Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 17934, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289434

ABSTRACT

Targeted radiopharmaceutical therapy with alpha-particle emitters (αRPT) is advantageous in cancer treatment because the short range and high local energy deposition of alpha particles enable precise radiation delivery and efficient tumor cell killing. However, these properties create sub-organ dose deposition effects that are not easily characterized by direct gamma-ray imaging (PET or SPECT). We present a computational procedure to determine the spatial distribution of absorbed dose from alpha-emitting radionuclides in tissues using digital autoradiography activity images from an ionizing-radiation quantum imaging detector (iQID). Data from 211At-radioimmunotherapy studies for allogeneic hematopoietic cell transplantation in a canine model were used to develop these methods. Nine healthy canines were treated with 16.9-30.9 MBq 211At/mg monoclonal antibodies (mAb). Lymph node biopsies from early (2-5 h) and late (19-20 h) time points (16 total) were obtained, with 10-20 consecutive 12-µm cryosections extracted from each and imaged with an iQID device. iQID spatial activity images were registered within a 3D volume for dose-point-kernel convolution, producing dose-rate maps. The accumulated absorbed doses for high- and low-rate regions were 9 ± 4 Gy and 1.2 ± 0.8 Gy from separate dose-rate curves, respectively. We further assess uptake uniformity, co-registration with histological pathology, and requisite slice numbers to improve microscale characterization of absorbed dose inhomogeneities in αRPT.


Subject(s)
Alpha Particles , Radiopharmaceuticals , Animals , Dogs , Alpha Particles/therapeutic use , Autoradiography , Radiopharmaceuticals/therapeutic use , Radiometry , Radioisotopes/therapeutic use , Antibodies, Monoclonal
2.
Cancer Res ; 76(22): 6669-6679, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27590740

ABSTRACT

Streptavidin (SA)-biotin pretargeted radioimmunotherapy (PRIT) that targets CD20 in non-Hodgkin lymphoma (NHL) exhibits remarkable efficacy in model systems, but SA immunogenicity and interference by endogenous biotin may complicate clinical translation of this approach. In this study, we engineered a bispecific fusion protein (FP) that evades the limitations imposed by this system. Briefly, one arm of the FP was an anti-human CD20 antibody (2H7), with the other arm of the FP an anti-chelated radiometal trap for a radiolabeled ligand (yttrium[Y]-DOTA) captured by a very high-affinity anti-Y-DOTA scFv antibody (C825). Head-to-head biodistribution experiments comparing SA-biotin and bispecific FP (2H7-Fc-C825) PRIT in murine subjects bearing human lymphoma xenografts demonstrated nearly identical tumor targeting by each modality at 24 hours. However, residual radioactivity in the blood and normal organs was consistently higher following administration of 1F5-SA compared with 2H7-Fc-C825. Consequently, tumor-to-normal tissue ratios of distribution were superior for 2H7-Fc-C825 (P < 0.0001). Therapy studies in subjects bearing either Ramos or Granta subcutaneous lymphomas demonstrated that 2H7-Fc-C825 PRIT is highly effective and significantly less myelosuppressive than 1F5-SA (P < 0.0001). All animals receiving optimal doses of 2H7-Fc-C825 followed by 90Y-DOTA were cured by 150 days, whereas the growth of tumors in control animals progressed rapidly with complete morbidity by 25 days. In addition to demonstrating reduced risk of immunogenicity and an absence of endogenous biotin interference, our findings offer a preclinical proof of concept for the preferred use of bispecific PRIT in future clinical trials, due to a slightly superior biodistribution profile, less myelosuppression, and superior efficacy. Cancer Res; 76(22); 6669-79. ©2016 AACR.


Subject(s)
Antibodies, Bispecific/metabolism , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/radiotherapy , Radioimmunotherapy/methods , Streptavidin/therapeutic use , Animals , Antibodies, Bispecific/analysis , Female , Humans , Lymphoma, B-Cell/pathology , Mice , Streptavidin/pharmacology
3.
Blood ; 127(3): 352-9, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26576864

ABSTRACT

Many patients with hematologic malignancies cannot tolerate hematopoietic cell transplantation (HCT), whereas others may not have a compatible human leukocyte antigen-matched donor. To overcome these limitations, we optimized a conditioning regimen employing anti-CD45 radioimmunotherapy (RIT) replacing total body irradiation (TBI) before haploidentical HCT in a murine model. Mice received 200 to 400 µCi (90)Y-anti-CD45 antibody (30F11), with or without fludarabine (5 days starting day -8), with cyclophosphamide (CY; days -2 and +2) for graft-versus-host disease prophylaxis, and 1.5 × 10(7) haploidentical donor bone marrow cells (day 0). Haploidentical bone marrow transplantation (BMT) with 300 µCi (90)Y-anti-CD45 RIT and CY, without TBI or fludarabine, led to mixed chimeras with 81.3 ± 10.6% mean donor origin CD8(+) cells detected 1 month after BMT, and remained stable (85.5 ± 11% mean donor origin CD8(+) cells) 6 months after haploidentical BMT. High chimerism levels were induced across multiple hematopoietic lineages 28 days after haploidentical BMT with 69.3 ± 14.1%, 75.6 ± 20.2%, and 88.5 ± 11.8% CD3(+) T cells, B220(+) B cells, and CD11b(+) myeloid cells, respectively. Fifty percent of SJL leukemia-bearing mice treated with 400 µCi (90)Y-DOTA-30F11, CY, and haploidentical BMT were cured and lived >200 days. Mice treated with 200 µCi (90)Y-DOTA-30F11 had a median overall survival of 73 days, while untreated leukemic mice had a median overall survival of 34 days (P < .001, Mantel-Cox test). RIT-mediated haploidentical BMT without TBI may increase treatment options for aggressive hematologic malignancies.


Subject(s)
Graft Survival/genetics , Graft Survival/immunology , Haplotypes , Immunoconjugates/administration & dosage , Leukocyte Common Antigens/antagonists & inhibitors , Radioimmunotherapy , Tissue Donors , Transplantation Conditioning , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Surface/metabolism , Bone Marrow Transplantation , Cell Lineage , Disease Models, Animal , Female , Graft Survival/drug effects , Graft Survival/radiation effects , Haplotypes/genetics , Haplotypes/immunology , Hematopoietic Stem Cell Transplantation , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Humans , Immunophenotyping , Leukemia/mortality , Leukemia/therapy , Male , Mice , Radioimmunotherapy/methods , Transplantation Chimera , Transplantation, Homologous , Whole-Body Irradiation
4.
J Nucl Med ; 56(11): 1766-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26338894

ABSTRACT

UNLABELLED: α-radioimmunotherapy targeting CD45 may substitute for total-body irradiation in hematopoietic cell transplantation (HCT) preparative regimens for lymphoma. Our goal was to optimize the anti-CD45 monoclonal antibody (mAb; CA12.10C12) protein dose for (211)At-radioimmunotherapy, extending the analysis to include intraorgan (211)At activity distribution and α-imaging-based small-scale dosimetry, along with immunohistochemical staining. METHODS: Eight normal dogs were injected with either a 0.75 (n = 5) or 1.00 (n = 3) mg/kg dose of (211)At-B10-CA12.10C12 (11.5-27.6 MBq/kg). Two were euthanized and necropsied 19-22 h after injection, and 6 received autologous HCT 3 d after (211)At-radioimmunotherapy, after lymph node and bone marrow biopsies at 2-4 and/or 19 h after injection. Blood was sampled to study toxicity and clearance; CD45 targeting was evaluated by flow cytometry. (211)At localization and small-scale dosimetry were assessed using two α-imaging systems: an α-camera and an ionizing-radiation quantum imaging detector (iQID) camera. RESULTS: (211)At uptake was highest in the spleen (0.31-0.61% injected activity [%IA]/g), lymph nodes (0.02-0.16 %IA/g), liver (0.11-0.12 %IA/g), and marrow (0.06-0.08 %IA/g). Lymphocytes in blood and marrow were efficiently targeted using either mAb dose. Lymph nodes remained unsaturated but displayed targeted (211)At localization in T lymphocyte-rich areas. Absorbed doses to blood, marrow, and lymph nodes were estimated at 3.1, 2.4, and 3.4 Gy/166 MBq, respectively. All transplanted dogs experienced transient hepatic toxicity. Liver enzyme levels were temporarily elevated in 5 of 6 dogs; one treated with 1.00 mg mAb/kg developed ascites and was euthanized 136 d after HCT. CONCLUSION: (211)At-anti-CD45 radioimmunotherapy with 0.75 mg mAb/kg efficiently targeted blood and marrow without severe toxicity. Dosimetry calculations and observed radiation-induced effects indicated that sufficient (211)At-B10-CA12.10C12 localization was achieved for efficient conditioning for HCT.


Subject(s)
Astatine/pharmacokinetics , Hematopoietic Stem Cell Transplantation/methods , Leukocyte Common Antigens , Radioimmunotherapy/methods , Radiopharmaceuticals/pharmacokinetics , Alpha Particles , Animals , Ascites/diagnostic imaging , Astatine/adverse effects , Biopsy , Bone Marrow/diagnostic imaging , Dogs , Drug Delivery Systems , Immunohistochemistry , Lymph Nodes/diagnostic imaging , Radiometry , Radionuclide Imaging , Radiopharmaceuticals/adverse effects , Spleen/diagnostic imaging , T-Lymphocytes/diagnostic imaging , Tissue Distribution
5.
Med Phys ; 42(7): 4094-105, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26133610

ABSTRACT

PURPOSE: Alpha-emitting radionuclides exhibit a potential advantage for cancer treatments because they release large amounts of ionizing energy over a few cell diameters (50-80 µm), causing localized, irreparable double-strand DNA breaks that lead to cell death. Radioimmunotherapy (RIT) approaches using monoclonal antibodies labeled with α emitters may thus inactivate targeted cells with minimal radiation damage to surrounding tissues. Tools are needed to visualize and quantify the radioactivity distribution and absorbed doses to targeted and nontargeted cells for accurate dosimetry of all treatment regimens utilizing α particles, including RIT and others (e.g., Ra-223), especially for organs and tumors with heterogeneous radionuclide distributions. The aim of this study was to evaluate and characterize a novel single-particle digital autoradiography imager, the ionizing-radiation quantum imaging detector (iQID) camera, for use in α-RIT experiments. METHODS: The iQID camera is a scintillator-based radiation detection system that images and identifies charged-particle and gamma-ray/x-ray emissions spatially and temporally on an event-by-event basis. It employs CCD-CMOS cameras and high-performance computing hardware for real-time imaging and activity quantification of tissue sections, approaching cellular resolutions. In this work, the authors evaluated its characteristics for α-particle imaging, including measurements of intrinsic detector spatial resolutions and background count rates at various detector configurations and quantification of activity distributions. The technique was assessed for quantitative imaging of astatine-211 ((211)At) activity distributions in cryosections of murine and canine tissue samples. RESULTS: The highest spatial resolution was measured at ∼20 µm full width at half maximum and the α-particle background was measured at a rate as low as (2.6 ± 0.5) × 10(-4) cpm/cm(2) (40 mm diameter detector area). Simultaneous imaging of multiple tissue sections was performed using a large-area iQID configuration (ø 11.5 cm). Estimation of the (211)At activity distribution was demonstrated at mBq/µg-levels. CONCLUSIONS: Single-particle digital autoradiography of α emitters has advantages over traditional film-based autoradiographic techniques that use phosphor screens, in terms of spatial resolution, sensitivity, and activity quantification capability. The system features and characterization results presented in this study show that the iQID is a promising technology for microdosimetry, because it provides necessary information for interpreting alpha-RIT outcomes and for predicting the therapeutic efficacy of cell-targeted approaches using α emitters.


Subject(s)
Autoradiography/instrumentation , Autoradiography/methods , Gamma Cameras , Radioimmunotherapy/instrumentation , Radioimmunotherapy/methods , Animals , Antigens, CD20/administration & dosage , Astatine , Dogs , Equipment Design , Female , Leukocyte Common Antigens/administration & dosage , Lymph Nodes/diagnostic imaging , Lymph Nodes/immunology , Lymph Nodes/radiation effects , Lymphoma, Non-Hodgkin/diagnostic imaging , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/radiotherapy , Mice , Mice, Nude , Neoplasm Transplantation , Phantoms, Imaging , Radiography , Software
6.
PLoS One ; 10(3): e0120561, 2015.
Article in English | MEDLINE | ID: mdl-25785845

ABSTRACT

PURPOSE: Pretargeted radioimmunotherapy (PRIT) is a multi-step method of selectively delivering high doses of radiotherapy to tumor cells while minimizing exposure to surrounding tissues. Yttrium-90 (90Y) and lutetium-177 (177Lu) are two of the most promising beta-particle emitting radionuclides used for radioimmunotherapy, which despite having similar chemistries differ distinctly in terms of radiophysical features. These differences may have important consequences for the absorbed dose to tumors and normal organs. Whereas 90Y has been successfully applied in a number of preclinical and clinical radioimmunotherapy settings, there have been few published pretargeting studies with 177Lu. We therefore compared the therapeutic potential of targeting either 90Y or 177Lu to human B-cell lymphoma xenografts in mice. METHODS: Parallel experiments evaluating the biodistribution, imaging, dosimetry, therapeutic efficacy, and toxicity were performed in female athymic nude mice bearing either Ramos (Burkitt lymphoma) or Granta (mantle cell lymphoma) xenografts, utilizing an anti-CD20 antibody-streptavidin conjugate (1F5-SA) and an 90Y- or 177Lu-labeled 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-biotin second step reagent. RESULTS: The two radionuclides displayed comparable biodistributions in tumors and normal organs; however, the absorbed radiation dose delivered to tumor was more than twice as high for 90Y (1.3 Gy/MBq) as for 177Lu (0.6 Gy/MBq). More importantly, therapy with 90Y-DOTA-biotin was dramatically more effective than with 177Lu-DOTA-biotin, with 100% of Ramos xenograft-bearing mice cured with 37 MBq 90Y, whereas 0% were cured using identical amounts of 177Lu-DOTA-biotin. Similar results were observed in mice bearing Granta xenografts, with 80% of the mice cured with 90Y-PRIT and 0% cured with 177Lu-PRIT. Toxicities were comparable with both isotopes. CONCLUSION: 90Y was therapeutically superior to 177Lu for streptavidin-biotin PRIT approaches in these human lymphoma xenograft models.


Subject(s)
Antigens, CD20/immunology , Immunoconjugates/therapeutic use , Lutetium/therapeutic use , Lymphoma/radiotherapy , Radioimmunotherapy/methods , Yttrium Radioisotopes/therapeutic use , Animals , Beta Particles/therapeutic use , Cell Line, Tumor , Cell Transformation, Neoplastic , Female , Humans , Immunoconjugates/adverse effects , Immunoconjugates/immunology , Lutetium/adverse effects , Lutetium/pharmacokinetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Nude , Radioimmunotherapy/adverse effects , Tissue Distribution , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/adverse effects , Yttrium Radioisotopes/pharmacokinetics
7.
Blood ; 125(13): 2111-9, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25628467

ABSTRACT

α-Emitting radionuclides deposit a large amount of energy within a few cell diameters and may be particularly effective for radioimmunotherapy targeting minimal residual disease (MRD). To evaluate this hypothesis, (211)At-labeled 1F5 monoclonal antibody (mAb) (anti-CD20) was studied in both bulky lymphoma tumor xenograft and MRD animal models. Superior treatment responses to (211)At-labeled 1F5 mAb were evident in the MRD setting. Lymphoma xenograft tumor-bearing animals treated with doses of up to 48 µCi of (211)At-labeled anti-CD20 mAb ([(211)At]1F5-B10) experienced modest responses (0% cures but two- to threefold prolongation of survival compared with negative controls). In contrast, 70% of animals in the MRD lymphoma model demonstrated complete eradication of disease when treated with (211)At-B10-1F5 at a radiation dose that was less than one-third (15 µCi) of the highest dose given to xenograft animals. Tumor progression among untreated control animals in both models was uniformly lethal. After 130 days, no significant renal or hepatic toxicity was observed in the cured animals receiving 15 µCi of [(211)At]1F5-B10. These findings suggest that α-emitters are highly efficacious in MRD settings, where isolated cells and small tumor clusters prevail.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , Astatine/therapeutic use , Immunoconjugates/therapeutic use , Lymphoma, B-Cell/radiotherapy , Animals , Female , Humans , Jurkat Cells , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Radioimmunotherapy , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Nucl Med ; 54(8): 1347-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23761919

ABSTRACT

UNLABELLED: Targeted α-therapy (TAT) appears to be an ideal therapeutic technique for eliminating malignant circulating, minimal residual, or micrometastatic cells. These types of malignancies are typically infraclinical, complicating the evaluation of potential treatments. This study presents a method of ex vivo activity quantification with an α-camera device, allowing measurement of the activity taken up by tumor cells in biologic structures a few tens of microns. METHODS: We examined micrometastases from a murine model of ovarian carcinoma after injection of a radioimmunoconjugate labeled with (211)At for TAT. At different time points, biologic samples were excised and cryosectioned. The activity level and the number of tumor cells were determined by combined information from 2 adjacent sections: one exposed to the α-camera and the other stained with hematoxylin and eosin. The time-activity curves for tumor cell clusters, comprising fewer than 10 cells, were derived for 2 different injected activities (6 and 1 MBq). RESULTS: High uptake and good retention of the radioimmunoconjugate were observed at the surface of tumor cells. Dosimetric calculations based on the measured time-integrated activity indicated that for an injected activity of 1 MBq, isolated tumor cells received at least 12 Gy. In larger micrometastases (≤ 100 µm in diameter), the activity uptake per cell was lower, possibly because of hindered penetration of radiolabeled antibodies; however, the mean absorbed dose delivered to tumor cells was above 30 Gy, due to cross-fire irradiation. CONCLUSION: Using the α-camera, we developed a method of ex vivo activity quantification at the cellular scale, which was further applied to characterize the behavior of a radiolabeled antibody administered in vivo against ovarian carcinoma. This study demonstrated a reliable measurement of activity. This method of activity quantification, based on experimentally measured data, is expected to improve the relevance of small-scale dosimetry studies and thus to accelerate the optimization of TAT.


Subject(s)
Alpha Particles , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/pathology , Radionuclide Imaging/instrumentation , Animals , Calibration , Cell Line, Tumor , Female , Humans , Mice , Neoplasm Metastasis , Radiation Dosage
9.
Cancer Biother Radiopharm ; 28(2): 108-14, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23230896

ABSTRACT

UNLABELLED: Abstract Purpose: Pretargeted radioimmunotherapy (PRIT) against intraperitoneal (i.p.) ovarian microtumors using avidin-conjugated monoclonal antibody MX35 (avidin-MX35) and (211)At-labeled, biotinylated, succinylated poly-l-lysine ((211)At-B-PLsuc) was compared with conventional radioimmunotherapy (RIT) using (211)At-labeled MX35 in a nude mouse model. METHODS: Mice were inoculated i.p. with 1×10(7) NIH:OVCAR-3 cells. After 3 weeks, they received PRIT (1.0 or 1.5 MBq), RIT (0.9 MBq), or no treatment. Concurrently, 10 additional animals were sacrificed and examined to determine disease progression at the start of therapy. Treated animals were analyzed with regard to presence of tumors and ascites (tumor-free fraction; TFF), 8 weeks after therapy. RESULTS: Tumor status at baseline was advanced: 70% of sacrificed animals exhibited ascites. The TFFs were 0.35 (PRIT 1.0 MBq), 0.45 (PRIT 1.5 MBq), and 0.45 (RIT). The 1.5-MBq PRIT group exhibited lower incidence of ascites and fewer tumors >1 mm than RIT-treated animals. CONCLUSIONS: PRIT was as effective as RIT with regard to TFF; however, the size distribution of tumors and presence of ascites indicated that 1.5-MBq PRIT was more efficient. Despite advanced disease in many animals at the time of treatment, PRIT demonstrated good potential to treat disseminated ovarian cancer.


Subject(s)
Alpha Particles/therapeutic use , Antibodies, Monoclonal/therapeutic use , Astatine/administration & dosage , Avidin/therapeutic use , Disease Models, Animal , Ovarian Neoplasms/radiotherapy , Radioimmunotherapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/immunology , Radionuclide Imaging , Tissue Distribution
10.
Curr Radiopharm ; 4(3): 248-60, 2011 Jul.
Article in English | MEDLINE | ID: mdl-22201711

ABSTRACT

Alpha-particle emitting radionuclides are attractive for targeted cancer therapies due to their physicochemical properties. Their high linear energy transfer (LET) and short particle range makes them particularly toxic at a microscopic level, which is ideal for treating disseminated micrometastases. However, their cytotoxic properties also place special demands on the pharmacokinetics of the tumor specific carrier vector, where high tumor-to-normal-tissue ratios are a prerequisite. Tumor specific antibodies are perhaps the most common vector for targeted therapy, but due to pharmacokinetics considerations antibodies will generally not meet the standard for α-particle radioimmunotherapy. However, the tumor specificity of monoclonal antibodies may be used in pretargeting techniques, strategies used to increase the selectivity of the radioactivity. The basic concept of pretargeting relies on a separate administration of a modified antibody and a radioactive ligand. The modified antibody is first injected and allowed to localize on the tumor. Then, the radiolabeled ligand is injected, which is a small molecule that rapidly localizes the modified antibody on tumor cells while non-localized ligand rapidly clears from the circulation, preferably through renal filtration. Several pretargeting strategies have been developed, in particular the avidin-biotin system and bispecific antibodies. Approaches under evaluation are the use of complementary DNA, morpholinos, and the use of infinite antigen binding. Preclinical and clinical studies of pretargeting have shown that favorable distribution of the radioactivity can be achieved, which may increase dose to the tumor as compared with the dose from directly labeled antibodies, and most important decrease the dose to normal tissues. This survey describes different pretargeting strategies, and includes a review of pretargeting with α emitting radionuclides.


Subject(s)
Alpha Particles/therapeutic use , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Humans
11.
Cancer ; 116(4 Suppl): 1101-10, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20127953

ABSTRACT

BACKGROUND: Pretargeting is an approach for enhancing the therapeutic index of radioimmunotherapy by separating the administrations of tumor-targeting substance and radiolabel. In this study, a pretargeting model system of avidin-conjugated monoclonal antibody trastuzumab and biotinylated, (211)At-labeled poly-L-lysine was constructed and analyzed in vitro. METHODS: Avidin activated by 4-(N-maleimidomethyl)cyclohexane-1-carboxylic acid 3-sulfo-N-hydroxysuccinimide ester sodium salt (sulfo-SMCC) and thiolated trastuzumab were incubated overnight at 4 degrees C. The monomeric fraction was extracted using size exclusion fast protein liquid chromatography (FPLC) and further purified on an iminobiotin affinity column. Poly-L-lysine was biotinylated with succinimidyl-6-(biotinamido)hexanoate (NHS-LC-biotin), followed by direct (211)At-labeling with N-succinimidyl-3-(trimethylstannyl)benzoate (m-MeATE), and succinylation with succinic anhydride. The avidin-trastuzumab conjugate was characterized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and FPLC, together with cell-binding and biotin-binding analyses. The labeled poly-L-lysine conjugate was assessed in terms of radiochemical purity and avidin binding. Furthermore, the full pretargeting system was evaluated in a tumor cell binding assay. RESULTS: The estimated size of the pretargeting molecule was 220 kDa, which corresponds to that of the expected avidin-trastuzumab monomer. Neither cell-binding ability (64%) nor biotin-binding ability (85%-95%) indicated any severe adverse effects from the chemical modifications. The radiochemical purity of the effector molecule was 92%-97%, and the avidin binding capacity was 91%-93%. The complete pretargeting assay resulted in a binding of 75.3 +/- 6.2% of added effector molecules to cells. CONCLUSIONS: The high binding of effector molecules to cells demonstrates a proof of concept for the synthesized molecules and pretargeting system, which will be further evaluated in vivo in future studies.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Astatine/therapeutic use , Avidin/administration & dosage , Peptides/therapeutic use , Radioimmunotherapy/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Biotinylation , In Vitro Techniques , Mice , Mice, Nude , Peptides/chemistry , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...