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










Publication year range
1.
Mol Cancer Ther ; 19(12): 2575-2584, 2020 12.
Article in English | MEDLINE | ID: mdl-33082277

ABSTRACT

Pretargeted radioimmunotherapy (PRIT) has been investigated as a multi-step approach to decrease relapse and toxicity for high-risk acute myeloid leukemia (AML). Relevant factors including endogenous biotin and immunogenicity, however, have limited the use of PRIT with an anti-CD45 antibody streptavidin conjugate and radiolabeled DOTA-biotin. To overcome these limitations we designed anti-murine and anti-human CD45 bispecific antibody constructs using 30F11 and BC8 antibodies, respectively, combined with an anti-yttrium (Y)-DOTA single-chain variable fragment (C825) to capture a radiolabeled ligand. The bispecific construct targeting human CD45 (BC8-Fc-C825) had high uptake in leukemia HEL xenografts [7.8 ± 0.02% percent injected dose/gram of tissue (% ID/g)]. Therapy studies showed that 70% of mice with HEL human xenografts treated with BC8-Fc-C825 followed by 44.4 MBq (1,200 µCi) of 90Y-DOTA-biotin survived at least 170 days after therapy, while all nontreated controls required euthanasia because of tumor progression by day 32. High uptake at sites of leukemia (spleen and bone marrow) was also seen with 30F11-IgG1-C825 in a syngeneic disseminated SJL murine leukemia model (spleen, 9.0 ± 1.5% ID/g and bone marrow, 8.1 ± 1.2% ID/g), with minimal uptake in all other normal organs (<0.5% ID/g) at 24 hours after 90Y-DOTA injections. SJL leukemia mice treated with the bispecific 30F11-IgG1-C825 and 29.6 MBq (800 µCi) of 90Y-DOTA-biotin had a survival advantage compared with untreated leukemic mice (median, 43 vs. 30 days, respectively; P < 0.0001). These data suggest bispecific antibody-mediated PRIT may be highly effective for leukemia therapy and translation to human studies.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Biotin/analogs & derivatives , Leukocyte Common Antigens/antagonists & inhibitors , Organometallic Compounds/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Bispecific/genetics , Biotin/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Genetic Engineering , Humans , Leukemia, Myeloid , Mice , Recombinant Fusion Proteins/genetics , Tissue Distribution , Xenograft Model Antitumor Assays
2.
Blood ; 131(6): 611-620, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29158362

ABSTRACT

Pretargeted radioimmunotherapy (PRIT) has demonstrated remarkable efficacy targeting tumor antigens, but immunogenicity and endogenous biotin blocking may limit clinical translation. We describe a new PRIT approach for the treatment of multiple myeloma (MM) and other B-cell malignancies, for which we developed an anti-CD38-bispecific fusion protein that eliminates endogenous biotin interference and immunogenic elements. In murine xenograft models of MM and non-Hodgkin lymphoma (NHL), the CD38-bispecific construct demonstrated excellent blood clearance and tumor targeting. Dosimetry calculations showed a tumor-absorbed dose of 43.8 Gy per millicurie injected dose of 90Y, with tumor-to-normal organ dose ratios of 7:1 for liver and 15:1 for lung and kidney. In therapy studies, CD38-bispecific PRIT resulted in 100% complete remissions by day 12 in MM and NHL xenograft models, ultimately curing 80% of mice at optimal doses. In direct comparisons, efficacy of the CD38 bispecific proved equal or superior to streptavidin (SA)-biotin-based CD38-SA PRIT. Each approach cured at least 75% of mice at the highest radiation dose tested (1200 µCi), whereas at 600- and 1000-µCi doses, the bispecific outperformed the SA approach, curing 35% more mice overall (P < .004). The high efficacy of bispecific PRIT, combined with its reduced risk of immunogenicity and endogenous biotin interference, make the CD38 bispecific an attractive candidate for clinical translation. Critically, CD38 PRIT may benefit patients with unresponsive, high-risk disease because refractory disease typically retains radiation sensitivity. We posit that PRIT might not only prolong survival, but possibly cure MM and treatment-refractory NHL patients.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Bispecific/therapeutic use , Leukemia, B-Cell/radiotherapy , Lymphoma, B-Cell/radiotherapy , Multiple Myeloma/radiotherapy , Radioimmunotherapy/methods , ADP-ribosyl Cyclase 1/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Humans , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/pathology , Mice, Nude , Molecular Targeted Therapy , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
3.
Cancer Res ; 77(14): 3885-3893, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28566329

ABSTRACT

Constitutive B-cell receptor signaling leads to overexpression of the antiapoptotic BCL-2 protein and is implicated in the pathogenesis of many types of B-cell non-Hodgkin lymphoma (B-NHL). The BCL-2 small-molecule inhibitor venetoclax shows promising clinical response rates in several lymphomas, but is not curative as monotherapy. Radiotherapy is a rational candidate for combining with BCL-2 inhibition, as DNA damage caused by radiotherapy increases the activity of pro-apoptotic BCL-2 pathway proteins, and lymphomas are exquisitely sensitive to radiation. We tested B-NHL responses to venetoclax combined with either external beam radiotherapy or radioimmunotherapy (RIT), which joins the selectivity of antibody targeting with the effectiveness of irradiation. We first tested cytotoxicity of cesium-137 irradiation plus venetoclax in 14 B-NHL cell lines representing five lymphoma subtypes. Combination treatment synergistically increased cell death in 10 of 14 lines. Lack of synergy was predicted by resistance to single-agent venetoclax and high BCL-XL expression. We then assessed the efficacy of external beam radiotherapy plus venetoclax in murine xenograft models of mantle cell (MCL), germinal-center diffuse large B-cell (GCB-DLBCL), and activated B-cell (ABC-DLBCL) lymphomas. In each model, external beam radiotherapy plus venetoclax synergistically increased mouse survival time, curing up to 10%. We finally combined venetoclax treatment of MCL and ABC-DLBCL xenografts with a pretargeted RIT (PRIT) system directed against the CD20 antigen. Optimal dosing of PRIT plus venetoclax cured 100% of mice with no detectable toxicity. Venetoclax combined with radiotherapy may be a promising treatment for a wide range of lymphomas Cancer Res; 77(14); 3885-93. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/radiotherapy , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Cesium Radioisotopes/pharmacology , Chemoradiotherapy , Female , Humans , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred NOD , Radiation Tolerance/drug effects , Radioimmunotherapy , Xenograft Model Antitumor Assays
4.
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
5.
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
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.
PLoS One ; 9(12): e113601, 2014.
Article in English | MEDLINE | ID: mdl-25460570

ABSTRACT

Radioimmunotherapy (RIT) for treatment of hematologic malignancies has primarily employed monoclonal antibodies (Ab) labeled with 131I or 90Y which have limitations, and alternative radionuclides are needed to facilitate wider adoption of RIT. We therefore compared the relative therapeutic efficacy and toxicity of anti-CD45 RIT employing 90Y and 177Lu in a syngeneic, disseminated murine myeloid leukemia (B6SJLF1/J) model. Biodistribution studies showed that both 90Y- and 177Lu-anti-murine CD45 Ab conjugates (DOTA-30F11) targeted hematologic tissues, as at 24 hours 48.8 ± 21.2 and 156 ± 14.6% injected dose per gram of tissue (% ID/g) of 90Y-DOTA-30F11 and 54.2 ± 9.5 and 199 ± 11.7% ID/g of 177Lu-DOTA-30F11 accumulated in bone marrow (BM) and spleen, respectively. However, 90Y-DOTA-30F11 RIT demonstrated a dose-dependent survival benefit: 60% of mice treated with 300 µCi 90Y-DOTA-30F11 lived over 180 days after therapy, and mice treated with 100 µCi 90Y-DOTA-30F11 had a median survival 66 days. 90Y-anti-CD45 RIT was associated with transient, mild myelotoxicity without hepatic or renal toxicity. Conversely, 177Lu- anti-CD45 RIT yielded no long-term survivors. Thus, 90Y was more effective than 177Lu for anti-CD45 RIT of AML in this murine leukemia model.


Subject(s)
Leukemia, Myeloid/immunology , Leukemia, Myeloid/radiotherapy , Leukocyte Common Antigens/immunology , Radioimmunotherapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Humans , Kidney/pathology , Kidney/radiation effects , Leukemia, Myeloid/pathology , Leukocyte Common Antigens/antagonists & inhibitors , Mice , Radionuclide Imaging , Spleen/pathology , Spleen/radiation effects , Tissue Distribution/immunology , Tissue Distribution/radiation effects , Treatment Outcome , Yttrium Radioisotopes/adverse effects , Yttrium Radioisotopes/therapeutic use
9.
Cancer Res ; 74(20): 5846-54, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25261237

ABSTRACT

Cerenkov radiation generated by positron-emitting radionuclides can be exploited for a molecular imaging technique known as Cerenkov luminescence imaging (CLI). Data have been limited, however, on the use of medium- to high-energy ß-emitting radionuclides of interest for cancer imaging and treatment. We assessed the use of CLI as an adjunct to determine localization of radioimmunoconjugates to hematolymphoid tissues. Radiolabeled (177)Lu- or (90)Y-anti-CD45 antibody (Ab; DOTA-30F11) was administered by tail vein injection to athymic mice bearing disseminated murine myeloid leukemia, with CLI images acquired at times afterward. Gamma counting of individual organs showed preferential uptake in CD45(+) tissues with significant retention of radiolabeled Ab in sites of leukemia (spleen and bone marrow). This result was confirmed in CLI images with 1.35 × 10(5) ± 2.2 × 10(4) p/s/cm(2)/sr and 3.45 × 10(3) ± 7.0 × 10(2) p/s/cm(2)/sr for (90)Y-DOTA-30F11 and (177)Lu-DOTA-30F11, respectively, compared with undetectable signal for both radionuclides using the nonbinding control Ab. Results showed that CLI allows for in vivo visualization of localized ß-emissions. Pixel intensity variability resulted from differences in absorbed doses of the associated energies of the ß-emitting radionuclide. Overall, our findings offer a preclinical proof of concept for the use of CLI techniques in tandem with currently available clinical diagnostic tools.


Subject(s)
Immunoconjugates , Leukemia/diagnostic imaging , Radiopharmaceuticals , Yttrium Radioisotopes , Animals , Cell Line, Tumor , Female , Luminescent Measurements , Lutetium , Mice , Phantoms, Imaging , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Spleen/metabolism , Tissue Distribution , Yttrium Radioisotopes/pharmacokinetics
10.
Cancer Res ; 74(4): 1179-89, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371230

ABSTRACT

The vast majority of patients with plasma cell neoplasms die of progressive disease despite high response rates to novel agents. Malignant plasma cells are very radiosensitive, but the potential role of radioimmunotherapy (RIT) in the management of plasmacytomas and multiple myeloma has undergone only limited evaluation. Furthermore, CD38 has not been explored as a RIT target despite its uniform high expression on malignant plasma cells. In this report, both conventional RIT (directly radiolabeled antibody) and streptavidin-biotin pretargeted RIT (PRIT) directed against the CD38 antigen were assessed as approaches to deliver radiation doses sufficient for multiple myeloma cell eradication. PRIT demonstrated biodistributions that were markedly superior to conventional RIT. Tumor-to-blood ratios as high as 638:1 were seen 24 hours after PRIT, whereas ratios never exceeded 1:1 with conventional RIT. (90)Yttrium absorbed dose estimates demonstrated excellent target-to-normal organ ratios (6:1 for the kidney, lung, liver; 10:1 for the whole body). Objective remissions were observed within 7 days in 100% of the mice treated with doses ranging from 800 to 1,200 µCi of anti-CD38 pretargeted (90)Y-DOTA-biotin, including 100% complete remissions (no detectable tumor in treated mice compared with tumors that were 2,982% ± 2,834% of initial tumor volume in control animals) by day 23. Furthermore, 100% of animals bearing NCI-H929 multiple myeloma tumor xenografts treated with 800 µCi of anti-CD38 pretargeted (90)Y-DOTA-biotin achieved long-term myeloma-free survival (>70 days) compared with none (0%) of the control animals.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal/therapeutic use , Heterocyclic Compounds/therapeutic use , Molecular Targeted Therapy/methods , Neoplasms, Plasma Cell/radiotherapy , Organometallic Compounds/therapeutic use , Radioimmunotherapy/methods , Animals , Cells, Cultured , Female , Humans , Mice , Mice, Nude , Mice, Transgenic , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/therapeutic use
11.
Blood ; 121(18): 3759-67, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23471305

ABSTRACT

Despite aggressive chemotherapy combined with hematopoietic stem cell transplantation (HSCT), many patients with acute myeloid leukemia (AML) relapse. Radioimmunotherapy (RIT) using monoclonal antibodies labeled with ß-emitting radionuclides has been explored to reduce relapse. ß emitters are limited by lower energies and nonspecific cytotoxicity from longer path lengths compared with α emitters such as (211)At, which has a higher energy profile and shorter path length. We evaluated the efficacy and toxicity of anti-CD45 RIT using (211)At in a disseminated murine AML model. Biodistribution studies in leukemic SJL/J mice showed excellent localization of (211)At-anti-murine CD45 mAb (30F11) to marrow and spleen within 24 hours (18% and 79% injected dose per gram of tissue [ID/g], respectively), with lower kidney and lung uptake (8.4% and 14% ID/g, respectively). In syngeneic HSCT studies, (211)At-B10-30F11 RIT improved the median survival of leukemic mice in a dose-dependent fashion (123, 101, 61, and 37 days given 24, 20, 12, and 0 µCi, respectively). This approach had minimal toxicity with nadir white blood cell counts >2.7 K/µL 2 weeks after HSCT and recovery by 4 weeks. These data suggest that (211)At-anti-CD45 RIT in conjunction with HSCT may be a promising therapeutic option for AML.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Astatine/therapeutic use , Bone Marrow Transplantation , Leukemia/therapy , Leukocyte Common Antigens/immunology , Radioimmunotherapy/methods , Animals , Combined Modality Therapy/methods , Disease Models, Animal , Female , Leukemia/mortality , Leukemia/pathology , Leukemia/radiotherapy , Mice , Neoplasm Metastasis , Survival Analysis , Tissue Distribution , Treatment Outcome , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...