Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood Adv ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38838232

RESUMO

Peripheral T cell lymphomas (PTCL) have a poor prognosis with current treatments. High-dose chemotherapy followed by autologous hematopoietic cell transplant (AHCT) is used as a consolidation strategy after achieving clinical remission with first-line therapy, as well as in chemosensitive relapse if allogeneic transplant is not an option. CD25 is a targetable protein often highly expressed in PTCL. In this phase 1 clinical trial, we tested the addition of beta-emitting 90Y-labeled chimeric anti-CD25 basiliximab (aTac) to BEAM (carmustine, etoposide, cytarabine, melphalan) as conditioning for AHCT in patients with PTCL. Twenty-three AHCT-eligible patients were enrolled, and 20 received therapeutic 90Y-aTac-BEAM AHCT. Radiation doses of 0.4, 0.5 and 0.6 mCi/kg were tested. With no observed dose-limiting toxicities, 0.6 mCi/kg was deemed the recommended phase 2 dose. The most prevalent adverse effect, grade 2 mucositis, was experienced by 80% of patients. As of this report, 6 (30%) of the treated patients had died, 5 due to progressive disease and 1 due to multiple organ failure [median time of death 17 mo (range: 9-21 mo)] post-AHCT. Median follow-up was 24 mo (range: 9-26 mo) overall and 24 mo (range: 13-26 mo) for surviving patients. For patients who received therapeutic 90Y-aTac-BEAM AHCT, the 2-year progression-free and overall survival were 59% (95% CI: 34-77%) and 68% (95% CI: 42-84%), respectively. 90Y-aTac-BEAM appears to be safe as an AHCT conditioning regimen for PTCL, with no increased toxicity over the toxicities historically seen with BEAM alone in this patient population. This trial was registered at www.clinicaltrials.gov as # NCT02342782.

2.
Cancer Biother Radiopharm ; 21(2): 88-100, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16706629

RESUMO

PURPOSE/OBJECTIVE: Previous radioimmunotherapy (RIT) clinical trials at this institution with (90)Y-labeled cT84.66 anti-CEA (carcinoembryonic antigen) evaluated the antibody conjugated to diethylenetriaminepentaacetic acid (DTPA). The aim of this phase I therapy trial was to evaluate cT84.66 conjugated to the macrocyclic chelate (90)Y-DOTA and labeled with (90)Y in a comparable patient population. EXPERIMENTAL DESIGN: Patients with metastatic CEA-producing cancers were entered in this trial. If antibody targeting to tumor was observed after the administration of (111)In-DTPA cT84.66, the patient then received the therapy infusion of (90)Y-DOTA-cT84.66 1 week later. Serial nuclear scans, blood and urine collections, and computed tomography (CT) scans were performed to assess antibody biodistribution, pharmacokinetics, toxicities, and antitumor effects. RESULTS: Thirteen (13) patients were treated in this study. Dose-limiting hematologic toxicity was experienced at initial starting activity levels of 12 and 8 mCi/m(2). Subsequent patients received systemic Ca-DTPA at 125 mg/m(2) every 12 hours for 3 days post-therapy to allow for a dose escalation to 16 mCi/m(2), where hematologic toxicity was observed with an associated maximum tolerated dose (MTD) of 13.4 mCi/m(2). Tumor doses ranged from 4.4 to 569 cGy/mCi, which translated to 97-12,500 cGy after a single infusion of (90)Y-DOTA-cT84.66. Human anti-chimeric antibody (HACA) response developed in 8 of 13 patients and prevented additional therapy in 4 patients. CONCLUSIONS: This study demonstrates the feasibility of using (90)Y-DOTA-cT84.66 for antibody-guided radiation therapy. Immunogenicity of the DOTA-conjugated cT84.66 antibody was not appreciably greater than that observed with (90)Y-DTPA-cT84.66 in previous trials. Dose-limiting hematopoietic toxicity with (90)Y-DOTA-cT84.66 decreased with Ca-DTPA infusions post-therapy and appears to be comparable to previously published results for (90)Y-DTPA-cT84.66. The highest antibody uptake and tumor doses were to small nodal lesions, which supports the predictions from preclinical and clinical data that RIT may be best applied in the minimal tumor burden setting.


Assuntos
Antígeno Carcinoembrionário/imunologia , Imunotoxinas/uso terapêutico , Neoplasias/imunologia , Neoplasias/radioterapia , Radioimunoterapia/métodos , Radioisótopos de Ítrio/uso terapêutico , Adulto , Idoso , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Antígeno Carcinoembrionário/biossíntese , Feminino , Compostos Heterocíclicos com 1 Anel/efeitos adversos , Compostos Heterocíclicos com 1 Anel/uso terapêutico , Humanos , Imunoglobulina G/imunologia , Imunotoxinas/efeitos adversos , Imunotoxinas/farmacocinética , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias/patologia , Radioimunoterapia/efeitos adversos , Radioisótopos de Ítrio/efeitos adversos , Radioisótopos de Ítrio/farmacocinética
3.
Clin Cancer Res ; 10(15): 5014-21, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15297402

RESUMO

PURPOSE: The chimeric T84.66 (cT84.66) minibody is a novel engineered antibody construct (V(L)-linker-V(H)-C(H)3; 80 kDa) that demonstrates bivalent and high affinity (4 x 10(10) m(-1)) binding to carcinoembryonic antigen (CEA). The variable regions (V(L) and V(H)) assemble to form the antigen-combining sites, and the protein forms dimers through self-association of the C(H)3 domains. In animal models, the minibody demonstrated high tumor uptake, approaching that of some intact antibodies, substantially faster clearance than intact chimeric T84.66, and superior tumor-to-blood ratios compared with the cT84.66 F(ab')(2) fragment, making it attractive for further evaluation as an imaging and therapy agent. The purpose of this pilot clinical study was to determine whether (123)I-cT84.66 minibody demonstrated tumor targeting and was well tolerated as well as to begin to evaluate its biodistribution, pharmacokinetics, and immunogenicity in patients with colorectal cancer. EXPERIMENTAL DESIGN: Ten patients with biopsy-proven colorectal cancer (6 newly diagnosed, 1 pelvic recurrence, 3 limited metastatic disease) were entered on this study. Each received 5-10 mCi (1 mg) of (123)I-labeled minibody i.v. followed by serial nuclear scans and blood and urine sampling over the next 48-72 h. Surgery was performed immediately after the last nuclear scan. RESULTS: Tumor imaging was observed with (123)I-labeled minibody in seven of the eight patients who did not receive neoadjuvant therapy before surgery. Two patients received neoadjuvant radiation and chemotherapy, which significantly reduced tumor size before surgery and minibody infusion. At surgery, no tumor was detected in one patient and only a 2-mm focus was seen in the second patient. (123)I-labeled minibody tumor targeting was not seen in either of these pretreated patients. Mean serum residence time of the minibody was 29.8 h (range, 10.9-65.4 h). No drug-related adverse reactions were observed. All 10 patients were evaluated for immune responses to the minibody, with no significant responses observed. CONCLUSION: This pilot study represents one of the first clinical efforts to evaluate an engineered intermediate-molecular-mass radiolabeled antibody construct directed against CEA. cT84.66 minibody demonstrates tumor targeting to colorectal cancer and a faster clearance in comparison with intact antibodies, making it appropriate for further evaluation as an imaging and therapy agent. The mean residence time of the minibody in patients is longer than predicted from murine models. We therefore plan to further evaluate its biodistribution and pharmacokinetic properties with minibody labeled with a longer-lived radionuclide, such as (111)In.


Assuntos
Anticorpos/química , Anticorpos/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Fragmentos de Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Radioisótopos do Iodo/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Cromatografia Líquida de Alta Pressão , Dimerização , Feminino , Humanos , Cinética , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Estrutura Terciária de Proteína , Radiometria , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único
4.
Clin Cancer Res ; 9(16 Pt 1): 5842-52, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14676105

RESUMO

PURPOSE: Targeted systemic radiation therapy using radiolabeled antibodies results in tumor doses sufficient to produce significant objective responses in the radiosensitive hematological malignancies. Although comparable doses to tumor are achieved with radioimmunotherapy (RIT) in solid tumors, results have been modest primarily because of their relative lack of radiosensitivity. For solid tumors, as with external beam radiotherapy, RIT should have a more important clinical role if combined with other systemic, potentially radiation-enhancing chemotherapy agents and if used as consolidative therapy in the minimal tumor burden setting. The primary objective of this trial was to evaluate the feasibility and toxicities of systemic 90Y-chimeric T84.66 (cT84.66) anti-carcinoembryonic antigen RIT in combination with continuous infusion 5-fluorouracil (5-FU). EXPERIMENTAL DESIGN: Patients with chemotherapy-refractory metastatic colorectal cancer were entered. The study was designed for each patient to receive 90Y-cT84.66 anti-carcinoembryonic antigen at 16.6 mCi/m2 as an i.v. bolus infusion combined with 5-FU delivered as a 5-day continuous infusion initiated 4 h before antibody infusion. Cohorts of patients were entered at 5-FU dose levels of 700, 800, 900, and 1000 mg/m2/day. Upon reaching the highest planned dose level of 5-FU, a final cohort received 90Y-cT84.66 at 20.6 mCi/m2 and 5-FU at 1000 mg/m2/day. For all patients, Ca-diethylenetriaminepentaacetic acid at 125 mg/m2 every 12 h was administered for the first 72 h after 90Y-cT84.66. Patients were eligible to receive up to three cycles of 90Y-cT84.66/5-FU every 6 weeks. RESULTS: Twenty-one patients were treated on this study. All had been heavily pretreated with 19 having previously received 5-FU and 16 having failed two to four chemotherapy regimens. A maximum-tolerated dose of 16.6 mCi/m2 90Y-cT84.66 combined with 1000 mg/m2/day 5-FU was reached. These dose levels are comparable with maximum-tolerated dose levels of each agent alone. Thirteen patients received one cycle and 8 patients two cycles of therapy. Hematopoietic toxicity was dose-limiting and reversible. RIT did not appear to increase nonhematopoietic toxicities associated with 5-FU. Two of 19 patients assayed developed a human anti-chimeric antibody immune response after the first cycle of therapy, which is significantly less than that observed in a previous trial evaluating 90Y-cT84.66 alone. No objective responses were observed. However, 11 patients with progressive disease entering the study demonstrated radiological stable disease of 3-8 months duration and 1 patient demonstrated a mixed response. CONCLUSIONS: Results from this trial are encouraging and demonstrate the feasibility and possible advantages of combining continuous infusion 5-FU with 90Y-cT84.66 RIT. The addition of 5-FU does not appear to significantly enhance hematological toxicities of the radiolabeled antibody. In addition, 5-FU reduces the development of human anti-chimeric antibody response, permitting multicycle therapy in a larger number of patients. Future efforts should continue to focus on integrating radiation therapy delivered by radiolabeled antibodies into established 5-FU regimens.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Fluoruracila/uso terapêutico , Radioimunoterapia , Proteínas Recombinantes de Fusão/uso terapêutico , Radioisótopos de Ítrio/uso terapêutico , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/secundário , Terapia Combinada , Estudos de Viabilidade , Humanos , Dose Máxima Tolerável
5.
Health Phys ; 107(2 Suppl 2): S163-5, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24949923

RESUMO

Initial radiation exposure levels X (0) at 1 m from the navel of thyroid cancer patients were measured for 165 individuals at the time of ingestion. Some 61 patients had previously signed informed consent so only those patients could be assayed with regard to body parameters. While the activity was in the stomach, resultant X (0) values were seen to be linearly correlated with the total (131)I activity (A) given orally. Yet large differences in X (0) were seen; e.g., at A = 7.4 GBq, variations of a factor of four were found between the largest and smallest exposure rates. Correlation analyses were performed between normalized rate X (0)A-1 and several patient physical parameters. These included age, sex, height, weight, and BMI (body mass index). Only weight and BMI had significant linear correlation (p < 0.05) with normalized exposure rate. In the former case, the correlation coefficient ρ (weight) was -0.296 (p = 0.02). Using BMI as the independent variable, ρ (BMI) was -0.386 (p = 0.0021). With further analysis of the BMI variation, 95% confidence intervals could be determined at various BMI levels. For example, at 28 kg m(-2), the normalized rate varied between 0.039 and 0.0446 µGy h(-1) MBq(-1)-approximately a ±6.5% variation on the mean value of 0.0419 µGy h(-1) MBq(-1) at this BMI. Given such clinical information, differences in normalized exposure rate can be reduced to values on the order of ±10% or less for BMI values over the clinically relevant interval 20 to 40 kg m(-2).


Assuntos
Exposição Ambiental/análise , Iodeto de Sódio/uso terapêutico , Neoplasias da Glândula Tireoide/radioterapia , Índice de Massa Corporal , Ingestão de Alimentos , Família , Feminino , Humanos , Radioisótopos do Iodo/efeitos adversos , Masculino , Exposição Ocupacional/análise , Radiometria , Iodeto de Sódio/efeitos adversos , Neoplasias da Glândula Tireoide/fisiopatologia
6.
Health Phys ; 103(2 Suppl 2): S131-5, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22739966

RESUMO

Patients receiving ¹³¹I-based therapies are generally restricted in leaving the medical institution. In the U.S., the U.S. Nuclear Regulatory Commission (U.S. NRC) has developed the rule that a ≤ 7 mR h⁻¹ reading at 1 m from the patient (or 33 mCi) is sufficient to allow unrestricted release. Because of home situations and other constraints, it is preferable that a patient-specific release level be determined by the radiation safety staff. Locally, the City of Hope has instituted a general release criterion of ≤ 2 mR h⁻¹ at 1 m. While contributing to a reduction in public exposure, this as low as reasonably achievable (ALARA) approach is difficult to justify on a cost basis due to the expense of maintaining the radioactive individual in a hospital room. Instead, it was determined that a motel-type room already on the campus be modified to allow the patient to remain on-site until at or below a locally permitted release level. By adding lead to the bathroom area and sealing the tile surfaces, the room may be converted for less than $5,000. Daily cost for the patient is $65. In comparing the use of this facility for thyroid cancer patients from 2006 to 2010, it was found that the public exposure at 1 m was reduced by approximately 70% as compared to release at the 7 mR h level. In addition, controlling the release reduces the likelihood of a radiation incident in the public environment such as on public transportation or in a hotel.


Assuntos
Materiais de Construção , Exposição Ambiental/análise , Exposição Ambiental/prevenção & controle , Habitação , Radioisótopos do Iodo/análise , Radioisótopos do Iodo/uso terapêutico , Proteção Radiológica/instrumentação , Desenho de Equipamento , Pacientes , Proteção Radiológica/métodos , Estados Unidos
7.
Cancer ; 94(4 Suppl): 1231-4, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11877750

RESUMO

BACKGROUND: Two types of correction for absorbed dose (D) estimates are described for clinical applications of internal emitters. The first is appropriate for legal and scientific reasons involving phantom-based estimates; the second is patient-specific and primarily intended for radioimmunotherapy (RIT). METHODS: The Medical Internal Radiation Dose (MIRD) relationship (D) = S A is used, where S is a geometric matrix factor and A is the integral of source organ activities. Internal consistency of the data and the size of organ systems in the humanoid phantom must be assured in both types of estimation. RESULTS: The first dose estimate correction (I) is one whereby computations refer to one or another standard (e.g., MIRD-type) phantom. In this case the S value remains as given, but the measured patient A data must be standardized. The correction factor is the phantom's ratio of organ mass to whole-body mass divided by the same ratio for the volunteer or patient. The second dose estimate correction (II) is patient-specific. While the A value is unchanged for this application, a correction term is provided for the phantom-derived S matrix. The dominant (nonpenetrating radiation) component of this correction factor can be obtained via the ratio of the patient to phantom organ masses. In both corrections, we recommend that true organ sizes, necessary in each method of estimation, be determined in a separate sequence of anatomic images. CONCLUSIONS: In both dose estimation corrections, true sizes of the patient's or volunteer's internal organs must be obtained. Correction due to organ mass size can be severalfold and is probably the dominant uncertainty in the internal emitter absorbed dose calculation process.


Assuntos
Radioimunoterapia/métodos , Absorção , Humanos , Imagens de Fantasmas , Doses de Radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA