Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Nucl Med ; 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38388517

RESUMO

[68Ga]Ga-DPI-4452, a first-in-class carbonic anhydrase IX-binding radiolabeled peptide, is the imaging agent of a theranostic pair with [177Lu]Lu-DPI-4452, developed for selecting and treating patients with carbonic anhydrase IX-expressing tumors. Here, [68Ga]Ga-DPI-4452 imaging characteristics, dosimetry, pharmacokinetics, and safety were assessed in 3 patients with clear cell renal cell carcinoma. Methods: After [68Ga]Ga-DPI-4452 administration, patients underwent serial full-body PET/CT imaging. Blood and urine were sampled. Safety was monitored for 7 d after injection. Results: Tumor uptake was observed at all time points (15 min to 4 h). Across 36 lesions, the SUVmax at 1 h after administration ranged from 6.8 to 211.6 (mean, 64.6 [SD, 54.8]). The kidneys, liver, and bone marrow demonstrated low activity. [68Ga]Ga-DPI-4452 was rapidly eliminated from blood and urine. No clinically significant toxicity was observed. Conclusion: [68Ga]Ga-DPI-4452 showed exceptional tumor uptake in patients with clear cell renal cell carcinoma, with very high tumor-to-background ratios and no significant adverse events, suggesting potential diagnostic and patient selection applications.

2.
J Nucl Med ; 54(6): 861-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23568366

RESUMO

UNLABELLED: The glutamic acid derivative (S)-4-(3-(18)F-Fluoropropyl)-l-glutamic acid ((18)F-FSPG, alias BAY 94-9392), a new PET tracer for the detection of malignant diseases, displayed promising results in non-small cell lung cancer patients. The aim of this study was to provide dosimetry estimates for (18)F-FSPG based on human whole-body PET/CT measurements. METHODS: (18)F-FSPG was prepared by a fully automated 2-step procedure and purified by a solid-phase extraction method. PET/CT scans were obtained for 5 healthy volunteers (mean age, 59 y; age range, 51-64 y; 2 men, 3 women). Human subjects were imaged for up to 240 min using a PET/CT scanner after intravenous injection of 299 ± 22.5 MBq of (18)F-FSPG. Image quantification, time-activity data modeling, estimation of normalized number of disintegrations, and production of dosimetry estimates were performed using the RADAR (RAdiation Dose Assessment Resource) method for internal dosimetry and in general concordance with the methodology and principles as presented in the MIRD 16 document. RESULTS: Because of the renal excretion of the tracer, the absorbed dose was highest in the urinary bladder wall and kidneys, followed by the pancreas and uterus. The individual organ doses (mSv/MBq) were 0.40 ± 0.058 for the urinary bladder wall, 0.11 ± 0.011 for the kidneys, 0.077 ± 0.020 for the pancreas, and 0.030 ± 0.0034 for the uterus. The calculated effective dose was 0.032 ± 0.0034 mSv/MBq. Absorbed dose to the bladder and the effective dose can be reduced significantly by frequent bladder-voiding intervals. For a 0.75-h voiding interval, the bladder dose was reduced to 0.10 ± 0.012 mSv/MBq, and the effective dose was reduced to 0.015 ± 0.0010 mSv/MBq. CONCLUSION: On the basis of the distribution and biokinetic data, the determined radiation dose for (18)F-FSPG was calculated to be 9.5 ± 1.0 mSv at a patient dose of 300 MBq, which is of similar magnitude to that of (18)F-FDG (5.7 mSv). The effective dose can be reduced to 4.5 ± 0.30 mSv (at 300 MBq), with a bladder-voiding interval of 0.75 h.


Assuntos
Glutamatos , Imagem Multimodal/métodos , Neoplasias/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Feminino , Glutamatos/efeitos adversos , Humanos , Marcação por Isótopo , Masculino , Pessoa de Meia-Idade , Radiometria , Segurança
3.
Blood ; 115(17): 3520-30, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20197554

RESUMO

Indoleamine 2,3-dioxygenase-1 (IDO1; IDO) mediates oxidative cleavage of tryptophan, an amino acid essential for cell proliferation and survival. IDO1 inhibition is proposed to have therapeutic potential in immunodeficiency-associated abnormalities, including cancer. Here, we describe INCB024360, a novel IDO1 inhibitor, and investigate its roles in regulating various immune cells and therapeutic potential as an anticancer agent. In cellular assays, INCB024360 selectively inhibits human IDO1 with IC(50) values of approximately 10nM, demonstrating little activity against other related enzymes such as IDO2 or tryptophan 2,3-dioxygenase (TDO). In coculture systems of human allogeneic lymphocytes with dendritic cells (DCs) or tumor cells, INCB024360 inhibition of IDO1 promotes T and natural killer (NK)-cell growth, increases IFN-gamma production, and reduces conversion to regulatory T (T(reg))-like cells. IDO1 induction triggers DC apoptosis, whereas INCB024360 reverses this and increases the number of CD86(high) DCs, potentially representing a novel mechanism by which IDO1 inhibition activates T cells. Furthermore, IDO1 regulation differs in DCs versus tumor cells. Consistent with its effects in vitro, administration of INCB024360 to tumor-bearing mice significantly inhibits tumor growth in a lymphocyte-dependent manner. Analysis of plasma kynurenine/tryptophan levels in patients with cancer affirms that the IDO pathway is activated in multiple tumor types. Collectively, the data suggest that selective inhibition of IDO1 may represent an attractive cancer therapeutic strategy via up-regulation of cellular immunity.


Assuntos
Células Dendríticas/imunologia , Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Neoplasias/imunologia , Linfócitos T/imunologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Antígeno B7-2/imunologia , Antígeno B7-2/metabolismo , Técnicas de Cocultura , Células Dendríticas/enzimologia , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Linfócitos T/enzimologia , Triptofano Oxigenase/imunologia , Triptofano Oxigenase/metabolismo
4.
Mol Cancer Ther ; 9(2): 489-98, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20124451

RESUMO

Malignant tumors arise, in part, because the immune system does not adequately recognize and destroy them. Expression of indoleamine-2,3-dioxygenase (IDO; IDO1), a rate-limiting enzyme in the catabolism of tryptophan into kynurenine, contributes to this immune evasion. Here we describe the effects of systemic IDO inhibition using orally active hydroxyamidine small molecule inhibitors. A single dose of INCB023843 or INCB024360 results in efficient and durable suppression of Ido1 activity in the plasma of treated mice and dogs, the former to levels seen in Ido1-deficient mice. Hydroxyamidines potently suppress tryptophan metabolism in vitro in CT26 colon carcinoma and PAN02 pancreatic carcinoma cells and in vivo in tumors and their draining lymph nodes. Repeated administration of these IDO1 inhibitors impedes tumor growth in a dose- and lymphocyte-dependent fashion and is well tolerated in efficacy and preclinical toxicology studies. Substantiating the fundamental role of tumor cell-derived IDO expression, hydroxyamidines control the growth of IDO-expressing tumors in Ido1-deficient mice. These activities can be attributed, at least partially, to the increased immunoreactivity of lymphocytes found in tumors and their draining lymph nodes and to the reduction in tumor-associated regulatory T cells. INCB024360, a potent IDO1 inhibitor with desirable pharmaceutical properties, is poised to start clinical trials in cancer patients.


Assuntos
Amidinas/farmacologia , Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Neoplasias/metabolismo , Triptofano/metabolismo , Animais , Linhagem Celular Tumoral , Cães , Feminino , Humanos , Sistema Imunitário , Imunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Cinurenina/farmacologia , Linfonodos/patologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Neoplasias/patologia
5.
Neoplasia ; 12(1): 28-38, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20072651

RESUMO

Cytokines in the bone marrow of multiple myeloma patients activate Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways in tumor cells and promote tumor growth, survival, and drug resistance. INCB16562 was developed as a novel, selective, and orally bioavailable small-molecule inhibitor of JAK1 and JAK2 markedly selective over JAK3. The specific cellular activity of the inhibitor was demonstrated by its potent and dose-dependent inhibition of cytokine-dependent JAK/STAT signaling and cell proliferation in the absence of effects on Bcr-Abl-expressing cells. Treatment of myeloma cells with INCB16562 potently inhibited interleukin-6 (IL-6)-induced phosphorylation of STAT3. Moreover, the proliferation and survival of myeloma cells dependent on IL-6 for growth, as well as the IL-6-induced growth of primary bone marrow-derived plasma cells from a multiple myeloma patient, were inhibited by INCB16562. Induction of caspase activation and apoptosis was observed and attributed, at least in part, to the suppression of Mcl-1 expression. Importantly, INCB16562 abrogated the protective effects of recombinant cytokines or bone marrow stromal cells and sensitized myeloma cells to cell death by exposure to dexamethasone, melphalan, or bortezomib. Oral administration of INCB16562 antagonized the growth of myeloma xenografts in mice and enhanced the antitumor activity of relevant agents in combination studies. Taken together, these data suggest that INCB16562 is a potent JAK1/2 inhibitor and that mitigation of JAK/STAT signaling by targeting JAK1 and JAK2 will be beneficial in the treatment of myeloma patients, particularly in combination with other agents.


Assuntos
Azepinas/farmacologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Células Estromais/efeitos dos fármacos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Azepinas/administração & dosagem , Azepinas/química , Western Blotting , Ácidos Borônicos/administração & dosagem , Bortezomib , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Humanos , Interleucina-6/farmacologia , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Melfalan/administração & dosagem , Camundongos , Camundongos SCID , Estrutura Molecular , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Pirazinas/administração & dosagem , Piridinas/administração & dosagem , Piridinas/química , Fator de Transcrição STAT3/metabolismo , Células Estromais/citologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Clin Cancer Res ; 15(22): 6891-900, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19887489

RESUMO

PURPOSE: Deregulation of the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway is a hallmark for the Philadelphia chromosome-negative myeloproliferative diseases polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We tested the efficacy of a selective JAK1/2 inhibitor in cellular and in vivo models of JAK2-driven malignancy. EXPERIMENTAL DESIGN: A novel inhibitor of JAK1/2 was characterized using kinase assays. Cellular effects of this compound were measured in cell lines bearing the JAK2V617F or JAK1V658F mutation, and its antiproliferative activity against primary polycythemiavera patient cells was determined using clonogenic assays. Antineoplastic activity in vivo was determined using a JAK2V617F-driven xenograft model, and effects of the compound on survival, organomegaly, body weight, and disease-associated inflammatory markers were measured. RESULTS: INCB16562 potently inhibited proliferation of cell lines and primary cells from PV patients carrying the JAK2V617F or JAK1V658F mutation by blocking JAK-STAT signaling and inducing apoptosis. In vivo, INCB16562 reduced malignant cell burden, reversed splenomegaly and normalized splenic architecture, improved body weight gains, and extended survival in a model of JAK2V617F-driven hematologic malignancy. Moreover, these mice suffered from markedly elevated levels of inflammatory cytokines, similar to advanced myeloproliferative disease patients, which was reversed upon treatment. CONCLUSIONS: These data showed that administration of the dual JAK1/2 inhibitor INCB16562 reduces malignant cell burden, normalizes spleen size and architecture, suppresses inflammatory cytokines, improves weight gain, and extends survival in a rodent model of JAK2V617F-driven hematologic malignancy. Thus, selective inhibitors of JAK1 and JAK2 represent a novel therapy for the patients with myeloproliferative diseases and other neoplasms associated with JAK dysregulation.


Assuntos
Inibidores Enzimáticos/farmacologia , Janus Quinase 1/genética , Janus Quinase 1/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação , Animais , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Concentração Inibidora 50 , Cinética , Camundongos , Transplante de Neoplasias , Policitemia Vera/tratamento farmacológico
7.
Hum Gene Ther ; 18(4): 312-22, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17408358

RESUMO

This study was done to aid in the design of a phase I gene therapy trial in patients with prostate cancer. We determined the dosimetric characteristics of our reporter gene system when coupled with intravenous administration of radioactive sodium pertechnetate (Na(99m) TcO(4)) and determined the feasibility of using human sodium iodide symporter (hNIS) as a reporter gene to study the dynamics of adenoviral transgene expression in a large animal tumor. A replication-competent Ad5-yCD/mutTK(SR39) rep-hNIS adenovirus was injected into the prostate gland of dogs for dosimetry purposes, and into a canine soft tissue sarcoma (STS) for imaging purposes. After resection of the prostate, the amount of (99m)TcO(4)() sequestered in the prostate was determined, the radiation dose absorbed by the prostate and nontarget critical organs was calculated, and hNIS reporter gene expression was imaged in the STS by single-photon emission computed tomography (SPECT). On the basis of the findings from 25 dogs, the amount of (99m)TcO (4)() sequestered in the prostate ranged from 13 to 276 muCi. Using the highest value observed, absorbed radiation dose to critical organs was calculated and found to be below U.S. Food and Drug Administration limits for diagnostic imaging. Also, (99m)TcO (4)() uptake was readily detected by SPECT and found to persist in vivo for at least 4 days. On the basis of our dosimetry calculations, up to five imaging procedures can be safely performed in humans after intraprostatic injection of the Ad5-yCD/mutTK(SR39)rep-hNIS adenovirus and the hNIS reporter gene system can be used to study the dynamics of adenoviral gene therapy vectors in large animal tumors.


Assuntos
Ensaios Clínicos Fase I como Assunto/normas , Genes Reporter , Neoplasias da Próstata/terapia , Interpretação de Imagem Radiográfica Assistida por Computador , Pertecnetato Tc 99m de Sódio/farmacocinética , Simportadores/genética , Animais , Cães , Terapia Genética , Vetores Genéticos , Humanos , Masculino , Projetos de Pesquisa , Pertecnetato Tc 99m de Sódio/administração & dosagem , Pertecnetato Tc 99m de Sódio/análise , Distribuição Tecidual
8.
J Med Chem ; 49(13): 3774-89, 2006 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-16789735

RESUMO

Potent nonpeptidic benzimidazole sulfonamide inhibitors of protein tyrosine phosphatase 1B (PTP1B) were derived from the optimization of a tripeptide containing the novel (S)-isothiazolidinone ((S)-IZD) phosphotyrosine (pTyr) mimetic. An X-ray cocrystal structure of inhibitor 46/PTP1B at 1.8 A resolution demonstrated that the benzimidazole sulfonamides form a bidentate H bond to Asp48 as designed, although the aryl group of the sulfonamide unexpectedly interacts intramolecularly in a pi-stacking manner with the benzimidazole. The ortho substitution to the (S)-IZD on the aryl ring afforded low nanomolar enzyme inhibitors of PTP1B that also displayed low caco-2 permeability and cellular activity in an insulin receptor (IR) phosphorylation assay and an Akt phosphorylation assay. The design, synthesis, and SAR of this novel series of benzimidazole sulfonamide containing (S)-IZD inhibitors of PTP1B are presented herein.


Assuntos
Benzimidazóis/síntese química , Oligopeptídeos/química , Fosfotirosina/química , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Sulfonamidas/síntese química , Tiazóis/síntese química , Benzimidazóis/química , Benzimidazóis/farmacologia , Linhagem Celular , Cristalografia por Raios X , Humanos , Modelos Moleculares , Mimetismo Molecular , Estrutura Molecular , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , Tiazóis/química , Tiazóis/farmacologia
10.
J Nucl Med ; 46(6): 1023-7, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15937315

RESUMO

UNLABELLED: The OLINDA/EXM version 1.0 personal computer code was created as a replacement for the widely used MIRDOSE3.1 code. This paper documents the basic function of the code and how it is similar to and different from the MIRDOSE software. METHODS: After creation of the code and alpha- and beta-testing phases, a premarket notification submission (510(k)) was filed with the Food and Drug Administration to permit marketing of the code. Permission was granted in June 2004, and the code is currently being distributed through Vanderbilt University. Not all of the technical details of the dosimetry methods have been shown here, as they have been previously documented. RESULTS: Agreement of doses between the MIRDOSE3.1 and OLINDA/EXM codes was good, within 1%-2% in most cases. CONCLUSION: The extensive testing of the OLINDA/EXM code, based on comparison with literature-established dose calculations and with the widely tested and accepted MIRDOSE3.1 code, should give users confidence in its output. The OLINDA/EXM code should be easy for MIRDOSE users to adopt and for new users to understand. It will be useful in standardizing and automating internal dose calculations, assessing doses in clinical trials with radiopharmaceuticals, making theoretic calculations for existing pharmaceuticals, teaching, and other purposes.


Assuntos
Medicina Nuclear/instrumentação , Planejamento da Radioterapia Assistida por Computador , Software , Doses de Radiação , Validação de Programas de Computador
11.
Cancer Biother Radiopharm ; 18(2): 165-78, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12804042

RESUMO

This was a 30-patient Phase II trial of reduced-dose (90)Y ibritumomab tiuxetan (Zevalin) RIT for patients with low-grade, follicular, or transformed B-cell NHL and mild thrombocytopenia. Patients were given an imaging dose of (111)In-labeled ibritumomab tiuxetan for dosimetry measurements. One week later, patients were administered a therapeutic dose of 0.3 mCi/kg (11 MBq/kg) (90)Y ibritumomab tiuxetan. Both (111)In- and (90)Y-labeled ibritumomab tiuxetan doses were preceded by an infusion of 250 mg/m(2) rituximab (Rituxan, MabThera) an unlabeled chimeric anti-CD20 antibody, to clear peripheral blood B cells and improve biodistribution of the radiolabeled antibody. For all 30 patients, normal organ and red marrow radiation absorbed doses were well below protocol-defined limits of 2000 cGy and 300 cGy, respectively. Median radiation absorbed doses were 48 cGy to red marrow (range: 6.5-95 cGy), 393 cGy to liver (range: 92-1581 cGy), 522 cGy to spleen (range: 165-1711 cGy), 162 cGy to lungs (41-295 cGy), and 14 cGy to kidneys (0.03-65 cGy). Though most correlative analyses were negative, certain analyses demonstrated a statistically significant correlation between the severity or duration of thrombocytopenia and pharmacokinetic or dosimetric parameters. These correlations were not consistent across the total patient population, and therefore, could not be exploited to predict hematologic toxicity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfoma não Hodgkin/radioterapia , Radioimunoterapia/métodos , Trombocitopenia/radioterapia , Radioisótopos de Ítrio/uso terapêutico , Adolescente , Antígenos CD20/imunologia , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos , Humanos , Linfoma não Hodgkin/diagnóstico por imagem , Radiometria , Terapia de Salvação , Trombocitopenia/diagnóstico por imagem , Distribuição Tecidual , Tomografia Computadorizada de Emissão , Resultado do Tratamento
12.
J Nucl Med ; 44(3): 465-74, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12621016

RESUMO

UNLABELLED: Ibritumomab tiuxetan is an anti-CD20 murine IgG1 kappa monoclonal antibody (ibritumomab) conjugated to the linker-chelator tiuxetan, which securely chelates (111)In for imaging or dosimetry and (90)Y for radioimmunotherapy (RIT). Dosimetry and pharmacokinetic data from 4 clinical trials of (90)Y-ibritumomab tiuxetan RIT for relapsed or refractory B-cell non-Hodgkin's lymphoma (NHL) were combined and assessed for correlations with toxicity data. METHODS: Data from 179 patients were available for analysis. Common eligibility criteria included <25% bone marrow involvement by NHL, no prior myeloablative therapy, and no prior RIT. The baseline platelet count was required to be > or = 100,000 cells/mm(3) for the reduced (90)Y-ibritumomab tiuxetan administered dose (7.4-11 MBq/kg [0.2-0.3 mCi/kg]) or > or = 150,000 cells/mm(3) for the standard (90)Y-ibritumomab tiuxetan administered dose (15 MBq/kg [0.4 mCi/kg]). Patients were given a tracer administered dose of 185 MBq (5 mCi) (111)In-ibritumomab tiuxetan on day 0, evaluated with dosimetry, and then a therapeutic administered dose of 7.4-15 MBq/kg (0.2-0.4 mCi/kg) (90)Y-ibritumomab tiuxetan on day 7. Both ibritumomab tiuxetan administered doses were preceded by an infusion of 250 mg/m(2) rituximab to clear peripheral B-cells and improve ibritumomab tiuxetan biodistribution. Residence times for (90)Y in blood and major organs were estimated from (111)In biodistribution, and the MIRDOSE3 computer software program was used, with modifications to account for patient-specific organ masses, to calculate radiation absorbed doses to organs and red marrow. RESULTS: Median radiation absorbed doses for (90)Y were 7.42 Gy to spleen, 4.50 Gy to liver, 2.11 Gy to lung, 0.23 Gy to kidney, 0.62 Gy (blood-derived method) and 0.97 Gy (sacral image-derived method) to red marrow, and 0.57 Gy to total body. The median effective blood half-life was 27 h, and the area under the curve (AUC) was 25 h. No patient failed to meet protocol-defined dosimetry safety criteria and all patients were eligible for treatment. Observed toxicity was primarily hematologic, transient, and reversible. Hematologic toxicity did not correlate with estimates of red marrow radiation absorbed dose, total-body radiation absorbed dose, blood effective half-life, or blood AUC. CONCLUSION: Relapsed or refractory NHL in patients with adequate bone marrow reserve and <25% bone marrow involvement by NHL can be treated safely with (90)Y-ibritumomab tiuxetan RIT on the basis of a fixed, weight-adjusted dosing schedule. Dosimetry and pharmacokinetic results do not correlate with toxicity.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfoma não Hodgkin/radioterapia , Radioimunoterapia , Radioisótopos de Ítrio/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Murinos , Antígenos CD20/imunologia , Medula Óssea/efeitos da radiação , Meia-Vida , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/uso terapêutico , Radioisótopos de Índio , Rim/efeitos da radiação , Pessoa de Meia-Idade , Doses de Radiação , Dosagem Radioterapêutica , Recidiva , Rituximab , Distribuição Tecidual , Radioisótopos de Ítrio/efeitos adversos , Radioisótopos de Ítrio/farmacocinética
14.
J Nucl Med ; 44(1): 67-76, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12515878

RESUMO

UNLABELLED: Calculated red marrow absorbed dose in patients receiving radioimmunotherapy (RAIT) has not been highly predictive of the dose-limiting hematologic toxicity observed in many patient populations studied. Because patients receiving the same red marrow dose often experience different grades of toxicity, other factors might help predict the different grades of toxicity observed. One such factor may be the plasma FLT3-L (FMS-related tyrosine kinase 3 ligand, hematopoiesis stimulatory cytokine) level, which has been shown to be a better indicator of recovery of progenitor cells and, thus, red marrow radiosensitivity (because during the recovery period the progenitor cells are hyperproliferative and potentially more radiosensitive) for patients treated with previous chemotherapy than peripheral blood counts. METHODS: Red marrow radiation doses were determined for 30 patients (20 male, 10 female; all without bone marrow or bone involvement; 19 had prior chemotherapy) after receiving (131)I RAIT (activity range, 2.1-8.9 GBq) for treatment of solid cancers known to produce carcinoembryonic antigen. Radiation dose estimates were calculated using 2 different methods of red marrow cumulated activity and red marrow-to-blood activity concentration ratio determinations for 2 dosimetric models: using both male and female and male-only masses and S values. Highest platelet toxicity grade at nadir (PTG), percentage platelet decrease (PPD) in counts, and platelet nadir (PN) counts were measured. FLT3-L levels (pg/mL) were determined by immunoassay before treatment; a normal FLT3-L level was assumed to be 80 pg/mL. The red marrow radiation doses (cGy) were adjusted for the patient's FLT3-L level when the patient's cytokine level exceeded the normal value. Marrow doses and FLT3-L-adjusted marrow doses were correlated with PTG, PPD, PN, and 1/PN. Administered activity, administered activity per unit body weight, and total body radiation dose were also correlated with these hematologic toxicity measures. RESULTS: All red marrow dose calculation schemes resulted in essentially the same correlations for a given hematologic toxicity measure. Poor correlations were observed between administered activity, administered activity per unit body weight, total body radiation dose, or red marrow radiation dose and PTG, PPD, PN, and 1/PN. All correlations improved greatly when the various predictors of toxicity were adjusted for the patient's FLT3-L level. The highest correlation observed was between red marrow dose or total body dose and 1/PN (r = 0.86). Using an unadjusted red marrow dose to predict toxicity >/= grade 3, there were 8 true-positive, but 13 false-positive cases with 9 true-negatives. However, using a FLT3-L-adjusted red marrow dose, there were 8 true-positives, but only 2 false-positives and 20 true-negatives. CONCLUSION: FLT3-L-adjusted red marrow radiation doses provide improved correlation with hematologic toxicity. Thus, elevated FLT3-L plasma levels before RAIT may indicate increased radiosensitivity of the bone marrow, and use of this measurement to adjust calculated red marrow or total body radiation doses may provide significantly better prediction of toxicity than radiation dose alone, leading to a patient-specific administered activity prescription that will deliver radiation doses to diseased tissues sufficient to produce an effective treatment outcome at acceptable toxicity levels.


Assuntos
Plaquetas/efeitos da radiação , Medula Óssea/efeitos da radiação , Neoplasias Ósseas/radioterapia , Proteínas de Membrana/sangue , Lesões por Radiação/prevenção & controle , Radioimunoterapia/efeitos adversos , Antígeno Carcinoembrionário , Neoplasias Colorretais/radioterapia , Relação Dose-Resposta à Radiação , Feminino , Humanos , Leucopenia/etiologia , Masculino , Lesões por Radiação/etiologia , Radioisótopos/administração & dosagem , Radioisótopos/efeitos adversos , Dosagem Radioterapêutica , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Estatística como Assunto , Neoplasias da Glândula Tireoide/radioterapia
15.
Cancer Biother Radiopharm ; 17(5): 535-43, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12470423

RESUMO

We have investigated several of the key model parameters and assumptions involved in the calculation of red marrow absorbed dose in order to better understand the sensitivity of the predicted results to changes in these model features and the subsequent effect on correlations of the red marrow absorbed dose values with observed hematologic toxicity. Red marrow dose calculations based on measured blood activity concentrations (to determine red marrow cumulated activity) and measured total body cumulated activity have a mass-independent and mass-dependent term. Adjustments for patient mass should be made in these calculations when patients' lean body masses are more than 10% different from that in the assumed standard models. The blood-based red marrow dose methodology has the potential to provide a reasonable estimate of red marrow dose as long as there is no specific uptake in red marrow or bone due to the presence of free radionuclide, disease, or retention of activity due to metabolism by the reticuloendothelial system. If these additional sources of red marrow dose are present, the blood-based methodology will significantly underestimate red marrow dose. For radiometals, such as in (90)Y-labeled antibodies, bone or red marrow uptake of free yttrium or catabolized (90)Y products may have a significant impact on the calculated dose, assuming fairly low amounts of free (90)Y or marrow activity uptake (5-10%), even in the absence of disease in red marrow and/or bone. This is also true for (131)I-labeled antibodies, although to a lesser extent due to typically reduced activity retention in the bone marrow in the absence of disease and lack of bone uptake of free radionuclide. Radiation dose calculations for the red marrow must be made as carefully as possible, taking into account all possible sources of radiation dose, and considering all sources of uncertainties, in order to give the best possible correlations of radiation dose with observed toxicity.


Assuntos
Medula Óssea/efeitos da radiação , Modelos Biológicos , Radioterapia/efeitos adversos , Doenças Ósseas/metabolismo , Osso e Ossos/metabolismo , Relação Dose-Resposta à Radiação , Humanos , Doses de Radiação , Radioisótopos/metabolismo
16.
J Nucl Med ; 43(9): 1241-4, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12215565

RESUMO

UNLABELLED: In calculations of absorbed doses from radioactive patients, the activity distribution in such patients is generally assumed to be an unattenuated point source and the dose to exposed individuals at a given distance is therefore calculated using the inverse square law. In many nuclear medicine patients, the activity distribution is widely dispersed and does not simulate a point source. In these cases, a line-source model is proposed to more accurately reflect this extended activity distribution. METHODS: Calculations of dose rate per unit activity were performed for a point source and for line sources of lengths of 20, 50, 70, 100, and 174 cm, and the ratios of line-source values to point-source values were calculated. In addition, radionuclide-independent conversion factors, to convert exposure rate constants to dose rates per unit activity, for these line-source lengths at various distances were determined. RESULTS: The calculated values, substantiated by published data, indicate that the inverse square law approximation is not valid for a line source until a certain distance is reached, dependent on the length of the line source. For the 20-, 50-, 70-, 100-, and 174-cm line sources, the dose rate values estimated by the inverse square law approximation are within approximately 10% of the values estimated using the line-source approach at distances of 20, 45, 60, 85, and 145 cm, respectively. At closer distances, use of the point-source model for a patient with an extended activity distribution will overestimate the radiation absorbed dose to exposed individuals, sometimes by a very significant amount. CONCLUSION: The line-source model is a more realistic and practical approach than the traditional point-source model for determining the dose to individuals exposed to radioactive patients with widespread activity distributions.


Assuntos
Doses de Radiação , Proteção Radiológica , Radioterapia , Exposição Ambiental , Humanos , Radiometria/métodos
17.
J Nucl Med ; 43(8): 1110-4, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163638

RESUMO

UNLABELLED: Given the relatively large tumor-absorbed doses reported for patients receiving radionuclide therapy, particularly radioimmunotherapy, and the relatively long pathlength of the nonpenetrating emissions of some radionuclides being used for these therapies, there exists the possibility of large absorbed doses to tissues adjacent to, surrounded by, or surrounding these tumors. Because tumors can occur adjacent to critical organs or tissues, such as arteries, nerves, pericardium, and the walls of the organs of the gastrointestinal tract, large absorbed doses to these normal tissues can lead to acute complications. METHODS: In this study, the Monte Carlo radiation transport code MCNP4b was used to simulate the deposition of energy from emissions of 2 radionuclides of interest, (131)I and (90)Y, to assess the possible magnitude of the absorbed doses in tissues adjacent to tumors. Mathematic models were constructed to simulate situations that might occur, such as tumor wrapped around a small cylinder (e.g., a nerve or artery), tumor against a tissue (e.g., the pericardium or wall of any gastrointestinal tract organ), and tumor surrounded by any soft tissue. Tumor masses of 10, 20, and 40 g were used in each model. Depth dose distributions were calculated using Monte Carlo simulations of the radiation transport in these geometric models. RESULTS: For tissues close to tumors containing (90)Y, the absorbed dose ranged from 24% of the absorbed dose in the tumor, for the case of tissues 1 mm from the tumor, to 103% of the absorbed dose in the tumor, for the case of small structures such as nerves or arteries surrounded by tumor. For tissues close to tumors containing (131)I, the absorbed dose ranged from 4% of the absorbed dose in the tumor, for the case of tissues 1 mm from the tumor, to 46% of the absorbed dose in the tumor, for the case of small structures such as nerves or arteries surrounded by tumor. CONCLUSION: This study showed that when absorbed doses to tumors are large, the absorbed dose to adjacent tissues can also be large, potentially causing unexpected toxicities.


Assuntos
Radioisótopos do Iodo/uso terapêutico , Neoplasias/radioterapia , Doses de Radiação , Dosagem Radioterapêutica , Radioisótopos de Ítrio/uso terapêutico , Simulação por Computador , Humanos , Método de Monte Carlo
18.
Cancer ; 94(4 Suppl): 1349-57, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11877765

RESUMO

BACKGROUND: Zevalin consists of a murine anti-CD20 monoclonal antibody (ibritumomab) conjugated to the linker-chelator tiuxetan, which securely chelates indium-111 ((111)In) for imaging and dosimetry and yttrium-90 ((90)Y) for radioimmunotherapy (RIT). Previous trials involving rituximab-naïve patients have demonstrated excellent targeting of Zevalin to CD20+ B-cell non-Hodgkin lymphoma with minimal uptake in normal organs. The purpose of this trial was to perform (111)In-Zevalin imaging in patients with rituximab-refractory tumors to determine normal organ dosimetry. METHODS: Twenty-seven patients were given an imaging dose of 5 mCi (185 MBq) (111)In-Zevalin on Day 0, evaluated with dosimetry, and then given a therapeutic dose of 0.4 mCi/kg (15 MBq/kg) (90)Y-Zevalin on Day 7. Both Zevalin doses were preceded by an infusion of 250 mg/m(2) rituximab to clear peripheral B cells and improve Zevalin biodistribution. Residence times for (90)Y in blood and major organs were estimated from (111)In biodistribution, and the MIRDOSE3 computer software program was used to calculate absorbed radiation doses to organs and red marrow. RESULTS: Median estimated absorbed radiation doses from (90)Y-Zevalin were 8.1 Gray (Gy) (range, 4.2-23.0 Gy) to the spleen, 5.1 Gy (range, 2.6-12.0 Gy) to the liver, 2.0 Gy (range, 1.4-5.3 Gy) to the lungs, 0.22 Gy (range, < 0.01-0.66 Gy) to the kidneys, and 0.74 Gy (range, 0.29-1.2 Gy) to the red marrow. These results are consistent with those from earlier Zevalin trials in rituximab-naïve patients. Hematologic toxicity was manageable and did not correlate with estimates of red marrow or total-body absorbed radiation dose. CONCLUSIONS: Zevalin treatment of rituximab-refractory follicular NHL patients at 0.4 mCi/kg resulted in acceptable estimates of absorbed radiation dose to organs, similar to those observed in other Zevalin-treated populations.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Linfoma não Hodgkin/tratamento farmacológico , Adulto , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Murinos , Antineoplásicos/efeitos adversos , Resistencia a Medicamentos Antineoplásicos , Humanos , Radioimunoterapia , Radiometria , Rituximab
19.
Health Phys ; 82(3): 367-72, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11845839

RESUMO

The U.S. NRC in 1997 removed its arbitrary 1.11 GBq (30 mCi) rule, which had been in existence for almost 50 y, and now many more patients receiving radionuclide therapy in nuclear medicine can be treated as outpatients. However, another problem has the potential to limit the short-lived reality of outpatient treatment unless nuclear medicine practitioners and the health physics community gets involved. Radioactive articles in the household trash of nuclear medicine patients are appearing at solid waste landfills that have installed radiation monitors to prevent the entry of any detectable radioactivity, and alarms are going off around the country. These monitors are set to alarm at extremely low activity levels. Some states may actually hold licensees responsible if a patient's radioactive household trash is discovered in a solid waste stream; this is another major reason [along with continued use of the 1.11 GBq (30 mCi) rule] why many licensees are still not releasing their radionuclide therapy patients. This is in spite of the fact that the radioactivity contained in released nuclear medicine therapy patients, let alone the much lower activity level contained in their potentially radioactive household wastes, poses a minimal hazard to the public health and safety or to the environment. Currently, there are no regulations governing the disposal of low-activity, rapidly-decaying radioactive materials found in the household trash of nuclear medicine patients, the performance of landfill radiation monitors, or the necessity of spectrometry equipment. Resources are, therefore, being unnecessarily expended by regulators and licensees in responding to radiation monitor alarms that are caused by these unregulated short-lived materials that may be mixed with municipal trash. Recommendations are presented that would have the effect of modifying the existing landfill regulations and practices so as to allow the immediate disposal of such wastes.


Assuntos
Resíduos Radioativos , Radioterapia/métodos , Gerenciamento de Resíduos , Humanos , Eliminação de Resíduos de Serviços de Saúde , Medicina Nuclear
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA