Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Nucl Med Mol Imaging ; 49(5): 1574-1583, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34837510

RESUMO

PURPOSE: Paediatric high-risk neuroblastoma has poor prognosis despite modern multimodality therapy. This phase I/II study aimed to determine the safety, dose-limiting toxicity (DLT), and efficacy of high-dose 131I-meta-iodobenzylguanidine (131I-mIBG) therapy combined with single high-dose chemotherapy (HDC) and haematopoietic stem cell transplantation (HSCT) in high-risk neuroblastoma in Japan. METHODS: Patients received 666 MBq/kg of 131I-mIBG and single HDC and HSCT from autologous or allogeneic stem cell sources. The primary endpoint was DLT defined as adverse events associated with 131I-mIBG treatment posing a significant obstacle to subsequent HDC. The secondary endpoints were adverse events/reactions, haematopoietic stem cell engraftment and responses according to the Response Evaluation Criteria in Solid Tumours version 1.1 (RECIST 1.1) and 123I-mIBG scintigraphy. Response was evaluated after engraftment. RESULTS: We enrolled eight patients with high-risk neuroblastoma (six females; six newly diagnosed and two relapsed high-risk neuroblastoma; median age, 4 years; range, 1-10 years). Although all patients had adverse events/reactions after high-dose 131I-mIBG therapy, we found no DLT. Adverse events and reactions were observed in 100% and 25% patients during single HDC and 100% and 12.5% patients during HSCT, respectively. No Grade 4 complications except myelosuppression occurred during single HDC and HSCT. The response rate according to RECIST 1.1 was observed in 87.5% (7/8) in stable disease and 12.5% (1/8) were not evaluated. Scintigraphic response occurred in 62.5% (5/8) and 37.5% (3/8) patients in complete response and stable disease, respectively. CONCLUSION: 131I-mIBG therapy with 666 MBq/kg followed by single HDC and autologous or allogeneic SCT is safe and efficacious in patients with high-risk neuroblastoma and has no DLT. TRIAL REGISTRATION NUMBER: jRCTs041180030. NAME OF REGISTRY: Feasibility of high-dose iodine-131-meta-iodobenzylguanidine therapy for high-risk neuroblastoma preceding myeloablative chemotherapy and haematopoietic stem cell transplantation (High-dose iodine-131-meta-iodobenzylguanidine therapy for high-risk neuroblastoma). URL OF REGISTRY: https://jrct.niph.go.jp/en-latest-detail/jRCTs041180030 . DATE OF ENROLMENT OF THE FIRST PARTICIPANT TO THE TRIAL: 12/01/2018.


Assuntos
3-Iodobenzilguanidina , Neuroblastoma , 3-Iodobenzilguanidina/administração & dosagem , 3-Iodobenzilguanidina/efeitos adversos , Criança , Pré-Escolar , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Radioisótopos do Iodo , Masculino , Neuroblastoma/radioterapia , Transplante Autólogo
2.
Am J Physiol Heart Circ Physiol ; 317(2): H226-H233, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31149841

RESUMO

Cardiac sympathetic overdrive provides inotropic support to the failing heart. However, as myocardial insult evolves, this compensatory response impairs contractile function and constitutes an independent mortality predictor and a primary target in the treatment of heart failure (HF). In this prospective, randomized, double-blind, controlled crossover trial, we proposed cervicothoracic transcutaneous electrical nerve stimulation (CTENS) as a nonpharmacological therapy on cardiac sympathetic activity in patients with HF. Seventeen patients with HF were randomly assigned to an in-home CTENS (30 min twice daily, 80-Hz frequency, and 150-µs pulse duration) or a control intervention (Sham) for 14 consecutive days. Following a 60-day washout phase, patients were crossed over to the opposite intervention. The heart-to-mediastinum ratio (HMR) and washout rate (WR) (indexes of sympathetic innervation density and activity from planar 123iodo-metaiodobenzylguanidine myocardial scintigraphy images, respectively), as well as blood pressure (BP) and heart rate (HR), were quantified before and after each intervention. HMR, BP, and HR did not change throughout the study. Nonetheless, CTENS reduced WR (CTENS -4 ± 10 vs. Sham +5 ± 15%, P = 0.03) when compared with Sham. When allocated in two independent groups, preserved (PCSI, HMR > 1.6, n = 10) and impaired cardiac sympathetic innervation (ICSI, HRM ≤1.6, n = 7), PCSI patients showed an important attenuation of WR (-11 ± 9 vs. Sham +8 ± 19%, P = 0.007) after CTENS. Nonetheless, neither Sham nor CTENS evoked changes in WR of the ICSI patients (P > 0.05). These findings indicate that CTENS attenuates the cardiac sympathetic overdrive in patients with HF and a preserved innervation constitutes an essential factor for this beneficial neuromodulatory impact. Clinical Trial Registration: URL: https://www.clinicaltrials.gov . Identifier: NCT03354689. NEW & NOTEWORTHY We found that short-term cervicothoracic transcutaneous electrical nerve stimulation (CTENS) attenuates cardiac sympathetic overdrive in patients with heart failure and a preserved autonomic innervation may constitute an essential factor to maximize this beneficial neuromodulatory effect. CTENS then emerges as an alternative noninvasive and nonpharmacological strategy to attenuate exaggerated cardiac sympathetic drive in patients with heart failure.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , Insuficiência Cardíaca/terapia , Coração/inervação , Radioisótopos do Iodo/administração & dosagem , Contração Miocárdica , Compostos Radiofarmacêuticos/administração & dosagem , Sistema Nervoso Simpático/fisiopatologia , Estimulação Elétrica Nervosa Transcutânea , Idoso , Pressão Sanguínea , Brasil , Estudos Cross-Over , Método Duplo-Cego , Feminino , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Sistema Nervoso Simpático/diagnóstico por imagem , Fatores de Tempo , Estimulação Elétrica Nervosa Transcutânea/efeitos adversos , Resultado do Tratamento
3.
J Hematol Oncol ; 10(1): 108, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28511709

RESUMO

BACKGROUND: In our previous SMC NB-2004 study of patients with high-risk neuroblastomas, which incorporated total-body irradiation (TBI) with second high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT), the survival rate was encouraging; however, short- and long-term toxicities were significant. In the present SMC NB-2009 study, only TBI was replaced with 131I-meta-iodobenzylguanidine (MIBG) treatment in order to reduce toxicities. METHODS: From January 2009 to December 2013, 54 consecutive patients were assigned to receive tandem HDCT/auto-SCT after nine cycles of induction chemotherapy. The CEC (carboplatin + etoposide + cyclophosphamide) regimen and the TM (thiotepa + melphalan) regimen with (for metastatic MIBG avid tumors) or without (for localized or MIBG non-avid tumors) 131I-MIBG treatment (18 or 12 mCi/kg) were used for tandem HDCT/auto-SCT. Local radiotherapy, differentiation therapy with 13-cis-retinoic acid, and immunotherapy with interleukin-2 were administered after tandem HDCT/auto-SCT. RESULTS: Fifty-two patients underwent the first HDCT/auto-SCT and 47 patients completed tandem HDCT/auto-SCT. There was no significant immediate toxicity during the 131I-MIBG infusion. Acute toxicities during the tandem HDCT/auto-SCT were less severe in the NB-2009 study than in the NB-2004 study. Late effects such as growth hormone deficiency, cataracts, and glomerulopathy evaluated at 3 years after the second HDCT/auto-SCT were also less significant in the NB-2009 study than in NB-2004 study. There was no difference in the 5-year event-free survival (EFS) between the two studies (67.5 ± 6.7% versus 58.3 ± 6.9%, P = 0.340). CONCLUSIONS: Incorporation of high-dose 131I-MIBG treatment into tandem HDCT/auto-SCT could reduce short- and long-term toxicities associated with TBI, without jeopardizing the survival rate. TRIAL REGISTRATION: ClinicalTrials.gov NCT03061656.


Assuntos
3-Iodobenzilguanidina/uso terapêutico , Antineoplásicos/uso terapêutico , Neuroblastoma/terapia , Transplante de Células-Tronco/métodos , 3-Iodobenzilguanidina/administração & dosagem , Adolescente , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/administração & dosagem , Carboplatina/uso terapêutico , Criança , Pré-Escolar , Terapia Combinada/métodos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Etoposídeo/administração & dosagem , Etoposídeo/uso terapêutico , Humanos , Quimioterapia de Indução/métodos , Lactente , Melfalan/administração & dosagem , Melfalan/uso terapêutico , Tiotepa/administração & dosagem , Tiotepa/uso terapêutico , Transplante Autólogo/métodos , Adulto Jovem
4.
Pediatr Blood Cancer ; 57(7): 1124-9, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21495159

RESUMO

BACKGROUND: Targeted radiotherapy with (131) I-Metaiodobenzylguanidine ((131) I-MIBG) is safe and effective therapy for patients with relapsed neuroblastoma, but anti-tumor activity is sometimes transient. The goal of this study was to determine the safety and efficacy of early (<100 days) second (131) I-MIBG treatment following an effective initial treatment. PROCEDURES: After an initial infusion of 18 mCi/kg (131) I-MIBG, patients with tumor response or stable disease (SD), and available hematopoietic stem cell product, were eligible for additional (131) I-MIBG therapy. Residual thrombocytopenia did not preclude patients from receiving additional treatment. Subsequent treatment was administered a minimum of 6 weeks and maximum 100 days from initial infusion, and subjects could receive subsequent therapy if the same criteria were met. RESULTS: Seventy-six heavily pretreated patients (median 4 prior chemotherapy regimens, range 1-8) with relapsed neuroblastoma were treated with (131) I-MIBG. Response rate to the first infusion was 30%, with 49% showing SD. Response rate among the 41 patients receiving a subsequent second infusion was 29%. After two treatments, 39% of patients experienced a reduction in overall disease burden. Four of five complete responses (CRs) to the initial infusion were maintained, despite all five having disease readily apparent on immediate post-second treatment (131) I-MIBG scanning. Hematologic toxicity was managed with early PBSC support after the second therapy (median: 15 days). CONCLUSIONS: Early second (131) I-MIBG safely reduces disease burden in patients with relapsed neuroblastoma. Patients with CR by conventional (123) I-MIBG scintigraphy may have substantial disease burden apparent on high-dose (131) I-MIBG scintigraphy, supporting consolidation with subsequent (131) I-MIBG therapy in cases of apparent complete remission. Pediatr Blood Cancer 2011; 57: 1124-1129. © 2011 Wiley Periodicals, Inc.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Radioisótopos do Iodo/administração & dosagem , Recidiva Local de Neoplasia/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , 3-Iodobenzilguanidina/efeitos adversos , Antineoplásicos/efeitos adversos , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Feminino , Humanos , Infusões Intravenosas , Radioisótopos do Iodo/efeitos adversos , Estimativa de Kaplan-Meier , Masculino , Estadiamento de Neoplasias , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Resultado do Tratamento
5.
Endocr Relat Cancer ; 18(1): 143-57, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21098082

RESUMO

[¹³¹I]meta-iodobenzylguanidine ([¹³¹I]MIBG) is the most commonly used treatment for metastatic pheochromocytoma and paraganglioma. It enters the chromaffin cells via the membrane norepinephrine transporter; however, its success has been modest. We studied the ability of histone deacetylase (HDAC) inhibitors to enhance [¹²³I]MIBG uptake by tumors in a mouse metastatic pheochromocytoma model. HDAC inhibitors are known to arrest growth, induce differentiation and apoptosis in various cancer cells, and further inhibit tumor growth. We report the in vitro and in vivo effects of two HDAC inhibitors, romidepsin and trichostatin A, on the uptake of [(3)H]norepinephrine, [¹²³I]MIBG, and [(18)F]fluorodopamine in a mouse model of metastatic pheochromocytoma. The effects of both inhibitors on norepinephrine transporter activity were assessed in mouse pheochromocytoma (MPC) cells by using the transporter-blocking agent desipramine and the vesicular-blocking agent reserpine. HDAC inhibitors increased [(3)H]norepinephrine, [¹²³I]MIBG, and [(18)F]fluorodopamine uptake through the norepinephrine transporter in MPC cells. In vivo, inhibitor treatment resulted in significantly increased uptake of [(18)F]fluorodopamine positron emission tomography (PET) in pheochromocytoma liver metastases (19.1 ± 3.2% injected dose per gram of tumor (%ID/g) compared to liver metastases in pretreatment scans 5.9 ± 0.6%; P<0.001). Biodistribution analysis after inhibitors treatment confirmed the PET results. The uptake of [(123)I]MIBG was significantly increased in liver metastases 9.5 ± 1.1% compared to 3.19 ± 0.4% in untreated control liver metastases (P<0.05). We found that HDAC inhibitors caused an increase in the amount of norepinephrine transporter expressed in tumors. HDAC inhibitors may enhance the therapeutic efficacy of [(131)I]MIBG treatment in patients with advanced malignant pheochromocytoma and paraganglioma.


Assuntos
3-Iodobenzilguanidina/farmacocinética , Neoplasias das Glândulas Suprarrenais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Dopamina/análogos & derivados , Inibidores de Histona Desacetilases/administração & dosagem , Norepinefrina/farmacocinética , Feocromocitoma , 3-Iodobenzilguanidina/administração & dosagem , Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Terapia Combinada , Dopamina/farmacocinética , Feminino , Radioisótopos de Flúor/administração & dosagem , Radioisótopos de Flúor/farmacocinética , Inibidores de Histona Desacetilases/uso terapêutico , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/farmacocinética , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Camundongos , Camundongos Knockout , Camundongos Nus , Transplante de Neoplasias , Norepinefrina/administração & dosagem , Feocromocitoma/diagnóstico por imagem , Feocromocitoma/tratamento farmacológico , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Cintilografia , Compostos Radiofarmacêuticos/farmacocinética , Regulação para Cima
6.
Cancer Biother Radiopharm ; 24(4): 469-75, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19694582

RESUMO

This is a first of many phase 1 study of Ultratrace Iobenguane I-131 (Ultratrace 131I-MIBG; Molecular Insight Pharmaceuticals, Inc., Cambridge, MA). High-specific-activity Ultratrace 131I-MIBG may provide improved efficacy and tolerability over carrier-added 131I-MIBG. We investigated the pharmacokinetics (PK), radiation dosimetry, and clinical safety in 11 patients with confirmed pheochromocytoma/paraganglioma (Pheo) or carcinoid tumors. A single 5.0-mCi (185 MBq) injection of Ultratrace 131I-MIBG, supplemented with 185 microg of unlabeled MIBG to simulate the amount of MIBG anticipated in a therapeutic dose, was administered. Over 120 hours postdose, blood and urine were collected for PK, and sequential whole-body planar imaging was performed. Patients were followed for adverse events for 2 weeks. Ultratrace 131I-MIBG is rapidly cleared from the blood and excreted in urine (80.3% +/- 2.8% of dose at 120 hours). For a therapeutic administration of 500 mCi (18.5 GBq), our estimate of the projected dose is 1.4 Gy for marrow and 10.4 Gy for kidneys. Safety results showed 12 mild adverse events, all considered unrelated to study drug, in 8 of 11 patients. These findings support the further development of Ultratrace 131I-MIBG for the treatment of neuroendocrine tumors, such as metastatic Pheo and carcinoid.


Assuntos
3-Iodobenzilguanidina/efeitos adversos , 3-Iodobenzilguanidina/farmacocinética , Neoplasias das Glândulas Suprarrenais/radioterapia , Radioisótopos do Iodo/efeitos adversos , Radioisótopos do Iodo/farmacocinética , Paraganglioma/radioterapia , Feocromocitoma/radioterapia , 3-Iodobenzilguanidina/administração & dosagem , Neoplasias das Glândulas Suprarrenais/metabolismo , Adulto , Relação Dose-Resposta à Radiação , Feminino , Humanos , Radioisótopos do Iodo/administração & dosagem , Masculino , Paraganglioma/metabolismo , Feocromocitoma/metabolismo , Doses de Radiação , Radiometria/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/efeitos adversos , Compostos Radiofarmacêuticos/farmacocinética
7.
Biol Blood Marrow Transplant ; 15(9): 1077-85, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19660720

RESUMO

We evaluated the feasibility and efficacy of using high-dose iodine-131-metaiodobenzylguanidine ((131)I-MIBG) followed by reduced-intensity conditioning (RIC) and transplantation of T cell-depleted haploidentical peripheral blood stem cells (designated haplo-SCT) to treat relapsing/refractory neuroblastoma (RRNB). Five RRNB patients were enrolled: 4 with relapse (3 after autologous SCT) and 1 with induction therapy failure. The preparative regimen included high-dose (131)I-MIBG on day -20, followed by fludarabine (Flu), thiotepa, and melphalan (Mel) from day -8 to -1. Granulocyte-colony stimulating factor (G-CSF)-mobilized, T cell-depleted haploidentical paternal stem cells were infused on day 0 together with cultured donor mesenchymal stem cells. A single dose of rituximab was given on day +1. After cessation of short immunosuppression (mycophenolate, OKT3), 4 children received donor lymphocyte infusion (DLI). (131)I-MIBG infusion and RIC were well tolerated. All patients engrafted. No primary acute graft-versus-host disease (aGVHD) was observed. Four children developed aGVHD after DLI and were successfully treated. Analysis of immunologic recovery showed fast reappearance of potentially immunocompetent natural killer (NK) and T cells, which might have acted as effector cells responsible for the graft-versus-tumor (GVT) effect. Two children are alive and well, with no evidence of disease 40 and 42 months after transplantation. One patient experienced late progression with new bone lesions (sternum) 38 months after haplo-SCT, and is being treated with local irradiation and reinstituted DLI. One patient rejected the graft, was rescued with autologous backup, and died of progressive disease 5 months after transplantation. Another child relapsed 7 months after transplantation and died 5 months later. High-dose (131)I-MIBG followed by RIC and haplo-SCT for RRNB is feasible and promising, because 2 of 5 children on that regimen achieved long-lasting remission. Further studies are needed to evaluate targeted therapy and immune-mediated tumor control in high-risk neuroblastoma.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , Transplante de Células-Tronco Hematopoéticas/métodos , Recidiva Local de Neoplasia/terapia , Neuroblastoma/terapia , Antineoplásicos/administração & dosagem , Técnicas de Cultura de Células , Criança , Pré-Escolar , Estudos de Coortes , Terapia Combinada , Feminino , Doença Enxerto-Hospedeiro/imunologia , Haplótipos , Humanos , Lactente , Masculino , Transplante de Células-Tronco Mesenquimais , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/radioterapia , Neuroblastoma/imunologia , Neuroblastoma/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Linfócitos T/imunologia , Quimeras de Transplante
8.
J Clin Oncol ; 27(25): 4162-8, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19636009

RESUMO

PURPOSE: To evaluate the safety and efficacy of high-dose [(131)I]metaiodobenzylguanidine ([(131)I]MIBG) in the treatment of malignant pheochromocytoma (PHEO) and paraganglioma (PGL). METHODS: Fifty patients with metastatic PHEO or PGL, age 10 to 64 years, were treated with [(131)I]MIBG doses ranging from 492 to 1,160 mCi (median, 12 mCi/kg). Cumulative [(131)I]MIBG administered ranged from 492 to 3,191 mCi. Autologous hematopoietic stem cells were collected and cryopreserved before treatment with [(131)I]MIBG greater than 12 mCi/kg or with a total dose greater than 500 mCi. Sixty-nine [(131)I]MIBG infusions were given, which included infusions to 35 patients treated once and infusions to 15 patients who received two or three treatments. Response was evaluated by [(123)I]MIBG scans, computed tomography/magnetic resonance imaging, urinary catecholamines/metanephrines, and chromogranin A. RESULTS: The overall complete response (CR) plus partial response (PR) rate in 49 evaluable patients was 22%. Additionally, 35% of patients achieved a CR or PR in at least one measure of response without progressive disease, and 8% of patients maintained stable disease for greater than 12 months. Thirty-five percent of patients experienced progressive disease within 1 year after therapy. The estimated 5-year overall survival rate was 64%. Toxicities included grades 3 to 4 neutropenia (87%) and thrombocytopenia (83%). Grades 3 to 4 nonhematologic toxicity included acute respiratory distress syndrome (n = 2), bronchiolitis obliterans organizing pneumonia (n = 2), pulmonary embolism (n = 1), fever with neutropenia (n = 7), acute hypertension (n = 10), infection (n = 2), myelodysplastic syndrome (n = 2), and hypogonadism (n = 4). CONCLUSION: Although serious toxicity may occur, the survival and response rates achieved with high-dose [(131)I]MIBG suggest its utility in the management of selected patients with metastatic PHEO and PGL.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , Neoplasias das Glândulas Suprarrenais/radioterapia , Radioisótopos do Iodo/administração & dosagem , Paraganglioma/radioterapia , Feocromocitoma/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , 3-Iodobenzilguanidina/efeitos adversos , Adolescente , Neoplasias das Glândulas Suprarrenais/diagnóstico , Neoplasias das Glândulas Suprarrenais/mortalidade , Neoplasias das Glândulas Suprarrenais/secundário , Adulto , Biomarcadores Tumorais/urina , Catecolaminas/urina , Criança , Cromogranina A/urina , Intervalo Livre de Doença , Relação Dose-Resposta à Radiação , Feminino , Humanos , Infusões Intravenosas , Radioisótopos do Iodo/efeitos adversos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Paraganglioma/diagnóstico , Paraganglioma/secundário , Feocromocitoma/diagnóstico , Feocromocitoma/mortalidade , Feocromocitoma/secundário , Modelos de Riscos Proporcionais , Estudos Prospectivos , Compostos Radiofarmacêuticos/efeitos adversos , Medição de Risco , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
9.
Pediatr Blood Cancer ; 44(3): 232-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15490487

RESUMO

BACKGROUND: (131)I Metaiodobenzylguanidine ((131)I-MIBG) is an effective targeted radiotherapeutic for neuroblastoma with response rates greater than 30% in refractory disease. Toxicity is mainly limited to myelosuppression. The aim of this study was to determine the response rate and hematologic toxicity of multiple infusions of (131)I-MIBG. PROCEDURE: Patients received two to four infusions of (131)I-MIBG at activity levels of 3-19 mCi/kg per infusion. Criteria for subsequent infusions were neutrophil recovery without stem cell support and lack of disease progression after the first infusion. RESULTS: Sixty-two infusions were administered to 28 patients, with 24 patients receiving two infusions, two patients receiving three infusions, and two patients receiving four infusions. All patients were heavily pre-treated, including 16 with prior myeloablative therapy. Eleven patients (39%) had overall disease response to multiple therapies, including eight patients with measurable responses to each of two or three infusions, and three with a partial response (PR) after the first infusion and stable disease after the second. The main toxicity was myelosuppression, with 78% and 82% of patients requiring platelet transfusion support after the first and second infusion, respectively, while only 50% had grade 4 neutropenia, usually transient. Thirteen patients did not recover platelet transfusion independence after their final MIBG infusion; stem cell support was given in ten patients. CONCLUSIONS: Multiple therapies with (131)I-MIBG achieved increasing responses, but hematologic toxicity, especially to platelets, was dose limiting. More effective therapy might be given using consecutive doses in rapid succession with early stem cell support.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , 3-Iodobenzilguanidina/toxicidade , Medula Óssea/efeitos dos fármacos , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/toxicidade , Neuroblastoma/radioterapia , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/toxicidade , Adolescente , Adulto , Anemia/induzido quimicamente , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Humanos , Infusões Intravenosas , Masculino , Neutropenia/induzido quimicamente , Trombocitopenia/induzido quimicamente , Resultado do Tratamento
10.
Expert Opin Investig Drugs ; 13(12): 1579-83, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15566315

RESUMO

The basic principles of treatment for phaeochromocytomas and paragangliomas are to block the effects of high catecholamines and make the patient safe for surgical removal of the tumour. The traditional preoperative medical preparation uses the non-selective alpha-adrenoceptor blocker phenoxybenzamine and a beta-adrenoceptor blocker, propranolol. Other agents have been used effectively, including selective alpha-adrenoceptor blockers, doxazosin and prazosin, and calcium channel antagonists. There have been no trial comparing regimens and there is some controversy as to the best regimen. Major advances have been made in laparoscopic and laparoscopic-assisted surgery. Cortical-sparing adrenalectomy has been used in some centres for familial phaeochromocytomas. High-dose [(131)I]-metaiodobenzylguanidine therapy and combined [(131)I]-metaiodobenzylguanidine and chemotherapy are promising new developments for the malignant disease. All patients should be followed indefinitely because the recurrence or malignancy rate is >or= 10% over a prolonged follow up.


Assuntos
3-Iodobenzilguanidina/uso terapêutico , Neoplasias das Glândulas Suprarrenais/terapia , Terapia Combinada/tendências , Feocromocitoma/terapia , 3-Iodobenzilguanidina/administração & dosagem , Neoplasias das Glândulas Suprarrenais/cirurgia , Catecolaminas/metabolismo , Humanos , Paraganglioma/terapia , Feocromocitoma/cirurgia
11.
J Clin Oncol ; 22(12): 2452-60, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15197208

RESUMO

PURPOSE: Iodine-131-metaiodobenzylguanidine ((131)I-MIBG) has been shown to be active against refractory neuroblastoma. The primary toxicity of (131)I-MIBG is myelosuppression, which might necessitate autologous hematopoietic stem-cell transplantation (AHSCT). The goal of this study was to determine risk factors for myelosuppression and the need for AHSCT after (131)I-MIBG treatment. PATIENTS AND METHODS: Fifty-three patients with refractory or relapsed neuroblastoma were treated with 18 mCi/kg (131)I-MIBG on a phase I/II protocol. The median whole-body radiation dose was 2.92 Gy. RESULTS: Almost all patients required at least one platelet (96%) or red cell (91%) transfusion and most patients (79%) developed neutropenia (< 0.5 x 10(3)/microL). Patients reached platelet nadir earlier than neutrophil nadir (P <.0001). Earlier platelet nadir correlated with bone marrow tumor, more extensive bone involvement, higher whole-body radiation dose, and longer time from diagnosis to (131)I-MIBG therapy (P

Assuntos
3-Iodobenzilguanidina/administração & dosagem , 3-Iodobenzilguanidina/efeitos adversos , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Neuroblastoma/tratamento farmacológico , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/efeitos adversos , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Radioisótopos do Iodo/uso terapêutico , Masculino , Neutropenia/induzido quimicamente , Neutropenia/terapia , Trombocitopenia/induzido quimicamente , Trombocitopenia/terapia , Transplante Autólogo
12.
Bone Marrow Transplant ; 27(6): 571-4, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11319584

RESUMO

Despite the use of aggressive chemotherapy, stage 4 high risk neuroblastoma still has very poor prognosis which is estimated at 25%. Metabolic radiotherapy with I(131) MIBG appears a feasible option to enhance the effects of chemotherapy. Seventeen patients having MIBG-positive residual disease received 4.1-11.1 mCi/kg of I(131) MIBG 7-10 days before initiating the high-dose chemotherapy cycle consisting of busulphan 16 mg/kg and melphalan 140 mg/m(2) followed by PBSC infusion. We compared the toxicity in these patients to that seen in 15 control subjects with neuroblastoma who underwent a PBSC transplant without MIBG therapy. We observed greater toxic involvement of the gastrointestinal system in children treated with I(131) MIBG: grade 2 or 3 mucositis developed in 13/17 patients treated with I(131) MIBG and in 9/15 treated without it. Grade 1-2 gastrointestinal toxicity occurred in 12/17 children given MIBG and in 5/15 of the controls. One child receiving I(131) MIBG developed transient interstitial pneumonia. Another child who also received I(131) MIBG after PBSC rescue developed fatal pneumonia after the third course of metabolic radiotherapy. Our experience indicates that MIBG can be included in the high-dose chemotherapy regimens followed by PBSC rescue for children with residual neuroblastoma taking up MIBG. Attention should be paid to avoiding lung complications. Prospective studies are needed to demonstrate the real efficacy of this treatment.


Assuntos
3-Iodobenzilguanidina/administração & dosagem , Antineoplásicos/administração & dosagem , Transplante de Células-Tronco Hematopoéticas , Neuroblastoma/terapia , Compostos Radiofarmacêuticos/administração & dosagem , Condicionamento Pré-Transplante/métodos , 3-Iodobenzilguanidina/toxicidade , Antineoplásicos/toxicidade , Criança , Pré-Escolar , Feminino , Seguimentos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Lactente , Recém-Nascido , Radioisótopos do Iodo , Doenças Pulmonares Intersticiais/etiologia , Masculino , Neuroblastoma/complicações , Neuroblastoma/mortalidade , Compostos Radiofarmacêuticos/toxicidade , Condicionamento Pré-Transplante/efeitos adversos , Resultado do Tratamento
13.
Int J Cancer ; 87(3): 412-22, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10897048

RESUMO

The clinical results of [(131)I]meta-iodobenzylguanidine (MIBG)-targeted radiotherapy in neuroblastoma patients is highly variable. To assess the therapeutic potential of [(131)I]MIBG, we used the SK-N-SH human neuroblastoma, xenografted in nude mice. The model was first characterized for basic parameters of MIBG handling in the host species. This demonstrated the presence of both strain- and nu/nu mutation-related differences in [(131)I]MIBG biodistribution. Fecal and urinary clearance rates of [(131)I]MIBG in mice roughly resemble those in humans, but mice metabolize MIBG more extensively. In both species, enzymatic deiodination in vivo was not an important metabolic route. Therapy with increasing [(131)I]MIBG doses (25-92 MBq) given as single i.v. injections resulted in proportionally increasing specific growth delay values (tumor regrowth delay/doubling time) of 1 to 5. Using gamma-camera scintigraphy for non-invasive dosimetry, the corresponding calculated absorbed tumor radiation doses ranged from 2 to 11 Gy. We also compared the therapeutic effects of a single [(131)I]MIBG administration with those resulting from a more protracted exposure by fractionating the dose in 2 to 6 injections or with high dose rate external-beam irradiation. No therapeutic advantage of a fractionated schedule was observed, and 5.5 Gy delivered by low dose-rate [(131)I]MIBG endo-irradiation was equi-effective with 5.0 Gy X-rays. The SK-N-SH neuroblastoma xenograft model thus appears suitable to evaluate possible treatment improvements to reach full potential of MIBG radiotherapy.


Assuntos
3-Iodobenzilguanidina/farmacocinética , Radioisótopos do Iodo/farmacocinética , Neuroblastoma/radioterapia , Compostos Radiofarmacêuticos/farmacocinética , 3-Iodobenzilguanidina/administração & dosagem , 3-Iodobenzilguanidina/uso terapêutico , 3-Iodobenzilguanidina/toxicidade , Animais , Biotransformação , Fracionamento da Dose de Radiação , Esquema de Medicação , Humanos , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/uso terapêutico , Radioisótopos do Iodo/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Transplante de Neoplasias , Neuroblastoma/diagnóstico por imagem , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Teleterapia por Radioisótopo , Radiometria/métodos , Cintilografia , Compostos Radiofarmacêuticos/uso terapêutico , Compostos Radiofarmacêuticos/toxicidade , Especificidade da Espécie , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA