Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Pediatr Blood Cancer ; 70 Suppl 4: e29974, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36184716

RESUMO

Neuroblastoma is the most common extracranial solid neoplasm in children. This manuscript provides consensus-based imaging recommendations for pediatric neuroblastoma patients at diagnosis and during follow-up.


Assuntos
Neuroblastoma , Ressonância de Plasmônio de Superfície , Criança , Humanos , Neuroblastoma/patologia , Diagnóstico por Imagem , Estadiamento de Neoplasias
2.
Pediatr Blood Cancer ; 66(8): e27763, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31012273

RESUMO

BACKGROUND: Radiation therapy (RT) confers local tumor control and survival advantages in some patients with osteosarcoma, yet pediatric and adolescent and young adult (AYA) population studies are limited. METHODS: Twenty-eight patients treated with curative-intent RT (median dose, 59.4 Gy; range, 40-76 Gy) at our institution from 1990 to 2017 were retrospectively identified. Cumulative incidence (CIN) of local failure (LF) was estimated by Gray's method and overall survival (OS) by the Kaplan-Meier method. Competing-risk regression and Cox proportional hazards models determined predictors of outcome. Toxicity was reported according to CTCAE v4.0. RESULTS: With a median follow-up of 99.1 months in living patients, nine patients (32.1%) developed LF. Estimated CINs of LF with competing risk of death at 5 years for the entire cohort, patients at initial diagnosis (n = 16), and recurrent/refractory patients (n = 12) were 32.7% (95% CI, 16.0-50.5%), 25.0% (95% CI, 7.3-48.0%), and 43.8% (95% CI, 13.6-71.0%), respectively (P = 0.31). Estimated 5-year OS was 42.6% (95% CI, 23.2-62.0%), 54.6% (95% CI, 29.5-79.6%), and 24.3% (95% CI, 0-52.2%), respectively (P = 0.15). No clinicopathologic features were significantly associated with LF, yet lack of chemotherapy or metastasis at the time of RT was independent significant prognostic factors of decreased OS. Eleven patients experienced RT-related morbidity, with two grade 3 toxicities and no grade 4/5 events. CONCLUSIONS: Curative-intent RT in pediatric and AYA patients was well tolerated and achieved a local tumor control rate of 75% in patients with primary disease. Local control rates were similar to those in primarily adult studies, with similar or lower doses.


Assuntos
Neoplasias Ósseas/radioterapia , Braquiterapia/mortalidade , Recidiva Local de Neoplasia/radioterapia , Osteossarcoma/radioterapia , Adolescente , Adulto , Neoplasias Ósseas/patologia , Criança , Feminino , Seguimentos , Humanos , Masculino , Recidiva Local de Neoplasia/patologia , Osteossarcoma/patologia , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Adulto Jovem
3.
Pediatr Dev Pathol ; 22(5): 492-498, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31072206

RESUMO

One-third of gastrointestinal stromal tumors (GISTs) that lack KIT or PDGFRA mutations show succinate dehydrogenase (SDH) mutations or promoter hypermethylation. Most SDH-deficient GISTs occur in the pediatric, adolescent, or young adult setting and have unique features including predilection for the stomach, multinodular plexiform architecture, epithelioid cytology, prominence of lymphovascular invasion, and predilection for nodal metastasis. Dedifferentiation in GIST is a rare histologic change which may occur de novo or secondary to imatinib therapy and is characterized by abrupt transition of well-differentiated (WD) GIST to a subclonal anaplastic process that shows loss of immunohistochemical marks (CD117, DOG1). We describe the case of a previously healthy 18-year-old man who presented with a large gastric wall mass that contained 2 distinct morphologic populations. The first was WD and characterized by sweeping fascicles of bland spindled cells. This population abruptly transitioned to dedifferentiated (DD) foci composed of large sheets of discohesive cells that displayed a spectrum of rhabdoid and epithelioid morphologies with marked pleomorphism and mitotic activity. Immunohistochemically, the tumor showed variable staining in the 2 components with diffuse DOG-1 and CD117 positivity in the WD component and complete absence in the DD foci. SDH-B staining was lost in both components. Whole exome and transcriptome analysis was performed on tissue from both components and both showed an SDHB mutation (c.286G>A) as well as unique mutational burden and copy number profiles. Herein, we describe the first case of a DD SDH-deficient GIST with morphologic, immunophenotypic, and molecular characterization.


Assuntos
Neoplasias Gastrointestinais/patologia , Tumores do Estroma Gastrointestinal/patologia , Succinato Desidrogenase/genética , Adolescente , Biomarcadores Tumorais/análise , Desdiferenciação Celular , Neoplasias Gastrointestinais/genética , Tumores do Estroma Gastrointestinal/genética , Humanos , Imuno-Histoquímica , Imunofenotipagem , Masculino , Succinato Desidrogenase/deficiência
4.
Pediatr Blood Cancer ; 70 Suppl 4: e30092, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36441588
5.
Pediatr Radiol ; 43(9): 1063-73, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23843130

RESUMO

The practice of contrast-enhanced ultrasound in children is in the setting of off-label use or research. The widespread practice of pediatric contrast-enhanced US is primarily in Europe. There is ongoing effort by the Society for Pediatric Radiology (SPR) and International Contrast Ultrasound Society (ICUS) to push for pediatric contrast-enhanced US in the United States. With this in mind, the main objective of this review is to describe the status of US contrast agent safety in non-cardiac applications in children. The five published studies using pediatric intravenous contrast-enhanced US comprise 110 children. There is no mention of adverse events in these studies. From a European survey 948 children can be added. In that survey six minor adverse events were reported in five children. The intravesical administration of US contrast agents for diagnosis of vesicoureteric reflux entails the use of a bladder catheter. Fifteen studies encompassing 2,951 children have evaluated the safety of intravesical US contrast agents in children. A European survey adds 4,131 children to this group. No adverse events could be attributed to the contrast agent. They were most likely related to the bladder catheterization. The existing data on US contrast agent safety in children are encouraging in promoting the widespread use of contrast-enhanced US.


Assuntos
Meios de Contraste/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Endossonografia/efeitos adversos , Extravasamento de Materiais Terapêuticos e Diagnósticos/etiologia , Injeções Intravenosas/efeitos adversos , Ultrassonografia de Intervenção/efeitos adversos , Administração Intravesical , Adolescente , Adulto , Criança , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Extravasamento de Materiais Terapêuticos e Diagnósticos/prevenção & controle , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Medição de Risco , Adulto Jovem
6.
J Clin Oncol ; 40(4): 335-344, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34871104

RESUMO

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.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Interleucina-2/uso terapêutico , Neuroblastoma/tratamento farmacológico , Adolescente , Fatores Etários , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Criança , Pré-Escolar , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/efeitos adversos , Humanos , Quimioterapia de Indução , Lactente , Interleucina-2/efeitos adversos , Masculino , Neuroblastoma/imunologia , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Intervalo Livre de Progressão , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Carga Tumoral/efeitos dos fármacos
7.
Pediatr Radiol ; 40(8): 1380-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20180103

RESUMO

BACKGROUND: Pancreatic metastases in childhood cancer have been rarely reported in the radiology literature although ample evidence exists in pathology reports for its occurrence in patients with alveolar rhabdomyosarcomas (RMS). OBJECTIVE: Assess the occurrence of pancreatic metastases in alveolar rhabdomyosarcomas, increase awareness of this association and reassess current staging protocols. MATERIALS AND METHODS: Three major oncology centers reviewed their records and imaging examinations. Patients' history and demographics, primary tumor site and histology, presence of tumor recurrence, and presence and location of other metastases were reviewed. RESULTS: Pancreatic metastases occurred in eight patients with alveolar RMS. Four of these presented at diagnosis and four with disease recurrence. In recurrent disease, the duration between the diagnosis of the primary tumor and pancreatic metastases varied from 8 months to 6 years (mean +/- SD: 2.38 +/- 2.49 years). In all patients who received PET scans, pancreatic metastases showed a marked FDG-uptake, but had variable detectability with CT. Pancreatic metastases were not associated with certain primary tumor locations or presence of other metastases, mandating an evaluation of the pancreas in all cases of alveolar rhabdomyosarcomas. CONCLUSION: Radiologists should be sensitized and actively evaluate the pancreas in patients with alveolar RMS. Optimizing CT and PET-CT protocols may increase the diagnostic yield.


Assuntos
Neoplasias Pancreáticas/complicações , Neoplasias Pancreáticas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Rabdomiossarcoma Alveolar/complicações , Rabdomiossarcoma Alveolar/diagnóstico por imagem , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Estadiamento de Neoplasias/métodos , Tomografia por Emissão de Pósitrons/tendências , Recidiva , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
8.
Eur J Radiol ; 133: 109376, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33166832

RESUMO

PURPOSE: To present a pictorial essay of paediatric primary synovial sarcomas from common and less documented anatomical locations. To review the literature for the imaging characteristics and prognostic factors of this rare but important childhood malignancy. METHOD: 24 primary synovial sarcoma cases (17 male, 7 female with an age range 4-21 years) were reviewed in a collaborative effort between St Jude Children's Research Hospital and Great Ormond Street Hospital for Children. Images from 19 cases were selected for inclusion, to demonstrate the spectrum of appearances across imaging modalities, in a range of different anatomical locations (upper limb, lower limb, chest/abdomen/pelvis, and head and neck). A literature review depicting the typical radiological features and the prognostic significance of these features, was also conducted. RESULTS AND CONCLUSIONS: Primary synovial sarcoma can occur in any anatomical location, but typically within the extremities and often in close association with joints. Rarer anatomical locations described in our essay include the gastrohepatic ligament and femoral nerve sheath. We detail the salient imaging characteristics, including the T2 'triple signal' pattern which is believed to be highly specific for this particular sarcoma and in many cases predicts a poor outcome. Other poor prognostic factors include haemorrhage, lack of calcification and tumour size >10 cm. A broad range of radiological appearances are described, and in some cases related to anatomical position and size, however the presence of a soft tissue mass close to a joint in a young patient are suggestive of this diagnosis.


Assuntos
Sarcoma Sinovial , Neoplasias de Tecidos Moles , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Sarcoma Sinovial/diagnóstico por imagem , Adulto Jovem
9.
Pathol Res Pract ; 216(6): 152942, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32299759

RESUMO

Lipofibromatosis (LPF) and lipofibromatosis-like neural tumor (LPF-NT) are histologically and prognostically similar neoplasms having differences in immunophenotype as well as molecular biology. In most cases, LPF-NT is driven by fusions in the NTRK gene, whereas LPF has been associated with fusions in a variety of receptor tyrosine kinases. The distinction between the driver fusion event holds clinical significance because of the profound clinical response to tropomyosin receptor kinase (Trk) inhibitors (larotrectinib) in the NTRK-driven tumors. Immunohistochemically, and consistent with its namesake, to-date all reported cases classified as LPF-NT have shown positivity for S100-protein staining. Consequently, as S100-protein staining is widely available, it represents a cost-effective screening tool for LPF-NT where the more specific studies such as the pan-Trk stain or fluorescence in situ hybridization for NTRK rearrangement are not available. Herein, we present a case of presumed LPF-NT harboring the recurrent NTRK1-LMNA fusion, but which was negative for S100-protein immunostaining and was previously classified as classical LPF. This case reveals a potential pitfall in distinguishing these rare subcutaneous tumors by S100-protein staining and highlights the challenges in reconciling the rapid and novel discoveries made in the field of diagnostic pathology.


Assuntos
Fibroma/diagnóstico , Lipoma/diagnóstico , Neoplasias de Tecidos Moles/diagnóstico , Pré-Escolar , Feminino , Fibroma/genética , Humanos , Lamina Tipo A/genética , Lipoma/genética , Fusão Oncogênica , Fenótipo , Receptor trkA/genética , Neoplasias de Tecidos Moles/genética
10.
Ultrasound Q ; 36(3): 206-217, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32890323

RESUMO

The Society of Radiologists in Ultrasound convened a panel of specialists in contrast-enhanced ultrasound (CEUS) to produce a white paper on noncardiac CEUS in North America. The panel met in Chicago, Illinois, on October 24 and 25, 2017. The recommendations are based on analysis of current literature and common practice strategies and are thought to represent a reasonable approach to introduce the advantages of this safe and noninvasive technique for the benefit of our patients. Characterization of liver nodules, and pediatric vascular and intravesicular applications comprise the approved indications for CEUS in the United States. They, along with the very successful off-label use of CEUS for the kidney, are included in this publication.Other off-label uses are presented with emphasis on their value and literature support in the online version.


Assuntos
Meios de Contraste , Aumento da Imagem/métodos , Neoplasias/diagnóstico por imagem , Ultrassonografia/métodos , Humanos , América do Norte
11.
Ultrasound Q ; 36(4S Suppl 1): S1-S39, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32956244

RESUMO

On October 24, 2017, in Chicago, the Society of Radiologists in Ultrasound convened a panel of specialists in contrast-enhanced ultrasound (CEUS) to arrive at a white paper regarding the use of CEUS in noncardiac applications in North America. Recommendations are based on analysis of the current literature and common practice strategies. They represent a reasonable approach to introduce the advantages of this safe and noninvasive technique for the benefit of our patients. Characterization of liver nodules with CEUS, as the approval indication worldwide, is the major focus of this endeavor. In addition, many off label uses are reviewed and literature supporting these indications provided.Key Points(1) Contrast-enhanced ultrasound (CEUS) allows cross-sectional imaging of the liver, kidneys and multiple other solid and hollow viscera, providing excellent characterization of identified focal mass lesions.(2) Performed with the injection of a microbubble contrast agent, CEUS provides a safe and readily available imaging technique which requires no ionizing radiation, making it appropriate for use in all ages, in those with renal insufficiency and when a portable examination is needed.(3) The CEUS can be considered in abdominal imaging whenever blood flow information is of value to diagnosis.(4) Dynamic real-time acquisition and the use of a purely intravascular contrast agent are the 2 most essential technical aspects of CEUS imaging which distinguish it from both computed tomography and magnetic resonance scan.


Assuntos
Meios de Contraste , Aumento da Imagem/métodos , Neoplasias Hepáticas/diagnóstico por imagem , Ultrassonografia/métodos , Humanos , Fígado/diagnóstico por imagem , América do Norte , Radiologistas , Sociedades Médicas
12.
J Clin Med ; 8(11)2019 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-31694285

RESUMO

Chronic blood transfusions in patients with sickle cell anemia (SCA) cause iron overload, which occurs with a degree of interpatient variability in serum ferritin and liver iron content (LIC). Reasons for this variability are unclear and may be influenced by genes that regulate iron metabolism. We evaluated the association of the copy number of the glutathione S-transferase M1 (GSTM1) gene and degree of iron overload among patients with SCA. We compared LIC in 38 children with SCA and ≥12 lifetime erythrocyte transfusions stratified by GSTM1 genotype. Baseline LIC was measured using magnetic resonance imaging (MRI), R2*MRI within 3 months prior to, and again after, starting iron unloading therapy. After controlling for weight-corrected transfusion burden (mL/kg) and splenectomy, mean pre-chelation LIC (mg/g dry liver dry weight) was similar in all groups: GSTM1 wild-type (WT) (11.45, SD±6.8), heterozygous (8.2, SD±4.52), and homozygous GSTM1 deletion (GSTM1-null; 7.8, SD±6.9, p = 0.09). However, after >12 months of chelation, GSTM1-null genotype subjects had the least decrease in LIC compared to non-null genotype subjects (mean LIC change for GSTM1-null = 0.1 (SD±3.3); versus -0.3 (SD±3.0) and -1.9 (SD±4.9) mg/g liver dry weight for heterozygous and WT, respectively, p = 0.047). GSTM1 homozygous deletion may prevent effective chelation in children with SCA and iron overload.

13.
Magn Reson Imaging ; 24(10): 1319-24, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17145403

RESUMO

PURPOSE/OBJECTIVE: This study aimed to develop objective models of radiation effects on musculature in children with soft tissue sarcoma using treatment dosimetry and clinical and quantitative magnetic resonance imaging (MRI) parameters that may be used to guide treatment planning or predict side effects. METHODS: In the initial 13 patients undergoing external beam radiation therapy (RT) on a Phase II study of conformal or intensity-modulated RT for the treatment of soft tissue sarcoma approved by an Institutional Review Board, we evaluated quantitative MRI changes in the musculature to assess radiation-related treatment effects. Patients with soft tissue sarcoma, including Ewing's sarcoma, had quantitative T1, T2 and dynamic enhanced MRI (DEMRI) performed before, during (Week 4) and after RT (Week 12). Regions of interest were selected in consistent locations within and outside the high-dose regions (on ipsilateral and contralateral sides when available). Mean RT dose, T1, T2 and DEMRI parameters were calculated and modeled using a mixed random coefficient dose model. RESULTS: The mean doses to the high- and low-dose regions were 56.4 Gy (41.8-75.3 Gy) and 13.0 Gy (0.1-37.5 Gy), respectively. Compared with tissues distant from the tumor bed, maximal enhancement was significantly increased in tissues adjacent to the tumor/tumor bed prior to RT (60.6 vs. 44.2, P=.045) and remained elevated after 12 weeks. T1 was significantly elevated in tissues adjacent to the tumor bed prior to RT (942.4 vs. 759.0, P=.0078). The slope of longitudinal change in T1 was greater for tissues that received low-dose irradiation than those that received high-dose irradiation (P=.0488). The effect of dose on the slope of T2 was different (P=.0333) when younger and older patients are compared. CONCLUSIONS: Acute affects of irradiation in muscle are quantifiable via MRI. These models provide evidence that quantifiable MRI parameters may be correlated with patient parameters of radiation dose and clinical factors including patient age. Long-term follow-up will be required to determine if acute changes correlate with clinically significant late effects.


Assuntos
Imageamento por Ressonância Magnética , Neoplasias Musculares/radioterapia , Músculo Esquelético/efeitos da radiação , Lesões por Radiação/diagnóstico , Sarcoma/radioterapia , Criança , Relação Dose-Resposta à Radiação , Humanos , Modelos Lineares , Modelos Biológicos , Neoplasias Musculares/patologia , Músculo Esquelético/patologia , Sarcoma/patologia
14.
Physiol Rep ; 4(7)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27081161

RESUMO

Measuring glomerular filtration rate (GFR) by dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) as part of standard of care clinicalMRIexams (e.g., in pediatric solid tumor patients) has the potential to reduce diagnostic burden. However, enthusiasm for this relatively newGFRtest may be curbed by the limited amount of cross-calibration studies with referenceGFRtechniques and the vast variety ofMRtracer model algorithms causing confusion on the choice of model. To advanceMRI-basedGFRquantification via improvedGFRmodeling and comparison with associated(99m)Tc-DTPA-GFR, 29 long-term Wilms' tumor survivors (19.0-43.3 years, [median 32.0 ± 6.0 years]) treated with nephrectomy, nonnephrotoxic chemotherapy ± radiotherapy underwentMRIwith Gd-DTPAadministration and a(99m)Tc-DTPA GFRtest. ForDCE-MRI-basedGFRestimation, a subject-specific two-compartment (SS-2C) model was developed that uses individual hematocrit values, automatically defines subject-specific uptake intervals, and fits tracer-uptake curves by incorporating these measures. The association between reference(99m)Tc-DTPA GFRandMR-GFRs obtained bySS-2C, three published 2C uptake, and inflow-outflow models was investigated via linear regression analysis. Uptake intervals varied from 64 sec to 141 sec [96 sec ± 21 sec] and hematocrit values ranged from 30% to 49% [41% ± 4%]; these parameters can therefore not be assumed as constants in 2C modeling. OurMR-GFRestimates using theSS-2C model showed accordingly the highest correlation with(99m)Tc-DTPA-GFRs (R(2) = 0.76,P < 0.001) compared with other models (R(2)-range: 0.36-0.66). In conclusion,SS-2C modeling ofDCE-MRIdata improved the association betweenGFRobtained by(99m)Tc-DTPAand Gd-DTPA DCE-MRIto such a degree that this approach could turn into a viable, diagnosticGFRassay without radiation exposure to the patient.


Assuntos
Meios de Contraste/administração & dosagem , Gadolínio DTPA/administração & dosagem , Taxa de Filtração Glomerular , Rim/fisiopatologia , Imageamento por Ressonância Magnética , Modelos Biológicos , Tumor de Wilms/diagnóstico , Adulto , Biomarcadores/sangue , Quimiorradioterapia Adjuvante , Quimioterapia Adjuvante , Creatinina/sangue , Feminino , Hematócrito , Humanos , Rim/diagnóstico por imagem , Modelos Lineares , Masculino , Nefrectomia , Projetos Piloto , Valor Preditivo dos Testes , Cintilografia , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/sangue , Pentetato de Tecnécio Tc 99m/administração & dosagem , Pentetato de Tecnécio Tc 99m/sangue , Fatores de Tempo , Resultado do Tratamento , Tumor de Wilms/sangue , Tumor de Wilms/diagnóstico por imagem , Tumor de Wilms/fisiopatologia , Tumor de Wilms/terapia , Adulto Jovem
15.
J Pediatr Surg ; 47(6): 1250-4, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22703801

RESUMO

BACKGROUND/PURPOSE: The purpose of this study is to determine if patients with osteosarcoma (OS) with metachronous metastatic pulmonary disease presenting with a single pulmonary nodule (SPN) on computed tomography (CT) were found to have other lesions at the time of thoracotomy. METHODS: Data were collected retrospectively on consecutive patients with OS treated at our institution from 1982 to 2007. Patients with no evidence of disease at the end of initial therapy who subsequently relapsed in the lung were identified. RESULTS: In our study, 16 (8%) of 198 patients with OS with metachronous metastatic pulmonary disease presented with a SPN on CT scan. In all patients, only 1 metastatic nodule for OS was found at the time of thoracotomy. The median time between diagnosis and first lung relapse was 23.8 months (range, 4-80 months). Eleven patients (68.7%) subsequently had a second lung relapse, but only 3 patients had involvement of the ipsilateral lung (mean time interval between first and second pulmonary relapses of 17 months; range, 2-44 months). Five-year overall survival from diagnosis was 56.2%. Seven patients (43.8%) died of disease progression. CONCLUSIONS: In our experience, patients with OS with metachronous metastatic pulmonary disease presenting with a SPN on CT were not found to have additional malignant lesions at the time of thoracotomy. Consideration should be given in this group of selected patients to use a minimally invasive approach to nodule removal with image-guided localization, if needed, rather than open thoracotomy because ipsilateral metastases are not likely to be found.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Pulmonares/secundário , Osteossarcoma/secundário , Nódulo Pulmonar Solitário/diagnóstico por imagem , Tomografia Computadorizada Espiral , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/cirurgia , Criança , Pré-Escolar , Terapia Combinada , Progressão da Doença , Intervalo Livre de Doença , Seguimentos , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/epidemiologia , Neoplasias Pulmonares/cirurgia , Osteossarcoma/diagnóstico por imagem , Osteossarcoma/tratamento farmacológico , Osteossarcoma/epidemiologia , Osteossarcoma/cirurgia , Seleção de Pacientes , Estudos Retrospectivos , Terapia de Salvação , Nódulo Pulmonar Solitário/cirurgia , Tennessee/epidemiologia , Cirurgia Torácica Vídeoassistida , Toracotomia , Procedimentos Desnecessários , Adulto Jovem
16.
Mol Cancer Ther ; 9(3): 761-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20197402

RESUMO

Ionizing radiation is an important component of multimodal therapy for alveolar rhabdomyosarcoma (ARMS). We sought to evaluate the ability of IFN-beta to enhance the activity of ionizing radiation. Rh-30 and Rh-41 ARMS cells were treated with IFN-beta and ionizing radiation to assess synergistic effects in vitro and as orthotopic xenografts in CB17 severe combined immunodeficient mice. In addition to effects on tumor cell proliferation and xenograft growth, changes in the tumor microenvironment including interstitial fluid pressure, perfusion, oxygenation, and cellular histology were assessed. A nonlinear regression model and isobologram analysis indicated that IFN-beta and ionizing radiation affected antitumor synergy in vitro in the Rh-30 cell line; the activity was additive in the Rh-41 cell line. In vivo continuous delivery of IFN-beta affected normalization of the dysfunctional tumor vasculature of both Rh-30 and Rh-41 ARMS xenografts, decreasing tumor interstitial fluid pressure, increasing tumor perfusion (as assessed by contrast-enhanced ultrasonography), and increasing oxygenation. Tumors treated with both IFN-beta and radiation were smaller than control tumors and those treated with radiation or IFN-beta alone. Additionally, treatment with high-dose IFN-beta followed by radiation significantly reduced tumor size compared with radiation treatment followed by IFN-beta. The combination of IFN-beta and ionizing radiation showed synergy against ARMS by sensitizing tumor cells to the cytotoxic effects of ionizing radiation and by altering tumor vasculature, thereby improving oxygenation. Therefore, IFN-beta and ionizing radiation may be an effective combination for treatment of ARMS.


Assuntos
Proliferação de Células/efeitos dos fármacos , Interferon beta/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Rabdomiossarcoma Alveolar/radioterapia , Carga Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/radioterapia , Consumo de Oxigênio/efeitos dos fármacos , Radiação Ionizante , Radiossensibilizantes/farmacologia , Proteínas Recombinantes/farmacologia , Rabdomiossarcoma Alveolar/irrigação sanguínea , Rabdomiossarcoma Alveolar/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Pediatr Surg ; 44(10): 1972-6, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19853757

RESUMO

PURPOSE: The incidence of pediatric nonrhabdomyosarcoma soft tissue sarcomas (NRSTSs) of the groin and axilla is unknown, and the optimal surgical approach to these patients is unclear. METHODS: We conducted a retrospective study of patients treated at St Jude Children's Research Hospital from January 1962 to March 2007 for NRSTSs of the groin and axilla. Demographic variables, tumor pathology, clinical management, and outcome were reviewed. RESULTS: Of the 300 patients treated for NRSTSs, only 10 had tumors of the axilla or groin (6 of whom had synovial sarcoma). Surgical interventions included wide resection of the tumor (n = 7), marginal resection (n = 1), subtotal resection (n = 1), and biopsy only (n = 1). Six patients underwent lymph node sampling; all were negative for tumor. Short- and long-term surgical complications were rare. Four patients received adjuvant chemotherapy (n = 3) and/or radiotherapy (n = 2). At a median follow-up of 8.5 years, 7 of the 10 were surviving free of disease. Two of these patients died of tumor progression (1 with metastases at diagnosis and 1 with an unresectable tumor at diagnosis), and one patient who was free of NRSTS died of secondary breast carcinoma. CONCLUSIONS: Pediatric NRSTSs of the axilla and groin are rare, but outcomes are similar to those of other patients with NRSTS. Wide local excision of the tumor with preservation of good limb function should be the surgical goal and may be sufficient therapy in some cases.


Assuntos
Axila/cirurgia , Virilha/cirurgia , Sarcoma/cirurgia , Adolescente , Adulto , Fatores Etários , Causas de Morte , Criança , Feminino , Humanos , Estudos Longitudinais , Masculino , Recidiva Local de Neoplasia/cirurgia , Avaliação de Resultados em Cuidados de Saúde , Estudos Retrospectivos , Rabdomiossarcoma/cirurgia , Fatores de Risco , Sarcoma Sinovial/cirurgia , Neoplasias de Tecidos Moles/cirurgia , Resultado do Tratamento
18.
J Pediatr Surg ; 44(1): 204-10; discussion 210-1, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19159744

RESUMO

PURPOSE: Osteoprotegerin (OPG) inhibits osteoclast activation and reduces osteolysis in bone tumors. We hypothesized that tumor-tropic neural progenitor cells (NPCs) engineered to express OPG would reduce neuroblastoma disease burden in the bone. METHODS: Stable expression of green fluorescent protein (NPC-GFP) and OPG (NPC-OPG) was established in human NPCs by lentivirus-mediated transduction. Bone disease was established by intrafemoral injection of luciferase-expressing human neuroblastoma (CHLA-255) cells into 20 SCID mice. Three weeks later, mice began receiving intravenous injection of 2 x 10(6) NPC-OPG or NPC-GFP (control) every 10 days x 3 doses. Disease was monitored with quantitative bioluminescence imaging and x-ray images, which were evaluated on a scale of 0 to 4. These studies were approved by the Institutional Animal Care and Use Committee. RESULTS: Osteoprotegerin treatment in vitro produced no direct toxicity to tumor cells. Coculture of tumor cells with bone marrow significantly increased activation of bone marrow-derived osteoclasts as assessed by tartrate-resistant acid phosphatase staining (156 +/- 10.8 osteoclasts per well) compared to bone marrow culture alone (91.67 +/- 4.7, P = .005). This increase was abrogated by adding OPG-containing media (68.3 +/- 2.8, P = .001). NPC-OPG slowed tumor progression (108-fold increase from pretreatment) compared to mice treated with NPC-GFP (538-fold), as judged by bioluminescence imaging. X-rays subjectively demonstrated less bone disease in NPC-OPG-treated mice (2.27 +/- 0.25) compared to NPC-GFP-treated mice (3.25 +/- 0.22, P = .04). CONCLUSIONS: Neural progenitor cell-mediated delivery of OPG slowed disease progression in a preclinical model of neuroblastoma bone metastasis. The decrease in bone disease was not from direct tumor cell toxicity but likely occurred indirectly through inhibition of osteoclast-directed bone resorption. Thus, targeted delivery of OPG by NPCs may be effective in the treatment of neuroblastoma bone metastasis.


Assuntos
Neoplasias Ósseas/patologia , Metástase Neoplásica/prevenção & controle , Neuroblastoma/patologia , Osteoprotegerina/farmacologia , Células-Tronco/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Proteínas de Fluorescência Verde/farmacologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID , Estatísticas não Paramétricas , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA