Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Clin Cancer Res ; 7(6): 1561-8, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11410491

RESUMEN

PURPOSE: Over 31,000 Americans die of androgen-independent metastatic prostate cancer each year. New strategies that do not involve hormonal manipulation but instead recognize the biochemical and molecular characteristics of prostate cancer are needed. Radioimmunotherapy (RIT) uses a tumor-specific monoclonal antibody to deliver systemic, targeted radiation to cancer. The objectives of this Phase I study of (111)In-2IT-BAD-m170 (for imaging) and (90)Y-2IT-BAD-m170 (for therapy) were to determine the toxicity and maximum tolerated dose (MTD), the specificity for targeting metastatic prostate cancer, and the efficacy for palliation of pain. EXPERIMENTAL DESIGN: M170 is a mouse monoclonal antibody that targets adenocarcinomas. Patients with adequate renal and liver function, rising prostate-specific antigen, and androgen-independent metastatic prostate cancer were eligible. After estimation of dosimetry and pharmacokinetics with (111)In-2IT-BAD-m170, a single dose of (90)Y-2IT-BAD-m170 (0.185, 0.370, 0.555, or 0.740 GBq/m(2)) was administered to cohorts of three patients. Pain was assessed objectively by questionnaires before and for 8 weeks after RIT; weekly prostate-specific antigen levels were obtained for 2 months after RIT. RESULTS: The MTD of (90)Y-2IT-BAD-m170 was 0.740 GBq/m(2) for patients that had up to 10% of the axial skeleton involved with prostate cancer. Toxicity was almost exclusively confined to reversible myelosuppression. Metastatic prostate cancer was targeted by (111)In-2IT-BAD-m170 in all 17 patients. The mean radiation dose delivered to 39 bone and 18 nodal metastases by (90)Y-2IT-BAD-m170 was 10.5 Gy/GBq (range 2.8-25.1). Thirteen of 17 patients reported pain before (90)Y-2IT-BAD-m170; 7 of these 13 had a partial or complete resolution of pain that lasted an average of 4.3 weeks. CONCLUSIONS: This study determined the MTD of (111)In/(90)Y-2IT-BAD-m170 in patients with metastatic prostate cancer. The drugs were well tolerated, targeted metastases, and temporarily palliated pain.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Terapia Combinada , Radioisótopos de Indio , Neoplasias de la Próstata/terapia , Radioinmunoterapia , Radioisótopos de Itrio/uso terapéutico , Adenocarcinoma/terapia , Anciano , Animales , Anticuerpos Monoclonales/farmacocinética , Estudios de Cohortes , Humanos , Radioisótopos de Indio/farmacocinética , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Dolor/tratamiento farmacológico , Antígeno Prostático Específico/biosíntesis , Radiometría , Factores de Tiempo , Resultado del Tratamiento , Radioisótopos de Itrio/farmacocinética
2.
Crit Rev Oncol Hematol ; 38(1): 25-35, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11255079

RESUMEN

Radioimmunotherapy (RIT) is a promising approach for treating metastatic breast cancer. Initial clinical trials using 131I radioimmunoconjugates, and more recent studies employing 90Y, have demonstrated objective, although transient, antitumor effects in heavily pretreated patients with minimal toxicity. Antibodies targeting unique epitopes of epithelial glycoprotein mucin (MUC-1) on breast cancer cell surfaces that have been studied in patients include BrE-3 (murine and humanized) and m170 (murine). Both antibodies react with at least 90% of breast cancers. In these and other RIT trials, myelosuppression has been the dose-limiting toxicity. However, this toxicity has been successfully circumvented with the use of autologous peripheral blood stem cell transplantation, and recent clinical trials have escalated 90Y doses up to 50 mCi/m(2). The therapeutic index indicates that using these agents with stem cell support should deliver 9000 to 18000 rads to metastatic tumors. Development of improved chelates that are readily metabolized in the liver may reduce doses to this organ, projected to be next in line as dose-limiting. Combination therapy will be required to produce durable benefits in metastatic breast cancer. Low dose taxanes are synergistic with RIT in preclinical studies and when administered in the optimal sequence could sensitize tumor cells to the continuous low dose radiation delivered by RIT, without increasing toxicity. The addition of systemically administered tumor targeting radiation therapy using RIT as part of combined modality therapy may enhance the rate of complete response and, in patients with minimal metastatic disease, could lead to curative therapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Metástasis de la Neoplasia/radioterapia , Radioinmunoterapia/métodos , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Femenino , Humanos , Radioisótopos de Itrio/uso terapéutico
3.
Clin Cancer Res ; 5(10 Suppl): 3219s-3223s, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10541367

RESUMEN

Single-agent radioimmunotherapy (RIT) has proven efficacy as a treatment for hematological malignancies, particularly non-Hodgkin's lymphoma. Although promising, RIT has been less effective for solid tumors, in part because they are less radiosensitive. Bone marrow transplantation permits the administration of larger radiopharmaceutical doses, but the results of bone marrow transplantation-supported RIT for solid tumors have been marginal. The purpose of this publication is to provide an overview of promising RIT strategies for solid tumors. It is apparent that combination therapy is required, but optimization of the radiopharmaceutical should be the first step. Metallic radionuclides provide higher tumor radiation doses but not necessarily an improved therapeutic index, that is, the ratio of tumor:normal tissue radiation doses. Biodegradable peptide linkers between the chelated metal and the antibody improve the therapeutic index. Further improvements depend on identification of synergistic therapies which recognize that: (a) continuous, low-dose radionuclide therapy acts through apoptosis; and (b) apoptosis is often blocked because most tumors have ineffective p53 and increased Bcl-2. Taxanes are particularly attractive as synergistic agents for RIT because they induce cell cycle arrest in the radiosensitive G2-M phase and p53-independent apoptosis. Optimal sequence and timing for combined modality RIT are critical to achieve synergy. Data from preclinical and clinical studies will be reviewed to illustrate the potential of these strategies.


Asunto(s)
Neoplasias/radioterapia , Radioinmunoterapia , Animales , Trasplante de Médula Ósea , Quelantes , Humanos , Ratones
4.
Clin Cancer Res ; 5(10 Suppl): 3243s-3248s, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10541370

RESUMEN

Radioimmunoconjugates of 170H.82 (m170), a panadenocarcinoma monoclonal antibody, are effective for imaging primary and metastatic breast cancer. To evaluate m170 as a targeting agent for therapy, we developed (111)In- and 90Y-2-iminothiolane-2-[p-(bromoacetamido)benzyl]-1,4,7,10 tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid-m170 immunoconjugates with 99% purity by molecular sieving and immunoreactivity comparable to unmodified antibody. (111)In-m170 pharmacokinetic studies were performed prior to each therapy to determine the maximum dose of 90Y-m170 that could be administered without exceeding a limit of 800 rad to the liver, lungs, or kidneys or 250 rad to the whole body or bone marrow for each of three cycles of treatment. Peripheral blood stem cells (PBSCs) were harvested and cyclosporin A (5 mg/kg twice daily) was administered as strategies to ameliorate myelosuppression and prevent the development of HAMA, respectively. An (111)In imaging/pharmacokinetic study was performed, and the 90Y dose was calculated and administered. The liver was the 90Y dose-limiting organ. The mean and range of calculated doses (in rad/mCi) for the five patients evaluated were as follows: whole body, 2.3 (2.1-2.4); liver, 17.8 (12.7-22.2); lung, 6.4 (4.8-7.2); kidney, 6.9 (6.3-11.5); marrow, 3.6 (1.9-4.4); and tumors (n = 25), 71.5 (14.1-141.5). Of the three patients treated, with doses of 37, 54, and 57 mCi of 90Y, one had a partial response, one had measurable tumor reduction but less than a partial response, and one had stable disease for more than 1 month. PBSCs prevented prolonged myelosuppression. The therapeutic responses, coupled with an absence, thus far, of significant adverse sequelae, suggest that this dosimetry-based approach combined with PBSCs may lead to effective therapy when higher 90Y doses are reached.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/terapia , Ciclosporina/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Inmunosupresores/uso terapéutico , Radioinmunoterapia , Dosificación Radioterapéutica , Radioisótopos de Itrio/uso terapéutico , Adulto , Animales , Femenino , Humanos , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Trasplante Autólogo
5.
Clin Cancer Res ; 4(10): 2483-90, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9796981

RESUMEN

Radioimmunotherapy using 131I-ChL6 antibody has shown promise in patients with breast cancer. To enhance this potential, a novel ChL6 immunoconjugate that is catabolizable and tightly binds 90Y and (111)In was developed. The immunoconjugate, 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA)-peptide-ChL6, consists of the macrocyclic chelator DOTA linked to ChL6 by a peptide that is preferentially catabolized in the liver. The pharmacokinetic and dosimetric properties of the radioimmunoconjugates (RICs) (111)In- and 90Y-DOTA-peptide-ChL6 and (111)In- and 90Y-2-iminothiolane (2-IT)-2-[p-(bromoacetamido)benzyl]-DOTA-ChL6 were compared in athymic mice bearing HBT3477 human breast cancer xenografts. Each of the RICs was stable in vivo and concentrated well in the xenografts. Liver concentration, cumulative radioactivity (activity over time), and radiation dose of the DOTA-peptide-ChL6 RICs were one-third to one-half of those of the corresponding 2-IT-2-[p(bromoacetamido)benzyl]-DOTA-ChL6 RICs. Indium-111 RICs were imperfect tracers for corresponding 90Y RICs, although their pharmacokinetics and radiation dosimetries were similar. The results of this study were consistent with previously published in vitro data, which indicated that the peptide linker of DOTA-peptide-ChL6 was catabolized by cathepsin B. The cumulative activities and radiation doses to the liver of DOTA-peptide-ChL6 RICs were one-half of those of corresponding RICs with the 2-IT linker. Preliminary data from pilot studies in patients with breast cancer are in accord with these observations. These novel DOTA-peptide RICs seem to have excellent clinical potential for radioimmunotherapy associated with marrow transplantation, for which liver radiation is likely to be dose limiting for 90Y.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Quelantes/farmacocinética , Inmunoconjugados/farmacocinética , Neoplasias Mamarias Experimentales/radioterapia , Radioinmunoterapia , Animales , Femenino , Humanos , Radioisótopos de Indio/uso terapéutico , Ratones , Trasplante de Neoplasias , Dosis de Radiación , Trasplante Heterólogo , Radioisótopos de Itrio/uso terapéutico
6.
Anticancer Res ; 18(6A): 4011-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9891439

RESUMEN

BACKGROUND: Paclitaxel (Taxol) has demonstrated synergistic enhancement of radioimmunotherapy (RIT) of breast cancer with Y-90 labeled antibody ChL6, in the xenografted mouse model. To determine the optimal sequence and timing of RIT and Taxol for a prospective clinical trial, efficacy and dosimetry in mice, and dosimetry in patients receiving RIT alone, were examined. MATERIALS AND METHODS: Mice bearing human breast cancer xenografts (HBT 3477) received i.v. Y-90-DOTA-peptide-ChL6 (260 microCi), and i.p. Taxol (300 or 600 micrograms) 72, 48, or 24 hours prior to RIT, or 6, 24, 48, or 72 hours after RIT. RESULTS: Taxol after RIT resulted in cure, CR, or PR of all mice (70/70 tumors) and demonstrated greater therapeutic enhancement (p = 0.001) than Taxol before RIT. Mice receiving 600 micrograms Taxol 48 hours after RIT achieved 88% cure (7/8 tumors). In mice, 57% and 42% of the radiation dose to tumor and marrow, respectively, was delivered from 48-336 hours after RIT; in patients receiving 90Y-DOTA-peptide-ChL6, the corresponding values were 56% and 22%. CONCLUSIONS: Taxol given approximately 48 hours after RIT provides coincident peak deposition of Taxol and Y-90 in tumor, and no Taxol in the marrow during the major radiation dose to marrow, resulting in therapeutic enhancement without observable additive toxicity. A clinical trial of low dose Taxol given after RIT to patients with metastatic breast cancer is planned.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Compuestos Heterocíclicos/uso terapéutico , Oligopéptidos/uso terapéutico , Paclitaxel/uso terapéutico , Radioinmunoterapia , Itrio/uso terapéutico , Animales , Terapia Combinada , Femenino , Compuestos Heterocíclicos/farmacocinética , Humanos , Ratones , Ratones Desnudos , Oligopéptidos/farmacocinética , Factores de Tiempo , Trasplante Heterólogo , Células Tumorales Cultivadas , Itrio/farmacocinética
7.
Cancer ; 80(12 Suppl): 2583-90, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9406713

RESUMEN

BACKGROUND: The beneficial effects of radioimmunotherapy (RIT) may result from activation of molecular pathways that lead to programmed cell death (apoptosis). The influences of sequence and timing of 90Y-DOTA-peptide-ChL6 antibody (90Y-ChL6) and anti-epidermal growth factor receptor antibody (ch225) or paclitaxel (Taxol; Bristol-Myers Squibb, Princeton, NJ) on efficacy and toxicity were examined. METHODS: Groups of human breast carcinoma (HBT 3477) tumored mice received paclitaxel (300 or 600 microg) or ch225 (70, 150, or 350 microg) at various intervals before or after 90Y-ChL6. Mortality, hematologic toxicity, weight loss, and therapeutic efficacy were evaluated. RESULTS: Mice receiving paclitaxel within 24 hours of 90Y-ChL6 had a 100% response rate; 48% were cured when paclitaxel was given 6 or 24 hours after 90Y-ChL6. When 150 microg ch225 was given 24 hours before 90Y-ChL6, the response and cure rates surpassed those of 90Y-ChL6 alone. Timing of administration was critical, with mortality rates as high as 80% in some groups receiving 350 microg ch225 and 90Y-ChL6. CONCLUSIONS: In this aggressive human breast carcinoma model, combined 90Y-ChL6 and paclitaxel had a high therapeutic index with many cures. Sequence of administration was critical in order for ch225 or paclitaxel, when combined with 90Y-ChL6, to enhance the response rate.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Mamarias Experimentales/terapia , Paclitaxel/uso terapéutico , Radioinmunoterapia , Radioisótopos de Itrio/uso terapéutico , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Terapia Combinada , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Pérdida de Peso
8.
Cancer ; 80(12 Suppl): 2728-32, 1997 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9406731

RESUMEN

BACKGROUND: In clinical trials of radiolabeled monoclonal antibodies targeting metastatic breast carcinoma, myelosuppression has been the initial dose-limiting toxicity. We previously have shown that mobilized peripheral blood stem cell transfusions ameliorate this toxicity of radioimmunotherapy (RIT). Because of the difficulty we experienced harvesting adequate numbers of precursor cells from some of these heavily pretreated patients, we have compared the mobilization results and clinical histories of the metastatic breast carcinoma patients referred for RIT with those of metastatic breast carcinoma patients referred for high dose chemotherapy (HDCT) with transplantation. METHODS: Mobilization of stem cells into the peripheral blood was accomplished using granulocyte-colony stimulating factor with or without chemotherapy. Granulocyte macrophage colony-forming assays (CFU-GM) and flow cytometric analysis for CD34 positive cells were used to evaluate the number of hematopoietic precursors mobilized and collected with each apheresis procedure. Clinical characteristics, including prior chemotherapy and external beam radiotherapy, tumor bulk, and performance status, were determined by chart review. RESULTS: Significantly fewer hematopoietic precursors (both CD34 positive cells and CFU-GM) were harvested per procedure from the six RIT patients compared with a group of six patients with metastatic breast carcinoma who had stem cells harvested prior to HDCT (P = 0.002). There was no significant difference between the groups with regard to age or prior radiotherapy. All the RIT patients had received more chemotherapy, had a less favorable performance status (1 vs. 0), and had measurable tumor, whereas all the HDCT patients had minimal residual disease. CONCLUSIONS: Patients enrolled in RIT studies had lower stem cell yields than metastatic breast carcinoma patients scheduled to receive HDCT with stem cell transplantation. Poor mobilization is unlikely to be due to the mobilizing regimen alone and may be related to the intensity of prior therapy and/or tumor bulk. Mobilizing adequate stem cells for multiple treatment cycles from patients on Phase I/II RIT trials may require new, more effective mobilizing regimens, but referral of patients earlier in their disease course, prior to chronic bone marrow damage and disease progression, is recommended strongly.


Asunto(s)
Neoplasias de la Mama/radioterapia , Movilización de Célula Madre Hematopoyética , Radioinmunoterapia , Adulto , Antineoplásicos/uso terapéutico , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Persona de Mediana Edad
9.
J Nucl Med ; 38(8): 1180-5, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9255145

RESUMEN

UNLABELLED: BrE-3 is a murine IgG1 monoclonal antibody that binds to 97% of human ductal breast cancer specimens. A previous study documented the ability of 111In-labeled 1,4-methyl-benzyl isothiocyanate diethylenetriamine pentaacetic acid (111In-MX-DTPA) BrE-3 to specifically target breast cancer tissue in patients, and the dosimetry derived from the pharmacokinetics suggested that a useful therapeutic index could be obtained with 90Y-MX-DTPA BrE-3. A Phase I maximum tolerated dose study was, therefore, initiated. METHODS: Six patients received 111In/90Y-MX-DTPA BrE-3, three of them receiving 6.25 and the other three receiving 9.25 mCi/m2 of 90Y. Pharmacokinetics, dosimetry, human anti-mouse antibody (HAMA), toxicity and clinical responses were evaluated. RESULTS: Three of six patients demonstrated a minor and transient, but objective tumor response, and none of the patients had significant toxicity. Tumor dosimetry ranged from 39 to 167 rad/mCi of 90Y (442-1887 rad/ dose). HAMA response occurred in five of six patients. CONCLUSION: Minimal toxicity, dosimetric calculations and clinical assessment indicate that a useful therapeutic index can be achieved with this therapy. Indium-111/yttrium-90-MX-DTPA BrE-3 can be safely administered to patients with metastatic breast cancer, and therapy doses yielded pharmacokinetics similar to those of tracer doses. Clinical responses, albeit transient, were achieved with single-dose therapy. Rapid onset of the HAMA response will hinder multicycle therapy, unless it is prevented with immunosuppressive drugs or the use of a "humanized" antibody. Further studies are needed to determine the optimal use of BrE-3 for radioimmunotherapy.


Asunto(s)
Neoplasias de la Mama/radioterapia , Radioisótopos de Indio/uso terapéutico , Radioinmunoterapia , Radioisótopos de Itrio/uso terapéutico , Quelantes/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , Ácido Pentético/análogos & derivados , Ácido Pentético/uso terapéutico , Dosificación Radioterapéutica
10.
Anticancer Res ; 17(3B): 1745-51, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9179228

RESUMEN

BACKGROUND: The biologically active, antiadenocarcinoma monoclonal antibody chimeric L6 (ChL6) was labeled with 131I and administered in cycles to patients with metastatic breast cancer who had failed standard therapy. The therapeutic potential, tumor targeting and maximum tolerated dose of 131I-ChL6 were studied. METHODS: Ten patients with L6 reactive breast cancer received an imaging dose of 131I-ChL6 followed 24 hours later by a therapy dose of 131I-ChL6 (20-70 mCi/m2). Patients received up to 4 monthly cycles unless they had significant myelosuppression, progression of disease, or a high human anti-mouse antibody titer. In vivo activation of effector cell function, complement levels and cytokine release were studied. RESULTS: All 10 patients had detectable cancer on the imaging study. In 7 patients with superficial cancer, the radiation dose was 120 to 3700 cGy/cycle; 5-30 times higher than the whole body dose. Therapy resulted in minimal acute or subacute toxicity. Dose limiting toxicities were neutropenia and thrombocytopenia. Six of 10 patients had clinically measurable tumor responses; 5 had responses that lasted more than one month (1.5-5 months). The MTD for 131I-ChL6, given in at least two monthly doses, was 60 mCi/m2. CONCLUSION: Objective clinical responses were seen in five of 10 patients treated with 131I-ChL6. The tumor response in heavily pretreated, advanced breast cancer may be related to the combined effects of targeted radiation and the biological activity of L6/ChL6.


Asunto(s)
Adenocarcinoma/radioterapia , Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/radioterapia , Radioisótopos de Yodo/uso terapéutico , Radioinmunoterapia , Adulto , Anciano , Animales , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunoglobulina G , Radioisótopos de Yodo/efectos adversos , Radioisótopos de Yodo/farmacocinética , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Proteínas Recombinantes de Fusión , Distribución Tisular
11.
Anticancer Res ; 17(3B): 1735-44, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9179227

RESUMEN

BACKGROUND: Radioimmunotherapy (RIT) using 131I-Chimeric L6 (ChL6) antibody has shown therapeutic promise for patients with breast cancer. To enhance this potential, a novel immunoconjugate was developed that targets adenocarcinomas like breast cancer and tightly binds yttrium-90 (90Y) for therapy and indium-111 (111In) for imaging. The radioimmunoconjugate consists of a macrocyclic chelator (DOTA) linked to ChL6 by a catabolizable peptide. 90Y-DOTA-peptide-ChL6 was designed to minimize the radiation dose to critical normal tissues, thereby improving the therapeutic index. MATERIALS AND METHODS: Three patients with incurable metastatic breast cancer received 90Y/111In-DOTA-peptide-ChL6 for 5 pharmacokinetics/dosimetry studies and one of these patients also received 2 therapy doses. Quantitative imaging of 111In and in vitro assay of 90Y and 111In in blood urine and bone marrow samples were obtained. RESULTS: 90Y/111In-DOTA-peptide-ChL6 was prepared at high purity and was stable in vivo. Assays of bone marrow revealed no evidence for escape of 90Y or 111In from the chelate. 111In imaging of tumors was excellent, providing a therapeutic index for tumor to marrow radiation as high as 229 to 1. 90Y and 111In provided comparable pharmacokinetics, as did tracer and therapeutic doses of radioimmunoconjugates. One patients that received 2 therapeutic doses of 90Y-DOTA-peptide-ChL6 showed regression of tumors and tumor markers. Toxicities were relatively minor and no anti-globulin response developed despite 5 immunoconjugate infusions. CONCLUSIONS: This first study in patients of radioimmunoconjugates with a catabolizable linker between the metal chelator and the antibody confirmed that these novel 90Y/111In-DOTA-peptide-ChL6 radioimmunoconjugates have significant potential. Tracer doses of 111In-DOTA-peptide-ChL6 for imaging predicted the behavior of therapeutic doses of 90Y-DOTA-peptide-ChL6. The latter radioimmunoconjugate induced regression of tumors and tumor markers without significant toxicity. When compared to earlier 131I-ChL6 dosimetry, 90Y-DOTA-peptide-ChL6 provided a therapeutic index several times better.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/radioterapia , Radioisótopos de Indio/uso terapéutico , Radioinmunodetección , Radioinmunoterapia , Radioisótopos de Itrio/uso terapéutico , Anticuerpos Monoclonales/farmacocinética , Biomarcadores de Tumor/análisis , Médula Ósea/diagnóstico por imagen , Neoplasias de la Mama/patología , Femenino , Humanos , Radioisótopos de Indio/farmacocinética , Tasa de Depuración Metabólica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Radioinmunodetección/efectos adversos , Radioinmunoterapia/efectos adversos , Proteínas Recombinantes de Fusión , Distribución Tisular , Tomografía Computarizada de Emisión , Tomografía Computarizada por Rayos X , Radioisótopos de Itrio/farmacocinética
12.
Cancer Res ; 55(23 Suppl): 5916s-5920s, 1995 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7493370

RESUMEN

Radioimmunotherapy (RAIT) using a humanized murine monoclonal antibody, chimeric L6 (ChL6), has produced objective tumor reduction in 50% of chemotherapy-refractory patients with metastatic breast cancer in our prior studies. Because myelosuppression limited dose escalation, we evaluated the ability of granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood progenitor cell (PBPC) transfusions to ameliorate this problem. 131I-labeled ChL6 was given at a starting dose of 150 mCi/m2 (2,5 times the maximum tolerated dose without PBPCs) for a planned three treatments. When blood radioactivity declined to less than 1 microCi/ml after treatment, PBPCs were transfused, and G-CSF was administered. Patient 1 had minimal myelosuppression, received two cycles of therapy, and then developed human antimonoclonal antibody (HAMA). Patient 2 had prolonged thrombocytopenia that resolved after additional PBPC transfusion. Progressive disease as well as HAMA prevented further treatment. Patient 3 received all three cycles of 150 mCi/m2 at 8-week intervals. Thrombocytopenia (< 25,000/microliter) occurred but was transient (0-7 days). Because HAMA developed in all prior patients who received G-CSF with ChL6 RAIT, including patients 1 and 2, who received PBPC, patient 3 was given cyclosporin for 14 days. She did not develop HAMA or significant toxicity following 3 cycles of RAIT. Cumulative radiation doses to her lungs and tumor were estimated at 3,100 and 11,200 cGy, respectively. For 9 months, she had a reduction in bone pain, a decline in serum tumor markers, and decreased tumor uptake of F-18-deoxyglucose on a positron emission scan. Her performance status improved, and she had no pulmonary toxicity. We conclude that: (a) PBPC transfusion can modify the myelotoxicity of RAIT and can permit repetitive dosing; (b) cyclosporin is a promising means to abrogate HAMA; and (c) fractionation of intensive-dose RAIT may increase the antitumor effect and reduce normal organ toxicity.


Asunto(s)
Neoplasias de la Mama/terapia , Trasplante de Células Madre Hematopoyéticas , Radioisótopos de Yodo/uso terapéutico , Radioinmunoterapia , Anticuerpos Monoclonales/uso terapéutico , Terapia Combinada , Femenino , Humanos , Dosificación Radioterapéutica
13.
J Clin Oncol ; 12(2): 342-6, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8113841

RESUMEN

PURPOSE: We investigated the role of high-dose chemotherapy and autologous bone marrow transplantation (ABMT) as the initial systemic treatment in patients with hormone-unresponsive metastatic breast cancer. We studied a regimen involving a split-course schedule using sequential administration of two pairs of alkylating agents separated by 5 days of rest. The rest period was intended to provide time for recovery from the treatment-immediate adverse effects, thereby allowing further dose escalation. PATIENTS AND METHODS: The treatment consisted of thiotepa 225 to 300 mg/m2/d (days - 11 to -9), cisplatin 50 to 100 mg/m2/d (days - 11 and -3), and cyclophosphamide 60 mg/kg/d (days - 3 and -2). Dose escalation was performed in the initial 15 patients before reaching dose-limiting toxicities. When feasible, responding patients received posttransplant irradiation to sites of residual or prior bulky disease. Patients with bone marrow or CNS involvement, prior pelvic irradiation, or age greater than 55 years were excluded. RESULTS: Thirty-nine patients with measurable or assessable tumor were enrolled: 23 with visceral metastases, 11 with only soft tissue disease, and five with skeletal involvement. Twenty-five patients had received no chemotherapy for metastatic disease before transplantation. The dose-limiting toxicities of this therapy were renal and gastrointestinal. Six patients died from complications: four of a fungal infection and two of hemorrhage. A complete response was achieved in 14 patients (36%), three of whom are free of disease at 79+, 55+, and 40+ months after transplantation. Ten of 25 patients not treated with standard-dose chemotherapy for metastatic disease achieved a complete response (40%). The three patients in continuous remission were in the untreated relapse group. CONCLUSION: This single high-dose treatment achieved a relatively high complete response rate in patients with metastatic breast cancer and may have cured some of them. On the other hand, the split-course dose schedule as tested here did not permit significant dose-intensification.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Trasplante de Médula Ósea , Neoplasias de la Mama/terapia , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cisplatino/administración & dosificación , Terapia Combinada , Ciclofosfamida/administración & dosificación , Esquema de Medicación , Femenino , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Análisis de Supervivencia , Resultado del Tratamiento
14.
Adv Exp Med Biol ; 353: 203-11, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7527179

RESUMEN

131I chimeric L6 (ChL6) monoclonal antibody (MoAb) therapy has been performed in 12 patients with advanced, metastatic breast cancer. The protocol was designed to determine the maximum tolerated dose (MTD) of radioimmunotherapy that could be administered at 4 intervals. Ten patients received 20-70 mCi/m2 of 131I ChL6. Two of the patients received granulocyte colony stimulating factor (GCSF) on days 10-20 post therapy. The MTD for two doses was 60 mCi/m2 and thrombocytopenia was the dose limiting toxicity in the absence of marrow reconstitution with stem cells. Two patients received 150 mCi/m2 with autologous peripheral blood stem cell support 7 and 9 days post treatment. The MTD has not been reached for 131I-ChL6 with autologous stem cell support. In the 12 patients treated with 131I ChL6, six patients (50%) had measurable tumor regressions greater than 30% of the sum of the largest two dimensional products for measurable tumors. Four of these 6 patients had a partial response (PR), i.e., > or = 50% reduction in tumor size. These therapeutic responses associated with modest clinical toxicity in heavily pretreated patients suggest that clinically relevant radioimmunotherapeutic approaches can be devised for metastatic breast cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/radioterapia , Radioisótopos de Yodo/uso terapéutico , Radioinmunoterapia , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Anticuerpos Antiidiotipos/biosíntesis , Enfermedades de la Médula Ósea/etiología , Enfermedades de la Médula Ósea/terapia , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoglobulina G/uso terapéutico , Radioisótopos de Yodo/administración & dosificación , Ratones , Metástasis de la Neoplasia , Inducción de Remisión , Terapia Recuperativa , Distribución Tisular , Resultado del Tratamiento
15.
J Clin Apher ; 9(3): 176-82, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7535760

RESUMEN

Modest success has been achieved with the use of high-dose cytotoxic therapy and bone marrow transplantation in solid tumors. Patient outcome can potentially be improved with further intensification of the therapy. The rapid hematologic recovery achieved with mobilized peripheral blood progenitor cells (PBPC) may reduce the toxicity of transplantation enabling the use of sequential courses of myeloablative therapy. We report on 42 patients with solid tumors enrolled in a tandem transplant protocol involving the use of PBPC mobilized with cyclophosphamide (4 g/m2), etoposide (1 g/m2), and granulocyte-colony-stimulating factor (G-CSF: 10 micrograms/kg/day). This regimen significantly increased the number of circulating progenitor cells; only 1-2 aphereses were sufficient to collect 2.5 x 10(8)/kg mononuclear cells, our goal for each transplant course. The median number of circulating colony-forming units (CFU) and CD34+ cells obtained for each transplant course were 70.3 x 10(4)/kg, and 11.7 x 10(6)/kg, respectively. There was a significant correlation between the numbers of CD34+ cells and CFU measured in the apheresis product (r = 0.49, P = .003). The first transplant regimen given to 38 patients consisted of thiotepa, carboplatin, and cyclophosphamide. The second transplant regimen given to 29 patients consisted of busulfan and etoposide. Hematologic recovery was comparable after each of the two transplant courses. The median time to neutrophil recovery over 0.5 x 10(9)/L and to platelet transfusion independence was 9 and 8 days, respectively. There was no difference in engraftment rates after transplant with PBPC only (n = 28 courses) compared to transplant with PBPC plus bone marrow (n = 39 courses).(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasias/terapia , Antígenos CD/análisis , Antígenos CD34 , Separación Celular/métodos , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos
17.
Bone Marrow Transplant ; 8(1): 41-5, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1912954

RESUMEN

We report the results of intensive therapy and autologous bone marrow transplantation (BMT) in 23 patients with malignant lymphoma (eight Hodgkin's disease and 15 non-Hodgkin's lymphoma) who failed primary therapy. All patients had evidence of disease prior to transplant therapy: 10 had never achieved a complete remission and 13 were in relapse. The preparative regimen included involved field radiation followed by fractionated total body irradiation and high dose cyclophosphamide. A complete remission was achieved in 15 patients, 11 of whom continue in unmaintained complete remission from 27 to 72 months after BMT (median follow-up of 52 months). Of the remaining patients, five did not achieve a complete remission and three died of early toxicity. The event-free survival of the entire group is 47%. Disease status at the time of BMT was significantly correlated with patient outcome. The event-free survival of 13 patients in whom there was no objective evidence of tumor growth on conventional dose therapy was 77% compared with only 10% in patients with tumors progressing on conventional dose therapy (p less than 0.002). All six patients transplanted in untreated relapse continue in unmaintained remission, suggesting that debulking chemotherapy may not be necessary before BMT. Alternative approaches are needed in patients whose tumors progress on conventional dose therapy.


Asunto(s)
Trasplante de Médula Ósea , Ciclofosfamida/uso terapéutico , Linfoma/terapia , Irradiación Corporal Total , Adolescente , Adulto , Niño , Terapia Combinada , Relación Dosis-Respuesta a Droga , Femenino , Enfermedad de Hodgkin/cirugía , Enfermedad de Hodgkin/terapia , Humanos , Linfoma/cirugía , Linfoma no Hodgkin/cirugía , Linfoma no Hodgkin/terapia , Masculino , Persona de Mediana Edad , Pronóstico
18.
J Biol Response Mod ; 9(6): 570-5, 1990 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1705960

RESUMEN

Interferons (IFN) have clinical efficacy in certain hematologic malignancies. Combining IFN with conventional cytotoxic agents has been proposed as a means of improving therapy for diseases such as chronic myelogenous leukemia (CML). In this study, we examined the effect of recombinant interferons alone and in combination with Ara-C on normal and leukemic human hematopoietic progenitor cells (CFU-GM) in vitro. Mononuclear cells from normal bone marrow, peripheral blood of patients with CML, or the acute nonlymphocytic leukemia cell line HL-60 were incubated with alpha-, beta-, or gamma-IFN (0-1,000 units/ml) followed by the addition of Ara-C. The survival of normal CFU-GM was significantly increased if cells were treated with IFN 1 h before 3 h of Ara-C exposure. Similar IFN pretreatment of CML and HL-60 progenitors failed to protect leukemic CFU-GM from Ara-C-induced toxicity. This selective protection of normal CFU-GM may have clinical application.


Asunto(s)
Citarabina/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Interferones/farmacología , Adulto , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Citarabina/toxicidad , Granulocitos/citología , Granulocitos/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Humanos , Técnicas In Vitro , Macrófagos/citología , Macrófagos/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos
19.
Exp Hematol ; 17(11): 1105-9, 1989 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2583254

RESUMEN

The effect of in vitro hyperthermia on normal human bone marrow granulocyte-macrophage progenitor cells (granulocyte-macrophage colony-forming units, CFU-GM) was compared to its effect on clonogenic acute nonlymphocytic leukemic (ANLL) cells. Mononuclear normal bone marrow cells, blasts from patients with ANLL, and HL-60 cells were incubated at room temperature (control) and at 42 degrees-44 degrees C for 0-120 min prior to assay in methylcellulose. The heat sensitivity of the leukemic cells was significantly greater than that of normal bone marrow progenitors. Two-h exposure to 43 degrees C, for example, resulted in survival of 52% of normal marrow CFU-GM, whereas only 3% of leukemic CFU-GM survived (p less than 0.001 for HL-60 cells and p less than 0.005 for patient blast cells). To determine the effect of hyperthermia on more primitive progenitors and on marrow stromal cells, long-term cultures of normal bone marrow were established using control and heat-treated cells. Generation of CFU-GM was detected in the nonadherent fraction of hyperthermia-treated samples throughout the 5-week culture period. Although stromal development was slightly delayed, hyperthermia-treated cells were able to establish stromal layers similar to control cells. These results indicate that normal bone marrow committed progenitor cells are more resistant to hyperthermia than are myeloid leukemic cells. Normal stromal cells and primitive cells assayed in long-term culture are also resistant to hyperthermia that is toxic for leukemic cells. Because of this differential sensitivity to heat, ex vivo hyperthermia may be applicable for removing residual leukemic cells from bone marrow harvested for autologous transplantation.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Hipertermia Inducida , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/fisiología , Células de la Médula Ósea , Supervivencia Celular , Hematopoyesis , Humanos , Leucemia Mieloide Aguda/terapia , Células Tumorales Cultivadas
20.
Blood ; 70(6): 1792-6, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3499944

RESUMEN

Prostaglandins of the E series (PGE) inhibit proliferation of normal bone marrow granulocyte/macrophage progenitors (CFU-GM). Circulating CFU-GM are known to differ from marrow CFU-GM in many characteristics, and in the present study, we compared the effect of PGE1 on circulating and bone marrow progenitors in normals and in patients with chronic myelogenous leukemia (CML). PGE1 caused a dose-dependent inhibition of normal marrow CFU-GM. Circulating CFU-GM were inhibited only at concentrations of 10(-5) mol/L or greater, and progenitor proliferation was, in fact, significantly stimulated at PGE1 concentrations between 10(-8) and 10(-6) mol/L. Bone marrow CFU-GM from patients with CML were inhibited in a manner similar to that of normal bone marrow. Circulating cells from patients with CML were, however, less sensitive to PGE1 inhibition than CML bone marrow cells and demonstrated a pattern intermediate between normal circulating and normal marrow progenitors. These studies suggest that peripheral blood and bone marrow contain different progenitor cell populations.


Asunto(s)
Alprostadil/farmacología , Granulocitos/citología , Hematopoyesis/efectos de los fármacos , Macrófagos/citología , Células Sanguíneas , Células de la Médula Ósea , Diferenciación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Factores Estimulantes de Colonias/farmacología , Sustancias de Crecimiento/farmacología , Humanos , Leucemia Mieloide/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA