogene Proteins, Fusion/genetics , Protein Kinases/drug effects , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/genetics
10.
Clin Cancer Res ; 25(21): 6320-6328, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31601569

ABSTRACT

PURPOSE: We sought to evaluate whether combining a humanized antidisialoganglioside mAb (hu14.18K322A) with induction chemotherapy improves early responses and outcomes in children with newly diagnosed high-risk neuroblastoma. PATIENTS AND METHODS: We conducted a prospective nonrandomized, single-arm, two-stage, phase II clinical trial. Six courses of induction chemotherapy were coadministered with hu14.18K322A and followed with granulocyte-macrophage colony-stimulating factor (GM-CSF) and low-dose IL2. Consolidation was performed with a busulfan/melphalan preparative regimen. An additional course of hu14.18K322A was administered with parent-derived natural killer cells, when available, during consolidation. Hu14.18K322A, GM-CSF, IL2, and isotretinoin were then administered. Secondary outcomes included reduced tumor volume and semiquantitative 123I-metaiodobenzylguanidine scoring [i.e., Curie scores (CS)] at the end of induction. RESULTS: Forty-two patients received hu14.18K322A and induction chemotherapy. This regimen was well tolerated, with continuous-infusion narcotics adjusted to patient tolerance. Partial responses (PR) or better after the first two chemoimmunotherapy courses occurred in 32 patients [76.2%; 95% confidence interval (CI), 60.6-88.0]. This was accompanied by primary tumor volume reductions (median, -76%; range, -100% to 5%). Of 35 patients with stage IV disease who completed induction, 31 had end-of-induction CSs of 2 or less. No patients experienced progression during induction. Two-year event-free survival (EFS) was 85.7% (95% CI, 70.9-93.3). CONCLUSIONS: Adding hu14.18K322A to induction chemotherapy produced early PR or better in most patients, reduced tumor volumes, improved CSs at the end of induction, and yielded an encouraging 2-year EFS. These results, if validated in a larger study, may change the standard of care for children with high-risk neuroblastoma.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Neuroblastoma/drug therapy , Adolescent , Adult , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Disease-Free Survival , Female , Gangliosides , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Induction Chemotherapy/adverse effects , Kaplan-Meier Estimate , Killer Cells, Natural/drug effects , Male , Neuroblastoma/pathology , Progression-Free Survival , Prospective Studies , Young Adult
11.
Pediatr Blood Cancer ; 66(6): e27678, 2019 06.
Article in English | MEDLINE | ID: mdl-30803146

ABSTRACT

Children with trisomy 18 are surviving longer and undergoing more aggressive life-sustaining therapy. This report describes two patients with trisomy 18 and hepatoblastoma (HB) successfully resected in the setting of significant pulmonary hypertension. Forty-four previously published cases of the association between HB and trisomy 18 are reviewed. With careful multidisciplinary preoperative planning, successful resection of HB in children with trisomy 18 who have significant pulmonary hypertension is feasible. Because HB and trisomy 18 are increasing in prevalence, the need for timely liver tumor resection in the setting of pulmonary hypertension will be more common.


Subject(s)
Anesthetics/administration & dosage , Hepatectomy/methods , Hepatoblastoma/surgery , Hypertension, Pulmonary/surgery , Liver Neoplasms/surgery , Trisomy 18 Syndrome/surgery , Female , Hepatoblastoma/complications , Hepatoblastoma/drug therapy , Hepatoblastoma/pathology , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Infant , Liver Neoplasms/complications , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Prognosis , Trisomy 18 Syndrome/complications , Trisomy 18 Syndrome/drug therapy , Trisomy 18 Syndrome/pathology
12.
Sci Rep ; 6: 20376, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26847534

ABSTRACT

The transition from a committed progenitor cell to one that is actively differentiating represents a process that is fundamentally important in skeletal myogenesis. Although the expression and functional activation of myogenic regulatory transcription factors (MRFs) are well known to govern lineage commitment and differentiation, exactly how the first steps in differentiation are suppressed in a proliferating myoblast is much less clear. We used cultured mammalian myoblasts and an RNA interference library targeting 571 kinases to identify those that may repress muscle differentiation in proliferating myoblasts in the presence or absence of a sensitizing agent directed toward CDK4/6, a kinase previously established to impede muscle gene expression. We identified 55 kinases whose knockdown promoted myoblast differentiation, either independently or in conjunction with the sensitizer. A number of the hit kinases could be connected to known MRFs, directly or through one interaction node. Focusing on one hit, Mtor, we validated its role to impede differentiation in proliferating myoblasts and carried out mechanistic studies to show that it acts, in part, by a rapamycin-sensitive complex that involves Raptor. Our findings inform our understanding of kinases that can block the transition from lineage commitment to a differentiating state in myoblasts and offer a useful resource for others studying myogenic differentiation.


Subject(s)
Muscle Development , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Checkpoints , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Mice , Microscopy, Fluorescence , Muscle Development/drug effects , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/metabolism , Piperazines/pharmacology , Protein Array Analysis , Protein Kinases/chemistry , Protein Kinases/genetics , Pyridines/pharmacology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Regulatory-Associated Protein of mTOR , Sequence Analysis, RNA , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...