Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Investig Med High Impact Case Rep ; 12: 23247096231224349, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38193433

RESUMO

Niacin, an important component of a balanced diet, is central to lipid metabolism. Occasionally used to treat hyperlipidemia, niacin is widely available without a prescription, making its use often unknown to treating physicians. Severe hepatotoxicity has been reported with niacin use. In the following report, we describe a case of hospitalization for acute decompensated cirrhosis with cholestatic morphology in a patient taking self-initiated large quantities of extended-release niacin. Despite medical management and support, the patient unfortunately expired on day 16 of hospitalization. Given ease of access and unclear long-term benefit in hyperlipidemia, the current case serves to raise awareness of niacin's potential hepatotoxicity through highlighting a severe outcome. Although mode of liver injury remains unknown, the use of extended-release niacin formulations and prolonged high-dose supplementation is associated with enhanced hepatotoxicity. Careful review and counseling of commonly available supplements remains an important task of both hospital and primary care physicians.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Colestase , Hiperlipidemias , Niacina , Humanos , Hiperlipidemias/tratamento farmacológico , Niacina/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Suplementos Nutricionais
2.
J Nucl Med ; 65(1): 109-116, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37945380

RESUMO

Although pancreatic ductal adenocarcinoma (PDAC) is associated with limited treatment options and poor patient outcomes, targeted α-particle therapy (TAT) represents a promising development in the field. TAT shows potential in treating metastatic cancers, including those that have become resistant to conventional treatments. Among the most auspicious radionuclides stands the in vivo α-generator 212Pb. Combined with the imaging-compatible radionuclide 203Pb, this theranostic match is a promising modality rapidly translating into the clinic. Methods: Using the pretargeting approach between a radiolabeled 1,2,4,5-tetrazine (Tz) tracer and a trans-cyclooctene (TCO) modified antibody, imaging and therapy with radiolead were performed on a PDAC tumor xenograft mouse model. For therapy, 3 cohorts received a single administration of 1.1, 2.2, or 3.7 MBq of the pretargeting agent, [212Pb]Pb-DO3A-PEG7-Tz, whereby administered activity levels were guided by dosimetric analysis. Results: The treated mice were holistically evaluated; minimal-to-mild renal tubular necrosis was observed. At the same time, median survival doubled for the highest-dose cohort (10.7 wk) compared with the control cohort (5.1 wk). Conclusion: This foundational study demonstrated the feasibility and safety of pretargeted TAT with 212Pb in PDAC while considering dose limitations and potential adverse effects.


Assuntos
Neoplasias Pancreáticas , Compostos Radiofarmacêuticos , Humanos , Animais , Camundongos , Compostos Radiofarmacêuticos/uso terapêutico , Chumbo , Medicina de Precisão , Linhagem Celular Tumoral , Radioisótopos , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/radioterapia
3.
J Hematol Oncol ; 15(1): 165, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371226

RESUMO

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.


Assuntos
Neoplasias do Sistema Nervoso Central , Neuroblastoma , Humanos , Animais , Camundongos , Distribuição Tecidual , Recidiva Local de Neoplasia/tratamento farmacológico , Anticorpos Monoclonais/efeitos adversos , Neoplasias do Sistema Nervoso Central/radioterapia , Neuroblastoma/radioterapia , Antígenos B7
4.
J Med Chem ; 61(17): 7589-7613, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30141927

RESUMO

Bile acid signaling and metabolism in the gastrointestinal tract have wide-ranging influences on systemic disease. G protein-coupled bile acid receptor 1 (GPBAR1, TGR5) is one of the major effectors in bile acid sensing, with demonstrated influence on metabolic, inflammatory, and proliferative processes. The pharmacologic utility of TGR5 agonists has been limited by systemic target-related effects such as excessive gallbladder filling and blockade of gallbladder emptying. Gut-restricted TGR5 agonists, however, have the potential to avoid these side effects and consequently be developed into drugs with acceptable safety profiles. We describe the discovery and optimization of a series of gut-restricted TGR5 agonists that elicit a potent response in mice, with minimal gallbladder-related effects. The series includes 12 (TGR5 EC50: human, 143 nM; mouse, 1.2 nM), a compound with minimal systemic availability that may have therapeutic value to patients with type 2 diabetes mellitus, nonalcoholic steatohepatitis, or inflammatory bowel disease.


Assuntos
Vesícula Biliar/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Tiazolidinas/química , Animais , Cães , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Fármacos Gastrointestinais/efeitos adversos , Fármacos Gastrointestinais/química , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/metabolismo , Relação Estrutura-Atividade
5.
Radiology ; 287(2): 667-675, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29388903

RESUMO

Purpose To assess the clinical safety, pharmacokinetics, and tumor imaging characteristics of fluorine 18-(2S,4R)-4-fluoroglutamine (FGln), a glutamine analog radiologic imaging agent. Materials and Methods This study was approved by the institutional review board and conducted under a U.S. Food and Drug Administration-approved Investigational New Drug application in accordance with the Helsinki Declaration and the Health Insurance Portability and Accountability Act. All patients provided written informed consent. Between January 2013 and October 2016, 25 adult patients with cancer received an intravenous bolus of FGln tracer (mean, 244 MBq ± 118, <100 µg) followed by positron emission tomography (PET) and blood radioassays. Patient data were summarized with descriptive statistics. FGln biodistribution and plasma amino acid levels in nonfasting patients (n = 13) were compared with those from patients who fasted at least 8 hours before injection (n = 12) by using nonparametric one-way analysis of variance with Bonferroni correction. Tumor FGln avidity versus fluorodeoxyglucose (FDG) avidity in patients with paired PET scans (n = 15) was evaluated with the Fisher exact test. P < .05 was considered indicative of a statistically significant difference. Results FGln PET depicted tumors of different cancer types (breast, pancreas, renal, neuroendocrine, lung, colon, lymphoma, bile duct, or glioma) in 17 of the 25 patients, predominantly clinically aggressive tumors with genetic mutations implicated in abnormal glutamine metabolism. Acute fasting had no significant effect on FGln biodistribution and plasma amino acid levels. FGln-avid tumors were uniformly FDG-avid but not vice versa (P = .07). Patients experienced no adverse effects. Conclusion Preliminary human FGln PET trial results provide clinical validation of abnormal glutamine metabolism as a potential tumor biomarker for targeted radiotracer imaging in several different cancer types. © RSNA, 2018 Online supplemental material is available for this article. Clinical trial registration no. NCT01697930.


Assuntos
Radioisótopos de Flúor/farmacocinética , Glutamina/análogos & derivados , Glutamina/metabolismo , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Tomografia por Emissão de Pósitrons , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica , Feminino , Radioisótopos de Flúor/metabolismo , Glutamina/farmacocinética , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/patologia , Distribuição Tecidual/efeitos dos fármacos , Estados Unidos , United States Food and Drug Administration
6.
Theranostics ; 6(12): 2267-2277, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27924162

RESUMO

The complementary nature of positron emission tomography (PET) and near-infrared fluorescence (NIRF) imaging makes the development of strategies for the multimodal PET/NIRF imaging of cancer a very enticing prospect. Indeed, in the context of colorectal cancer, a single multimodal PET/NIRF imaging agent could be used to stage the disease, identify candidates for surgical intervention, and facilitate the image-guided resection of the disease. While antibodies have proven to be highly effective vectors for the delivery of radioisotopes and fluorophores to malignant tissues, the use of radioimmunoconjugates labeled with long-lived nuclides such as 89Zr poses two important clinical complications: high radiation doses to the patient and the need for significant lag time between imaging and surgery. In vivo pretargeting strategies that decouple the targeting vector from the radioactivity at the time of injection have the potential to circumvent these issues by facilitating the use of positron-emitting radioisotopes with far shorter half-lives. Here, we report the synthesis, characterization, and in vivo validation of a pretargeted strategy for the multimodal PET and NIRF imaging of colorectal carcinoma. This approach is based on the rapid and bioorthogonal ligation between a trans-cyclooctene- and fluorophore-bearing immunoconjugate of the huA33 antibody (huA33-Dye800-TCO) and a 64Cu-labeled tetrazine radioligand (64Cu-Tz-SarAr). In vivo imaging experiments in mice bearing A33 antigen-expressing SW1222 colorectal cancer xenografts clearly demonstrate that this approach enables the non-invasive visualization of tumors and the image-guided resection of malignant tissue, all at only a fraction of the radiation dose created by a directly labeled radioimmunoconjugate. Additional in vivo experiments in peritoneal and patient-derived xenograft models of colorectal carcinoma reinforce the efficacy of this methodology and underscore its potential as an innovative and useful clinical tool.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Carcinoma/diagnóstico por imagem , Neoplasias Colorretais/diagnóstico por imagem , Radioisótopos de Cobre/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Animais , Anticorpos Antineoplásicos/química , Radioisótopos de Cobre/química , Modelos Animais de Doenças , Corantes Fluorescentes/química , Xenoenxertos , Camundongos
7.
J Am Soc Nephrol ; 26(5): 1138-49, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25404658

RESUMO

In CKD, phosphate retention arising from diminished GFR is a key early step in a pathologic cascade leading to hyperthyroidism, metabolic bone disease, vascular calcification, and cardiovascular mortality. Tenapanor, a minimally systemically available inhibitor of the intestinal sodium-hydrogen exchanger 3, is being evaluated in clinical trials for its potential to (1) lower gastrointestinal sodium absorption, (2) improve fluid overload-related symptoms, such as hypertension and proteinuria, in patients with CKD, and (3) reduce interdialytic weight gain and intradialytic hypotension in ESRD. Here, we report the effects of tenapanor on dietary phosphorous absorption. Oral administration of tenapanor or other intestinal sodium-hydrogen exchanger 3 inhibitors increased fecal phosphorus, decreased urine phosphorus excretion, and reduced [(33)P]orthophosphate uptake in rats. In a rat model of CKD and vascular calcification, tenapanor reduced sodium and phosphorus absorption and significantly decreased ectopic calcification, serum creatinine and serum phosphorus levels, circulating phosphaturic hormone fibroblast growth factor-23 levels, and heart mass. These results indicate that tenapanor is an effective inhibitor of dietary phosphorus absorption and suggest a new approach to phosphate management in renal disease and associated mineral disorders.


Assuntos
Calcinose/prevenção & controle , Trato Gastrointestinal/efeitos dos fármacos , Isoquinolinas/uso terapêutico , Fósforo/urina , Insuficiência Renal Crônica/tratamento farmacológico , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Sulfonamidas/uso terapêutico , Animais , Calcinose/etiologia , Modelos Animais de Doenças , Trato Gastrointestinal/metabolismo , Isoquinolinas/farmacologia , Masculino , Distribuição Aleatória , Ratos Sprague-Dawley , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Sulfonamidas/farmacologia
8.
J Nucl Med ; 51(7): 1123-30, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20554722

RESUMO

UNLABELLED: The development of novel multimodality imaging agents and techniques represents the current frontier of research in the field of medical imaging science. However, the combination of nuclear tomography with optical techniques has yet to be established. Here, we report the use of the inherent optical emissions from the decay of radiopharmaceuticals for Cerenkov luminescence imaging (CLI) of tumors in vivo and correlate the results with those obtained from concordant immuno-PET studies. METHODS: In vitro phantom studies were used to validate the visible light emission observed from a range of radionuclides including the positron emitters (18)F, (64)Cu, (89)Zr, and (124)I; beta-emitter (131)I; and alpha-particle emitter (225)Ac for potential use in CLI. The novel radiolabeled monoclonal antibody (89)Zr-desferrioxamine B [DFO]-J591 for immuno-PET of prostate-specific membrane antigen (PSMA) expression was used to coregister and correlate the CLI signal observed with the immuno-PET images and biodistribution studies. RESULTS: Phantom studies confirmed that Cerenkov radiation can be observed from a range of positron-, beta-, and alpha-emitting radionuclides using standard optical imaging devices. The change in light emission intensity versus time was concordant with radionuclide decay and was also found to correlate linearly with both the activity concentration and the measured PET signal (percentage injected dose per gram). In vivo studies conducted in male severe combined immune deficient mice bearing PSMA-positive, subcutaneous LNCaP tumors demonstrated that tumor-specific uptake of (89)Zr-DFO-J591 could be visualized by both immuno-PET and CLI. Optical and immuno-PET signal intensities were found to increase over time from 24 to 96 h, and biodistribution studies were found to correlate well with both imaging modalities. CONCLUSION: These studies represent the first, to our knowledge, quantitative assessment of CLI for measuring radiotracer uptake in vivo. Many radionuclides common to both nuclear tomographic imaging and radiotherapy have the potential to be used in CLI. The value of CLI lies in its ability to image radionuclides that do not emit either positrons or gamma-rays and are, thus, unsuitable for use with current nuclear imaging modalities. Optical imaging of Cerenkov radiation emission shows excellent promise as a potential new imaging modality for the rapid, high-throughput screening of radiopharmaceuticals.


Assuntos
Diagnóstico por Imagem , Luminescência , Compostos Radiofarmacêuticos , Partículas alfa , Animais , Anticorpos Monoclonais , Partículas beta , Desferroxamina , Avaliação Pré-Clínica de Medicamentos , Elétrons , Feminino , Humanos , Masculino , Camundongos , Transplante de Neoplasias/diagnóstico por imagem , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Imagens de Fantasmas , Tomografia por Emissão de Pósitrons , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/imunologia , Radioisótopos , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual , Zircônio
9.
Nucl Med Biol ; 32(8): 891-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16253815

RESUMO

86Y is an attractive PET radionuclide due to its intermediate half-life. (86)Y was produced via the 86Sr(p,n)86Y nuclear reaction. Enriched SrCO3 or SrO was irradiated with 2-6 microA of beam current for <4 h on a CS-15 cyclotron. It was shown that the SrO target could withstand at least 6 microA of beam current, a significant improvement over a maximum of 2 microA on the SrCO3 target. Average yields of 4.5 mCi/microA.h were achieved with SrO, which represent 71% of the theoretical yield, compared to 2.3 mCi/microA.h with SrCO3. The radioisotopic contaminants were (86m)Y (220%), 87Y (0.27%), (87m)Y (0.43%) and 88Y (0.024%). 86Y was isolated in an electrochemical cell consisting of three Pt electrodes. The solution was electrolyzed at 2000 mA (40 min) using two Pt plate electrodes. A second electrolysis (230 mA for 20 min) was performed using one Pt plate and a Pt wire. On average, 97.1% of the 86Y was recollected on the Pt wire after a second electrolysis. The (86)Y was collected from the Pt wire using 2.8 M HNO3/EtOH (3:1). After evaporation, 86Y was reconstituted in 100 microl of 0.1 M HCl. Target materials were recovered as SrCO3 and then converted to SrO by thermal decomposition at 1150 degrees C. Specific activity of 86Y was determined to be 29+/-19 mCi/microg via titration of 86Y(OAc)3 with DOTA or DTPA. We have established techniques for the routine, economical production of high purity, high specific activity 86Y on a small biomedical cyclotron that are translatable to other institutions.


Assuntos
Ciclotrons , Estabilidade de Medicamentos , Compostos Radiofarmacêuticos/análise , Compostos Radiofarmacêuticos/síntese química , Coloração e Rotulagem/métodos , Radioisótopos de Ítrio/análise , Radioisótopos de Ítrio/química , Engenharia Biomédica/métodos , Avaliação Pré-Clínica de Medicamentos , Cinética , Garantia da Qualidade dos Cuidados de Saúde/métodos , Controle de Qualidade
10.
Nucl Med Biol ; 32(1): 21-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15691658

RESUMO

64Cu-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM) is a promising radiotherapy agent for the treatment of hypoxic tumors. In an attempt to elucidate the radiobiological basis of 64Cu-ATSM radiotherapy, we have investigated the cellular response patterns in vitro cell line models. Cells were incubated with 64Cu-ATSM, and the dose-response curves were obtained by performing a clonogenic survival assay. Radiation-induced damage in DNA was evaluated using the alkali comet assay and apoptotic cells were detected using Annexin V-FITC and propidium iodide staining methods. Washout rate and subcellular distribution of 64Cu in cells were investigated to further assess the effectiveness of 64Cu-ATSM therapy on a molecular basis. A direct comparison of subcellular localization of Cu-ATSM was made with the flow tracer analog Cu-pyruvladehyde-bis(N4-methylthiosemicarbazone). In this study, 64Cu-ATSM was shown to reduce the clonogenic survival rate of tumor cells in a dose-dependent manner. Under hypoxic conditions, cells took up 64Cu-ATSM and radioactive 64Cu was highly accumulated in the cells. In the 64Cu-ATSM-treated cells, DNA damage by the radiation emitted from 64Cu was detected, and inhibition of cell proliferation and induction of apoptosis was observed at 24 and 36 h after the treatment. The typical features of postmitotic apoptosis induced by radiation were observed following 64Cu-ATSM treatment. The majority of the 64Cu taken up into the cells remained in the postmitochondrial supernatant (the cellular residue after removal of the nuclei and mitochondria), which indicates that the beta- particle emitted from 64Cu may be as effective as the Auger electrons in 64Cu-ATSM therapy. These data allow us to postulate that 64Cu-ATSM will be able to attack the hypoxic tumor cells directly, as well as potentially affecting the peripheral nonhypoxic regions indirectly by the beta- particle decay of 64Cu.


Assuntos
Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/radioterapia , Radioisótopos de Cobre/farmacocinética , Radioisótopos de Cobre/uso terapêutico , Compostos Organometálicos/farmacocinética , Compostos Organometálicos/uso terapêutico , Tiossemicarbazonas/farmacocinética , Tiossemicarbazonas/uso terapêutico , Animais , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Complexos de Coordenação , Avaliação Pré-Clínica de Medicamentos , Taxa de Depuração Metabólica , Camundongos , Compostos Radiofarmacêuticos/farmacocinética , Compostos Radiofarmacêuticos/uso terapêutico , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA