Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Hematol Oncol ; 15(1): 165, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371226

ABSTRACT

BACKGROUND: The prognosis for metastatic and recurrent tumors of the central nervous system (CNS) remains dismal, and the need for newer therapeutic targets and modalities is critical. The cell surface glycoprotein B7H3 is expressed on a range of solid tumors with a restricted expression on normal tissues. We hypothesized that compartmental radioimmunotherapy (cRIT) with the anti-B7H3 murine monoclonal antibody omburtamab injected intraventricularly could safely target CNS malignancies. PATIENTS AND METHODS: We conducted a phase I trial of intraventricular 131I-omburtamab using a standard 3 + 3 design. Eligibility criteria included adequate cerebrospinal fluid (CSF) flow, no major organ toxicity, and for patients > dose level 6, availability of autologous stem cells. Patients initially received 74 MBq radioiodinated omburtamab to evaluate dosimetry and biodistribution followed by therapeutic 131I-omburtamab dose-escalated from 370 to 2960 MBq. Patients were monitored clinically and biochemically for toxicity graded using CTCAEv 3.0. Dosimetry was evaluated using serial CSF and blood sampling, and serial PET or gamma-camera scans. Patients could receive a second cycle in the absence of grade 3/4 non-hematologic toxicity or progressive disease. RESULTS: Thirty-eight patients received 100 radioiodinated omburtamab injections. Diagnoses included metastatic neuroblastoma (n = 16) and other B7H3-expressing solid tumors (n = 22). Thirty-five patients received at least 1 cycle of treatment with both dosimetry and therapy doses. Acute toxicities included < grade 4 self-limited headache, vomiting or fever, and biochemical abnormalities. Grade 3/4 thrombocytopenia was the most common hematologic toxicity. Recommended phase 2 dose was 1850 MBq/injection. The median radiation dose to the CSF and blood by sampling was 1.01 and 0.04 mGy/MBq, respectively, showing a consistently high therapeutic advantage for CSF. Major organ exposure was well below maximum tolerated levels. In patients developing antidrug antibodies, blood clearance, and therefore therapeutic index, was significantly increased. In patients receiving cRIT for neuroblastoma, survival was markedly increased (median PFS 7.5 years) compared to historical data. CONCLUSIONS: cRIT with 131I-omburtamab is safe, has favorable dosimetry and may have a therapeutic benefit as adjuvant therapy for B7-H3-expressing leptomeningeal metastases. TRIAL REGISTRATION: clinicaltrials.gov NCT00089245, August 5, 2004.


Subject(s)
Central Nervous System Neoplasms , Neuroblastoma , Humans , Animals , Mice , Tissue Distribution , Neoplasm Recurrence, Local/drug therapy , Antibodies, Monoclonal/adverse effects , Central Nervous System Neoplasms/radiotherapy , Neuroblastoma/radiotherapy , B7 Antigens
2.
J Nucl Med ; 62(9): 1192-1199, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34475242

ABSTRACT

Whereas benign pheochromocytomas and paragangliomas are often successfully cured by surgical resection, treatment of metastatic disease can be challenging in terms of both disease control and symptom control. Fortunately, several options are available, including chemotherapy, radiation therapy, and surgical debulking. Radiolabeled metaiodobenzylguanidine (MIBG) and somatostatin receptor imaging have laid the groundwork for use of these radiopharmaceuticals as theranostic agents. 131I-MIBG therapy of neuroendocrine tumors has a long history, and the recent approval of high-specific-activity 131I-MIBG for metastatic or inoperable pheochromocytoma or paraganglioma by the U.S. Food and Drug Administration has resulted in general availability of, and renewed interest in, this treatment. Although reports of peptide receptor radionuclide therapy of pheochromocytoma and paraganglioma with 90Y- or 177Lu-DOTA conjugated somatostatin analogs have appeared in the literature, the approval of 177Lu-DOTATATE in the United States and Europe, together with National Comprehensive Cancer Network guidelines suggesting its use in patients with metastatic or inoperable pheochromocytoma and paraganglioma, has resulted in renewed interest. These agents have shown evidence of efficacy as palliative treatments in patients with metastatic or inoperable pheochromocytoma or paraganglioma. In this continuing medical education article, we discuss the therapy of pheochromocytoma and paraganglioma with 131I-MIBG and 90Y- or 177Lu-DOTA-somatostatin analogs.


Subject(s)
Paraganglioma , Pheochromocytoma , Positron-Emission Tomography , Radionuclide Imaging
3.
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
4.
Pediatr Blood Cancer ; 63(5): 801-7, 2016 May.
Article in English | MEDLINE | ID: mdl-26773712

ABSTRACT

BACKGROUND: Although (131) I-metaiodobenzylguanidine ((131) I-MIBG) therapy is increasingly used for children with high-risk neuroblastoma, a paucity of lead-lined rooms limits its wider use. We implemented radiation safety procedures to comply with New York City Department of Health and Mental Hygiene regulations for therapeutic radioisotopes and administered (131) I-MIBG using rolling lead shields. PROCEDURE: Patients received 0.67 GBq (18 mCi)/kg/dose (131) I-MIBG on an IRB-approved protocol (NCT00107289). Radiation safety procedures included private room with installation of rolling lead shields to maintain area dose rates ≤0.02 mSv/hr outside the room, patient isolation until dose rate <0.07 mSv/hr at 1 m, and retention of a urinary catheter with collection of urine in lead boxes. Parents were permitted in the patient's room behind lead shields, trained in radiation safety principles, and given real-time radiation monitors. RESULTS: Records on 16 (131) I-MIBG infusions among 10 patients (age 2-11 years) were reviewed. Mean ± standard deviation (131) I-MIBG administered was 17.67 ± 11.14 (range: 6.11-40.59) GBq. Mean maximum dose rates outside treatment rooms were 0.013 ± 0.008 mSv/hr. Median time-to-discharge was 3 days post-(131) I-MIBG. Exposure of medical staff and parents was below regulatory limits. Cumulative whole-body dose received by the physician, nurse, and radiation safety officer during treatment was 0.098 ± 0.058, 0.056 ± 0.045, 0.055 ± 0.050 mSv, respectively. Cumulative exposure to parents was 0.978 ± 0.579 mSv. Estimated annual radiation exposure for inpatient nurses was 0.096 ± 0.034 mSv/nurse. Thyroid bioassay scans on all medical personnel showed less than detectable activity. Contamination surveys were <200 dpm/100 cm(2) . CONCLUSIONS: The use of rolling lead shields and implementation of specific radiation safety procedures allows administration of high-dose (131) I-MIBG and may broaden its use without dedicated lead-lined rooms.


Subject(s)
Iodine Radioisotopes/administration & dosage , Neuroblastoma/radiotherapy , Radiation Exposure/standards , Radiation Protection , Adult , Child , Child, Preschool , Female , Humans , Male , Radiotherapy Dosage , Time Factors
5.
J Nucl Med ; 52(8): 1173-80, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21764796

ABSTRACT

UNLABELLED: Humanized A33 (huA33) is a promising monoclonal antibody that recognizes A33 antigen, which is present in more than 95% of colorectal cancers and in normal bowel. In this study, we took advantage of quantitative PET to evaluate (124)I huA33 targeting, biodistribution, and safety in patients with colorectal cancer. We also determined the biodistribution of (124)I-huA33 when a large dose of human intravenous IgG (IVIG) was administered to manipulate the Fc receptor or when (124)I-huA33 was given via hepatic arterial infusion (HAI). METHODS: We studied 25 patients with primary or metastatic colorectal cancer; 19 patients had surgical exploration or resection. Patients received a median of 343 MBq (44.4-396 MBq) and 10 mg of (124)I-huA33. Nineteen patients received the antibody intravenously and 6 patients via HAI, and 5 patients also received IVIG. RESULTS: Ten of 12 primary tumors were visualized in 11 patients. The median concentration in primary colon tumors was 0.016% injected dose per gram, compared with 0.004% in normal colon. The PET-based median ratio of hepatic tumor uptake to normal-liver uptake was 3.9 (range, 1.8-22.2). Quantitation using PET, compared with well counting of serum and tissue, showed little difference. Prominent uptake in bowel hindered tumor identification in some patients. Pharmacokinetics showed that patients receiving IVIG had a significantly shorter serum half-time (41.6 ± 14.0 h) than those without (65.2 ± 9.8 h). There were no differences in clearance rates among the intravenous group, IVIG group, and HAI group, nor was there any difference in serum area under the curve, maximum serum concentration, or volume of distribution. Weak titers of human-antihuman antibodies were observed in 6 of 25 patients. No acute side effects or significant toxicities were associated with huA33. CONCLUSION: Good localization of (124)I-huA33 in colorectal cancer with no significant toxicity has been observed. PET-derived (124)I concentrations agreed well with those obtained by well counting of surgically resected tissue and blood, confirming the quantitative accuracy of (124)I-huA33 PET. The HAI route had no advantage over the intravenous route. No clinically significant changes in blood clearance were induced by IVIG.


Subject(s)
Colorectal Neoplasms/immunology , Iodine Radioisotopes/pharmacology , Membrane Glycoproteins/chemistry , Positron-Emission Tomography/methods , Aged , Area Under Curve , Colon/diagnostic imaging , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/therapy , Female , Humans , Immunoglobulins, Intravenous/metabolism , Immunoglobulins, Intravenous/pharmacokinetics , Male , Middle Aged , Neoplasm Metastasis , Prospective Studies , Radioimmunotherapy/methods , Treatment Outcome
6.
J Clin Invest ; 117(9): 2422-30, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17717602

ABSTRACT

The long plasma half-life of IgG, while allowing for enhanced tumor uptake of tumor-targeted IgG conjugates, also results in increased background activity and normal-tissue toxicity. Therefore, successful therapeutic uses of conjugated antibodies have been limited to the highly sensitive and readily accessible hematopoietic tumors. We report a therapeutic strategy to beneficially alter the pharmacokinetics of IgG antibodies via pharmacological inhibition of the neonatal Fc receptor (FcRn) using high-dose IgG therapy. IgG-treated mice displayed enhanced blood and whole-body clearance of radioactivity, resulting in better tumor-to-blood image contrast and protection of normal tissue from radiation. Tumor uptake and the resultant therapeutic response was unaltered. Furthermore, we demonstrated the use of this approach for imaging of tumors in humans and discuss its potential applications in cancer imaging and therapy. The ability to reduce the serum persistence of conjugated IgG antibodies after their infusion can enhance their therapeutic index, resulting in improved therapeutic and diagnostic efficacy.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Neoplasms/pathology , Neoplasms/therapy , Receptors, Fc/metabolism , Actinium/chemistry , Animals , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/adverse effects , Immunotherapy , Indium Radioisotopes/chemistry , Injections, Intravenous , Iodine Radioisotopes , Mice , Neoplasms/immunology , Neoplasms/metabolism , Positron-Emission Tomography , Time Factors
7.
Nucl Med Commun ; 27(1): 31-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16340721

ABSTRACT

BACKGROUND: Although the majority of pheochromocytomas (PHEO) are benign, a subset is malignant. Computed tomography (CT) and magnetic resonance imaging (MRI) localize PHEO with high sensitivity but, because of limited specificity, [(131)I]- or [(123)I]-metaiodobenzylguanidine ([(131)I]- or [(123)I]-MIBG) is often used as a complementary agent. 6-[18F]-fluorodopamine ([18F]-DA) has been developed as a radiopharmaceutical for the targeting of noradrenergic pathways, and has been shown to result in a better detection rate of PHEO sites than MIBG; however, [18F]-DA has shown a lack of accumulation in some patients with metastatic PHEO. METHODS: Five patients with widespread metastatic PHEO who had CT and MRI evidence of metastatic disease (one man and four women; age range, 25-64 years), and who underwent imaging with [(123)I]-MIBG, [18F]-DA and 2-[18F]-fluoro-2-deoxy-D-glucose ([18F]-FDG), were evaluated retrospectively. Tomographic imaging was performed and positron emission tomography (PET) images were inspected visually and quantitatively. RESULTS: All five patients had [(123)I]-MIBG scans that grossly underestimated the extent of disease when compared with conventional CT and MRI. All lesions seen on [(123)I]-MIBG scans were detected on [18F]-DA scans, which also detected additional lesions. Nonetheless, [18F]-DA also failed to detect numerous lesions seen on CT and MRI. In all of these cases, [18F]-FDG PET showed lesions that were not detected on either [(123)I]-MIBG or [18F]-DA scans. CONCLUSIONS: When [(123)I]-MIBG or [18F]-DA fails to localize lesions seen on conventional imaging studies, [18F]-FDG may be recommended as an ancillary test for the diagnosis and localization of metastatic PHEO. This is particularly important in patients with aggressive PHEO.


Subject(s)
3-Iodobenzylguanidine , Dopamine/analogs & derivatives , Fluorodeoxyglucose F18 , 3-Iodobenzylguanidine/pharmacokinetics , Adult , Dopamine/pharmacokinetics , False Negative Reactions , Female , Fluorine Radioisotopes/pharmacokinetics , Fluorodeoxyglucose F18/pharmacokinetics , Humans , Iodine Radioisotopes/pharmacokinetics , Male , Middle Aged , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity
8.
J Clin Endocrinol Metab ; 89(5): 2214-21, 2004 May.
Article in English | MEDLINE | ID: mdl-15126544

ABSTRACT

Conventional imaging modalities cannot localize the source of ACTH in 30-50% of patients with Cushing's syndrome (CS) caused by ectopic ACTH secretion (EAS). We prospectively evaluated whether [(18)F]fluorodeoxyglucose (FDG) positron emission tomography (PET) or [(111)In]-diethylenetriaminepentaacetate-D-Phe-pentetreotide (OCT) at higher than standard doses of radionuclide (18 mCi; H-OCT), can detect these tumors. Seventeen patients with presumed EAS based on inferior petrosal sinus sampling results underwent routine anatomical imaging studies [computed tomography (CT) and magnetic resonance imaging (MRI)] and OCT scintigraphy with 6 mCi (L-OCT). Research studies included FDG-PET in all patients and H-OCT if L-OCT was negative. ACTH-secreting tumors were localized in 13 patients and were occult in four. Nine of 17 CT, six of 16 MRI, six of 17 FDG-PET, eight of 17 L-OCT, and one of nine H-OCT studies were true positives. The sensitivity of CT and combined H- and L-OCT scintigraphy was higher (both 53%; 95% confidence interval, 29-76%) than that of MRI (37%; 95% confidence interval, 16-64%) or FDG-PET (35%; 95% confidence interval, 15-61%). FDG-PET did not detect tumors that were occult on CT/MRI. L-OCT was a useful complementary modality to CT and MRI. As H-OCT identified a tumor in one patient with otherwise negative imaging, it should be considered only when other imaging modalities fail to localize the ACTH-secreting tumor in patients with EAS.


Subject(s)
ACTH Syndrome, Ectopic , Cushing Syndrome/etiology , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Tomography, Emission-Computed , Adult , Aged , Female , Fluorodeoxyglucose F18 , Humans , Indium Radioisotopes , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasms/complications , Neoplasms/diagnosis , Pentetic Acid , Radiopharmaceuticals , Tomography, X-Ray Computed
9.
Blood ; 100(1): 208-16, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12070029

ABSTRACT

We used a pretargeting technique to treat a nonobese diabetic/severe combined immunodeficient murine model of human adult T-cell leukemia with an anti-Tac antibody-streptavidin (HAT-SA) conjugate, which recognizes CD25, followed by bismuth 213 ((213)Bi)-1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA)- biotin. In the 3-step pretargeting radioimmunotherapy protocol, HAT-SA (140 or 400 microg) was administered intravenously (i.v.) to bind to the interleukin 2 receptor alpha (IL-2R alpha; CD25)-expressing tumor cells. After 24 hours, 100 microg of a synthetic clearing agent was administered i.v. to remove unbound circulating HAT-SA conjugate from the circulation. Four hours later, (213)Bi-DOTA-biotin was administered i.v. for therapy. Tumor growth was significantly inhibited in 3 trials by using 250 microCi (9.25 MBq) of (213)Bi-DOTA-biotin with a pretargeting technique as monitored by serum levels of soluble IL-2R alpha and/or human beta-2-microglobulin (P <.05, t test) and by survival of tumor-bearing mice in the treatment groups (P <.02, log rank test) as compared with the control groups. No prolongation of survival was observed with a nonspecific antibody-SA conjugate or in the absence of the radionuclide. Additionally, no prolongation of survival resulted from administration of (213)Bi directly linked to intact HAT. Furthermore, there was no prolongation of survival when the beta-emitting radionuclide yttrium 90 instead of the alpha-emitting radionuclide (213)Bi was used. The pretargeting approach with (213)Bi inhibited tumor growth more effectively than did immunotherapy with unmodified HAT. The best results were obtained with combination therapy that involved (213)Bi-DOTA-biotin with a pretargeting technique supplemented by 4 weekly doses of HAT. The findings of this study support the use of this combination approach in a clinical trial in patients with IL-2R alpha-expressing leukemias.


Subject(s)
Bismuth , Leukemia-Lymphoma, Adult T-Cell/radiotherapy , Radioimmunotherapy/methods , Radioisotopes , Alpha Particles , Animals , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/toxicity , Biotin , Bismuth/administration & dosage , Bismuth/pharmacokinetics , Bismuth/toxicity , Heterocyclic Compounds, 1-Ring , Mice , Mice, Inbred NOD , Neoplasms, Experimental/therapy , Radioisotopes/administration & dosage , Radioisotopes/pharmacokinetics , Radioisotopes/toxicity , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/toxicity , Receptors, Interleukin-2/immunology , Streptavidin , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL