Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 141(3): 244-259, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36206490

RESUMO

Acute myeloid leukemia (AML) is an aggressive blood cancer with poor prognosis. FMS-like tyrosine kinase receptor-3 (FLT3) is one of the major oncogenic receptor tyrosine kinases aberrantly activated in AML. Although protein tyrosine phosphatase PRL2 is highly expressed in some subtypes of AML compared with normal human hematopoietic stem and progenitor cells, the mechanisms by which PRL2 promotes leukemogenesis are largely unknown. We discovered that genetic and pharmacological inhibition of PRL2 significantly reduce the burden of FLT3-internal tandem duplications-driven leukemia and extend the survival of leukemic mice. Furthermore, we found that PRL2 enhances oncogenic FLT3 signaling in leukemia cells, promoting their proliferation and survival. Mechanistically, PRL2 dephosphorylates the E3 ubiquitin ligase CBL at tyrosine 371 and attenuates CBL-mediated ubiquitination and degradation of FLT3, leading to enhanced FLT3 signaling in leukemia cells. Thus, our study reveals that PRL2 enhances oncogenic FLT3 signaling in leukemia cells through dephosphorylation of CBL and will likely establish PRL2 as a novel druggable target for AML.


Assuntos
Leucemia Mieloide Aguda , Ubiquitina-Proteína Ligases , Humanos , Animais , Camundongos , Ubiquitina-Proteína Ligases/metabolismo , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo , Mutação
2.
Pediatr Blood Cancer ; 71(3): e30817, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38189770

RESUMO

BACKGROUND: Ramucirumab is a monoclonal antibody that binds the extracellular domain of vascular endothelial growth factor receptor (VEGFR-2) and prevents binding of VEGF ligands. Based on population pharmacokinetic (PK) analysis and correlation with efficacy in adults, a target steady state trough concentration (Css,min ) ≥ 50 µg/mL was established. PROCEDURES: This phase 1 trial (ADVL1416) used a rolling six design and a PK primary endpoint to define the recommended phase 2 dose (RP2D) of ramucirumab in children with recurrent/refractory solid tumors. Two dose levels (DL) were planned (DL1: 8 mg/kg, DL2: 12 mg/kg administered intravenously [IV] every 2 weeks). Toxicity during the initial 6 weeks was used to assess maximum tolerated dose (MTD). Cycle 1 Day 42 trough (Cmin ) ≥ 50 µg/mL was the target concentration for the PK endpoint. At the RP2D, cohorts for PK expansion and children with central nervous tumors were planned. RESULTS: Twenty-nine patients were enrolled; 28 were eligible; median age [range] = 13.5 [1-21] years; 22 were evaluable for the PK endpoint. Dose-limiting proteinuria occurred at both DLs; however, the MTD was not exceeded. At DL2 (12 mg/kg), the median Day 42 Cmin (n = 16) was 87.8 µg/mL; 15 of 16 patients achieved a Cmin  ≥ 50 µg/mL. CONCLUSION: Ramucirumab was well tolerated in children and adolescents with solid tumors. The RP2D for ramucirumab was 12 mg/kg IV every 2 weeks. This trial demonstrates the feasibility of incorporating a primary PK endpoint to determine dose escalation and the RP2D in children. Studies of ramucirumab in children with selected solid tumors are ongoing.


Assuntos
Neoplasias do Sistema Nervoso Central , Neoplasias , Adulto , Criança , Humanos , Adolescente , Ramucirumab , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Neoplasias/patologia , Anticorpos Monoclonais/uso terapêutico , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Dose Máxima Tolerável
3.
Pediatr Blood Cancer ; 66(9): e27829, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31136068

RESUMO

BCR-ABL1-positive leukemias have historically been classified as either chronic myelogenous leukemia or Ph+ acute lymphoblastic leukemia. Recent analyses suggest there may be a wider range of subtypes. We report a patient with BCR-ABL1 fusion positive T-cell ALL with a previously undescribed cell distribution of the fusion gene. The examination of sorted cells by fluorescence in situ hybridization showed the BCR-ABL1 fusion in the malignant T cells and a subpopulation of the nonmalignant B cells, but not nonmalignant T cells or myeloid or CD34+ progenitor cells providing evidence that the fusion may have occurred in an early lymphoid progenitor.


Assuntos
Proteínas de Fusão bcr-abl , Células Progenitoras Linfoides , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Adolescente , Citometria de Fluxo , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Hibridização In Situ , Células Progenitoras Linfoides/enzimologia , Células Progenitoras Linfoides/patologia , Masculino , Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia
4.
Pediatr Hematol Oncol ; 33(1): 39-48, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26900730

RESUMO

Fever and neutropenia (FN) is a common precipitant for hospitalization among children with cancer, but hospital utilization trends are not well described. This study describes national trends for hospital discharges for FN among children with cancer for the year 2012, compared with the authors' previous analysis from 2009. Data were analyzed from the Kids' Inpatient Database (KID), an all-payer US hospital database, for 2012. Pediatric patients with cancer who had a discharge for FN were identified using age ≤19 years, urgent or emergent admit type, nontransferred, and a combination of International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) codes for fever and neutropenia. The authors evaluated factors associated with a "short length of stay" (SLOS). Sampling weights were used to permit national inferences. In 2012, children with cancer accounted for 1.8% of pediatric hospital discharges (n = 120,675), with 12.2% (n = 13,456) of cancer-related discharges meeting FN criteria. Two fifths of FN discharges had a SLOS, which accounted for $91 million (2015 US$) in hospital charges. The majority had no serious infections; most common infections were viral infection (9.6%) or upper respiratory infection (9.6%). Factors significantly associated with SLOS included having a diagnosis of ear infection (odds ratio [OR] = 1.54, 95% confidence interval [CI]: 1.16-2.03), soft tissue sarcoma (OR = 1.47, CI: 1.10-1.95), and Hodgkin lymphoma (OR = 1.51, CI: 1.09-2.10), as compared with not having those diagnoses. SLOS admissions continue to be rarely associated with serious infections, but contribute substantially to the burden of hospitalization for pediatric FN. Implementation of risk stratification schemas to identify patients who meet low-risk criteria may decrease financial burden.


Assuntos
Bases de Dados Factuais , Febre/epidemiologia , Neoplasias/epidemiologia , Neutropenia/epidemiologia , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Febre/terapia , Humanos , Lactente , Masculino , Neoplasias/terapia , Neutropenia/terapia , Alta do Paciente , Estados Unidos
5.
Mol Cancer Res ; 22(1): 94-103, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37756563

RESUMO

Receptor tyrosine kinase KIT is frequently activated in acute myeloid leukemia (AML). While high PRL2 (PTP4A2) expression is correlated with activation of SCF/KIT signaling in AML, the underlying mechanisms are not fully understood. We discovered that inhibition of PRL2 significantly reduces the burden of oncogenic KIT-driven leukemia and extends leukemic mice survival. PRL2 enhances oncogenic KIT signaling in leukemia cells, promoting their proliferation and survival. We found that PRL2 dephosphorylates CBL at tyrosine 371 and inhibits its activity toward KIT, leading to decreased KIT ubiquitination and enhanced AKT and ERK signaling in leukemia cells. IMPLICATIONS: Our studies uncover a novel mechanism that fine-tunes oncogenic KIT signaling in leukemia cells and will likely identify PRL2 as a novel therapeutic target in AML with KIT mutations.


Assuntos
Leucemia Mieloide Aguda , Monoéster Fosfórico Hidrolases , Animais , Camundongos , Leucemia Mieloide Aguda/genética , Mutação , Monoéster Fosfórico Hidrolases/genética , Fosforilação , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais/genética
6.
Lancet Oncol ; 13(12): 1218-24, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23099009

RESUMO

BACKGROUND: Plexiform neurofibromas are slow-growing chemoradiotherapy-resistant tumours arising in patients with neurofibromatosis type 1 (NF1). Currently, there are no viable therapeutic options for patients with plexiform neurofibromas that cannot be surgically removed because of their proximity to vital body structures. We undertook an open-label phase 2 trial to test whether treatment with imatinib mesylate can decrease the volume burden of clinically significant plexiform neurofibromas in patients with NF1. METHODS: Eligible patients had to be aged 3-65 years, and to have NF1 and a clinically significant plexiform neurofibroma. Patients were treated with daily oral imatinib mesylate at 220 mg/m(2) twice a day for children and 400 mg twice a day for adults for 6 months. The primary endpoint was a 20% or more reduction in plexiform size by sequential volumetric MRI imaging. Clinical data were analysed on an intention-to-treat basis; a secondary analysis was also done for those patients able to take imatinib mesylate for 6 months. This trial is registered with ClinicalTrials.gov, number NCT01673009. FINDINGS: Six of 36 patients (17%, 95% CI 6-33), enrolled on an intention-to-treat basis, had an objective response to imatinib mesylate, with a 20% or more decrease in tumour volume. Of the 23 patients who received imatinib mesylate for at least 6 months, six (26%, 95% CI 10-48) had a 20% or more decrease in volume of one or more plexiform tumours. The most common adverse events were skin rash (five patients) and oedema with weight gain (six). More serious adverse events included reversible grade 3 neutropenia (two), grade 4 hyperglycaemia (one), and grade 4 increases in aminotransferase concentrations (one). INTERPRETATION: Imatinib mesylate could be used to treat plexiform neurofibromas in patients with NF1. A multi-institutional clinical trial is warranted to confirm these results. FUNDING: Novartis Pharmaceuticals, the Indiana University Simon Cancer Centre, and the Indiana University Herman B Wells Center for Pediatric Research.


Assuntos
Antineoplásicos/uso terapêutico , Neurofibroma Plexiforme/tratamento farmacológico , Neurofibromatose 1/complicações , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Adolescente , Adulto , Benzamidas , Criança , Pré-Escolar , Feminino , Humanos , Mesilato de Imatinib , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neurofibroma Plexiforme/complicações , Neurofibroma Plexiforme/patologia , Adulto Jovem
7.
Front Pediatr ; 11: 1183295, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37292376

RESUMO

Background: The survival rates for pediatric patients with relapsed and refractory tumors are poor. Successful treatment strategies are currently lacking and there remains an unmet need for novel therapies for these patients. We report here the results of a phase 1 study of talimogene laherparepvec (T-VEC) and explore the safety of this oncolytic immunotherapy for the treatment of pediatric patients with advanced non-central nervous system tumors. Methods: T-VEC was delivered by intralesional injection at 106 plaque-forming units (PFU)/ml on the first day, followed by 108 PFU/ml on the first day of week 4 and every 2 weeks thereafter. The primary objective was to evaluate the safety and tolerability as assessed by the incidence of dose-limiting toxicities (DLTs). Secondary objectives included efficacy indicated by response and survival per modified immune-related response criteria simulating the Response Evaluation Criteria in Solid Tumors (irRC-RECIST). Results: Fifteen patients were enrolled into two cohorts based on age: cohort A1 (n = 13) 12 to ≤21 years old (soft-tissue sarcoma, n = 7; bone sarcoma, n = 3; neuroblastoma, n = 1; nasopharyngeal carcinoma, n = 1; and melanoma, n = 1) and cohort B1 (n = 2) 2 to <12 years old (melanoma, n = 2). Overall, patients received treatment for a median (range) of 5.1 (0.1, 39.4) weeks. No DLTs were observed during the evaluation period. All patients experienced at least one treatment-emergent adverse event (TEAE), and 53.3% of patients reported grade ≥3 TEAEs. Overall, 86.7% of patients reported treatment-related TEAEs. No complete or partial responses were observed, and three patients (20%) overall exhibited stable disease as the best response. Conclusions: T-VEC was tolerable as assessed by the observation of no DLTs. The safety data were consistent with the patients' underlying cancer and the known safety profile of T-VEC from studies in the adult population. No objective responses were observed. Trial Registration: ClinicalTrials.gov: NCT02756845. https://clinicaltrials.gov/ct2/show/NCT02756845.

8.
Semin Oncol Nurs ; 37(3): 151162, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34148742

RESUMO

OBJECTIVES: Phase 1 clinical trials are essential in the development of novel therapies for childhood cancers. Children with cancer can participate in phase 1 clinical trials when no known curative therapy remains. Understanding the experiences of children and their families in these clinical trials can help ensure that participation supports the children's and parents' well-being. This article explores the specific aspects of pediatric oncology phase 1 trials that parents found particularly challenging. DATA SOURCES: This qualitative, empirical phenomenology study considered 11 parents' experiences during the time their child with cancer participated in a phase 1 clinical trial. The primary study results were previously reported. This article reports parents' insights into the processes and procedures that occurred as part of participation in a pediatric oncology phase 1 trial. CONCLUSION: Parents' experiences during the phase 1 clinical trials were primarily positive. However, data analysis revealed five aspects of these trials that were challenging for families: learning about clinical trials, being referred to another institution, research-only procedures, adhering to trial requirements, and oral medications. IMPLICATIONS FOR NURSING PRACTICE: Although experiences during phase 1 clinical trials were positive overall, opportunities to enhance children's and parents' experiences warrant attention. Enhancing the education provided to families during recruitment and minimizing the logistical burdens associated with trial requirements through care coordination may alleviate challenges experienced by children and parents.


Assuntos
Neoplasias , Pais , Criança , Humanos , Oncologia , Neoplasias/terapia , Relações Profissional-Família , Pesquisa Qualitativa
9.
Eur J Clin Pharmacol ; 66(1): 77-86, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19798490

RESUMO

PURPOSE: Until recently, ondansetron was approved for the prevention of nausea and vomiting only in patients older than 2 years. However, as the use of ondansetron in patients younger than 2 years had been documented, characterization of ondansetron pharmacokinetics in this younger pediatric age group was warranted. METHODS: The pharmacokinetics of intravenously administered ondansetron were evaluated in oncology and surgical patients aged 1-48 months. Pooled data from 124 patients, including 745 pharmacokinetic samples, were analyzed using nonlinear mixed-effects modeling. RESULTS: Ondansetron pharmacokinetics were described by a two-compartment model. Body-size effects on ondansetron disposition were accounted for via standard allometric relationships, normalized to 10.4 kg. A maturation process with a half-life of approximately 4 months was incorporated to describe a decrease in clearance (CL) in infants. Clearance [95% confidence interval (CI)] for a typical patient was 1.53 (1.34-1.78) L/h/kg(0.75) with an interindividual variability of 56.8%. Ondansetron CL was reduced by 31%, 53%, and 76% for the typical 6-month-, 3-month-, and 1-month-old patient, respectively. Simulations showed that an ondansetron dose of 0.1 mg/kg in children younger than 6 months produced exposure similar to a 0.15-mg/kg dose in older children. CONCLUSIONS: The population pharmacokinetic analysis of ondansetron allows for characterization of individual patients based on body weight and age. It is recommended that patients younger than 4 months receiving ondansetron be closely monitored.


Assuntos
Anestesia Geral/efeitos adversos , Antieméticos/farmacocinética , Antineoplásicos/efeitos adversos , Náusea/induzido quimicamente , Ondansetron/farmacocinética , Antagonistas da Serotonina/farmacocinética , Antieméticos/administração & dosagem , Antieméticos/uso terapêutico , Pré-Escolar , Simulação por Computador , Feminino , Humanos , Lactente , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Náusea/prevenção & controle , Ondansetron/administração & dosagem , Ondansetron/uso terapêutico , Antagonistas da Serotonina/administração & dosagem , Antagonistas da Serotonina/uso terapêutico
10.
J Clin Oncol ; 23(25): 6172-80, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16135484

RESUMO

PURPOSE: Epidermal growth factor receptor is expressed in pediatric malignant solid tumors. We conducted a phase I trial of gefitinib, an epidermal growth factor receptor tyrosine kinase inhibitor, in children with refractory solid tumors. PATIENTS AND METHODS: Gefitinib (150, 300, 400, or 500 mg/m2) was administered orally to cohorts of three to six patients once daily continuously until disease progression or significant toxicity. Pharmacokinetic studies were performed during course one (day 1 through 28). RESULTS: Of the 25 enrolled patients, 19 (median age, 15 years) were fully evaluable for toxicity and received 54 courses. Dose-limiting toxicity was rash in two patients treated with 500 mg/m2 and elevated ALT and AST in one patient treated with 400 mg/m2. The maximum-tolerated dose was 400 mg/m2/d. The most frequent non-dose-limiting toxicities were grade 1 or 2 dry skin, anemia, diarrhea, nausea, and vomiting. One patient with Ewing's sarcoma had a partial response. Disease stabilized for 8 to > or = 60 weeks in two patients with Wilms' tumor and two with brainstem glioma (one exophytic). At 400 mg/m2, the median peak gefitinib plasma concentration was 2.2 microg/mL (range, 1.2 to 3.6 microg/mL) and occurred at a median of 2.3 hours (range, 2.0 to 8.3 hours) after drug administration. The median apparent clearance and median half-life were 14.8 L/h/m2 (range, 3.8 to 24.8 L/h/m2) and 11.7 hours (range, 5.6 to 22.8 hours), respectively. Gefitinib systemic exposures were comparable with those associated with antitumor activity in adults. CONCLUSION: Oral gefitinib is well tolerated in children. Development of the drug in combination with cytotoxic chemotherapy will be pursued.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Neoplasias/tratamento farmacológico , Quinazolinas/efeitos adversos , Quinazolinas/farmacocinética , Administração Oral , Adolescente , Adulto , Fatores Etários , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Criança , Pré-Escolar , Receptores ErbB/antagonistas & inibidores , Feminino , Gefitinibe , Humanos , Lactente , Masculino , Quinazolinas/administração & dosagem , Quinazolinas/uso terapêutico , Resultado do Tratamento
11.
Curr Opin Mol Ther ; 7(5): 461-6, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16248281

RESUMO

Fanconi anemia (FA) is a genetic disease characterized by progressive, fatal bone marrow failure, congenital anomalies and predisposition to cancer. Although stem cell transplantation is therapeutic, human leukocyte antigen-identical sibling donors are available to a minority of patients. In murine models and human cells in vitro, gene transfer corrects the FA cellular phenotype of chromosomal breakage in response to DNA-damaging agents, suggesting therapeutic use of gene transfer is possible. However, disease-specific characteristics make application of viral vector technology difficult. Multiple studies are currently underway to develop a gene therapy approach for treating this disease, including phase I trials.


Assuntos
Anemia de Fanconi/terapia , Técnicas de Transferência de Genes , Terapia Genética , Células-Tronco Hematopoéticas , Ensaios Clínicos como Assunto , Anemia de Fanconi/genética , Terapia Genética/efeitos adversos , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Humanos , Técnicas In Vitro
12.
Blood Coagul Fibrinolysis ; 23(1): 39-44, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22089938

RESUMO

Postthrombotic syndrome (PTS) is a chronic morbidity of venous thromboembolism (VTE) in children. Information about the evolution of PTS is lacking in children. Present study was aimed to evaluate the time-course of extremity PTS in children who were serially followed in a hematology clinic. This retrospective cohort study included 69 consecutive children with documented VTEs that presented with symptoms of extremity VTE: 67 extremity VTEs with or without extension to vena cava, 2 inferior vena cava VTEs. Severity of PTS was assessed using modified Villalta scale. Median age of the cohort was 12.6 years (interquartile range 1.6-15 years) while median follow-up was 28.7 months (interquartile range 13.3-33.4 months. PTS prevalence was 46.8% [95% confidence interval (CI) 37.9-57.7%]. Lower extremity VTE was associated with development of PTS compared to upper extremity VTE regardless of catheter use (P = 0.002). The time-course of PTS fluctuated in 11 of 33 children (33%; 95% CI 20-47%) at a median interval of 12 months from diagnosis of VTE (range 4-14 months): three progressed from mild/moderate to severe, one improved from moderate to mild, seven fluctuated between mild and moderate. Recurrence and incomplete resolution of VTE were associated with variability in PTS severity (P < 0.05). In summary, this study suggested that almost 50% of study cohort developed PTS, and the time-course of PTS was not static in one third of children. Future research should focus on identifying the predictors contributing to the worsening of PTS and developing risk-stratified treatment interventions so as to improve the outcome of children with VTE.


Assuntos
Síndrome Pós-Trombótica/patologia , Tromboembolia Venosa/patologia , Adolescente , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Lactente , Masculino , Síndrome Pós-Trombótica/tratamento farmacológico , Prevalência , Recidiva , Estudos Retrospectivos , Fatores de Risco
13.
J Pediatr Hematol Oncol ; 27(8): 441-3, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16096528

RESUMO

Li-Fraumeni syndrome and the LF-like syndrome, rare heritable conditions that predispose to the development of malignancy, are associated with germline mutations of the tumor suppressor gene p53. The authors describe a 14-month-old boy who presented with synchronous rhabdomyosarcoma and adrenal cortical carcinoma and a novel mutation of the p53 gene. Analysis of exons 2 through 11 of the p53 gene using the polymerase chain reaction and DNA sequencing revealed a mutation of codon 273. Although codon 273 is a known hotspot region for p53 mutation, the patient's mutation, R273H, has not been associated with development of adrenal cortical carcinoma.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Carcinoma/genética , Genes p53 , Mutação em Linhagem Germinativa , Síndrome de Li-Fraumeni/complicações , Neoplasias Primárias Múltiplas/genética , Rabdomiossarcoma/genética , Neoplasias do Córtex Suprarrenal/patologia , Carcinoma/patologia , Códon , Humanos , Lactente , Síndrome de Li-Fraumeni/genética , Masculino , Neoplasias Primárias Múltiplas/patologia , Rabdomiossarcoma/patologia , Fatores de Risco
14.
Am J Hematol ; 78(3): 181-7, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15726607

RESUMO

Data were reviewed on treatment patterns, outcome, and hospital charges for children with idiopathic thrombocytopenic purpura (ITP). Records of 186 children with ITP, seen between January 1997 and April 2001, were reviewed. Hospital charges for initial management and first re-treatment were calculated by combining physician, hospital, and pathology charges. Anti-D immune globulin [anti-D IG] was used in 32.3%, intravenous immune globulin [IVIG] in 22.6%, steroids in 22.6%, combination therapies in 8%, and 14.5% were observed. Two patients had CNS bleeding, one with intraventricular hemorrhage at diagnosis, and the other with a parietal bleed 1 year from diagnosis. There was no significant differences in time to reach platelet counts of 20, 50, or 150 (x 10(9)/L) across different treatment groups. There was no significant difference in median charges for the IVIG and anti-D IG groups for the initial treatment of ITP. However, the IVIG was significantly more expensive than steroids or observation. Charges for the anti-D IG group were higher than the observation group but not the steroid group. After drug charges were excluded, patients in the IVIG group had statistically higher charges compared to patients in anti-D IG group. Almost half the patients were re-treated. There was no significant difference between anti-D IG, IVIG, and steroid groups when initial and re-treatment charges were combined. The observation group remained least expensive. Outcome for children with ITP is similar regardless of initial management. There is not a statistically significant difference in hospital charges between patients treated with anti-D IG and IVIG. The IVIG-treated group tends to be more costly, but this is not due to drug charges. Re-treatment is common and decreases the difference in patient charges among initial therapies.


Assuntos
Honorários e Preços , Imunoglobulinas Intravenosas/uso terapêutico , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Imunoglobulina rho(D)/uso terapêutico , Esteroides/uso terapêutico , Adolescente , Criança , Pré-Escolar , Quimioterapia Combinada , Honorários Farmacêuticos , Feminino , Preços Hospitalares , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Imunoglobulinas Intravenosas/economia , Lactente , Masculino , Contagem de Plaquetas , Púrpura Trombocitopênica Idiopática/sangue , Púrpura Trombocitopênica Idiopática/economia , Estudos Retrospectivos , Imunoglobulina rho(D)/administração & dosagem , Imunoglobulina rho(D)/economia , Esteroides/administração & dosagem , Esteroides/economia , Resultado do Tratamento
15.
Curr Hematol Rep ; 2(4): 335-40, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12901331

RESUMO

Fanconi anemia is a hereditary syndrome of bone marrow failure, congenital anomalies, and a predisposition to malignancy. Most patients die from bone marrow failure. Cells from patients display a heightened sensitivity to DNA cross-linking agents with increased chromosomal breakage and increased cytotoxicity. Bone marrow from patients with Fanconi anemia have decreased numbers of hematopoietic progenitors when grown in culture. Transfer of the normal Fanconi anemia cDNA into cells from patients corrects the laboratory abnormalities, suggesting that gene transfer may prevent or reverse the bone marrow failure. Advances in gene transfer into human hematopoietic cells make this approach seem feasible. However, decreased numbers of stem cell targets may represent a significant obstacle. In addition, new insights on potential toxicities related to gene transfer have heightened a cautious approach. Fanconi anemia represents a prototype disorder for gene therapy and highlights the difficulties in adapting this technology to human disease.


Assuntos
Proteínas de Ciclo Celular , Anemia de Fanconi/terapia , Terapia Genética/métodos , Animais , Medula Óssea/patologia , Proteínas de Ligação a DNA , Proteína do Grupo de Complementação A da Anemia de Fanconi , Proteína do Grupo de Complementação D2 da Anemia de Fanconi , Proteína do Grupo de Complementação F da Anemia de Fanconi , Proteína do Grupo de Complementação G da Anemia de Fanconi , Proteínas de Grupos de Complementação da Anemia de Fanconi , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Proteínas Nucleares , Proteínas/genética , Proteínas de Ligação a RNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa