Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Pediatr Blood Cancer ; 63(3): 436-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26506090

ABSTRACT

BACKGROUND: (131) I-metaiodobenzylguanidine ((131) I-MIBG) is a targeted radiopharmaceutical for patients with neuroblastoma. Despite its tumor-specific uptake, the treatment with (131) I-MIBG results in whole-body radiation exposure. Our aim was to correlate whole-body radiation dose (WBD) from (131) I-MIBG with tumor response, toxicities, and other clinical factors. METHODS: This retrospective cohort analysis included 213 patients with high-risk neuroblastoma treated with (131) I-MIBG at UCSF Benioff Children's Hospital between 1996 and 2015. WBD was determined from radiation exposure rate measurements. The relationship between WBD ordered tertiles and variables were analyzed using Cochran-Mantel-Haenszel test of trend, Kruskal-Wallis test, and one-way analysis of variance. Correlation between WBD and continuous variables was analyzed using Pearson correlation and Spearman rank correlation. RESULTS: WBD correlated with (131) I-MIBG administered activity, particularly with (131) I-MIBG per kilogram (P < 0.001). Overall response rate did not differ significantly among the three tertiles of WBD. Correlation between response by relative Curie score and WBD was of borderline significance, with patients receiving a lower WBD showing greater reduction in osteomedullary metastases by Curie score (rs = 0.16, P = 0.049). There were no significant ordered trends among tertiles in any toxicity measures (grade 4 neutropenia, thrombocytopenia < 20,000/µl, and grade > 1 hypothyroidism). CONCLUSIONS: This study showed that (131) I-MIBG activity per kilogram correlates with WBD and suggests that activity per kilogram will predict WBD in most patients. Within the range of activities prescribed, there was no correlation between WBD and either response or toxicity. Future studies should evaluate tumor dosimetry, rather than just WBD, as a tool for predicting response following therapy with (131) I-MIBG.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Iodine Radioisotopes/therapeutic use , Neuroblastoma/radiotherapy , Radiopharmaceuticals/therapeutic use , 3-Iodobenzylguanidine/toxicity , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Iodine Radioisotopes/toxicity , Male , Middle Aged , Radiopharmaceuticals/toxicity , Radiotherapy Dosage , Retrospective Studies , Statistics, Nonparametric , Whole-Body Irradiation , Young Adult
2.
J Nucl Med ; 53(7): 1146-54, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22689924

ABSTRACT

UNLABELLED: Targeted radiotherapy using (131)I-metaiodobenzylguanidine ((131)I-MIBG) has produced remissions in some neuroblastoma patients. We previously reported that combining (131)I-MIBG with the topoisomerase I inhibitor topotecan induced long-term DNA damage and supraadditive toxicity to noradrenaline transporter (NAT)-expressing cells and xenografts. This combination treatment is undergoing clinical evaluation. This present study investigated the potential of poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP-1) inhibition, in vitro and in vivo, to further enhance (131)I-MIBG/topotecan efficacy. METHODS: Combinations of topotecan and the PARP-1 inhibitor PJ34 were assessed for synergism in vitro by combination-index analysis in SK-N-BE(2c) (neuroblastoma) and UVW/NAT (NAT-transfected glioma) cells. Three treatment schedules were evaluated: topotecan administered 24 h before, 24 h after, or simultaneously with PJ34. Combinations of PJ34 and (131)I-MIBG and of PJ34 and (131)I-MIBG/topotecan were also assessed using similar scheduling. In vivo efficacy was measured by growth delay of tumor xenografts. We also assessed DNA damage by γH2A.X assay, cell cycle progression by fluorescence-activated cell sorting analysis, and PARP-1 activity in treated cells. RESULTS: In vitro, only simultaneous administration of topotecan and PJ34 or PJ34 and (131)I-MIBG induced supraadditive toxicity in both cell lines. All scheduled combinations of PJ34 and (131)I-MIBG/topotecan induced supraadditive toxicity and increased DNA damage in SK-N-BE(2c) cells, but only simultaneous administration induced enhanced efficacy in UVW/NAT cells. The PJ34 and (131)I-MIBG/topotecan combination treatment induced G(2) arrest in all cell lines, regardless of the schedule of delivery. In vivo, simultaneous administration of PJ34 and (131)I-MIBG/topotecan significantly delayed the growth of SK-N-BE(2c) and UVW/NAT xenografts, compared with (131)I-MIBG/topotecan therapy. CONCLUSION: The antitumor efficacy of topotecan, (131)I-MIBG, and (131)I-MIBG/topotecan combination treatment was increased by PARP-1 inhibition in vitro and in vivo.


Subject(s)
3-Iodobenzylguanidine/toxicity , Antineoplastic Agents/toxicity , Norepinephrine Plasma Membrane Transport Proteins/physiology , Poly(ADP-ribose) Polymerase Inhibitors , Radiopharmaceuticals/toxicity , Topotecan/toxicity , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/therapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chemistry, Pharmaceutical , Combined Modality Therapy , DNA Breaks/drug effects , DNA Breaks/radiation effects , Female , Flow Cytometry , Histones/metabolism , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neuroblastoma/drug therapy , Neuroblastoma/radiotherapy , Neuroblastoma/therapy , Norepinephrine Plasma Membrane Transport Proteins/genetics , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Tumor Stem Cell Assay
3.
Drug Chem Toxicol ; 33(1): 17-9, 2010.
Article in English | MEDLINE | ID: mdl-20001216

ABSTRACT

Metaiodobenzylguanidine (MIBG) is an analog of norepinephrine. I-131-labeled MIBG has been thought to be safe and effective in the evaluation of neuroendocrine tumors, mainly in neuroblastoma and pheochromocytoma. This article describes the acute toxicity of MIBG in imprinting control region (ICR) mice. Treated mice were administered with MIBG at dose levels of 75, 150, and 300ng/kg with dose volumes of 20mL/kg. The control mice were administered 20mL/kg of vehicle control. The mice were observed for 14 days. Observations included general demeanor, clinical signs, mortality, body weights/total body-weight gains, and gross necropsy findings. None of the animals died during the 14-day study period. There was no difference in body weights among all treated and control mice.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/toxicity , Adrenal Gland Neoplasms/pathology , Injections, Intravenous/adverse effects , Animals , Body Burden , Combined Modality Therapy , Dose-Response Relationship, Radiation , Iodine Radioisotopes , Lethal Dose 50 , Mice , Mice, Inbred ICR , Neuroblastoma/pathology , Norepinephrine , Organ Size , Pheochromocytoma/pathology , Radiation Dosage , Radiopharmaceuticals , Radiotherapy Dosage/standards
4.
Pediatr Blood Cancer ; 44(3): 232-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15490487

ABSTRACT

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.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/toxicity , Bone Marrow/drug effects , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/toxicity , Neuroblastoma/radiotherapy , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/toxicity , Adolescent , Adult , Anemia/chemically induced , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infusions, Intravenous , Male , Neutropenia/chemically induced , Thrombocytopenia/chemically induced , Treatment Outcome
5.
Bone Marrow Transplant ; 27(6): 571-4, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11319584

ABSTRACT

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.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , Antineoplastic Agents/administration & dosage , Hematopoietic Stem Cell Transplantation , Neuroblastoma/therapy , Radiopharmaceuticals/administration & dosage , Transplantation Conditioning/methods , 3-Iodobenzylguanidine/toxicity , Antineoplastic Agents/toxicity , Child , Child, Preschool , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Infant, Newborn , Iodine Radioisotopes , Lung Diseases, Interstitial/etiology , Male , Neuroblastoma/complications , Neuroblastoma/mortality , Radiopharmaceuticals/toxicity , Transplantation Conditioning/adverse effects , Treatment Outcome
6.
Int J Cancer ; 87(3): 412-22, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10897048

ABSTRACT

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.


Subject(s)
3-Iodobenzylguanidine/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Neuroblastoma/radiotherapy , Radiopharmaceuticals/pharmacokinetics , 3-Iodobenzylguanidine/administration & dosage , 3-Iodobenzylguanidine/therapeutic use , 3-Iodobenzylguanidine/toxicity , Animals , Biotransformation , Dose Fractionation, Radiation , Drug Administration Schedule , Humans , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/therapeutic use , Iodine Radioisotopes/toxicity , Mice , Mice, Inbred C3H , Mice, Nude , Neoplasm Transplantation , Neuroblastoma/diagnostic imaging , Neuroblastoma/metabolism , Neuroblastoma/pathology , Radioisotope Teletherapy , Radiometry/methods , Radionuclide Imaging , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/toxicity , Species Specificity , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured/transplantation
7.
Br J Cancer ; 79(5-6): 802-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10070872

ABSTRACT

meta-iodobenzylguanidine (MIBG) radiolabelled with iodine-131 is used for diagnosis and treatment of neuroadrenergic neoplasms such as phaeochromocytoma and neuroblastoma. In addition, non-radiolabelled MIBG, administered i.v., is used in several clinical studies. These include palliation of the carcinoid syndrome, in which MIBG proved to be effective in 60% of the patients. Oral MIBG administration might be convenient to maintain palliation and possibly improve the percentage of responders. We have, therefore, investigated the feasibility of oral administration of MIBG in an animal model. Orally administered MIBG demonstrated a bioavailability of 59%, with a maximal tolerated dose of 60 mg kg(-1). The first and only toxicity encountered was a decrease in renal function, measured by a reduced clearance of [51Cr]EDTA and accompanied by histological tubular damage. Repeated MIBG administration of 40 mg kg(-1) for 5 sequential days or of 20 mg kg(-1) for two courses of 5 sequential days with a 2-day interval did not affect renal clearance and was not accompanied by histological abnormalities in kidney, stomach, intestines, liver, heart, lungs, thymus, salivary glands and testes. Because of a sufficient bioavailability in absence of gastrointestinal toxicity, MIBG is considered suitable for further clinical investigation of repeated oral administration in patients.


Subject(s)
3-Iodobenzylguanidine/pharmacokinetics , 3-Iodobenzylguanidine/toxicity , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , 3-Iodobenzylguanidine/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Biological Availability , Injections, Intravenous , Iodine Radioisotopes , Kidney/physiology , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C3H , Tissue Distribution
8.
Br J Cancer ; 79(5-6): 793-801, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10070871

ABSTRACT

Tumour-selective acidification is of potential interest for enhanced therapeutic gain of pH sensitive drugs. In this study, we investigated the feasibility of a tumour-selective reduction of the extracellular and intracellular pH and their effect on the tumour response of selected anti-cancer drugs. In an in vitro L1210 leukaemic cell model, we confirmed enhanced cytotoxicity of chlorambucil at low extracellular pH conditions. In contrast, the alkylating drugs melphalan and cisplatin, and bioreductive agents mitomycin C and its derivative EO9, required low intracellular pH conditions for enhanced activation. Furthermore, a strong and pH-independent synergism was observed between the pH-equilibrating drug nigericin and melphalan, of which the mechanism is unclear. In radiation-induced fibrosarcoma (RIF-1) tumour-bearing mice, the extracellular pH was reduced by the mitochondrial inhibitor m-iodobenzylguanidine (MIBG) or its analogue benzylguanidine (BG) plus glucose. To simultaneously reduce the intracellular pH, MIBG plus glucose were combined with the ionophore nigericin or the Na+/H+ exchanger inhibitor amiloride and the Na+-dependent HCO3-/Cl- exchanger inhibitor 4,4'-diisothiocyanostilbene-2,2'-disulphonic acid (DIDS). Biochemical studies confirmed an effective reduction of the extracellular pH to approximately 6.2, and anti-tumour responses to the interventions indicated a simultaneous reduction of the intracellular pH below 6.6 for at least 3 h. Combined reduction of extra- and intracellular tumour pH with melphalan increased the tumour regrowth time to 200% of the pretreatment volume from 5.7 +/- 0.6 days for melphalan alone to 8.1 +/- 0.7 days with pH manipulation (P < 0.05). Mitomycin C related tumour growth delay was enhanced by the combined interventions from 3.8 +/- 0.5 to 5.2 +/- 0.5 days (P < 0.05), but only in tumours of relatively large sizes. The interventions were non-toxic alone or in combination with the anti-cancer drugs and did not affect melphalan biodistribution. In conclusion, we have developed non-toxic interventions for sustained and selective reduction of extra- and intracellular tumour pH which potentiated the tumour responses to selected anti-cancer drugs.


Subject(s)
3-Iodobenzylguanidine/toxicity , 3-Iodobenzylguanidine/therapeutic use , Antineoplastic Agents/toxicity , Antineoplastic Agents/therapeutic use , Guanidines/toxicity , Hydrogen-Ion Concentration , Leukemia L1210/drug therapy , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Amiloride/pharmacology , Animals , Cell Division/drug effects , Cell Survival/drug effects , Chlorambucil/toxicity , Chromium Radioisotopes , Doxorubicin/toxicity , Drug Synergism , Edetic Acid/pharmacokinetics , Glucose/pharmacology , Kidney/drug effects , Kidney/physiology , Melphalan/toxicity , Mice , Mitomycin/toxicity , Tumor Cells, Cultured
9.
Cancer Chemother Pharmacol ; 42(1): 37-45, 1998.
Article in English | MEDLINE | ID: mdl-9619756

ABSTRACT

meta-Iodobenzylguanidine (MIBG) is a multipotent drug used in its radiolabeled form as a tumor-seeking radiopharmaceutical in the diagnosis and treatment of pheochromocytoma and neuroblastoma. Nonradiolabeled MIBG has also proved to be effective in the palliation of carcinoid syndromes and, on a predosing schedule, in enhancing the relative tumor uptake of a subsequent [131I]-MIBG dose in tumors of neuroadrenergic origin. In addition, MIBG is under investigation as an inhibitor of mitochondrial respiration and, as such, for its use in tumor-specific acidification. In this report we describe the side effects of nonradiolabeled MIBG on kidney function in mice. High doses of MIBG (40 mg/kg) reduced renal blood perfusion as measured by 86Rb distribution by 50%, which could be antagonized by the bioamine receptor blockers prazosin and cyproheptadine. MIBG also induced reversible renal damage as evidenced from a decrease in [51Cr]-ethylenediaminetetraacetic acid (EDTA) clearance and from histological damage, which was most pronounced in the distal tubuli. These effects were unrelated to reduced perfusion, however, and could not be antagonized by bioamine receptor blockers, Ca2+-channel blockers, or diuretics. Clearance effects of MIBG were mimicked by N-nitro-L-arginine methyl ester (L-NAME), a known inhibitor of nitric oxide synthase (NOS), and MIBG itself (100 microM) also inhibited NOS in vitro, suggesting that NOS inhibition by MIBG may have contributed to the observed reduction in renal clearance. The MIBG analog benzylguanidine (BG), which is equipotent in terms of mitochondrial inhibition, did not affect renal clearance, thus excluding mitochondrial inhibition as the main mechanism of MIBG-induced damage. MIBG, however, was much more cytotoxic than BG to kidney tubular cells in primary cultures. Although the renal effects of high-dose MIBG were reversible, alterations in the pharmacokinetics of concomitant medications by a temporary reduction in renal function should be taken into account in its clinical application.


Subject(s)
3-Iodobenzylguanidine/toxicity , Antineoplastic Agents/toxicity , Kidney/drug effects , 3-Iodobenzylguanidine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Glucose/metabolism , Glucose/pharmacology , Male , Mice , Mice, Inbred C3H , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...