Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Vet Comp Oncol ; 21(1): 45-53, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36149359

ABSTRACT

The objective of this report is to describe the use and outcome of stereotactic body radiation therapy (SBRT) for treatment of pheochromocytomas in 8 dogs. Pheochromocytomas are an uncommon but challenging tumour to manage. Adrenalectomy is the standard of care for treatment of pheochromocytomas in both animals and humans; however, unpredictable catecholamine secretion from the tumour and vascular and local invasion of the tumour and thrombi can pose life-threatening perioperative and anaesthetic risks. SBRT has been investigated as an alternative to adrenalectomy in human patients with pheochromocytomas. Eight dogs with clinical signs, an adrenal mass, and cytology and/or urine normetanephrine/creatinine ratios consistent with pheochromocytoma were treated with SBRT in lieu of adrenalectomy. Three dogs presented with acute hemoabdomen. Seven dogs had caval tumour invasion, 3 with extension into the right atrium. Following SBRT, all dogs had complete resolution of clinical signs and reduced urine normetanephrine/creatinine ratio and/or tumour size. No significant anaesthetic complications were encountered. Acute radiation toxicity was limited to grade I gastrointestinal signs in 3 dogs and resolved within 1-2 days of symptomatic therapy. Five of 8 dogs were alive at the time of follow up, with a median follow up time of 25.8 months. SBRT resulted in a favourable outcome and mitigated the life-threatening risks of adrenalectomy in these 8 dogs. SBRT may be a safe and effective alternative to adrenalectomy for pheochromocytomas in dogs with non-resectable tumours, or for owners averse to the risks of surgery.


Subject(s)
Adrenal Gland Neoplasms , Anesthetics , Dog Diseases , Pheochromocytoma , Radiosurgery , Humans , Dogs , Animals , Adrenalectomy/veterinary , Adrenalectomy/methods , Pheochromocytoma/radiotherapy , Pheochromocytoma/surgery , Pheochromocytoma/veterinary , Radiosurgery/veterinary , Creatinine , Normetanephrine , Dog Diseases/radiotherapy , Dog Diseases/surgery , Adrenal Gland Neoplasms/radiotherapy , Adrenal Gland Neoplasms/surgery , Adrenal Gland Neoplasms/veterinary , Retrospective Studies
2.
Clin Cancer Res ; 27(11): 2989-2995, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33685867

ABSTRACT

Targeted radionuclide therapies (TRT) using 131I-metaiodobenzylguanidine (131I-MIBG) and peptide receptor radionuclide therapy (177Lu or 90Y) represent several of the therapeutic options in the management of metastatic/inoperable pheochromocytoma/paraganglioma. Recently, high-specific-activity-131I-MIBG therapy was approved by the FDA and both 177Lu-DOTATATE and 131I-MIBG therapy were recommended by the National Comprehensive Cancer Network guidelines for the treatment of metastatic pheochromocytoma/paraganglioma. However, a clinical dilemma often arises in the selection of TRT, especially when a patient can be treated with either type of therapy based on eligibility by MIBG and somatostatin receptor imaging. To address this problem, we assembled a group of international experts, including oncologists, endocrinologists, and nuclear medicine physicians, with substantial experience in treating neuroendocrine tumors with TRTs to develop consensus and provide expert recommendations and perspectives on how to select between these two therapeutic options for metastatic/inoperable pheochromocytoma/paraganglioma. This article aims to summarize the survival outcomes of the available TRTs; discuss personalized treatment strategies based on functional imaging scans; address practical issues, including regulatory approvals; and compare toxicities and risk factors across treatments. Furthermore, it discusses the emerging TRTs.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Adrenal Gland Neoplasms/secondary , Iodine Radioisotopes/therapeutic use , Lutetium/therapeutic use , Octreotide/analogs & derivatives , Organometallic Compounds/therapeutic use , Paraganglioma/radiotherapy , Paraganglioma/secondary , Pheochromocytoma/radiotherapy , Pheochromocytoma/secondary , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Radiotherapy/methods , Adrenal Gland Neoplasms/diagnostic imaging , Humans , Octreotide/therapeutic use , Paraganglioma/diagnostic imaging , Pheochromocytoma/diagnostic imaging , Positron Emission Tomography Computed Tomography
3.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-31614368

ABSTRACT

CONTEXT: Prognosis of metastatic pheochromocytoma/paraganglioma following 131-Iodine metaiodobenzylguanidine (MIBG) is incompletely characterized due to small samples and shorter follow-up in these rare, often indolent tumors. OBJECTIVE: To describe long-term survival, frequency, and prognostic impact of imaging, biochemical, and symptomatic response to 131-I MIBG. DESIGN: Retrospective chart and imaging review at a tertiary referral center. PATIENTS: Six hundred sixty-eight person-years of follow-up in 125 patients with metastatic pheochromocytoma/paraganglioma with progression through prior multimodal treatment. INTERVENTION: Median 18 800 MBq 131-I MIBG. MAIN OUTCOME MEASURES: Overall survival, Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST) imaging response, symptomatic response per chart review, and biochemical response (20% change over 2 consecutive assays of catecholamines, vanillylmandelic acid, metanephrines, or chromogranin A). RESULTS: Median survival standard deviation [SD] from diagnosis was 11.5 years [2.4]; following metastasis, 6.5 years [0.8]; post treatment, 4.3 years [0.7]. Among 88 participants with follow-up imaging, 1% experienced complete response, 33% partial response, 53% stability, and 13% progression. Fifty-one percent showed subsequent progression, median progression-free survival [SD] of 2.0 years [0.6]. Stability/response vs progression at first imaging follow-up (3-6 months) predicted improved survival, 6.3 vs 2.4 years (P = 0.021). Fifty-nine percent of 54 patients demonstrated biochemical response. Fifty percent of these relapsed, with median time to laboratory progression [SD] of 2.8 years [0.7]. Biochemical response did not predict extended survival. Seventy-five percent of 83 patients reported improvement in pretreatment symptoms, consisting primarily of pain (42%), fatigue (27%), and hypertension (14%). Sixty-one percent of these patients experienced subsequent symptomatic progression at median [SD] 1.8 years [0.4]. Symptomatic response did not predict extended survival. CONCLUSIONS: Imaging, symptomatic, and laboratory response to multimodal treatment including high-dose 131-I MIBG were achieved on long-term follow-up in metastatic pheochromocytoma or paraganglioma. Imaging response at 3 to 6 months was prognostic.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/therapeutic use , Adrenal Gland Neoplasms/secondary , Adult , Disease Progression , Female , Follow-Up Studies , Humans , Male , Middle Aged , Paraganglioma/pathology , Pheochromocytoma/pathology , Prognosis , Retrospective Studies , Survival Rate
4.
Jpn J Radiol ; 29(6): 449-51, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21786102

ABSTRACT

A 63-year-old man underwent computed tomography (CT) using intravenous low-osmolar iodine contrast medium (LOCM) 6 days after undergoing high-dose (131)I-MIBG therapy for metastatic pheochromocytoma. Immediately after the CT examination, his blood pressure increased to 260/160 mmHg (from 179/101 mmHg before the examination). Phentolamine mesilate was administered, and the blood pressure rapidly went back to normal. Although hypertensive crisis after administration of LOCM is rare, this case suggests that high-dose (131)IMIBG therapy may be a risk factor for hypertensive crisis after administration of intravenous LOCM.


Subject(s)
Adrenal Gland Neoplasms/diagnostic imaging , Contrast Media/adverse effects , Hypertensive Encephalopathy/chemically induced , Iopamidol/adverse effects , Pheochromocytoma/diagnostic imaging , Radiopharmaceuticals/therapeutic use , Tomography, X-Ray Computed/adverse effects , 3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Humans , Male , Middle Aged , Pheochromocytoma/radiotherapy , Radionuclide Imaging
5.
Cancer Biother Radiopharm ; 24(5): 519-25, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19877881

ABSTRACT

Metaiodobenzylguanidine (MIBG), radioiodinated with (131)I, has been available for 25 years. Its role in the United States is limited to diagnostic imaging, whereas its therapeutic application in patients with neuroendocrine tumors for whom surgical treatment would not lead to a cure, has been approved in Europe. (131)I-MIBG treatments can be a valuable addition to the current gamut of treatment options for patients with metastatic neuroendocrine tumors, especially given the limited role for other systemic treatments, such as chemotherapy. There are basically two treatment strategies: one or two high-dose treatments or continuous low-dose treatments. (131)I-MIBG could induce symptomatic relief in the vast majority of patients treated, both following high-dose treatment and low-dose maintenance treatment. Biochemical responses can be observed in about half of the patients, whereas radiographic responses are described in roughly one third of the patients. Several articles suggested a survival benefit to patients treated with (131)I-MIBG. Side-effects of the treatment mainly consist of myelotoxicity, nausea, and hypothyroidism. Future developments are focused on the use of high-specific-activity (131)I-MIBG in high doses. The role of (131)I-MIBG in relation to other treatments remains to be established, although treatment (131)I-MIBG seems to be at least as effective as other systemic treatments, with limited side-effects.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Iodine Radioisotopes/therapeutic use , Neuroendocrine Tumors/radiotherapy , Adult , Aged , Algorithms , Carcinoid Tumor/radiotherapy , Carcinoma, Medullary/radiotherapy , Drug Interactions , Humans , Male , Medical Oncology/methods , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/therapeutic use , Thyroid Neoplasms/radiotherapy
6.
Cancer Biother Radiopharm ; 24(4): 469-75, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19694582

ABSTRACT

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.


Subject(s)
3-Iodobenzylguanidine/adverse effects , 3-Iodobenzylguanidine/pharmacokinetics , Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/adverse effects , Iodine Radioisotopes/pharmacokinetics , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , 3-Iodobenzylguanidine/administration & dosage , Adrenal Gland Neoplasms/metabolism , Adult , Dose-Response Relationship, Radiation , Female , Humans , Iodine Radioisotopes/administration & dosage , Male , Paraganglioma/metabolism , Pheochromocytoma/metabolism , Radiation Dosage , Radiometry/methods , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics
7.
J Clin Oncol ; 27(25): 4162-8, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19636009

ABSTRACT

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.


Subject(s)
3-Iodobenzylguanidine/administration & dosage , Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/administration & dosage , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/administration & dosage , 3-Iodobenzylguanidine/adverse effects , Adolescent , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/mortality , Adrenal Gland Neoplasms/secondary , Adult , Biomarkers, Tumor/urine , Catecholamines/urine , Child , Chromogranin A/urine , Disease-Free Survival , Dose-Response Relationship, Radiation , Female , Humans , Infusions, Intravenous , Iodine Radioisotopes/adverse effects , Kaplan-Meier Estimate , Male , Middle Aged , Paraganglioma/diagnosis , Paraganglioma/secondary , Pheochromocytoma/diagnosis , Pheochromocytoma/mortality , Pheochromocytoma/secondary , Proportional Hazards Models , Prospective Studies , Radiopharmaceuticals/adverse effects , Risk Assessment , Time Factors , Treatment Outcome , Young Adult
8.
Ann N Y Acad Sci ; 1073: 465-90, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17102115

ABSTRACT

Thirty patients with malignant pheochromocytoma (PHEO) or paraganglioma (PGL) were treated with high-dose 131I-MIBG. Patients were 11-62 (mean 39) years old: 19 patients males and 11 females. Nineteen patients had PGL, three of which were multifocal. Six PGLs were nonsecretory. Eleven patients had PHEO. All 30 patients had prior surgery. Fourteen patients were refractory to prior radiation or chemotherapy before 131I-MIBG. Peripheral blood stem cells (PBSCs) were collected and cryopreserved. 131I-MIBG was synthesized on-site, by exchange-labeling 131I with 127I-MIBG in a solid-phase Cu2+-catalyzed exchange reaction. 131I-MIBG was infused over 2 h via a peripheral IV. Doses ranged from 557 mCi to 1185 mCi (7.4 mCi/kg to 18.75 mCi/kg). Median dose was 833 mCi (12.55 mCi/kg). Marrow hypoplasia commenced 3 weeks after 131I-MIBG therapy. After the first 131I-MIBG therapy, 19 patients required platelet transfusions; 19 received GCSF; 12 received epoeitin or RBCs. Four patients received a PBSC infusion. High-dose 131I-MIBG resulted in the following overall tumor responses in 30 patients: 4 sustained complete remissions (CRs); 15 sustained partial remissions (PRs); 1 sustained stable disease (SD); 5 progressive disease (PD); 5 initial PRs or SD but relapsed to PD. Twenty-three of the 30 patients remain alive; deaths were from PD (5), myelodysplasia (1), and unrelated cause (1). Overall predicted survival at 5 years is 75% (Kaplan Meier estimate). For patients with metastatic PHEO or PGL, who have good *I-MIBG uptake on diagnostic scanning, high-dose 131I-MIBG therapy was effective in producing a sustained CR, PR, or SD in 67% of patients, with tolerable toxicity.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , 3-Iodobenzylguanidine/adverse effects , Adolescent , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/metabolism , Adult , Child , Diagnosis, Differential , Dose-Response Relationship, Radiation , Female , Humans , Iodine Radioisotopes/adverse effects , Male , Middle Aged , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , Pheochromocytoma/metabolism
9.
Ann N Y Acad Sci ; 1073: 498-504, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17102117

ABSTRACT

Malignant pheochromocytomas/paragangliomas (PCs/PGs) often have distant metastases to the skeleton, liver, and lungs. Radionuclide therapy is valuable for treatment of disseminated tumor disease and could be used as adjuvant therapy after surgery. Patients with local and/or distant metastases of PC/PG should be investigated preoperatively by scintigraphy using both 123I-MIBG and 111In-DTPA-D-Phe1-octreotide, to evaluate the possibilities for radionuclide therapy (i.e., a dosimetric estimation of radiation dose to the tumor tissue versus critical normal tissues). Individual patient dose-planning should be performed. For patients in whom positive therapeutic effects are anticipated radionuclide therapy can be applied. Therapy with both 131I-MIBG and 177Lu-octreotate might be favorable in individual patients with lesions visualized by both metaiodobenzylguanidine (MIBG) and octreotide scintigraphy with enhanced therapeutic effects and reduced side effects.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Pheochromocytoma/radiotherapy , Humans
10.
Surgery ; 134(6): 956-62; discussion 962-3, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14668728

ABSTRACT

INTRODUCTION: Iodine 131-meta-iodobenzylguanidine ((131)I-MIBG) has been applied to the palliative treatment of metastatic pheochromocytoma in small studies. We report our institutional experience for the treatment of metastatic pheochromocytoma and paraganglioma. METHODS: We performed a retrospective review of 33 patients with metastatic pheochromocytoma (n=22) and paraganglioma (n=11) treated at our institution with (131)I-MIBG over a 10-year period. RESULTS: Patients received a mean dose of 388+/-131 mCi (131)I-MIBG. Median survival after treatment was 4.7 years. Most patients experienced a symptomatic response leading to an improved survival (4.7 years vs 1.8 years, P<.01). Patients with a measurable hormone response demonstrated an increased survival in comparison to those with no response (4.7 years vs 2.6 years, P=.01). Patients who received a high dose (>500 mCi) as their initial therapy also had improved survival (3.8 years vs 2.8 years, P=.02). CONCLUSION: These data support (131)I-MIBG treatment for select patients with metastatic pheochromocytoma. In our experience, prolonged survival was best predicted by symptomatic and hormone response to (131)I-MIBG treatment. An initial dose of 500 mCi may be optimal. The benefit of (131)I-MIBG treatment for metastatic pheochromocytoma must also be weighed against its side effects.


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Paraganglioma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/therapeutic use , Adult , Aged , Dose-Response Relationship, Radiation , Female , Humans , Male , Middle Aged , Retrospective Studies , Survival Analysis
11.
Int J Cancer ; 90(6): 312-25, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11180134

ABSTRACT

[(131)I]Metaiodobenzylguanidine ([(131)I]MIBG) targeted radiotherapy is effective in debulking childhood neuroblastoma. The high-energy beta-emitter [(131)I]MIBG is, however, not very well suited to treat submillimeter tumors. The [(125)I]MIBG emission is more fully absorbed in small target volumes and therefore advocated for treatment of microscopic neuroblastoma. We investigated whether i.v. [(125)I]MIBG can have a therapeutic advantage over i.v. [(131)I]MIBG in realistic animal models. We used BALB/c nu/nu mice, bearing neuroadrenergic xenografts which differ in MIBG handling, i.e., extragranular vs. granular MIBG storage in the SK-N-SH human neuroblastoma and PC12 rat pheochromocytoma, respectively. Groups of 4-9 animals were treated with 10-100 MBq radioiodinated MIBG. Responses were calibrated against the effect of 4-5 Gy of external beam X-rays. SUBCUTANEOUS XENOGRAFTS: Due to the more extensive MIBG accumulation, the estimated MIBG exposure of the PC12 tumor was nearly 20-fold higher compared with the SK-N-SH xenograft which corresponded with a marked, i.e., nine-fold increased tumor growth delay after radioiodinated MIBG therapy. Both xenografts were equally sensitive to high-dose rate local irradiation. In neuroblastoma as well as pheochromocytoma, the therapeutic efficacy of [(131)I]MIBG was 6 times higher compared to the [(125)I]MIBG which is in reasonable agreement with the reported "131-I over 125-I" ratio of approximately 9 for the calculated absorbed radiation doses per unit of radioactivity. Apparently, the neuroblastoma was not relatively more sensitive to the (ultra)short range emitter [(125)I]MIBG than the pheochromocytoma, indicating that its therapeutic efficacy is independent of the intracellular MIBG storage mode. MICROSCOPIC TUMORS: The pheochromocytoma model consisted of widespread disease after i.v. cell injection with survival as endpoint. For the neuroblastoma, we induced focal intrahepatic microscopic tumors by intrasplenic injection and evaluated total liver weights 26 days after therapy. Theoretically, the therapeutic potential of [(125)I]MIBG at the cellular level should be at least as high as [(131)I]MIBG, but we failed to show any effect of [(125)I]MIBG therapy in both models. In contrast, measurable responses were obtained with [(131)I]MIBG, but these were lower than in the s.c. tumors when related to the responses induced by external X-rays. In conclusion, [(131)I]MIBG is decreasingly effective in microscopic disease and can therefore not be curative as a single agent. Our results strongly argue against the clinical use of [(125)I]MIBG and indicate that conventional total body irradiation was superior to [(131)I]MIBG for microscopic neuroblastoma. Int. J. Cancer (Radiat. Oncol. Invest.) 90, 312-325 (2000).


Subject(s)
3-Iodobenzylguanidine/therapeutic use , Adrenal Gland Neoplasms/radiotherapy , Antineoplastic Agents/therapeutic use , Iodine Radioisotopes/therapeutic use , Neuroblastoma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/therapeutic use , 3-Iodobenzylguanidine/adverse effects , Animals , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , PC12 Cells , Rats , Transplantation, Heterologous
12.
Zentralbl Chir ; 122(6): 494-7, 1997.
Article in German | MEDLINE | ID: mdl-9334119

ABSTRACT

Malignant pheochromocytomas are rare. Although 5-year survival is less than 50%, the prognosis varies. Some patients, even those with extensive metastases, have been followed up for many years. If the tumor tissue's uptake is adequate (> 5 Gy/100 mCi) the therapeutic use of 131I-meta-iodobenzylguanidin (131I-MIBG) is at present the therapy of first choice. The use of cytostatic chemotherapy should be limited to patients with rapidly progressive disease.


Subject(s)
Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Iodobenzenes/therapeutic use , Pheochromocytoma/radiotherapy , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/mortality , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant , Humans , Neoplasm Metastasis , Pheochromocytoma/drug therapy , Pheochromocytoma/mortality , Survival Rate , Treatment Outcome
13.
Q J Nucl Med ; 39(4 Suppl 1): 55-7, 1995 Dec.
Article in English | MEDLINE | ID: mdl-9002750

ABSTRACT

The avidity of many metastatic pheochromocytomas and neuroblastomas for metaiodobenzylguanidine (MIBG) observed at diagnostic scintigraphy has led to attempts to treat these lesions with large doses of MIBG. We and others have achieved therapeutic responses with 131I-MIBG (usually partial) in about a third of malignant pheochromocytomas. A small but important subgroup of advanced, poor prognosis neuroblastomas which have been resistant to all other therapies have also shown responses including occasional long-term survival (> 5 years) and apparent complete responses to 131I-MIBG. Because the physical properties of 131I are suboptimal for the delivery of therapeutic radiation to bone marrow micrometastases, a frequent problem in neuroblastoma, we have performed preliminary studies in poor prognosis Stage III and VI neuroblastoma using 125I-MIBG which has more satisfactory emissions. This has led to prolonged tumor stabilization and survival (> 19 to > 52 months) in 5 of 10 patients. MIBG radiopharmaceutical treatment of neuroendocrine tumor patients must still be considered an experimental but nevertheless promising treatment modality.


Subject(s)
Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Iodobenzenes/therapeutic use , Neuroblastoma/radiotherapy , Pheochromocytoma/radiotherapy , Radiopharmaceuticals/therapeutic use , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/diagnostic imaging , Adult , Bone Marrow Neoplasms/radiotherapy , Bone Marrow Neoplasms/secondary , Child , Humans , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/chemistry , Iodobenzenes/administration & dosage , Neoplasm Recurrence, Local/radiotherapy , Neoplasm Staging , Neoplasm, Residual/radiotherapy , Neuroblastoma/diagnostic imaging , Neuroblastoma/secondary , Pheochromocytoma/diagnostic imaging , Pheochromocytoma/secondary , Prognosis , Radionuclide Imaging , Radiopharmaceuticals/administration & dosage , Radiotherapy Dosage , Remission Induction , Survival Rate
14.
Baillieres Clin Endocrinol Metab ; 7(2): 491-507, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8489488

ABSTRACT

MIBG radiolabelled with 131I or 123I is a radiopharmaceutical which is concentrated in neuroendocrine tumours, particularly phaeochromocytomas and neuroblastomas. This permits non-invasive whole-body scintigraphic screening for benign and malignant, familial and sporadic, intra-adrenal and extra-adrenal phaeochromocytomas and primary and metastatic neuroblastomas, with high sensitivity (85-90%) and specificity (> 95%). MIBG is also concentrated in presynaptic terminals of adrenergic, autonomically innervated organs such as the heart, and may be used as a non-invasive in vivo probe to study this system. Large doses of 131I-MIBG and 125I-MIBG have been used experimentally to selectively deliver therapeutic doses of radiotherapy to malignant phaeochromocytomas and refractory advanced neuroblastomas.


Subject(s)
Catecholamines/metabolism , Iodine Radioisotopes , Iodobenzenes/therapeutic use , Neuroblastoma , Pheochromocytoma , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/diagnostic imaging , Adrenal Gland Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Iodine Radioisotopes/therapeutic use , Neuroblastoma/diagnostic imaging , Neuroblastoma/metabolism , Neuroblastoma/radiotherapy , Pheochromocytoma/diagnostic imaging , Pheochromocytoma/metabolism , Pheochromocytoma/radiotherapy , Radionuclide Imaging
15.
Eur J Nucl Med ; 15(9): 618-23, 1989.
Article in English | MEDLINE | ID: mdl-2598959

ABSTRACT

We have recently reported evidence that the calcium antagonist nifedipine can improve the tumour retention of 131I-metaidobenzylguanidine (131I-MIBG) in patients with malignant phaeochromocytoma. During studies of the pharmacological modification of tumour MIBG kinetics, it is important to distinguish clearly between a direct effect on MIBG cellular retention by a pharmaceutical, and secondary effects due, for example, to a change in glomerular filtration rate (GFR). In order to provide the fundamental kinetic data required for the numerical modelling of the effect of nifedipine on tumour MIBG kinetics, we have investigated the influence of GFR on MIBG plasma and renal kinetics. The 123I-MIBG plasma curve and MIBG renal plasma clearance rate were studied in ten patients, ranging from subjects without biochemical or scintigraphic evidence of phaeochromocytoma to individuals with widely disseminated metastatic disease. GFR was measured using the 99mTc-DTPA plasma clearance method. In four cases, the studies were repeated with the patients taking oral nifedipine. Statistically significant correlations were found between GFR and the MIBG plasma concentration. MIBG renal plasma clearance rate and the early (0 to 5 min) renal excretion of MIBG. The data permit the evaluation of the plasma integral during the first few min following bolus injection, a quantity important in the numerical modelling of tumour kinetics. GFR was found to have a major influence on whole-body MIBG kinetics, but there was also evidence of the effect of the metastatic tumour burden.


Subject(s)
Glomerular Filtration Rate , Iodine Radioisotopes/therapeutic use , Iodobenzenes/pharmacokinetics , Pheochromocytoma/radiotherapy , 3-Iodobenzylguanidine , Adult , Combined Modality Therapy , Glomerular Filtration Rate/drug effects , Humans , Iodobenzenes/therapeutic use , Nifedipine/therapeutic use , Pheochromocytoma/drug therapy , Pheochromocytoma/metabolism
16.
Eur J Nucl Med ; 14(7-8): 345-8, 1988.
Article in English | MEDLINE | ID: mdl-3181183

ABSTRACT

Following a case report that oral nifedipine can suppress the secretion of noradrenaline by phaeochromocytoma, we examined the effect of nifedipine on the tumour kinetics of tracer 131I-meta-iodobenzylguanidine (131I-mIBG) in five patients referred for mIBG radionuclide therapy for disseminated malignant phaeochromocytoma. In one subject a striking modification of mIBG kinetics was found that resulted in a doubling of the absorbed dose to tumour while the patient was taking nifedipine. At the same time, urinary excretion of noradrenaline was suppressed by a factor of three. The effect of nifedipine in this patient was confirmed when tracer studies were repeated nine months later. The changes in tumour kinetics were shown to be due to prolonged retention of mIBG rather than increased tumour blood flow or alteration of the curve of mIBG plasma concentration as a function of time.


Subject(s)
Adrenal Gland Neoplasms/radiotherapy , Iodine Radioisotopes/therapeutic use , Iodobenzenes/pharmacokinetics , Nifedipine/pharmacology , Pheochromocytoma/radiotherapy , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/metabolism , Humans , Iodobenzenes/therapeutic use , Nifedipine/therapeutic use , Pheochromocytoma/metabolism
17.
Semin Nucl Med ; 15(2): 132-53, 1985 Apr.
Article in English | MEDLINE | ID: mdl-3890187

ABSTRACT

Radioiodinated meta-iodobenzylguanidine, a recently developed radiopharmaceutical, has been shown to permit safe, noninvasive, sensitive, and specific scintigraphic location of pheochromocytomas of all types. The technique is especially efficacious in the case of extraadrenal primary lesions and locally recurrent and metastatic tumors. In addition to being taken up by pheochromocytomas, meta-iodobenzylguanidine may be used to image neuroblastomas, nonfunctioning paragangliomas, and carcinoid tumors. Lesions with high 131I-meta-iodobenzylguanidine uptake may respond to treatment with large doses of this radiopharmaceutical.


Subject(s)
Adrenal Gland Neoplasms/diagnostic imaging , Iodine Radioisotopes , Iodobenzenes , Pheochromocytoma/diagnostic imaging , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/radiotherapy , Adult , Aged , Child , Female , Humans , Iodine Radioisotopes/therapeutic use , Iodobenzenes/therapeutic use , Male , Middle Aged , Pheochromocytoma/radiotherapy , Radionuclide Imaging
18.
Cardiology ; 72 Suppl 1: 137-42, 1985.
Article in English | MEDLINE | ID: mdl-4053121

ABSTRACT

The newly developed radiopharmaceutical, 131I-metaiodobenzylguanidine (131I-MIBG), has been shown to be efficacious for the location of intra- and extra-adrenal, primary pheochromocytomas and metastatic, malignant pheochromocytomas (11.4% false-negative and 1.8% false-positive in patients with proven pheochromocytomas). Preliminary experience in selected patients with malignant pheochromocytoma suggest that therapy using large doses of 131I-MIBG results in partial tumor regression and improvement in catecholamine hypersecretion in some cases.


Subject(s)
Adrenal Gland Neoplasms/diagnostic imaging , Pheochromocytoma/diagnostic imaging , 3-Iodobenzylguanidine , Adrenal Gland Neoplasms/radiotherapy , Adrenal Glands/diagnostic imaging , Catecholamines/metabolism , Humans , Iodine Radioisotopes/therapeutic use , Iodobenzenes/therapeutic use , Pheochromocytoma/radiotherapy , Radionuclide Imaging
SELECTION OF CITATIONS
SEARCH DETAIL