64 results on '"Raubitschek AA"'
Search Results
2. Phase II trial of a transplantation regimen of yttrium-90 ibritumomab tiuxetan and high-dose chemotherapy in patients with non-Hodgkin's lymphoma.
- Author
-
Krishnan A, Nademanee A, Fung HC, Raubitschek AA, Molina A, Yamauchi D, Rodriguez R, Spielberger RT, Falk P, Palmer JM, Forman SJ, Krishnan, Amrita, Nademanee, Auayporn, Fung, Henry C, Raubitschek, Andrew A, Molina, Arturo, Yamauchi, Dave, Rodriguez, Roberto, Spielberger, Ricardo T, and Falk, Peter
- Published
- 2008
3. Estimating residence times and their associated errors in patient absorbed-dose circulation.
- Author
-
Kaplan DD, Williams LE, Clarke KG, Odom-Maryon TL, Liu A, Lopatin G, Raubitschek AA, and Wong JYC
- Published
- 1997
4. A shielded, automated injector for energetic beta-emitting radionuclides.
- Author
-
Williams LE, Ettinger LM, Cofresi E, Raubitschek AA, and Wong JYC
- Published
- 1995
5. Tumor Uptake of 64 Cu-DOTA-Trastuzumab in Patients with Metastatic Breast Cancer.
- Author
-
Mortimer JE, Bading JR, Park JM, Frankel PH, Carroll MI, Tran TT, Poku EK, Rockne RC, Raubitschek AA, Shively JE, and Colcher DM
- Subjects
- Adult, Aged, Biological Transport, Breast Neoplasms diagnostic imaging, Female, Humans, Middle Aged, Neoplasm Metastasis, Positron Emission Tomography Computed Tomography, Receptor, ErbB-2 metabolism, Trastuzumab, Antibodies, Monoclonal, Humanized metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Organometallic Compounds metabolism
- Abstract
The goal of this study was to characterize the relationship between tumor uptake of
64 Cu-DOTA-trastuzumab as measured by PET/CT and standard, immunohistochemistry (IHC)-based, histopathologic classification of human epidermal growth factor receptor 2 (HER2) status in women with metastatic breast cancer (MBC). Methods: Women with biopsy-confirmed MBC and not given trastuzumab for 2 mo or more underwent complete staging, including18 F-FDG PET/CT. Patients were classified as HER2-positive (HER2+) or -negative (HER2-) based on fluorescence in situ hybridization (FISH)-supplemented immunohistochemistry of biopsied tumor tissue. Eighteen patients underwent64 Cu-DOTA-trastuzumab injection, preceded in 16 cases by trastuzumab infusion (45 mg). PET/CT was performed 21-25 (day 1) and 47-49 (day 2) h after64 Cu-DOTA-trastuzumab injection. Radiolabel uptake in prominent lesions was measured as SUVmax Average intrapatient SUVmax (max > pt ) was compared between HER2+ and HER2- patients. Results: Eleven women were HER2+ (8 immunohistochemistry 3+; 3 immunohistochemistry 2+/FISH amplified), whereas 7 were HER2- (3 immunohistochemistry 2+/FISH nonamplified; 4 immunohistochemistry 1+). Medianmax > pt for day 1 and day 2 was 6.6 and 6.8 g/mL for HER 2+ and 3.7 and 4.3 g/mL for HER2- patients ( P < 0.005 either day). The distributions ofmax > pt overlapped between the 2 groups, and interpatient variability was greater for HER2+ than HER2- disease ( P < 0.005 and 0.001, respectively, on days 1 and 2). Conclusion: By 1 d after injection, uptake of64 Cu-DOTA-trastuzumab in MBC is strongly associated with patient HER2 status and is indicative of binding to HER2. The variability within and among HER2+ patients, as well as the overlap between the HER2+ and HER2- groups, suggests a role for64 Cu-DOTA-trastuzumab PET/CT in optimizing treatments that include trastuzumab., (© 2018 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2018
- Full Text
- View/download PDF
6. Low Dose Focused Ultrasound Induces Enhanced Tumor Accumulation of Natural Killer Cells.
- Author
-
Sta Maria NS, Barnes SR, Weist MR, Colcher D, Raubitschek AA, and Jacobs RE
- Subjects
- Adenocarcinoma immunology, Adenocarcinoma pathology, Animals, Carcinoembryonic Antigen immunology, Colorectal Neoplasms immunology, Colorectal Neoplasms pathology, Heterografts, Humans, Killer Cells, Natural, Mice, Mice, Inbred NOD, Neoplasm Transplantation, Adenocarcinoma therapy, Colorectal Neoplasms therapy, High-Energy Shock Waves therapeutic use, Microbubbles therapeutic use
- Abstract
Natural killer (NK) cells play a vital antitumor role as part of the innate immune system. Efficacy of adoptive transfer of NK cells depends on their ability to recognize and target tumors. We investigated whether low dose focused ultrasound with microbubbles (ldbFUS) could facilitate the targeting and accumulation of NK cells in a mouse xenograft of human colorectal adenocarcinoma (carcinoembryonic antigen (CEA)-expressing LS-174T implanted in NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice) in the presence of an anti-CEA immunocytokine (ICK), hT84.66/M5A-IL-2 (M5A-IL-2). Human NK cells were labeled with an FDA-approved ultra-small superparamagnetic iron oxide particle, ferumoxytol. Simultaneous with the intravenous injection of microbubbles, focused ultrasound was applied to the tumor. In vivo longitudinal magnetic resonance imaging (MRI) identified enhanced accumulation of NK cells in the ensonified tumor, which was validated by endpoint histology. Significant accumulation of NK cells was observed up to 24 hrs at the tumor site when ensonified with 0.50 MPa peak acoustic pressure ldbFUS, whereas tumors treated with at 0.25 MPa showed no detectable NK cell accumulation. These clinically translatable results show that ldbFUS of the tumor mass can potentiate tumor homing of NK cells that can be evaluated non-invasively using MRI.
- Published
- 2015
- Full Text
- View/download PDF
7. Functional imaging of human epidermal growth factor receptor 2-positive metastatic breast cancer using (64)Cu-DOTA-trastuzumab PET.
- Author
-
Mortimer JE, Bading JR, Colcher DM, Conti PS, Frankel PH, Carroll MI, Tong S, Poku E, Miles JK, Shively JE, and Raubitschek AA
- Subjects
- Adult, Aged, Breast Neoplasms metabolism, Female, Humans, Liver metabolism, Middle Aged, Neoplasm Metastasis, Radiometry, Reproducibility of Results, Time Factors, Tomography, X-Ray Computed, Trastuzumab, Antibodies, Monoclonal, Humanized, Breast Neoplasms diagnostic imaging, Breast Neoplasms pathology, Copper Radioisotopes, Organometallic Compounds, Positron-Emission Tomography, Receptor, ErbB-2 metabolism
- Abstract
Unlabelled: Women with human epidermal growth factor receptor 2 (HER2)-positive breast cancer are candidates for treatment with the anti-HER2 antibody trastuzumab. Assessment of HER2 status in recurrent disease is usually made by core needle biopsy of a single lesion, which may not represent the larger tumor mass or other sites of disease. Our long-range goal is to develop PET of radiolabeled trastuzumab for systemically assessing tumor HER2 expression and identifying appropriate use of anti-HER2 therapies. The purpose of this study was to evaluate PET/CT of (64)Cu-DOTA-trastuzumab for detecting and measuring tumor uptake of trastuzumab in patients with HER2-positive metastatic breast cancer., Methods: Eight women with biopsy-confirmed HER2-positive metastatic breast cancer and no anti-HER2 therapy for 4 mo or longer underwent complete staging, including (18)F-FDG PET/CT. For 6 of the 8 patients, (64)Cu-DOTA-trastuzumab injection (364-512 MBq, 5 mg of trastuzumab) was preceded by trastuzumab infusion (45 mg). PET/CT (PET scan duration 1 h) was performed 21-25 (day 1) and 47-49 (day 2) h after (64)Cu-DOTA-trastuzumab injection. Scan fields of view were chosen on the basis of (18)F-FDG PET/CT. Tumor detection sensitivity and uptake analyses were limited to lesions identifiable on CT; lesions visualized relative to adjacent tissue on PET were considered PET-positive. Radiolabel uptake in prominent lesions was measured as maximum single-voxel standardized uptake value (SUVmax)., Results: Liver uptake of (64)Cu was reduced approximately 75% with the 45-mg trastuzumab predose, without significant effect on tumor uptake. The study included 89 CT-positive lesions. Detection sensitivity was 77%, 89%, and 93% for day 1, day 2, and (18)F-FDG, respectively. On average, tumor uptake was similar for (64)Cu-DOTA-trastuzumab and (18)F-FDG (SUVmax and range, 8.1 and 3.0-22.5 for day 1 [n = 48]; 8.9 and 0.9-28.9 for day 2 [n = 38]; 9.7 and 3.3-25.4 for (18)F-FDG [n = 56]), but same-lesion SUVmax was not correlated between the 2 radiotracers. No toxicities were observed, and estimated radiation dose from (64)Cu-DOTA-trastuzumab was similar to (18)F-FDG., Conclusion: (64)Cu-DOTA-trastuzumab visualizes HER2-positive metastatic breast cancer with high sensitivity and is effective in surveying disseminated disease. A 45-mg trastuzumab predose provides a (64)Cu-DOTA-trastuzumab biodistribution favorable for tumor imaging. (64)Cu-DOTA-trastuzumab PET/CT warrants further evaluation for assessing tumor HER2 expression and individualizing treatments that include trastuzumab.
- Published
- 2014
- Full Text
- View/download PDF
8. Serial diffusion MRI to monitor and model treatment response of the targeted nanotherapy CRLX101.
- Author
-
Ng TS, Wert D, Sohi H, Procissi D, Colcher D, Raubitschek AA, and Jacobs RE
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Lymphoma drug therapy, Mice, Mice, Nude, Models, Biological, Nanomedicine, Treatment Outcome, Tumor Burden, Xenograft Model Antitumor Assays, Antineoplastic Agents, Phytogenic administration & dosage, Camptothecin administration & dosage, Cyclodextrins administration & dosage, Diffusion Magnetic Resonance Imaging, Lymphoma pathology
- Abstract
Purpose: Targeted nanotherapies are being developed to improve tumor drug delivery and enhance therapeutic response. Techniques that can predict response will facilitate clinical translation and may help define optimal treatment strategies. We evaluated the efficacy of diffusion-weighted magnetic resonance imaging to monitor early response to CRLX101 (a cyclodextrin-based polymer particle containing the DNA topoisomerase I inhibitor camptothecin) nanotherapy (formerly IT-101), and explored its potential as a therapeutic response predictor using a mechanistic model of tumor cell proliferation., Experimental Design: Diffusion MRI was serially conducted following CRLX101 administration in a mouse lymphoma model. Apparent diffusion coefficients (ADCs) extracted from the data were used as treatment response biomarkers. Animals treated with irinotecan (CPT-11) and saline were imaged for comparison. ADC data were also input into a mathematical model of tumor growth. Histological analysis using cleaved-caspase 3, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, Ki-67, and hematoxylin and eosin (H&E) were conducted on tumor samples for correlation with imaging results., Results: CRLX101-treated tumors at day 2, 4, and 7 posttreatment exhibited changes in mean ADC = 16 ± 9%, 24 ± 10%, 49 ± 17%, and size (TV) = -5 ± 3%, -30 ± 4%, and -45 ± 13%, respectively. Both parameters were statistically greater than controls [p(ADC) ≤ 0.02, and p(TV) ≤ 0.01 at day 4 and 7], and noticeably greater than CPT-11-treated tumors (ADC = 5 ± 5%, 14 ± 7%, and 18 ± 6%; TV = -15 ± 5%, -22 ± 13%, and -26 ± 8%). Model-derived parameters for cell proliferation obtained using ADC data distinguished CRLX101-treated tumors from controls (P = 0.02)., Conclusions: Temporal changes in ADC specified early CRLX101 treatment response and could be used to model image-derived cell proliferation rates following treatment. Comparisons of targeted and nontargeted treatments highlight the utility of noninvasive imaging and modeling to evaluate, monitor, and predict responses to targeted nanotherapeutics., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
9. Quantitative, simultaneous PET/MRI for intratumoral imaging with an MRI-compatible PET scanner.
- Author
-
Ng TS, Bading JR, Park R, Sohi H, Procissi D, Colcher D, Conti PS, Cherry SR, Raubitschek AA, and Jacobs RE
- Subjects
- Algorithms, Animals, Antibodies, Autoradiography, Calibration, Copper Radioisotopes, Fluorodeoxyglucose F18, Image Processing, Computer-Assisted, Mice, Mice, Inbred C57BL, Phantoms, Imaging, Radiopharmaceuticals, Reproducibility of Results, Magnetic Resonance Imaging instrumentation, Magnetic Resonance Imaging methods, Neoplasms diagnostic imaging, Neoplasms pathology, Positron-Emission Tomography instrumentation, Positron-Emission Tomography methods
- Abstract
Unlabelled: Noninvasive methods are needed to explore the heterogeneous tumor microenvironment and its modulation by therapy. Hybrid PET/MRI systems are being developed for small-animal and clinical use. The advantage of these integrated systems depends on their ability to provide MR images that are spatially coincident with simultaneously acquired PET images, allowing combined functional MRI and PET studies of intratissue heterogeneity. Although much effort has been devoted to developing this new technology, the issue of quantitative and spatial fidelity of PET images from hybrid PET/MRI systems to the tissues imaged has received little attention. Here, we evaluated the ability of a first-generation, small-animal MRI-compatible PET scanner to accurately depict heterogeneous patterns of radiotracer uptake in tumors., Methods: Quantitative imaging characteristics of the MRI-compatible PET (PET/MRI) scanner were evaluated with phantoms using calibration coefficients derived from a mouse-sized linearity phantom. PET performance was compared with a commercial small-animal PET system and autoradiography in tumor-bearing mice. Pixel and structure-based similarity metrics were used to evaluate image concordance among modalities. Feasibility of simultaneous PET/MRI functional imaging of tumors was explored by following (64)Cu-labeled antibody uptake in relation to diffusion MRI using cooccurrence matrix analysis., Results: The PET/MRI scanner showed stable and linear response. Activity concentration recovery values (measured and true activity concentration) calculated for 4-mm-diameter rods within linearity and uniform activity rod phantoms were near unity (0.97 ± 0.06 and 1.03 ± 0.03, respectively). Intratumoral uptake patterns for both (18)F-FDG and a (64)Cu-antibody acquired using the PET/MRI scanner and small-animal PET were highly correlated with autoradiography (r > 0.99) and with each other (r = 0.97 ± 0.01). On the basis of these data, we performed a preliminary study comparing diffusion MRI and radiolabeled antibody uptake patterns over time and visualized movement of antibodies from the vascular space into the tumor mass., Conclusion: The MRI-compatible PET scanner provided tumor images that were quantitatively accurate and spatially concordant with autoradiography and the small-animal PET examination. Cooccurrence matrix approaches enabled effective analysis of multimodal image sets. These observations confirm the ability of the current simultaneous PET/MRI system to provide accurate observations of intratumoral function and serve as a benchmark for future evaluations of hybrid instrumentation.
- Published
- 2012
- Full Text
- View/download PDF
10. Site-specific conjugation of monodispersed DOTA-PEGn to a thiolated diabody reveals the effect of increasing peg size on kidney clearance and tumor uptake with improved 64-copper PET imaging.
- Author
-
Li L, Crow D, Turatti F, Bading JR, Anderson AL, Poku E, Yazaki PJ, Carmichael J, Leong D, Wheatcroft D, Raubitschek AA, Hudson PJ, Colcher D, and Shively JE
- Subjects
- Animals, Binding Sites, Copper Radioisotopes chemistry, Copper Radioisotopes pharmacokinetics, Female, Kidney diagnostic imaging, Mice, Mice, Nude, Molecular Structure, Neoplasm Transplantation, Neoplasms diagnostic imaging, Tissue Distribution, Heterocyclic Compounds, 1-Ring chemistry, Heterocyclic Compounds, 1-Ring pharmacokinetics, Kidney metabolism, Neoplasms metabolism, Polyethylene Glycols chemistry, Polyethylene Glycols pharmacokinetics, Positron-Emission Tomography, Radiopharmaceuticals chemistry, Radiopharmaceuticals pharmacokinetics, Sulfhydryl Compounds chemistry
- Abstract
Optimal PET imaging of tumors with radiolabeled engineered antibodies requires, among other parameters, matching blood clearance and tumor uptake with the half-life of the engineered antibody. Although diabodies have favorable molecular sizes (50 kDa) for rapid blood clearance (t(1/2) = 30-60 min) and are bivalent, thereby increasing tumor uptake, they exhibit substantial kidney uptake as their major route of clearance, which is especially evident when they are labeled with the PET isotope (64)Cu (t(1/2) = 12 h). To overcome this drawback, diabodies may be conjugated to PEG, a modification that increases the apparent molecular size of the diabody and reduces kidney uptake without adversely affecting tumor uptake or the tumor to blood ratio. We show here that site-specific attachment of monodispersed PEGn of increasing molecular size (n = 12, 24, and 48) can uniformly increase the apparent molecular size of the PEG-diabody conjugate, decrease kidney uptake, and increase tumor uptake, the latter due to the increased residence time of the conjugate in the blood. Since the monodispersed PEGs were preconjugated to the chelator DOTA, the conjugates were able to bind radiometals such as (111)In and (64)Cu that can be used for SPECT and PET imaging, respectively. To allow conjugation of the DOTA-PEG to the diabody, the DOTA-PEG incorporated a terminal cysteine conjugated to a vinyl sulfone moiety. In order to control the conjugation chemistry, we have engineered a surface thiolated diabody that incorporates two cysteines per monomer (four per diabody). The thiolated diabody was expressed and purified from bacterial fermentation and only needs to be reduced prior to conjugation to the DOTA-PEGn-Cys-VS. This novel imaging agent (a diabody with DOTA-PEG48-Cys-VS attached to introduced thiols) gave up to 80%ID/g of tumor uptake with a tumor to blood ratio (T/B) of 8 at 24 h when radiolabeled with (111)In and 37.9% ID/g of tumor uptake (T/B = 8) at 44 h when radiolabeled with (64)Cu in PET imaging in an animal model. Tumor uptake was significantly improved from the 50% ID/g at 24 h observed with diabodies that were pegylated on surface lysine residues. Importantly, there was no loss of immunoreactivity of the site-specific Cys-conjugated diabody to its antigen (TAG-72) compared to the parent, unconjugated diabody. We propose that thiolated diabodies conjugated to DOTAylated monodisperse PEGs have the potential for superior SPECT and PET imaging in a clinical setting.
- Published
- 2011
- Full Text
- View/download PDF
11. Mathematical modeling of chimeric TCR triggering predicts the magnitude of target lysis and its impairment by TCR downmodulation.
- Author
-
James SE, Greenberg PD, Jensen MC, Lin Y, Wang J, Budde LE, Till BG, Raubitschek AA, Forman SJ, and Press OW
- Subjects
- Antigens, CD20 immunology, B-Lymphocyte Subsets immunology, B-Lymphocyte Subsets metabolism, B-Lymphocyte Subsets pathology, Cell Line, Transformed, Cell Line, Tumor, Cell Survival genetics, Cell Survival immunology, Dose-Response Relationship, Immunologic, Humans, Jurkat Cells, Linear Models, Mutant Chimeric Proteins biosynthesis, Predictive Value of Tests, Receptors, Antigen, T-Cell biosynthesis, Sialic Acid Binding Ig-like Lectin 2 immunology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology, Cytotoxicity, Immunologic genetics, Down-Regulation genetics, Down-Regulation immunology, Models, Immunological, Mutant Chimeric Proteins antagonists & inhibitors, Mutant Chimeric Proteins genetics, Receptors, Antigen, T-Cell antagonists & inhibitors, Receptors, Antigen, T-Cell genetics
- Abstract
We investigated relationships among chimeric TCR (cTCR) expression density, target Ag density, and cTCR triggering to predict lysis of target cells by cTCR(+) CD8(+) T human cells as a function of Ag density. Triggering of cTCR and canonical TCR by Ag could be quantified by the same mathematical equation, but cTCR represented a special case in which serial triggering was abrogated. The magnitude of target lysis could be predicted as a function of cTCR triggering, and the predicted minimum cTCR density required for maximal target lysis by CD20-specific cTCR was experimentally tested. cTCR density below approximately 20,000 cTCR/cell impaired target lysis, but increasing cTCR expression above this density did not improve target lysis or Ag sensitivity. cTCR downmodulation to densities below this critical minimum by interaction with Ag-expressing targets limited the sequential lysis of targets in a manner that could be predicted based on the number of cTCRs remaining. In contrast, acute inhibition of lysis of primary, intended targets (e.g., leukemic B cells) due to the presence of an excess of secondary targets (e.g., normal B cells) was dependent on the Ag density of the secondary target but occurred at Ag densities insufficient to promote significant cTCR downmodulation, suggesting a role for functional exhaustion rather than insufficient cTCR density. This suggests increasing cTCR density above a critical threshold may enhance sequential lysis of intended targets in isolation, but will not overcome the functional exhaustion of cTCR(+) T cells encountered in the presence of secondary targets with high Ag density.
- Published
- 2010
- Full Text
- View/download PDF
12. ImmunoPET imaging of B-cell lymphoma using 124I-anti-CD20 scFv dimers (diabodies).
- Author
-
Olafsen T, Sirk SJ, Betting DJ, Kenanova VE, Bauer KB, Ladno W, Raubitschek AA, Timmerman JM, and Wu AM
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Murine-Derived, Dimerization, Female, Humans, Immunologic Factors chemistry, Immunologic Factors immunology, Iodine Radioisotopes, Lymphoma, B-Cell immunology, Mice, Mice, Inbred Strains, Rituximab, Single-Chain Antibodies immunology, Antibodies, Monoclonal chemistry, Lymphoma, B-Cell diagnostic imaging, Positron-Emission Tomography, Single-Chain Antibodies chemistry
- Abstract
Rapid clearing engineered antibody fragments for immunoPET promise high sensitivity at early time points. Here, tumor targeting of anti-CD20 diabodies (scFv dimers) for detection of low-grade B-cell lymphomas were evaluated. In addition, the effect of linker length on oligomerization of the diabody was investigated. Four rituximab scFv variants in the V(L)-V(H) orientation with different linker lengths between the V domains (scFv-1, scFv-3, scFv-5, scFv-8), plus the scFv-5 with a C-terminal cysteine (Cys-Db) for site-specific modification were generated. The scFv-8 and Cys-Db were radioiodinated with (124)I for PET imaging, and biodistribution of (131)I-Cys-Db was carried out at 2, 4 10 and 20 h. The five anti-CD20 scFv variants were expressed as fully functional dimers. Shortening the linker to three or one residue did not produce higher order of multimers. Both (124)I-labeled scFv-8 and Cys-Db exhibited similar tumor targeting at 8 h post injection, with significantly higher uptakes than in control tumors (P < 0.05). At 20 h, less than 1% ID/g of (131)I-labeled Cys-Db was present in tumors and tissues. Specific tumor targeting and high contrast images were achieved with the anti-CD20 diabodies. These agents extend the repertoire of reagents that can potentially be used to improve detection of low-grade lymphomas.
- Published
- 2010
- Full Text
- View/download PDF
13. Minibodies and Multimodal Chromatography Methods: A Convergence of Challenge and Opportunity.
- Author
-
Gagnon P, Cheung CW, Lepin EJ, Wu AM, Sherman MA, Raubitschek AA, and Yazaki PJ
- Abstract
This case study describes early phase purification process development for a recombinant anticancer minibody produced in mammalian cell culture. The minibody did not bind to protein A. Cation-exchange, anion-exchange, hydrophobic-interaction, and hydroxyapatite (eluted by phosphate gradient) chromatographic methods were scouted, but the minibody coeluted with BSA to a substantial degree on each. Hydroxyapatite eluted with a sodium chloride gradient separated BSA and also removed a dimeric contaminant, but BSA consumed so much binding capacity that this proved impractical as a capture tool. Capto MMC media proved capable of supporting adequate capture and significant dimer removal, although both loading and elution selectivity varied dramatically with the amount of supernatant applied to the column. An anion-exchange step was included to fortify overall virus and DNA removal. These results illustrate the value of multimodal chromatography methods when affinity chromatography methods are lacking and conventional alternatives prove inadequate.
- Published
- 2010
14. Recombinant anti-CD20 antibody fragments for small-animal PET imaging of B-cell lymphomas.
- Author
-
Olafsen T, Betting D, Kenanova VE, Salazar FB, Clarke P, Said J, Raubitschek AA, Timmerman JM, and Wu AM
- Subjects
- Animals, Antibodies, Monoclonal, Murine-Derived, Disease Models, Animal, Female, Humans, Immunoglobulin Fragments immunology, Lymphoma, B-Cell immunology, Mice, Mice, Inbred C3H, Reproducibility of Results, Rituximab, Sensitivity and Specificity, Antibodies, Monoclonal immunology, Lymphoma, B-Cell diagnostic imaging, Positron-Emission Tomography methods, Positron-Emission Tomography veterinary, Radiopharmaceuticals immunology
- Abstract
Unlabelled: The CD20 cell surface antigen is expressed at high levels by over 90% of B-cell non-Hodgkin lymphomas (NHL) and is the target of the anti-CD20 monoclonal antibody rituximab. To provide more sensitive, tumor-specific PET imaging of NHL, we sought to develop PET agents targeting CD20., Methods: Two recombinant anti-CD20 rituximab fragments, a minibody (scFv-C(H)3 dimer; 80 kDa) and a modified scFv-Fc fragment (105 kDa), designed to clear rapidly, were generated. Both fragments were radiolabeled with (124)I, and the minibody was additionally labeled with (64)Cu (radiometal) after conjugation to 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA). The radioiodinated fragments and the radiometal-labeled minibody were evaluated in mice as small-animal PET imaging agents for the in vivo imaging of human CD20-expressing lymphomas., Results: Rapid and specific localization to CD20-positive tumors was observed with the radioiodinated fragments. However, the tumor uptake levels and blood activities differed, resulting in different levels of contrast in the images. The better candidate was the minibody, with superior uptake (2-fold higher than that obtained with scFv-Fc) in CD20-positive tumors and low uptake in CD20-negative tumors. Ratios of CD20-positive tumors to CD20-negative tumors at 21 h were 7.0 +/- 3.1 (mean +/- SD) and 3.9 +/- 0.7 for the minibody and scFv-Fc, respectively. The ratio achieved with the (64)Cu-DOTA-minibody at 19 h was about 5-fold lower because of higher residual background activity in CD20-negative tumors., Conclusion: A radioiodinated minibody and a radioiodinated scFv-Fc fragment produced excellent, high-contrast images in vivo. These new immunoPET agents may prove useful for imaging CD20-positive lymphomas in preclinical models and in humans with NHL.
- Published
- 2009
- Full Text
- View/download PDF
15. Rapid and efficient production of radiolabeled antibody conjugates using vacuum diafiltration guided by mathematical modeling.
- Author
-
Bartlett DW, Colcher D, and Raubitschek AA
- Subjects
- Animals, Chelating Agents chemistry, Humans, Models, Theoretical, Vacuum, Antibodies, Monoclonal, Filtration methods, Heterocyclic Compounds, 1-Ring chemistry, Immunoconjugates chemistry, Isotope Labeling methods, Radioimmunotherapy methods, Radiopharmaceuticals chemistry
- Abstract
Increasing interest in the use of radiolabeled antibodies for cancer imaging and therapy drives the need for more efficient production of the antibody conjugates. Here, we illustrate a method for rapid and efficient production of radiolabeled antibody conjugates using vacuum diafiltration guided by mathematical modeling. We apply this technique to the production of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated antibodies at the milligram and gram production scale and achieve radiolabeling efficiencies >95% using In-111. Using vacuum diafiltration, antibody-chelate conjugation and purification can be accomplished within the same vessel, and the entire process can be completed in <24 h. Vacuum diafiltration also offers safer and gentler processing conditions by eliminating the need to keep the retentate vessel under positive pressure through applied gas pressure or shear-inducing restriction points in the retentate flow path. Experimental data and mathematical model calculations suggest there exists a weak binding affinity (approximately 10(4)M(-1)) between the charged chelate molecules (e.g., DOTA) and the antibodies that slows the removal of excess chelate during purification. By analyzing the radiolabeling efficiency as a function of the number of diavolumes, we demonstrate the importance of balancing the removal of free chelate with the introduction of metal contaminants from the diafiltration buffer and also illustrate how to optimize radiolabeling of antibody conjugates under a variety of operating conditions. This methodology is applicable to the production of antibody conjugates in general.
- Published
- 2008
- Full Text
- View/download PDF
16. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane.
- Author
-
James SE, Greenberg PD, Jensen MC, Lin Y, Wang J, Till BG, Raubitschek AA, Forman SJ, and Press OW
- Subjects
- Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cell Membrane chemistry, Epitopes, T-Lymphocyte chemistry, Flow Cytometry, Humans, Leukemia, B-Cell therapy, Lymphoma, B-Cell therapy, Receptors, Antigen, T-Cell genetics, Cytotoxicity, Immunologic, Epitopes, T-Lymphocyte immunology, Immunotherapy methods, Receptors, Antigen, T-Cell chemistry, Receptors, Antigen, T-Cell immunology
- Abstract
We have targeted CD22 as a novel tumor-associated Ag for recognition by human CTL genetically modified to express chimeric TCR (cTCR) recognizing this surface molecule. CD22-specific cTCR targeting different epitopes of the CD22 molecule promoted efficient lysis of target cells expressing high levels of CD22 with a maximum lytic potential that appeared to decrease as the distance of the target epitope from the target cell membrane increased. Targeting membrane-distal CD22 epitopes with cTCR(+) CTL revealed defects in both degranulation and lytic granule targeting. CD22-specific cTCR(+) CTL exhibited lower levels of maximum lysis and lower Ag sensitivity than CTL targeting CD20, which has a shorter extracellular domain than CD22. This diminished sensitivity was not a result of reduced avidity of Ag engagement, but instead reflected weaker signaling per triggered cTCR molecule when targeting membrane-distal epitopes of CD22. Both of these parameters were restored by targeting a ligand expressing the same epitope, but constructed as a truncated CD22 molecule to approximate the length of a TCR:peptide-MHC complex. The reduced sensitivity of CD22-specific cTCR(+) CTL for Ag-induced triggering of effector functions has potential therapeutic applications, because such cells selectively lysed B cell lymphoma lines expressing high levels of CD22, but demonstrated minimal activity against autologous normal B cells, which express lower levels of CD22. Thus, our results demonstrate that cTCR signal strength, and consequently Ag sensitivity, can be modulated by differential choice of target epitopes with respect to distance from the cell membrane, allowing discrimination between targets with disparate Ag density.
- Published
- 2008
- Full Text
- View/download PDF
17. Targeting, imaging, and therapy using a humanized antiprostate stem cell antigen (PSCA) antibody.
- Author
-
Olafsen T, Gu Z, Sherman MA, Leyton JV, Witkosky ME, Shively JE, Raubitschek AA, Morrison SL, Wu AM, and Reiter RE
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal chemistry, Antigens, Neoplasm, Antineoplastic Agents pharmacology, Cell Line, Tumor, GPI-Linked Proteins, Humans, Hybridomas metabolism, Kinetics, Male, Mice, Molecular Sequence Data, Neoplasm Transplantation, Prostatic Neoplasms metabolism, Sequence Homology, Amino Acid, Immunotherapy methods, Membrane Glycoproteins immunology, Neoplasm Proteins immunology, Prostatic Neoplasms pathology
- Abstract
The murine 1G8 (micro1G8) monoclonal antibody directed against prostate stem cell antigen (PSCA) prevents prostate tumor establishment, growth, and metastasis in murine models. To further delineate in vivo targeting properties, micro1G8 was radiolabeled with In-111 and evaluated in nude mice bearing PC3-PSCA xenografts. Tumor activity ranged from 11.8% to 17.1% injected dose per gram (ID/g) at 24 to 96 hours postinjection. To extend the clinical applicability of micro1G8, a chimeric 1G8 antibody was produced that exhibited specific binding to PSCA and significant antitumor effect over micro1G8 in established LAPC-9 prostate cancer xenografts (P=0.0014). However, low expression yields and instability prompted us to humanize 1G8 by grafting the complementary determining regions onto the stable, human Fv framework of anti-p185 4D5v8 (trastuzumab). Two humanized 1G8 (hu1G8) versions (A and B) that differed in the number of murine residues present in the C-terminal half of CDR-H2, were produced. Biacore binding studies demonstrated affinities of 1.47 nM for micro1G8 and 3.74 nM for hu2B3-B, representing a 2.5-fold reduction. Tumor targeting of version B radioiodinated with I was evaluated by serial microPET imaging. Specific tumor targeting of I-hu1G8-B to PC3-PSCA [12.7 (+/-1.6)% ID/g at 94 h] and LAPC-9 [6.6 (+/-0.9)% ID/g at 168 h) xenografts was observed. Inhibition of tumor growth by hu1G8-B was demonstrated in mice bearing low-expressing SW-780-PSCA bladder carcinoma xenografts. In this model, the micro1G8 was ineffective, whereas the hu1G8-B exhibited approximately 50% inhibitory effect. These data support further development of hu1G8 anti-PSCA antibody for targeted imaging and therapy for tumors of urogenital origin.
- Published
- 2007
- Full Text
- View/download PDF
18. Radioiodinated versus radiometal-labeled anti-carcinoembryonic antigen single-chain Fv-Fc antibody fragments: optimal pharmacokinetics for therapy.
- Author
-
Kenanova V, Olafsen T, Williams LE, Ruel NH, Longmate J, Yazaki PJ, Shively JE, Colcher D, Raubitschek AA, and Wu AM
- Subjects
- Animals, Cell Line, Tumor, Colonic Neoplasms metabolism, Female, Heterocyclic Compounds, 1-Ring chemistry, Heterocyclic Compounds, 1-Ring pharmacokinetics, Humans, Immunoglobulin Fragments immunology, Isotope Labeling, Mice, Mice, Inbred BALB C, Mice, Nude, Tissue Distribution, Transplantation, Heterologous, Carcinoembryonic Antigen immunology, Immunoconjugates pharmacokinetics, Immunoglobulin Fragments metabolism, Indium Radioisotopes pharmacokinetics, Iodine Radioisotopes pharmacokinetics, Radiopharmaceuticals pharmacokinetics
- Abstract
Antibody fragments with optimized pharmacokinetic profiles hold potential for detection and therapy of tumor malignancies. We studied the behavior of three anti-carcinoembryonic antigen (CEA) single-chain Fv-Fc (scFv-Fc) variants (I253A, H310A, and H310A/H435Q; Kabat numbering system) that exhibited differential serum persistence. Biodistribution studies done on CEA-positive tumor xenografted mice revealed that the 111In-labeled I253A fragment with the slowest clearance kinetics (T1/2beta, 27.7 h) achieved the highest tumor uptake (44.6% ID/g at 24 h), whereas the radiometal-labeled H310A/H435Q fragment with the most rapid elimination (T1/2beta, 7.05 h) reached a maximum of 28.0% ID/g at 12 h postinjection. The H310A protein was characterized by both intermediate serum half-life and tumor uptake. The 111In-based biodistribution studies showed that all three fragments were eliminated primarily through the liver, and hepatic radiometal activity correlated with the rate of fragment clearance. The 111In-labeled H310A/H435Q protein exhibited the highest liver uptake (23.5% ID/g at 24 h). Metabolism of the 125I-labeled scFv-Fc proteins resulted in low normal organ activity. Finally, the 125I/111In biodistribution data allowed for dose estimations, which suggest the 131I-labeled scFv-Fc H310A/H435Q as a promising candidate for radioimmunotherapy.
- Published
- 2007
- Full Text
- View/download PDF
19. A phase I trial of (90)Y-DOTA-anti-CEA chimeric T84.66 (cT84.66) radioimmunotherapy in patients with metastatic CEA-producing malignancies.
- Author
-
Wong JY, Chu DZ, Williams LE, Liu A, Zhan J, Yamauchi DM, Wilczynski S, Wu AM, Yazaki PJ, Shively JE, Leong L, and Raubitschek AA
- Subjects
- Adult, Aged, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal therapeutic use, Carcinoembryonic Antigen biosynthesis, Female, Heterocyclic Compounds, 1-Ring adverse effects, Heterocyclic Compounds, 1-Ring therapeutic use, Humans, Immunoglobulin G immunology, Immunotoxins adverse effects, Immunotoxins pharmacokinetics, Male, Middle Aged, Neoplasm Metastasis, Neoplasms pathology, Radioimmunotherapy adverse effects, Yttrium Radioisotopes adverse effects, Yttrium Radioisotopes pharmacokinetics, Carcinoembryonic Antigen immunology, Immunotoxins therapeutic use, Neoplasms immunology, Neoplasms radiotherapy, Radioimmunotherapy methods, Yttrium Radioisotopes therapeutic use
- Abstract
Purpose/objective: Previous radioimmunotherapy (RIT) clinical trials at this institution with (90)Y-labeled cT84.66 anti-CEA (carcinoembryonic antigen) evaluated the antibody conjugated to diethylenetriaminepentaacetic acid (DTPA). The aim of this phase I therapy trial was to evaluate cT84.66 conjugated to the macrocyclic chelate (90)Y-DOTA and labeled with (90)Y in a comparable patient population., Experimental Design: Patients with metastatic CEA-producing cancers were entered in this trial. If antibody targeting to tumor was observed after the administration of (111)In-DTPA cT84.66, the patient then received the therapy infusion of (90)Y-DOTA-cT84.66 1 week later. Serial nuclear scans, blood and urine collections, and computed tomography (CT) scans were performed to assess antibody biodistribution, pharmacokinetics, toxicities, and antitumor effects., Results: Thirteen (13) patients were treated in this study. Dose-limiting hematologic toxicity was experienced at initial starting activity levels of 12 and 8 mCi/m(2). Subsequent patients received systemic Ca-DTPA at 125 mg/m(2) every 12 hours for 3 days post-therapy to allow for a dose escalation to 16 mCi/m(2), where hematologic toxicity was observed with an associated maximum tolerated dose (MTD) of 13.4 mCi/m(2). Tumor doses ranged from 4.4 to 569 cGy/mCi, which translated to 97-12,500 cGy after a single infusion of (90)Y-DOTA-cT84.66. Human anti-chimeric antibody (HACA) response developed in 8 of 13 patients and prevented additional therapy in 4 patients., Conclusions: This study demonstrates the feasibility of using (90)Y-DOTA-cT84.66 for antibody-guided radiation therapy. Immunogenicity of the DOTA-conjugated cT84.66 antibody was not appreciably greater than that observed with (90)Y-DTPA-cT84.66 in previous trials. Dose-limiting hematopoietic toxicity with (90)Y-DOTA-cT84.66 decreased with Ca-DTPA infusions post-therapy and appears to be comparable to previously published results for (90)Y-DTPA-cT84.66. The highest antibody uptake and tumor doses were to small nodal lesions, which supports the predictions from preclinical and clinical data that RIT may be best applied in the minimal tumor burden setting.
- Published
- 2006
- Full Text
- View/download PDF
20. Optimizing radiolabeled engineered anti-p185HER2 antibody fragments for in vivo imaging.
- Author
-
Olafsen T, Kenanova VE, Sundaresan G, Anderson AL, Crow D, Yazaki PJ, Li L, Press MF, Gambhir SS, Williams LE, Wong JY, Raubitschek AA, Shively JE, and Wu AM
- Subjects
- Animals, Breast Neoplasms diagnostic imaging, Breast Neoplasms immunology, Breast Neoplasms metabolism, Carcinoembryonic Antigen immunology, Copper Radioisotopes pharmacokinetics, Female, Humans, Immunoconjugates chemistry, Immunoglobulin Fragments chemistry, Indium Radioisotopes chemistry, Indium Radioisotopes pharmacokinetics, Kidney diagnostic imaging, Kidney immunology, Kidney metabolism, Mice, Mice, Nude, Neoplasms immunology, Neoplasms metabolism, Positron-Emission Tomography, Radiopharmaceuticals chemistry, Radiopharmaceuticals pharmacokinetics, Receptor, ErbB-2 chemistry, Receptor, ErbB-2 metabolism, Tissue Distribution, Immunoconjugates pharmacokinetics, Immunoglobulin Fragments metabolism, Neoplasms diagnostic imaging, Receptor, ErbB-2 immunology
- Abstract
We have recently described the in vivo properties of an iodinated anti-p185HER2 engineered antibody fragment [minibody (scFv-C(H)3)2; 80 kDa], made from the internalizing 10H8 monoclonal antibody. Although the 10H8 minibody showed excellent binding to the target in vitro, only modest tumor uptake [5.6 +/- 1.7% injected dose per gram (ID/g) of tissue] was achieved in nude mice bearing MCF7/HER2 breast cancer tumors. Here, in an attempt to improve targeting, the 10H8 minibody was conjugated to 1,4,7,10-tetraazacyclododecane-N, N', N'', N'''-tetraacetic acid (DOTA), radiometal labeled, and evaluated in vivo. The tumor uptake of 111In-DOTA 10H8 minibody was 5.7 +/- 0.1% ID/g, similar to the radioiodinated 10H8 minibody. However, in addition to the expected liver clearance, the kidneys had unexpectedly high activity (34.0 +/- 4.0% ID/g). A minibody derived from a second anti-p185(HER2) antibody (trastuzumab; hu4D5v8) was also made. Tumor uptakes, evaluated by quantitative microPET using 64Cu-DOTA hu4D5v8 minibody, were 4.2 +/- 0.5% ID/g. Furthermore, in non-tumor-bearing mice, 111In-DOTA hu4D5v8 minibody exhibited similar elevated uptake in the kidneys (28.4 +/- 6.5% ID/g). Immunohistochemical staining of kidneys from non-tumor-bearing mice showed strong specific staining of the proximal tubules, and Western blot analysis of kidney lysate confirmed the presence of cross-reactive antigen. To further improve tumor uptake and normal tissue distribution, a larger hu4D5v8 fragment [(scFv-C(H)2-C(H)3)2; 105 kDa] was made, engineered to exhibit rapid clearance kinetics. This fragment, when evaluated by microPET, exhibited improved tumor targeting (12.2 +/- 2.4% ID/g) and reduced kidney uptake (13.1 +/- 1.5% ID/g). Thus, by manipulating the size and format of anti-p185(HER2) antibody fragments, the kidney activity was reduced and high or low expression of p185HER2 in xenografts could be distinguished by microPET imaging.
- Published
- 2005
- Full Text
- View/download PDF
21. Tailoring the pharmacokinetics and positron emission tomography imaging properties of anti-carcinoembryonic antigen single-chain Fv-Fc antibody fragments.
- Author
-
Kenanova V, Olafsen T, Crow DM, Sundaresan G, Subbarayan M, Carter NH, Ikle DN, Yazaki PJ, Chatziioannou AF, Gambhir SS, Williams LE, Shively JE, Colcher D, Raubitschek AA, and Wu AM
- Subjects
- Animals, Carcinoembryonic Antigen metabolism, Cell Line, Tumor, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms metabolism, Female, Humans, Immunoglobulin Fragments biosynthesis, Immunoglobulin Fragments isolation & purification, Mice, Mice, Inbred BALB C, Mice, Nude, Multiple Myeloma metabolism, Positron-Emission Tomography, Tissue Distribution, Transplantation, Heterologous, Carcinoembryonic Antigen immunology, Immunoconjugates pharmacokinetics, Immunoglobulin Fragments metabolism, Iodine Radioisotopes pharmacokinetics, Radiopharmaceuticals pharmacokinetics
- Abstract
Antibody fragments are recognized as promising vehicles for delivery of imaging and therapeutic agents to tumor sites in vivo. The serum persistence of IgG1 and fragments with intact Fc region is controlled by the protective neonatal Fc receptor (FcRn) receptor. To modulate the half-life of engineered antibodies, we have mutated the Fc-FcRn binding site of chimeric anti-carcinoembryonic antigen (CEA) antibodies produced in a single-chain Fv-Fc format. The anti-CEA T84.66 single-chain Fv-Fc format wild-type and five mutants (I253A, H310A, H435Q, H435R, and H310A/H435Q, Kabat numbering system) expressed well in mammalian cell culture. After purification and characterization, effective in vitro antigen binding was shown by competition ELISA. Biodistribution studies in BALB/c mice using (125)I- and (131)I-labeled fragments revealed blood clearance rates from slowest to fastest as follows: wild-type > H435R > H435Q > I253A > H310A > H310A/H435Q. The terminal half-lives of the mutants ranged from 83.4 to 7.96 hours, whereas that of the wild-type was approximately 12 days. Additionally, (124)I-labeled wild-type, H435Q, I253A, H310A, and H310A/H435Q variants were evaluated in LS174T xenografted athymic mice by small animal positron emission tomography imaging, revealing localization to the CEA-positive xenografts. The slow clearing wild-type and H435Q constructs required longer to localize to the tumor and clear from the circulation. The I253A and H310A fragments showed intermediate behavior, whereas the H310A/H435Q variant quickly localized to the tumor site, rapidly cleared from the animal circulation and produced clear images. Thus, attenuating the Fc-FcRn interaction provides a way of controlling the antibody fragment serum half-life without compromising expression and tumor targeting.
- Published
- 2005
22. Pilot trial evaluating an 123I-labeled 80-kilodalton engineered anticarcinoembryonic antigen antibody fragment (cT84.66 minibody) in patients with colorectal cancer.
- Author
-
Wong JY, Chu DZ, Williams LE, Yamauchi DM, Ikle DN, Kwok CS, Liu A, Wilczynski S, Colcher D, Yazaki PJ, Shively JE, Wu AM, and Raubitschek AA
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Chromatography, High Pressure Liquid, Dimerization, Female, Humans, Kinetics, Male, Mice, Middle Aged, Neoplasm Metastasis, Pilot Projects, Protein Structure, Tertiary, Radiometry, Time Factors, Tomography, Emission-Computed, Single-Photon, Antibodies chemistry, Antibodies therapeutic use, Carcinoembryonic Antigen immunology, Colorectal Neoplasms therapy, Immunoglobulin Fragments therapeutic use, Immunotherapy methods, Iodine Radioisotopes therapeutic use
- Abstract
Purpose: The chimeric T84.66 (cT84.66) minibody is a novel engineered antibody construct (V(L)-linker-V(H)-C(H)3; 80 kDa) that demonstrates bivalent and high affinity (4 x 10(10) m(-1)) binding to carcinoembryonic antigen (CEA). The variable regions (V(L) and V(H)) assemble to form the antigen-combining sites, and the protein forms dimers through self-association of the C(H)3 domains. In animal models, the minibody demonstrated high tumor uptake, approaching that of some intact antibodies, substantially faster clearance than intact chimeric T84.66, and superior tumor-to-blood ratios compared with the cT84.66 F(ab')(2) fragment, making it attractive for further evaluation as an imaging and therapy agent. The purpose of this pilot clinical study was to determine whether (123)I-cT84.66 minibody demonstrated tumor targeting and was well tolerated as well as to begin to evaluate its biodistribution, pharmacokinetics, and immunogenicity in patients with colorectal cancer., Experimental Design: Ten patients with biopsy-proven colorectal cancer (6 newly diagnosed, 1 pelvic recurrence, 3 limited metastatic disease) were entered on this study. Each received 5-10 mCi (1 mg) of (123)I-labeled minibody i.v. followed by serial nuclear scans and blood and urine sampling over the next 48-72 h. Surgery was performed immediately after the last nuclear scan., Results: Tumor imaging was observed with (123)I-labeled minibody in seven of the eight patients who did not receive neoadjuvant therapy before surgery. Two patients received neoadjuvant radiation and chemotherapy, which significantly reduced tumor size before surgery and minibody infusion. At surgery, no tumor was detected in one patient and only a 2-mm focus was seen in the second patient. (123)I-labeled minibody tumor targeting was not seen in either of these pretreated patients. Mean serum residence time of the minibody was 29.8 h (range, 10.9-65.4 h). No drug-related adverse reactions were observed. All 10 patients were evaluated for immune responses to the minibody, with no significant responses observed., Conclusion: This pilot study represents one of the first clinical efforts to evaluate an engineered intermediate-molecular-mass radiolabeled antibody construct directed against CEA. cT84.66 minibody demonstrates tumor targeting to colorectal cancer and a faster clearance in comparison with intact antibodies, making it appropriate for further evaluation as an imaging and therapy agent. The mean residence time of the minibody in patients is longer than predicted from murine models. We therefore plan to further evaluate its biodistribution and pharmacokinetic properties with minibody labeled with a longer-lived radionuclide, such as (111)In.
- Published
- 2004
- Full Text
- View/download PDF
23. Humanization of the anti-CEA T84.66 antibody based on crystal structure data.
- Author
-
Yazaki PJ, Sherman MA, Shively JE, Ikle D, Williams LE, Wong JY, Colcher D, Wu AM, and Raubitschek AA
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal genetics, Humans, Immunoglobulin Constant Regions genetics, Immunoglobulin Constant Regions immunology, Indium Radioisotopes, Mice, Molecular Sequence Data, Protein Structure, Tertiary, Sequence Alignment, Antibodies, Monoclonal immunology, Carcinoembryonic Antigen immunology
- Abstract
Chimeric T84.66 (cT84.66) is a monoclonal antibody (mAb) of high specificity and affinity for the tumor-associated carcinoembryonic antigen (CEA). Radiolabeled cT84.66 has demonstrated utility in the clinic as a reagent for the radioimmunoscintigraphy and radioimmunotherapy of CEA-positive colorectal and breast malignancies. To extend the therapeutic efficacy of T84.66, humanization by complementary determining region (CDR) grafting was employed. CDR grafting is a well-established technique, though often a series of framework back-mutations is required to restore high affinity. Recently, the crystal structure of the T84.66 diabody (scFv dimer) derived from the murine T84.66 mAb was determined, facilitating the humanization process by the availability of crystal structure data for both the graft donor and graft acceptor. A search of the Protein Data Bank revealed close structural similarity (r.m.s.d. of 1.07 A) between the Fv of T84.66 and the Fv of 4D5v8, a humanized anti-p185HER2 antibody marketed as Herceptin (Trastuzumab). This resulted in two humanized versions of the T84.66 M5A and M5B mAbs that differed only in the number of murine residues present in the C-terminal half of CDR-H2. Biochemical analysis and animal biodistribution studies were conducted to evaluate the humanized mAbs. The M5A, M5B and cT84.66 mAbs showed sub-nanomolar affinity for CEA and as radiolabeled mAbs exhibited specific tumor localization in tumor bearing mice. The T84.66 M5A mAb was selected for clinical development due to a slightly higher tumor uptake and a larger content of human residues, and was renamed hT84.66. A limited-scale production and animal imaging study have demonstrated hT84.66's ability to support clinical trials. Planned clinical trials will determine the effective utilization of this structure-based approach in the development of a promising new therapeutic.
- Published
- 2004
- Full Text
- View/download PDF
24. Characterization of engineered anti-p185HER-2 (scFv-CH3)2 antibody fragments (minibodies) for tumor targeting.
- Author
-
Olafsen T, Tan GJ, Cheung CW, Yazaki PJ, Park JM, Shively JE, Williams LE, Raubitschek AA, Press MF, and Wu AM
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Tumor, Flow Cytometry, Humans, Immunohistochemistry, Mice, Mice, Nude, Molecular Sequence Data, Immunoglobulin Fragments chemistry, Immunoglobulin Fragments therapeutic use, Mammary Neoplasms, Experimental therapy, Protein Engineering, Receptor, ErbB-2 immunology
- Abstract
An engineered antibody fragment (minibody; scFv-C(H)3gamma(1) dimer, M(r) 80 000) specific for carcinoembryonic antigen (CEA) has previously demonstrated excellent tumor targeting coupled with rapid clearance in vivo. In this study, variable (V) genes from the anti- p185(HER-2) 10H8 antibody were similarly assembled and expressed. Four constructs were made: first, the V genes were assembled in both orientations (V(L)-linker-V(H) and V(H)-linker-V(L)) as single chain Fvs (scFvs). Then each scFv was fused to the human IgG1 C(H)3 domain, either by a two amino acid linker (ValGlu) that resulted in a non-covalent, hingeless minibody, or by IgG1 hinge and a GlySer linker peptide to produce a covalent, hinge-minibody. The constructs, expressed in NS0 mouse myeloma cells at levels of 20-60 mg/l, demonstrated binding to the human p185(HER-2) overexpressing breast cancer cell line, MCF7/HER2. Binding affinities (K(D) approximately 2-4 nM) were equivalent to that for the parental 10H8 mAb (K(D) approximately 1.6 nM). Radioiodinated 10H8 hinge-minibody was evaluated in athymic mice, bearing MCF7/HER2 xenografts. Maximum tumor uptake was 5.6 (+/-1.65)% injected dose/g (ID/g) at 12 h, which was lower than that of the anti-CEA minibody, whereas the blood clearance (beta-phase, 5.62 h) was similar. Thus, minibodies with different specificities display similar pharmacokinetics, while tumor uptake may vary depending on the antigen-antibody system.
- Published
- 2004
- Full Text
- View/download PDF
25. Covalent disulfide-linked anti-CEA diabody allows site-specific conjugation and radiolabeling for tumor targeting applications.
- Author
-
Olafsen T, Cheung CW, Yazaki PJ, Li L, Sundaresan G, Gambhir SS, Sherman MA, Williams LE, Shively JE, Raubitschek AA, and Wu AM
- Subjects
- Animals, Antibodies chemistry, Base Sequence, Binding, Competitive, Cysteine chemistry, Dimerization, Electrophoresis, Polyacrylamide Gel, Female, Mice, Mice, Nude, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Neoplasm Transplantation, Neoplasms metabolism, Peptides chemistry, Protein Structure, Tertiary, Sulfhydryl Compounds, Time Factors, Tomography, Emission-Computed, Carcinoembryonic Antigen chemistry, Disulfides
- Abstract
An engineered anti-carcinoembryonic antigen (CEA) diabody (scFv dimer, 55 kDa) was previously constructed from the murine anti-CEA T84.66 antibody. Tumor targeting, imaging and biodistribution studies in nude mice bearing LS174T xenografts with radiolabeled anti-CEA diabody demonstrated rapid tumor uptake and fast blood clearance, which are favorable properties for an imaging agent. Current radiolabeling approaches result in random modification of the protein surface, which may impair immunoreactivity especially for smaller antibody fragments. Site-specific conjugation approaches can direct modifications to reactive groups located away from the binding site. Here, cysteine residues were introduced into the anti-CEA diabody at three different locations, to provide specific thiol groups for chemical modification. One version (with a C-terminal Gly-Gly-Cys) existed exclusively as a disulfide-bonded dimer. This cysteine-modified diabody (Cys-diabody) retained high binding to CEA and demonstrated tumor targeting and biodistribution properties identical to the non-covalent diabody. Furthermore, following reduction of the disulfide bond, the Cys-diabody could be chemically modified using a thiol-specific bifunctional chelating agent, for radiometal labeling. Thus, the Cys-diabody provides a covalently linked alternative to conventional diabodies, which can be reduced and modified site-specifically. This format will provide a versatile platform for targeting a variety of agents to CEA-positive tumors.
- Published
- 2004
- Full Text
- View/download PDF
26. 124I-labeled engineered anti-CEA minibodies and diabodies allow high-contrast, antigen-specific small-animal PET imaging of xenografts in athymic mice.
- Author
-
Sundaresan G, Yazaki PJ, Shively JE, Finn RD, Larson SM, Raubitschek AA, Williams LE, Chatziioannou AF, Gambhir SS, and Wu AM
- Subjects
- Animals, Cell Line, Tumor, Colorectal Neoplasms metabolism, Female, Humans, Immunoconjugates pharmacokinetics, Isotope Labeling methods, Metabolic Clearance Rate, Mice, Mice, Nude, Neoplasm Transplantation, Protein Engineering methods, Quality Control, Radioimmunoassay methods, Radiopharmaceuticals pharmacokinetics, Tissue Distribution, Transplantation, Heterologous, Antibodies, Monoclonal pharmacokinetics, Antigens metabolism, Carcinoembryonic Antigen metabolism, Colorectal Neoplasms diagnostic imaging, Glioma diagnostic imaging, Iodine Radioisotopes pharmacokinetics, Tomography, Emission-Computed methods
- Abstract
Unlabelled: Prolonged clearance kinetics have hampered the development of intact antibodies as imaging agents, despite their ability to effectively deliver radionuclides to tumor targets in vivo. Genetically engineered antibody fragments display rapid, high-level tumor uptake coupled with rapid clearance from the circulation in the athymic mouse/LS174T xenograft model. The anticarcinoembryonic antigen (CEA) T84.66 minibody (single-chain Fv fragment [scFv]-C(H)3 dimer, 80 kDa) and T84.66 diabody (noncovalent dimer of scFv, 55 kDa) exhibit pharmacokinetics favorable for radioimmunoimaging. The present work evaluated the minibody or diabody labeled with (124)I, for imaging tumor-bearing mice using a high-resolution small-animal PET system., Methods: Labeling was conducted with 0.2-0.3 mg of protein and 65-98 MBq (1.7-2.6 mCi) of (124)I using an iodination reagent. Radiolabeling efficiencies ranged from 33% to 88%, and immunoreactivity was 42% (diabody) or >90% (minibody). In vivo distribution was evaluated in athymic mice bearing paired LS174T human colon carcinoma (CEA-positive) and C6 rat glioma (CEA-negative) xenografts. Mice were injected via the tail vein with 1.9-3.1 MBq (53-85 microCi) of (124)I-minibody or with 3.1 MBq (85 microCi) of (124)I-diabody and imaged at 4 and 18 h by PET. Some mice were also imaged using (18)F-FDG 2 d before imaging with (124)I-minibody., Results: PET images using (124)I-labeled minibody or diabody showed specific localization to the CEA-positive xenografts and relatively low activity elsewhere in the mice, particularly by 18 h. Target-to-background ratios for the LS174T tumors versus soft tissues using (124)I-minibody were 3.05 at 4 h and 11.03 at 18 h. Similar values were obtained for the (124)I-diabody (3.95 at 4 h and 10.93 at 18 h). These results were confirmed by direct counting of tissues after the final imaging. Marked reduction of normal tissue activity, especially in the abdominal region, resulted in high-contrast images at 18 h for the (124)I-anti-CEA diabody. CEA-positive tumors as small as 11 mg (<3 mm in diameter) could be imaged, and (124)I-anti-CEA minibodies, compared with (18)F-FDG, demonstrated highly specific localization., Conclusion: (124)I labeling of engineered antibody fragments provides a promising new class of tumor-specific probes for PET imaging of tumors and metastases.
- Published
- 2003
27. A Phase I trial of 90Y-anti-carcinoembryonic antigen chimeric T84.66 radioimmunotherapy with 5-fluorouracil in patients with metastatic colorectal cancer.
- Author
-
Wong JY, Shibata S, Williams LE, Kwok CS, Liu A, Chu DZ, Yamauchi DM, Wilczynski S, Ikle DN, Wu AM, Yazaki PJ, Shively JE, Doroshow JH, and Raubitschek AA
- Subjects
- Antimetabolites, Antineoplastic therapeutic use, Colorectal Neoplasms metabolism, Colorectal Neoplasms secondary, Combined Modality Therapy, Feasibility Studies, Humans, Maximum Tolerated Dose, Antibodies, Monoclonal therapeutic use, Carcinoembryonic Antigen immunology, Colorectal Neoplasms therapy, Fluorouracil therapeutic use, Radioimmunotherapy, Recombinant Fusion Proteins therapeutic use, Yttrium Radioisotopes therapeutic use
- Abstract
Purpose: Targeted systemic radiation therapy using radiolabeled antibodies results in tumor doses sufficient to produce significant objective responses in the radiosensitive hematological malignancies. Although comparable doses to tumor are achieved with radioimmunotherapy (RIT) in solid tumors, results have been modest primarily because of their relative lack of radiosensitivity. For solid tumors, as with external beam radiotherapy, RIT should have a more important clinical role if combined with other systemic, potentially radiation-enhancing chemotherapy agents and if used as consolidative therapy in the minimal tumor burden setting. The primary objective of this trial was to evaluate the feasibility and toxicities of systemic 90Y-chimeric T84.66 (cT84.66) anti-carcinoembryonic antigen RIT in combination with continuous infusion 5-fluorouracil (5-FU)., Experimental Design: Patients with chemotherapy-refractory metastatic colorectal cancer were entered. The study was designed for each patient to receive 90Y-cT84.66 anti-carcinoembryonic antigen at 16.6 mCi/m2 as an i.v. bolus infusion combined with 5-FU delivered as a 5-day continuous infusion initiated 4 h before antibody infusion. Cohorts of patients were entered at 5-FU dose levels of 700, 800, 900, and 1000 mg/m2/day. Upon reaching the highest planned dose level of 5-FU, a final cohort received 90Y-cT84.66 at 20.6 mCi/m2 and 5-FU at 1000 mg/m2/day. For all patients, Ca-diethylenetriaminepentaacetic acid at 125 mg/m2 every 12 h was administered for the first 72 h after 90Y-cT84.66. Patients were eligible to receive up to three cycles of 90Y-cT84.66/5-FU every 6 weeks., Results: Twenty-one patients were treated on this study. All had been heavily pretreated with 19 having previously received 5-FU and 16 having failed two to four chemotherapy regimens. A maximum-tolerated dose of 16.6 mCi/m2 90Y-cT84.66 combined with 1000 mg/m2/day 5-FU was reached. These dose levels are comparable with maximum-tolerated dose levels of each agent alone. Thirteen patients received one cycle and 8 patients two cycles of therapy. Hematopoietic toxicity was dose-limiting and reversible. RIT did not appear to increase nonhematopoietic toxicities associated with 5-FU. Two of 19 patients assayed developed a human anti-chimeric antibody immune response after the first cycle of therapy, which is significantly less than that observed in a previous trial evaluating 90Y-cT84.66 alone. No objective responses were observed. However, 11 patients with progressive disease entering the study demonstrated radiological stable disease of 3-8 months duration and 1 patient demonstrated a mixed response., Conclusions: Results from this trial are encouraging and demonstrate the feasibility and possible advantages of combining continuous infusion 5-FU with 90Y-cT84.66 RIT. The addition of 5-FU does not appear to significantly enhance hematological toxicities of the radiolabeled antibody. In addition, 5-FU reduces the development of human anti-chimeric antibody response, permitting multicycle therapy in a larger number of patients. Future efforts should continue to focus on integrating radiation therapy delivered by radiolabeled antibodies into established 5-FU regimens.
- Published
- 2003
28. Reduction of kidney uptake in radiometal labeled peptide linkers conjugated to recombinant antibody fragments. Site-specific conjugation of DOTA-peptides to a Cys-diabody.
- Author
-
Li L, Olafsen T, Anderson AL, Wu A, Raubitschek AA, and Shively JE
- Subjects
- Animals, Colonic Neoplasms pathology, Cross-Linking Reagents chemistry, Drug Design, Humans, Immunoconjugates administration & dosage, Immunoconjugates chemistry, Immunoglobulin Fragments chemistry, Indium Radioisotopes administration & dosage, Indium Radioisotopes pharmacokinetics, Mice, Mice, Nude, Neoplasm Transplantation, Organ Specificity, Peptides chemistry, Radiopharmaceuticals administration & dosage, Radiopharmaceuticals chemical synthesis, Recombinant Proteins chemistry, Structure-Activity Relationship, Tissue Distribution, Transplantation, Heterologous, Immunoconjugates pharmacokinetics, Kidney metabolism, Radiopharmaceuticals pharmacokinetics
- Abstract
Arano and co-workers (Arano et al. (1999) Cancer Res. 59, 128-134) have synthesized peptides with an N-terminal radioiodinated hippuric acid and a C-terminal lysine linked to antibody fragments via the epsilon-amino group of lysine that show reduced kidney uptake compared to antibody fragments directly radioiodinated. This approach takes advantage of the lysine specific carboxypeptidase activity of the kidney brush border enzymes that cleave off the radiolabeled peptide linker from the antibody fragment prior to uptake by proximal tubule cells. On the basis of their approach, we have synthesized a tetrapeptide with an N-terminal DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and a C-terminal (N(epsilon)-maleoyl)lysine that was site-specifically conjugated to an anti-CEA diabody (Yazaki et al. (2001) Bioconjugate Chem. 12, 220-228) that was engineered to contain a C-terminal cysteine (Cys-diabody). Biodistributions of the In-111-radiolabeled conjugate in nude mice show significantly reduced kidney uptake (a maximum of 82%ID/g at 6 h) compared to In-111 radiolabeled DOTA-diabody (184%ID/g at 6 h) in which DOTA was conjugated to endogenous lysine residues using DOTA-active ester chemistry. To further reduce kidney uptake, a homologous compound with a C-terminal (N(epsilon)-amino-1,6-hexane-bis-vinyl sulfone)lysine was synthesized and site-specifically conjugated to the Cys-diabody. Biodistributions of this In-111-labeled conjugate reduced kidney uptake to 54%ID/g at 6 h. To explore the effect of the relative positions of the chelate vs the cys-diabody on kidney uptake, we also synthesized a tetrapeptide with an N-terminal bromoacetate for conjugation to Cys-diabody and a C-terminal (N(epsilon)-amidino-propyl-3-thio-vinylsulfonyl-DO3A)lysine. This peptide essentially reverses the positions of the chelate and Cys-diabody attachment points on the peptide, while retaining the linker length on the epsilon-amino group of the lysine. In this case, biodistributions of the In-111-radiolabeled conjugate in nude mice showed high kidney uptake (189%ID/g at 6 h), comparable to that obtained with the In-111-radiolabeled active ester conjugated DOTA-diabody (184%ID/g at 6 h). We conclude that the peptide linker strategy of Arano and co-workers to reduce kidney uptake can be successfully applied to chelate/radiometal complexes and requires that the chelate/radiometal be located at the N-terminus of the peptide and the antibody fragment attachment site on the epsilon-amino group of the lysine. Furthermore, we demonstrated a role for the attachment chemistry to the epsilon-amino group of the lysine on the magnitude of kidney uptake.
- Published
- 2002
- Full Text
- View/download PDF
29. The two types of correction of absorbed dose estimates for internal emitters.
- Author
-
Williams LE, Liu A, Yamauchi DM, Lopatin G, Raubitschek AA, and Wong JY
- Subjects
- Absorption, Humans, Phantoms, Imaging, Radiation Dosage, Radioimmunotherapy methods
- Abstract
Background: Two types of correction for absorbed dose (D) estimates are described for clinical applications of internal emitters. The first is appropriate for legal and scientific reasons involving phantom-based estimates; the second is patient-specific and primarily intended for radioimmunotherapy (RIT)., Methods: The Medical Internal Radiation Dose (MIRD) relationship (D) = S A is used, where S is a geometric matrix factor and A is the integral of source organ activities. Internal consistency of the data and the size of organ systems in the humanoid phantom must be assured in both types of estimation., Results: The first dose estimate correction (I) is one whereby computations refer to one or another standard (e.g., MIRD-type) phantom. In this case the S value remains as given, but the measured patient A data must be standardized. The correction factor is the phantom's ratio of organ mass to whole-body mass divided by the same ratio for the volunteer or patient. The second dose estimate correction (II) is patient-specific. While the A value is unchanged for this application, a correction term is provided for the phantom-derived S matrix. The dominant (nonpenetrating radiation) component of this correction factor can be obtained via the ratio of the patient to phantom organ masses. In both corrections, we recommend that true organ sizes, necessary in each method of estimation, be determined in a separate sequence of anatomic images., Conclusions: In both dose estimation corrections, true sizes of the patient's or volunteer's internal organs must be obtained. Correction due to organ mass size can be severalfold and is probably the dominant uncertainty in the internal emitter absorbed dose calculation process., (Copyright 2002 American Cancer Society.)
- Published
- 2002
- Full Text
- View/download PDF
30. Vinyl sulfone bifunctional derivatives of DOTA allow sulfhydryl- or amino-directed coupling to antibodies. Conjugates retain immunoreactivity and have similar biodistributions.
- Author
-
Li L, Tsai SW, Anderson AL, Keire DA, Raubitschek AA, and Shively JE
- Subjects
- Animals, Antibodies immunology, Chelating Agents chemistry, Cysteine chemistry, Drug Stability, Indium Radioisotopes chemistry, Isotope Labeling, Mice, Mice, Nude, Tissue Distribution, Amino Acids chemistry, Antibodies chemistry, Heterocyclic Compounds, 1-Ring chemistry, Heterocyclic Compounds, 1-Ring immunology, Sulfhydryl Compounds chemistry, Sulfones chemistry
- Abstract
We have synthesized a bifunctional vinyl sulfone-cysteineamido derivative of DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) that can be conjugated to the sulfhydryls of mildly reduced recombinant antibody (chimeric anti-CEA antibody cT84.66) at pH 7 or to the amino groups of lysine residues at pH 9. The conjugation is sulfhydryl specific at pH 7 (case 1), and amino specific at pH 9 (case 2) as long as the antibody has no free sulhydryl groups. At a molar ratio of 50 BCA (bifunctional chelating agent) to mAb, the number of chelates conjugated is 0.8 for case 1, and 4.6 for case 2. The resulting conjugates can be radiolabeled with (111)In to high specific activity (5 mCi/mg) with high efficiency (>95%) at 43 degrees C in 60 min. The radiolabeled conjugates retained >95% immunoreactivity and are stable in serum containing 1mM DTPA over 3 d. When the radiolabeled conjugates were injected into nude mice bearing LS174T human colon tumor xenografts, over 40% ID/g accumulated in tumors during the period 24-72h. Tumor-to-blood ratios were 4.5, 3.5, and 2.5 for the sulfhydryl coupled conjugate at 24, 48, and 72 h, respectively, and 2.7, 2.5, and 2.3 for the amino-coupled conjugate at the same time points. For other organs the biodistributions were nearly identical whether the conjugates were attached via sulfhydryl or amino groups. These novel BCAs are easy to synthesize, offer versatile conjugation options, and give equivalent biodistributions that result in high tumor uptake and good tumor-to-blood ratios.
- Published
- 2002
- Full Text
- View/download PDF
31. Multimerization of a chimeric anti-CD20 single-chain Fv-Fc fusion protein is mediated through variable domain exchange.
- Author
-
Wu AM, Tan GJ, Sherman MA, Clarke P, Olafsen T, Forman SJ, and Raubitschek AA
- Subjects
- Amino Acid Sequence, Animals, Antibodies chemistry, Antibodies genetics, Antibodies isolation & purification, Antigens, CD20 chemistry, Cloning, Molecular, Dimerization, Immunoglobulin Fragments genetics, Immunoglobulin Fragments immunology, Mice, Molecular Sequence Data, Papain, Pepsin A, Protein Conformation, Protein Isoforms, Protein Structure, Tertiary, Recombinant Fusion Proteins chemistry, Tumor Cells, Cultured, Antibodies immunology, Antigens, CD20 immunology
- Abstract
A series of single-chain anti-CD20 antibodies was produced by fusing single-chain Fv (scFv) with human IgG1 hinge and Fc regions, designated scFv-Fc. The initial scFv-Fc construct was assembled using an 18 amino acid (aa) linker between the antibody light- and heavy-chain variable regions, with the Cys residue in the upper hinge region (Kabat 233) mutagenized to Ser. Anti-CD20 scFv-Fc retained specific binding to CD20-positive cells and was active in mediating complement-dependent cytolysis. Size-exclusion HPLC analysis revealed that the purified scFv-Fc included multimeric as well as monomeric components. Variant scFv-Fcs were constructed incorporating four different hinges between the scFv and Fc regions, or three different linkers in the scFv domain. All formed multimers, with the highest level of multimerization found in the scFv-Fc with the shortest linker (8 aa). Elimination of an unusual salt bridge between residues L38 and H89 in the V(L)-V(H) domain interface failed to reduce the formation of higher order forms. Structural analysis of the scFv-Fc constructed with 18 or 8 aa linkers by pepsin or papain cleavage suggested the proteins contained a form in which scFv units had cross-paired to form a 'diabody'. Thus, domain exchange or cross-pairing appears to be the basis of the observed multimerization.
- Published
- 2001
- Full Text
- View/download PDF
32. Mammalian expression and hollow fiber bioreactor production of recombinant anti-CEA diabody and minibody for clinical applications.
- Author
-
Yazaki PJ, Shively L, Clark C, Cheung CW, Le W, Szpikowska B, Shively JE, Raubitschek AA, and Wu AM
- Subjects
- Antibodies, Monoclonal biosynthesis, Antibodies, Monoclonal genetics, Antibodies, Monoclonal isolation & purification, Chromatography, Ion Exchange, Clinical Trials as Topic, Humans, Immunoglobulin Fragments genetics, Neoplasms diagnostic imaging, Radionuclide Imaging, Recombinant Proteins biosynthesis, Recombinant Proteins immunology, Recombinant Proteins isolation & purification, Bioreactors, Carcinoembryonic Antigen immunology, Immunoglobulin Fragments biosynthesis, Immunoglobulin Fragments isolation & purification
- Abstract
Genetically engineered radiolabeled antibody fragments have shown great promise for the radioimmunoscintigraphy of cancer. Retaining the exquisite specificity of monoclonal antibodies yet smaller in molecular size, antibody fragments display rapid tumor targeting and blood clearance, a more uniform distribution in the tumor, and present a lower potential to elicit an immune response. However, one of the factors that has limited clinical evaluation of these antibody-derived proteins has been the difficulty in expressing and purifying the quantities necessary for clinical trials. This study outlines the capability of mammalian expression for the production of recombinant antibody fragments intended for clinical use. Two anti-carcinoembryonic antigen antibody fragments, the T84.66/212 Flex minibody (scFv-C(H)3) and the T84.66 diabody (scFv dimer) have been previously expressed and have shown excellent radioimaging properties in tumor bearing animals. To proceed toward human studies, these high affinity recombinant fragments and a second minibody version, the T84.66/GS18 Flex minibody, were expressed using a high-level mammalian expression system. Production of all three antibody fragments in a small-scale hollow fiber bioreactor resulted in 137-307 mg of crude antibody harvest. A purification protocol that employed ceramic hydroxyapatite and anion exchange chromatography resulted in 50-150 mg of purified T84.66 diabody and T84.66 minibody. The development of this level of research grade material established conditions for clinical production as well as provided material to complete pre-clinical studies and undertake protein crystallization studies. Scale-up for clinical studies produced 3.4 g of the T84.66 minibody in the harvest. A portion of this material was purified yielding 180 mg of highly purified T84.66 minibody intended for pilot radioimmunoscintigraphy studies of carcinoembryonic antigen (CEA) positive disease.
- Published
- 2001
- Full Text
- View/download PDF
33. Tumor targeting of radiometal labeled anti-CEA recombinant T84.66 diabody and t84.66 minibody: comparison to radioiodinated fragments.
- Author
-
Yazaki PJ, Wu AM, Tsai SW, Williams LE, Ikler DN, Wong JY, Shively JE, and Raubitschek AA
- Subjects
- Animals, Chelating Agents chemistry, Chromatography, High Pressure Liquid, Heterocyclic Compounds, 1-Ring chemistry, Humans, Immunoconjugates chemistry, Immunoglobulin Fragments immunology, Iodine Radioisotopes chemistry, Iodine Radioisotopes metabolism, Kidney metabolism, Liver metabolism, Mice, Mice, Nude, Recombinant Proteins immunology, Recombinant Proteins metabolism, Recombinant Proteins pharmacokinetics, Spleen metabolism, Carcinoembryonic Antigen immunology, Immunoconjugates metabolism, Immunoglobulin Fragments metabolism, Indium Radioisotopes chemistry, Indium Radioisotopes metabolism, Neoplasms diagnostic imaging, Radioimmunodetection
- Abstract
Recombinant antibody fragments offer potential advantages over intact monoclonal antibodies in the radioimmunoscintigraphy (RIS) of solid tumors. Due to their smaller molecular size, antibody fragments have shown rapid tumor targeting and blood clearance, a more uniform tumor distribution and a lower potential to elicit a human immune response. Previously, we have expressed two genetically engineered antibody fragments, the T84.66 diabody (scFv dimer) and the T84.66 minibody (scFv-CH3 dimer), specific to carcinoembryonic antigen (CEA). When radioiodinated, both antibody fragments exhibited rapid tumor targeting and rapid blood clearance in xenografted mice. To extend and optimize their future clinical RIS utility with radiometals, these antibody fragments were conjugated with the macrocycle 1,4,7,10-tetraazacyclododecane N,N',N' ',N' "-tetraacetic acid (DOTA) and labeled with 111In. Tumor targeting and biodistribution studies were carried out in athymic mice xenografted with a human colorectal tumor cell line, LS174T. The [111In]T84.66 diabody (55 kDa) exhibited very rapid tumor targeting with 12.5 +/- 0.4% injected dose per gram (% ID g(-1) +/- standard error) at 2 h and reached a maximum of 13.3 +/- 0.9% ID g(-1) at 6 h. However, kidney uptake was observed to reached a peak of 183.5 +/- 21.0% ID g(-1) at 6 h, a result similar to that reported by others for other low molecular weight fragments labeled with radiometals. Preadministration of an oral dose of D-lysine resulted in a 59% lowering of the renal accumulation at 6 h, but was accompanied by a 31% reduction of tumor uptake to 9.2 +/- 1.2% ID g(-1). The second recombinant antibody fragment, the [111In]T84.66 minibody (80 kDa), displayed rapid tumor targeting of 14.2 +/- 6.1% ID g(-1) at 2 h, and reached a maximum activity of 24.5 +/- 6.1% ID g(-1) by 12 h. Renal uptake achieved a plateau of 12-13% ID g(-1) which cleared to 7.2% ID g(-1) at 72 h. However, hepatic uptake was elevated and reached a maximum of 26.0 +/- 1.0% ID g(-1) at 12 h in these xenograft-bearing mice. Experiments in nontumor bearing mice showed a reduction of hepatic activity at 12 h to 16.6 +/- 1.5% ID g(-1), indicative of an intrinsic hepatic accumulation of the [111In]DOTA-T84.66 minibody or metabolites. While the anti-CEA [111In]DOTA-T84.66 diabody and T84.66 minibody retain the rapid tumor targeting properties of the radioiodinated form, the normal organ accumulation (kidneys and liver, respectively) of the [111In]DOTA forms appeared problematic for RIS and RIT applications. Development of alternative blocking strategies or new metabolizable chelates are under investigation to enhance the utility of the radiometal form of these and other promising recombinant antibody fragments.
- Published
- 2001
- Full Text
- View/download PDF
34. Metabolism and renal clearance of 111In-labeled DOTA-conjugated antibody fragments.
- Author
-
Tsai SW, Li L, Williams LE, Anderson AL, Raubitschek AA, and Shively JE
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal, Murine-Derived, Antineoplastic Agents immunology, Antineoplastic Agents metabolism, Chromatography, Electrophoresis, Polyacrylamide Gel, Humans, Immunoconjugates chemistry, Immunoglobulin Fragments chemistry, Indium Radioisotopes chemistry, Kidney anatomy & histology, Kidney chemistry, Mice, Mice, Nude, Rituximab, Tissue Distribution, Chelating Agents chemistry, Heterocyclic Compounds, 1-Ring chemistry, Immunoconjugates metabolism, Immunoglobulin Fragments metabolism, Indium Radioisotopes metabolism, Kidney metabolism
- Abstract
Radiometal-labeled antibody fragments are promising reagents for radioimmunotherapy due to their high tumor uptake and rapid pharmacokinetics, but their therapeutic potentials are limited by high uptake and retention in the kidney. Identification of metabolic products is a first step in designing rationale approaches to lower kidney uptake. Previous studies in rats have shown that 111In-labeled DTPA-conjugated antibody fragments (via lysine residues) were degraded to an DTPA-epsilon-amino-lysine derivative and retained in the lysosomal compartments of the liver and kidney [Rogers et al. (1995) Cancer Res. 55, 5714s-5720s]. To determine the metabolic profile of another widely used metal-chelate, [111In]DOTA conjugated to lysines in antibody fragments via active ester chemistry, we analyzed kidney homogenates from nude mice injected with an [111In]DOTA-Fab generated enzymatically from the anti-lymphoma intact antibody Rituxan. The major kidney metabolite was identified as [111In]DOTA-epsilon-amino-lysine by comparison to an authentic synthetic standard. This end product was also identified in the urine, along with relatively small amounts of [111In]DOTA-Fab. Since injection of [111In]DOTA-epsilon-amino-lysine into nude mice resulted in rapid clearance into the urine without kidney retention, it is likely that the renal retention observed was due to kidney uptake of [111In]DOTA-Fab, followed by lysosomal degradation to [111In]DOTA-epsilon-amino-lysine, which is only slowly cleared from this compartment. This observation is supported by autoradiographs of the kidney showing rapid localization of radioactivity into the distal regions of the kidney cortex. To extend this analysis to clinical trials, we have also analyzed urine taken from a patient injected with the intact antibody [111In]DOTA-cT84.66. In that example, we found that the major radioactive species was also [111In]DOTA-epsilon-amino-lysine.
- Published
- 2001
- Full Text
- View/download PDF
35. Numerical selection of optimal tumor imaging agents with application to engineered antibodies.
- Author
-
Williams LE, Wu AM, Yazaki PJ, Liu A, Raubitschek AA, Shively JE, and Wong JY
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal genetics, Antibodies, Monoclonal pharmacokinetics, Antibodies, Neoplasm chemistry, Antibodies, Neoplasm genetics, Antibody Affinity, Antigen-Antibody Reactions, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms pathology, Drug Design, Female, Fluorine Radioisotopes pharmacokinetics, Humans, Immunoglobulin Fab Fragments chemistry, Immunoglobulin Fab Fragments genetics, Immunoglobulin Fragments chemistry, Immunoglobulin Fragments genetics, Immunoglobulin G chemistry, Immunoglobulin G genetics, Iodine Radioisotopes pharmacokinetics, Mice, Molecular Weight, Neoplasm Transplantation, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins pharmacokinetics, Tissue Distribution, Transplantation, Heterologous, Tumor Cells, Cultured pathology, Antibodies, Monoclonal immunology, Antibodies, Neoplasm immunology, Antigens, Neoplasm immunology, Carcinoembryonic Antigen immunology, Immunoglobulin Fab Fragments immunology, Immunoglobulin Fragments immunology, Immunoglobulin G immunology, Neoplasms diagnostic imaging, Protein Engineering, Radioimmunodetection, Radiopharmaceuticals pharmacokinetics, Recombinant Fusion Proteins immunology
- Abstract
Three analytic indicators were used to compare five members of a monoclonal antibody (Mab) family. The cognates consisted of the genetically engineered intact chimeric IgGI (cT84.66) and related engineered fragments [scFv, diabody, minibody, F(ab')2] reactive against the same epitope of carcinoembryonic antigen (CEA). All analyses were based on radioiodinated Mabs targeting to colorectal xenografts of LS174T tumors in nude mice. Affinity constants were evaluated initially. A second indicator was the imaging figure of merit (IFOM) which determines how rapidly a statistically significant tumor image can be acquired. Finally, deconvolution was used to determine tumor temporal response to an arterial bolus. This last analysis gave the possible tumor accumulation in the absence of normal tissue sequestration. Affinities were all in excess of 10(8) M-1 and were highest for the divalent Mabs. Using the IFOM criterion, an 131I label was best suited as a radiolabel for the intact (IgG) T84.66, while an 123I label indicated optimal imaging with either minibody or F(ab')2. Deconvolution analyses showed that divalent members behaved similarly while the univalent member (scFv) had a tumor residence time smaller by an order of magnitude. The diabody had the largest impulse response function, but renal uptake may limit its present usefulness.
- Published
- 2001
- Full Text
- View/download PDF
36. A phase I radioimmunotherapy trial evaluating 90yttrium-labeled anti-carcinoembryonic antigen (CEA) chimeric T84.66 in patients with metastatic CEA-producing malignancies.
- Author
-
Wong JYC, Chu DZ, Yamauchi DM, Williams LE, Liu A, Wilczynski S, Wu AM, Shively JE, Doroshow JH, and Raubitschek AA
- Subjects
- Animals, Antibodies, Monoclonal pharmacokinetics, Bone Marrow radiation effects, Humans, Immunoglobulin G metabolism, Liver radiation effects, Lung Neoplasms therapy, Mice, Pentetic Acid pharmacology, Radioisotopes pharmacokinetics, Recombinant Fusion Proteins metabolism, Thyroid Neoplasms therapy, Time Factors, Yttrium Radioisotopes pharmacokinetics, Antibodies, Monoclonal therapeutic use, Carcinoembryonic Antigen blood, Colorectal Neoplasms radiotherapy, Colorectal Neoplasms therapy, Lung Neoplasms radiotherapy, Radioimmunotherapy methods, Radioisotopes therapeutic use, Thyroid Neoplasms radiotherapy, Yttrium Radioisotopes therapeutic use
- Abstract
Chimeric T84.66 (cT84.66) is a genetically engineered human/murine chimeric IgG, with high affinity and specificity to carcinoembryonic antigen (CEA). The purpose of this Phase I dose escalation therapy trial was to evaluate the toxicities, biodistribution, pharmacokinetics, tumor targeting, immunogenicity, and organ and tumor absorbed dose estimates of cT84.66 labeled with 90Y. Patients with metastatic CEA-producing malignancies were first administered 5 mCi 111In-labeled DTPA-cT84.66 (5 mg), followed by administration of the therapy dose of 90Y-labeled DTPA-cT84.66 1 week later. The therapy infusion was immediately followed by a 72-h administration of DTPA at 250 mg/m2/24 h. Dose levels of administered activity ranged from 5 to 22 mCi/m2 with three to six patients per level. Serial nuclear scans, blood samples, and 24-h urine collections were performed out to 5 days after infusion. Human antichimeric antibody response was assayed out to 6 months. Patients were administered up to 3 cycles of therapy every 6 weeks. Radiation absorbed doses to organs were estimated using a five compartment model and MIRDOSE3. Twenty-two patients received at least one cycle of therapy, with one individual receiving two cycles and two receiving three cycles of therapy. All were heavily pretreated and had progressive disease prior to entry in this trial. Reversible leukopenia and thrombocytopenia were the primary dose-limiting toxicities observed. Maximum tolerated dose was reached at 22 mCi/ m2. In general, patients with liver metastases demonstrated more rapid blood clearance of the antibody. Thirteen patients developed an immune response to the antibody. Average radiation doses to marrow, liver, and whole body were 2.6, 29, and 1.9 cGy/mCi 90Y, respectively. Dose estimates to tumor ranged from 66 to 1670 cGy (8.7 to 52.2 cGy/mCi 90Y) for each cycle of therapy delivered. Although no major responses were observed, three patients demonstrated stable disease of 12-28 weeks duration and two demonstrated a mixed response. In addition, a 41-100% reduction in tumor size was observed with five tumor lesions. 90Y-labeled cT84.66 was well tolerated, with reversible thrombocytopenia and leukopenia being dose limiting. Patients with extensive hepatic involvement by tumor demonstrated unfavorable biodistribution for therapy with rapid blood clearance and poor tumor targeting. Average tumor doses when compared with red marrow doses indicated a favorable therapeutic ratio. Stable disease and mixed responses were observed in this heavily pretreated population with progressive disease. This trial represents an important step toward further improving the therapeutic potential of this agent through refinements in the characteristics of the antibody and the treatment strategies used. Future trials will focus on the use of peripheral stem cell support to allow for higher administered activities and the use of combined modality strategies with radiation-enhancing chemotherapy drugs. Further efforts to reduce immunogenicity through humanization of the antibody are also planned. Finally, novel engineered, lower molecular weight, faster clearing constructs derived from cT84.66 continue to be evaluated in preclinical models as potential agents for radioimmunotherapy.
- Published
- 2000
37. High-resolution microPET imaging of carcinoembryonic antigen-positive xenografts by using a copper-64-labeled engineered antibody fragment.
- Author
-
Wu AM, Yazaki PJ, Tsai Sw, Nguyen K, Anderson AL, McCarthy DW, Welch MJ, Shively JE, Williams LE, Raubitschek AA, Wong JY, Toyokuni T, Phelps ME, and Gambhir SS
- Subjects
- Animals, Antibody Specificity, Copper Radioisotopes, Female, Humans, Immunoglobulin Fragments genetics, Immunoglobulin Variable Region genetics, Immunoglobulin Variable Region immunology, Isotope Labeling, Mice, Mice, Nude, Neoplasm Transplantation, Rats, Recombinant Proteins genetics, Recombinant Proteins immunology, Time Factors, Tomography, Emission-Computed methods, Transplantation, Heterologous, Tumor Cells, Cultured, Carcinoembryonic Antigen immunology, Immunoglobulin Fragments immunology
- Abstract
Rapid imaging by antitumor antibodies has been limited by the prolonged targeting kinetics and clearance of labeled whole antibodies. Genetically engineered fragments with rapid access and high retention in tumor tissue combined with rapid blood clearance are suitable for labeling with short-lived radionuclides, including positron-emitting isotopes for positron-emission tomography (PET). An engineered fragment was developed from the high-affinity anticarcinoembryonic antigen (CEA) monoclonal antibody T84.66. This single-chain variable fragment (Fv)-C(H)3, or minibody, was produced as a bivalent 80 kDa dimer. The macrocyclic chelating agent 1,4,7, 10-tetraazacyclododecane-N,N',N", N"'-tetraacetic acid (DOTA) was conjugated to the anti-CEA minibody for labeling with copper-64, a positron-emitting radionuclide (t(1/2) = 12.7 h). In vivo distribution was evaluated in athymic mice bearing paired LS174T human colon carcinoma (CEA positive) and C6 rat glioma (CEA negative) xenografts. Five hours after injection with (64)Cu-DOTA-minibody, microPET imaging showed high uptake in CEA-positive tumor (17.9% injected dose per gram +/- 3.79) compared with control tumor (6.0% injected dose per gram +/- 1.0). In addition, significant uptake was seen in liver, with low uptake in other tissues. Average target/background ratios relative to neighboring tissue were 3-4:1. Engineered antibody fragments labeled with positron-emitting isotopes such as copper-64 provide a new class of agents for PET imaging of tumors.
- Published
- 2000
- Full Text
- View/download PDF
38. Biodistribution and radioimmunotherapy of human breast cancer xenografts with radiometal-labeled DOTA conjugated anti-HER2/neu antibody 4D5.
- Author
-
Tsai SW, Sun Y, Williams LE, Raubitschek AA, Wu AM, and Shively JE
- Subjects
- Animals, Antibodies, Monoclonal analysis, Breast Neoplasms, Female, Humans, Indium Radioisotopes, Mammary Neoplasms, Experimental pathology, Mice, Mice, Nude, Neoplasm Transplantation, Tissue Distribution, Transplantation, Heterologous, Tumor Cells, Cultured, Yttrium Radioisotopes, Antibodies, Monoclonal therapeutic use, Chelating Agents, Mammary Neoplasms, Experimental radiotherapy, Organometallic Compounds therapeutic use, Radioimmunotherapy, Receptor, ErbB-2 immunology
- Abstract
HER2/neu oncogene encodes a 185 kDa trans-membrane protein which is overexpressed in 20-30% of breast and ovarian cancers and portends a poor prognosis. We have studied the targeting and therapy of this oncoprotein with 4D5, a murine monoclonal antibody which recognizes a distinct epitope on the extracelluar domain of HER2/neu. We conjugated the antibody with an active ester of the macrocyclic chelating agent DOTA, radiolabeled the conjugate with either (111)In or (90)Y, and studied the antibody distribution and therapy, respectively, in athymic mice bearing xenografts of MCF7/HER2/neu, a human breast cancer cell line transfected with the HER2/neu oncogene. For the biodistribution of (111)In-labeled DOTA-4D5, a high specificity of tumor localization (30% ID/g) was seen with a tumor-to-blood ratio of greater than 2 at 48 h postinjection. Compared to a previously published study with (125)I-labeled 4D5 in beige nude mice bearing NIH3T3/HER2/neu xenografts [De Santes et al. (1992) Cancer Res. 52, 1916-1923], (111)In-labeled 4D5 antibody gave superior antibody uptake in tumor (30% ID/g vs 17% ID/g at 48h). In the therapy study, treatment of the nude mice bearing MCF7/HER2/neu xenografts with 100 microCi (3 microg) of (90)Y-labeled DOTA-4D5 caused a 3-fold reduction of tumor growth compared to untreated controls (injected with human serum albumin) in 40 days. Treatment of animals with 100 microCi of nonspecific antibody (90)Y-labeled DOTA-Leu16 (3 microg) had no tumor growth inhibition. Treatment with unlabeled DOTA-4D5 (3 microg) had a slight effect on tumor growth compared to untreated controls. When analyzed at the level of single animals, no effect was seen in seven of nine animals; however, in two of the animals, tumor growth inhibition was observed. Although a cold antibody therapeutic effect was unexpected at this dose level (3 microg), it may be possible that in some animals that 3 microg of antibody of (90)Y-labeled DOTA-4D5 augmented tumor growth reduction. To further explore the effects of cold antibody treatment alone, animals were treated with 100 or 400 microg of unlabeled 4D5 administered in two doses. These animals showed a 1.7-1.8-fold reduction in tumor growth over 28 days, a result less than that obtained with RIT only.
- Published
- 2000
- Full Text
- View/download PDF
39. Truncation of blood curves to enhance imaging and therapy with monoclonal antibodies.
- Author
-
Williams LE, Liu A, Wu AM, Yazaki PJ, Yamauchi DM, Lopatin G, Raubitschek AA, and Wong JY
- Subjects
- Animals, Antibodies, Monoclonal blood, Biophysical Phenomena, Biophysics, Humans, Mice, Models, Biological, Neoplasms blood, Neoplasms diagnostic imaging, Neoplasms radiotherapy, Radiopharmaceuticals therapeutic use, Antibodies, Monoclonal therapeutic use, Radioimmunodetection, Radioimmunotherapy, Radiopharmaceuticals blood
- Abstract
Targeting of monoclonal antibody (Mab) to solid tumor sites is a function of the blood curve of activity versus time. It has been suggested that the blood curve be artificially reduced to approach zero so that the contrast between tumor and blood uptake is maximized. We analyzed tumor uptake as a function of the time tc of blood curve truncation. By using a convolution approach, we were able to find the optimal times for setting the blood curve to zero in either diagnostic or therapeutic animal examples. Two iodinated cT84.66 anti-CEA engineered fragments, diabody and minibody, were considered using previous data from nude mouse studies involving the LS174T colorectal tumor model. Figures of merit (FOMs) were used to compare ordinary and truncated blood curves and their associated tumor accumulations. Using a 1231 label, it was seen that the appropriate time for diagnostic truncation occurred when tumor uptake, as measured, was a maximum. The corresponding point for therapy (with 1311 as a label) was at infinite time. We also demonstrated that the use of traditional indices led to ambiguities in the choice of truncation times. The traditional therapy index, the ratio of the integral of the tumor uptake to the integral of the blood uptake, was found to be a numerical constant independent of tc. This ratio was proved to be the integral of the tumor impulse response function. Use of such convolution techniques to assess truncation of the perfused material is probably also applicable to multistep processes as well as to lesion targeting with other tumor-specific pharmaceuticals.
- Published
- 2000
- Full Text
- View/download PDF
40. Evaluating changes in stable chromosomal translocation frequency in patients receiving radioimmunotherapy.
- Author
-
Wong JY, Wang J, Liu A, Odom-Maryon T, Shively JE, Raubitschek AA, and Williams LE
- Subjects
- Chromosomes, Human, Pair 3 radiation effects, Chromosomes, Human, Pair 4 radiation effects, Dose-Response Relationship, Radiation, Humans, In Situ Hybridization, Fluorescence, Male, Regression Analysis, Bone Marrow radiation effects, Prostatic Neoplasms genetics, Prostatic Neoplasms radiotherapy, Radioimmunotherapy adverse effects, Translocation, Genetic
- Abstract
Purpose: The lack of any consistent correlation between radioimmunotherapy (RIT) dose and observed hematologic toxicity has made it difficult to validate RIT radiation dose estimates to marrow. Stable chromosomal translocations (SCT) which result after radiation exposure may be a biologic parameter that more closely correlates with RIT radiation dose. Increases in the frequency of SCT are observed after radiation exposure and are highly correlated with absorbed radiation dose. SCT are cumulative after multiple radiation doses and conserved through an extended number of cell divisions. The purpose of this study was to evaluate whether increases in SCT frequency were detectable in peripheral lymphocytes after RIT and whether the magnitude of these increases correlated with estimated radiation dose to marrow and whole body., Methods and Materials: Patients entered in a Phase I dose escalation therapy trial each received 1-3 intravenous cycles of the radiolabeled anti- carcinoembryonic antigen (CEA) monoclonal antibody, 90Y-chimeric T84.66. Five mCi of 111In-chimeric T84.66 was co-administered for imaging and biodistribution purposes. Blood samples were collected immediately prior to the start of therapy and 5-6 weeks after each therapy cycle. Peripheral lymphocytes were harvested after 72 hours of phytohemagglutinin stimulation and metaphase spreads prepared. Spreads were then stained by fluorescence in situ hybridization (FISH) using commercially available chromosome paint probes to chromosomes 3 and 4. Approximately 1000 spreads were evaluated for each chromosome sample. Red marrow radiation doses were estimated using the AAPM algorithm and blood clearance curves., Results: Eighteen patients were studied, each receiving at least one cycle of therapy ranging from 5-22 mCi/m2. Three patients received 2 cycles and two patients received 3 cycles of therapy. Cumulative estimated marrow doses ranged from 9.2 to 310 cGy. Increases in SCT frequencies were observed after each cycle for both chromosomes 3 and 4 in 16 of 18 patients and in at least one chromosome for the remaining 2 patients. Cumulative increases in SCT frequencies ranged from 0.001 to 0.046 with no major differences observed between chromosomes 3 and 4. A linear correlation between cumulative marrow dose and increases in SCT frequencies was observed for chromosome 3 (R2 = 0.63) and chromosome 4 (R2 = 0.80). A linear correlation was also observed between increases in SCT frequency and whole body radiation dose or administered activity (R2 = 0.67-0.89). There was less correlation between observed decrease in wbc or platelet counts and marrow dose, whole body dose, or administered activity (R2 = 0.28-0.43)., Conclusions: Increases in SCT frequency were detectable in peripheral lymphocytes after low dose-rate RIT irradiation. A linear correlation was observed between increases in SCT and marrow dose, whole body dose, and administered activity. This correlation provides one of the strongest radiation dose-response and activity-response relationships observed with RIT. The detection of SCT may therefore have application as an in situ integrating biodosimeter after RIT. This biologic parameter should prove useful in comparing effects on marrow for different therapeutic radionuclides and in comparing effects of RIT and external beam radiation doses on a cGy per cGy basis. As a result, this should allow for a more direct comparison between different methods of irradiation and in further refinement of radioimmunotherapy dose estimates and dosimetry methodology.
- Published
- 2000
- Full Text
- View/download PDF
41. Initial clinical experience evaluating Yttrium-90-chimeric T84.66 anticarcinoembryonic antigen antibody and autologous hematopoietic stem cell support in patients with carcinoembryonic antigen-producing metastatic breast cancer.
- Author
-
Wong JY, Somlo G, Odom-Maryon T, Williams LE, Liu A, Yamauchi D, Wu AM, Yazaki P, Wilczynski S, Shively JE, Forman S, Doroshow JH, and Raubitschek AA
- Subjects
- Animals, Breast Neoplasms metabolism, Carcinoembryonic Antigen biosynthesis, Combined Modality Therapy, Female, Humans, Mice, Pentetic Acid therapeutic use, Transplantation, Autologous, Breast Neoplasms therapy, Carcinoembryonic Antigen immunology, Hematopoietic Stem Cell Transplantation, Immunoglobulin G therapeutic use, Radioimmunoassay adverse effects, Recombinant Fusion Proteins therapeutic use, Yttrium Radioisotopes therapeutic use
- Abstract
cT84.66 is a human/murine IgG1 with high affinity and specificity for carcinoembryonic antigen (CEA). An earlier Phase I trial defined the maximum tolerated dose for 90Y-diethylenetriaminepentaacetic acid (DTPA)-cT84.66 at 22 mCi/m2. Dose-limiting toxicities were reversible leukopenia and thrombocytopenia. The purpose of this Phase I trial was to evaluate the feasibility and toxicities of administering higher activities of 90Y-DTPA-cT84.66 with stem cell support in patients with CEA-producing breast cancer. Patients with CEA-producing breast cancer refractory to standard therapies underwent peripheral stem cell collection followed by infusion of 111indium-DTPA-cT84.66. Those patients demonstrating tumor targeting received a single therapy dose of 90Y-DTPA-cT84.66, followed by Ca-DTPA infusion for 72 h posttherapy. Stem cells were reinfused following a divided schedule. To date, seven patients have been accrued to this trial. Each patient received an imaging dose of (111)In-cT84.66. Six patients demonstrated tumor imaging and received a single cycle of 90Y-cT84.66 at 15 mCi/m2 (three patients) and 22.5 mCi/m2 (three patients). One patient did not demonstrate tumor imaging and was not treated. At these administered activities, 90Y-cT84.66 was well tolerated. No dose-limiting toxicities have been observed. All patients demonstrated hematopoietic recovery after stem cell infusion. One patient demonstrated stable disease for 4 months; one patient had stable disease and reduction of bone pain for 3 months; and a third patient experienced >50% reduction of an ovarian metastasis, resolution of malignant pleural effusion, stable pleural metastases, and stable bone scan for 14 months. Preliminary results from this ongoing Phase I trial are promising and demonstrate the feasibility and potential for antitumor effects of stem cell supported 90Y-cT84.66 therapy in patients with CEA-producing breast cancers.
- Published
- 1999
42. A method for patient-specific absorbed dose estimation for internal beta emitters.
- Author
-
Williams LE, Liu A, Raubitschek AA, and Wong JY
- Subjects
- Beta Particles, Humans, Radioimmunotherapy, Radiotherapy Dosage, Yttrium Radioisotopes therapeutic use
- Abstract
The purpose of the study was to determine a technique for estimating patient-specific absorbed radiation doses in radioimmunotherapy and other internal emitter therapies. Beta Radiation sources were considered, with 90Y being the radionuclide of primary interest. Organ uptake of activity was determined using a merged set of computed tomography and planar nuclear images. Estimation of local absorbed dose was accomplished using a voxel source kernel. Voxel size was 0.2 x 0.2 x 0.5 cm; dimensions were from the digital resolution of the nuclear and computed tomography data sets. Dose-volume histograms were also obtained due to the voxel nature of the estimations. Organ dose estimates were made for two patients receiving the chimeric anticarcinoembryonic antigen antibody cT84.66. Considerable variation was observed when comparing the voxel kernel results with medical internal radiation dosimetry values obtained via the MIRDOSE3 program. Primary uncertainty in the organ dose estimates was determined to be due to the variation in organ mass. By correcting the S values in that program by the organ mass ratio, we found generally good agreement between our method and MIRDOSE3. We conclude that patient-specific absorbed doses can be estimated for 90Y-labeled antibodies.
- Published
- 1999
43. A radionuclide therapy treatment planning and dose estimation system.
- Author
-
Liu A, Williams LE, Lopatin G, Yamauchi DM, Wong JY, and Raubitschek AA
- Subjects
- Humans, Phantoms, Imaging, Radiometry, Radiotherapy Dosage, Software, Radioimmunotherapy, Radiotherapy Planning, Computer-Assisted
- Abstract
Unlabelled: An object-oriented software system is described for estimating internal emitter absorbed doses using a set of computer modules operating within a personal computer environment. The system is called the Radionuclide Treatment Planning and Absorbed Dose Estimation System (RTDS). It is intended for radioimmunotherapy applications, although other forms of internal emitter therapy may also be considered., Methods: Four software modules interact through a database backend. Clinical, demographic and image data are directly entered into the database. Modules include those devoted to clinical imaging (nuclear, CT and MR), activity determination, organ compartmental modeling and absorbed dose estimation., Results: Both standard phantom (Medical Internal Radiation Dose [MIRD]) and patient-specific absorbed doses are estimated. All modules interact with the database backend so that changes in one process do not influence other operations. Results of the modular operations are written to the database as computations are completed. Dose-volume histograms are an intrinsic part of the output for patient-specific absorbed dose estimates. A sample dose estimate for a potential 90Y monoclonal antibody is described., Conclusion: A four-module software system has been implemented to estimate MIRD phantom and patient-specific absorbed doses. Computations of the doses and their statistical distribution for a pure beta emitter such as 90Y take approximately 1 min on a 300 MHz personal computer.
- Published
- 1999
44. Intrapatient consistency of imaging biodistributions and their application to predicting therapeutic doses in a phase I clinical study of 90Y-based radioimmunotherapy.
- Author
-
Clarke KG, Odom-Maryon TL, Williams LE, Liu A, Lopatin G, Chou J, Farino GM, Raubitschek AA, and Wong JY
- Subjects
- Adult, Aged, Female, Humans, Indium Radioisotopes blood, Indium Radioisotopes urine, Male, Middle Aged, Reproducibility of Results, Time Factors, Tissue Distribution, Yttrium Radioisotopes blood, Yttrium Radioisotopes urine, Radioimmunotherapy methods, Radiotherapy Planning, Computer-Assisted, Yttrium Radioisotopes therapeutic use
- Abstract
Intrapatient variation in the biodistribution of the chimeric monoclonal antibody cT84.66 was assessed in 19 patients having a variety of carcinoembryonic antigen (CEA) positive tumors. The two studies, including whole-body imaging and blood and urine specimen collections, were conducted within 14 days of each other using (111)In-cT84.66 at a fixed total protein dose of 5 mg per patient per study. An initial pretherapy infusion of (111)In-cT84.66 was administered followed by a therapy coinfusion of (111)In-ct84.66 and 90Y-cT84.66 A closed five-compartment model was used to integrate source organ activity curves as residence time inputs into the MIRDOSE3 program. Normal organ absorbed doses were estimated for 90Y-cT84.66, the corresponding radiotherapeutic agent. For the two (111)In-cT84.66 biodistributions, all data were modeled with a R2 value of between 0.72 and 1.00 with the exception of the urine data taken during therapy. This was due to the need of diethylenetriaminepentaacetic acid during the therapy phase because of the possibility that yttrium might escape from the chelator attached to the antibody. With the assurance that the biodistributions were reproducible, we were able to estimate the 90Y-cT84.66 absorbed doses on a per-patient basis. Concordance coefficients showing the agreement between the imaging and therapy phase dose estimates were between the 0.60 and 0.99 levels for liver, spleen, red marrow, total body, and other organ systems. Median results were: 27, 17, and 2.7 rad/mCi of 90Y-cT84.66 for liver, spleen, and red marrow, respectively. Because of decreases in platelets and white cells as the amount of 90Y was increased, dose-limiting toxicity was found at 22 mCi/m2. We conclude that patient biodistributions were consistent over time to 14 days so as to allow absorbed dose estimation in a radioimmunotherapy trial involving the cT84.66 anti-CEA antibody.
- Published
- 1999
- Full Text
- View/download PDF
45. Dose escalation trial of indium-111-labeled anti-carcinoembryonic antigen chimeric monoclonal antibody (chimeric T84.66) in presurgical colorectal cancer patients.
- Author
-
Wong JY, Chu DZ, Yamauchi D, Odom-Maryon TL, Williams LE, Liu A, Esteban JM, Wu AM, Primus FJ, Beatty JD, Shively JE, and Raubitschek AA
- Subjects
- Adult, Aged, Animals, Antibodies, Monoclonal adverse effects, Carcinoembryonic Antigen immunology, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms pathology, Female, Humans, Immunoglobulin G adverse effects, Indium Radioisotopes adverse effects, Indium Radioisotopes pharmacokinetics, Liver Neoplasms diagnostic imaging, Liver Neoplasms secondary, Lymphatic Metastasis, Male, Metabolic Clearance Rate, Mice, Middle Aged, Neoplasm Metastasis, Pilot Projects, Radiography, Radioimmunotherapy, Radionuclide Imaging, Radiopharmaceuticals adverse effects, Radiopharmaceuticals pharmacokinetics, Radiopharmaceuticals therapeutic use, Radiotherapy Dosage, Radiotherapy, Adjuvant, Sensitivity and Specificity, Tissue Distribution, Colorectal Neoplasms radiotherapy, Colorectal Neoplasms surgery, Indium Radioisotopes therapeutic use
- Abstract
Unlabelled: Chimeric T84.66 (cT84.66) is a high-affinity (1.16x10(11) M(-1)) IgG1 monoclonal antibody against carcinoembryonic antigen (CEA). The purpose of this pilot trial was to evaluate the tumor-targeting properties, biodistribution, pharmacokinetics and immunogenicity of 111In-labeled cT84.66 as a function of administered antibody protein dose., Methods: Patients with CEA-producing colorectal cancers with localized disease or limited metastatic disease who were scheduled to undergo definitive surgical resection were each administered a single intravenous dose of 5 mg of isothiocyanatobenzyl diethylenetriaminepentaacetic acid-cT84.66, labeled with 5 mCi of 111In. Before receiving the radiolabeled antibody, patients received unlabeled diethylenetriaminepentaacetic acid-cT84.66. The amount of unlabeled antibody was 0, 20 or 100 mg, with five patients at each level. Serial blood samples, 24-hr urine collections and nuclear images were collected until 7 days postinfusion. Human antichimeric antibody response was assessed up to 6 mo postinfusion., Results: Imaging of at least one known tumor site was performed in all 15 patients. Fifty-two lesions were analyzed, with an imaging sensitivity rate of 50.0% and a positive predictive value of 76.9%. The antibody detected tumors that were not detected by conventional means in three patients, resulting in a modification of surgical management. Interpatient variations in serum clearance rates were observed and were secondary to differences in clearance and metabolic rates of antibody and antibody:antigen complexes by the liver. Antibody uptake in primary tumors, metastatic sites and regional metastatic lymph nodes ranged from 0.4% to 134% injected dose/kg, resulting in estimated 90Y-cT84.66 radiation doses ranging from 0.3 to 193 cGy/mCi. Thirteen patients were evaluated 1-6 mo after infusion for human antichimeric antibody, and none developed a response. No major differences in tumor imaging, tumor uptake, pharmacokinetics or organ biodistribution were observed with increasing protein doses, although a trend toward increasing blood uptake and decreasing liver uptake was observed with increasing protein dose., Conclusion: Chimeric T84.66 demonstrated tumor targeting comparable to other radiolabeled intact anti-CEA monoclonal antibodies. Its immunogenicity after single administration was lower than murine monoclonal antibodies. These properties make 111In-cT84.66, or a lower molecular weight derivative, attractive for further evaluation as an imaging agent. Yttrium-90 dosimetry estimates predict potentially cytotoxic radiation doses to select tumor sites, which makes 90Y-cT84.66 also appropriate for further evaluation in Phase I radioimmunotherapy trials. Although clinically important changes in biodistribution, pharmacokinetics and tumor targeting with increasing protein doses of 111In-cT84.66 were not demonstrated, the results do suggest that antibody clearance from the blood is driven by hepatic uptake and metabolism, with more rapid blood clearance seen in patients with liver metastases. These patients with rapid clearance and potentially unfavorable biodistribution for imaging and therapy may, therefore, be a more appropriate subset in which to evaluate the role of administering higher protein doses. This underscores the need to further identify, characterize and understand those factors that influence the biodistribution and clearance of radiolabeled anti-CEA antibodies, to allow for better selection of patients for therapy and rational planning of radioimmunotherapy.
- Published
- 1998
46. Monte Carlo-assisted voxel source kernel method (MAVSK) for internal beta dosimetry.
- Author
-
Liu A, Williams LE, Wong JY, and Raubitschek AA
- Subjects
- Humans, Yttrium Radioisotopes, Monte Carlo Method, Neoplasms radiotherapy, Radioimmunotherapy methods, Radiotherapy Planning, Computer-Assisted
- Abstract
A method is described for the determination of patient-specific organ beta doses given a known cumulated internal radioactivity distribution. A voxel source kernel for 90Y analogous to the point source function was simulated. Dose to each organ of interest could then be estimated by convolving the voxel source kernel with the patient's 3-D volume with known radioactivity assigned to each voxel. The dose calculation on eight organs took less than 1 min per patient using a Sun Sparc10 workstation.
- Published
- 1998
- Full Text
- View/download PDF
47. Biodistribution of 111In- and 90Y-labeled DOTA and maleimidocysteineamido-DOTA conjugated to chimeric anticarcinoembryonic antigen antibody in xenograft-bearing nude mice: comparison of stable and chemically labile linker systems.
- Author
-
Williams LE, Lewis MR, Bebb GG, Clarke KG, Odom-Maryon TL, Shively JE, and Raubitschek AA
- Subjects
- Animals, Bone Marrow metabolism, Bone and Bones metabolism, Colorectal Neoplasms metabolism, Drug Stability, Humans, Indium Radioisotopes pharmacokinetics, Isotope Labeling, Mice, Mice, Nude, Tissue Distribution, Transplantation, Heterologous, Yttrium Radioisotopes pharmacokinetics, Antibodies, Monoclonal chemistry, Carcinoembryonic Antigen immunology, Chelating Agents pharmacokinetics, Heterocyclic Compounds pharmacokinetics, Heterocyclic Compounds, 1-Ring, Maleimides pharmacokinetics, Neoplasm Transplantation
- Abstract
Biodistributions of two radiometal chelate conjugates of the human/murine chimeric anticarcinoembryonic antigen monoclonal antibody cT84.66 were obtained in nude mice bearing LS174T human colorectal carcinoma xenografts. Derivatives of the macrocyclic chelating agent 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA) were covalently attached to the antibody by a stable amide linkage and by a maleimidocysteineamido side chain (MC-DOTA) that has been shown to be chemically labile at physiological temperature and pH. Biodistributions of both 111In and 90Y labels were obtained in these studies. At common biodistribution time points, it was found that the 111In label had greater uptake in the liver than 90Y for both conjugates. No significant differences were found with respect to bone uptake of 90Y using either chelate. Blood curves were generally lower at comparable time points for MC-DOTA, indicative of faster clearance as compared to DOTA. Tumor uptake was high for both conjugates (57-68% ID/g at 48 h), with a longer tumor residence time in the case of the DOTA conjugate, probably a result of its longer blood circulation times. We conclude that bone uptake of 90Y would be minimal if either DOTA or MC-DOTA were used as the bifunctional chelator. This would imply preference for these macrocyclic ligands if radiation doses to the bone marrow would be considered to be dominated by skeletal uptakes. Alternatively, if bone marrow radiation dose is dominated by circulating antibody, the chemically labile linker system employed by the MC-DOTA conjugate offers the advantage of enhanced blood clearance.
- Published
- 1998
- Full Text
- View/download PDF
48. Pharmacokinetic modeling and absorbed dose estimation for chimeric anti-CEA antibody in humans.
- Author
-
Odom-Maryon TL, Williams LE, Chai A, Lopatin G, Liu A, Wong YC, Chou J, Clarke KG, and Raubitschek AA
- Subjects
- Adult, Aged, Animals, Female, Humans, Male, Mice, Middle Aged, Models, Theoretical, Pilot Projects, Radiation Dosage, Recombinant Fusion Proteins pharmacokinetics, Recombinant Fusion Proteins therapeutic use, Tissue Distribution, Carcinoembryonic Antigen immunology, Indium Radioisotopes pharmacokinetics, Indium Radioisotopes therapeutic use, Radioimmunodetection, Radioimmunotherapy, Yttrium Radioisotopes pharmacokinetics, Yttrium Radioisotopes therapeutic use
- Abstract
Unlabelled: The objective of this article was to model pharmacokinetic data from clinical diagnostic studies involving the 111In-labeled monoclonal antibody (MAb) chimeric T84.66, against carcinoembryonic antigen. Model-derived results based on the 111In-MAb blood, urine and digital imaging data were used to predict 90Y-MAb absorbed radiation doses and to guide treatment planning for future therapy trials. Fifteen patients with at least one carcinoembryonic antigen-positive lesion were evaluated. We report the kinetic parameter estimates and absorbed 111In-MAb dose and projected 90Y-MAb doses for each patient as well as describe our approach and rationale for modeling an extensive set of pharmacokinetic data., Methods: The ADAPT II software package was used to create three- and five-compartment models of uptake against time in the patient population. The "best-fit" model was identified using ordinary least squares. Areas under the curve were calculated using the modeled curves and input into MIRDOSE3 to estimate absorbed radiation doses for each patient., Results: A five-compartment model best described the liver, whole body, blood and urine data for a subcohort of nine patients with digital imaging data. A three-compartment model best described the blood and urine data for all 15 clinical patients accrued in the clinical trial. For the subcohort, the largest projected 90Y-MAb doses were delivered to the liver (mean, 24.78 rad/mCi; range, 15.02-37.07 rad/mCi), with red marrow estimates on the order of 3.32 rad/mCi (range, 1.24-5.55) of 90Y. Corresponding estimates for the 111In-MAb were 3.18 (range, 2.09-4.43) and 0.55 (range, 0.34-0.74), respectively., Conclusion: The three- and five-compartment models presented here were successfully used to represent the blood, urine and imaging data. This was evidenced by the small standard errors for the kinetic parameter estimates and R2 values close to 1. As planned future therapeutic trials will involve stem cell support to alleviate hematological toxicities, the development of an approach for estimating doses to other major organs is crucial.
- Published
- 1997
49. Clinical evaluation of indium-111-labeled chimeric anti-CEA monoclonal antibody.
- Author
-
Wong JY, Thomas GE, Yamauchi D, Williams LE, Odom-Maryon TL, Liu A, Esteban JM, Neumaier M, Dresse S, Wu AM, Primus FJ, Shively JE, and Raubitschek AA
- Subjects
- Adult, Aged, Animals, Female, Humans, Indium Radioisotopes pharmacokinetics, Male, Mice, Middle Aged, Pilot Projects, Predictive Value of Tests, Recombinant Fusion Proteins pharmacokinetics, Recombinant Fusion Proteins therapeutic use, Sensitivity and Specificity, Tissue Distribution, Yttrium Radioisotopes pharmacokinetics, Yttrium Radioisotopes therapeutic use, Carcinoembryonic Antigen immunology, Indium Radioisotopes therapeutic use, Radioimmunodetection, Radioimmunotherapy
- Abstract
Unlabelled: Chimeric T84.66 (cT84.66) is a high-affinity (1.16 x 10[11] M[-1]) IgG1 monoclonal antibody (MAb) against carcinoembryonic antigen (CEA). This pilot trial evaluated the tumor-targeting properties, biodistribution, pharmacokinetics and immunogenicity of 111In-labeled cT84.66., Methods: Patients with CEA-producing metastatic malignancies were administered a single intravenous dose of 5 mCi 111In-diethylenetriaminepentaacetic acid-cT84.66. Serial blood samples, 24-hr urine collections and nuclear images were collected up to 7 days postinfusion. Human antichimeric antibody response was assessed up to 6 mo postinfusion., Results: Imaging of at least one known tumor site was observed in 14 of 15 (93%) patients. Seventy-four lesions were analyzed with an imaging sensitivity rate of 45.1% and a positive predictive value of 94.1%. In one patient, two additional bone metastases developed within 6 mo of antibody administration at sites initially felt to be falsely positive on scan. One patient developed a human antichimeric antibody response predominantly to the murine portion of the antibody. The antibody cleared serum with a median T(1/2alpha) of 6.53 hr and a T(1/2beta) of 90.87 hr. Interpatient variations in serum clearance rates were observed and were secondary to differences in clearance and metabolic rates of antibody-antigen complexes by the liver. One patient demonstrated very rapid clearance of antibody by the liver, which compromised antibody localization to the primary tumor. Antibody uptake in primary and metastatic tumors ranged from 0.5% to 10.5% injected dose/kg, resulting in estimated radiation doses ranging from 0.97 to 21.3 cGy/mCi 90Y. Antibody uptake in regional lymph nodes ranged from 1.3% to 377% injected dose/kg, resulting in estimated radiation doses ranging from 2.0 to 617 cGy/mCi 90Y., Conclusion: Chimeric T84.66 demonstrated tumor targeting that was comparable to that of other radiolabeled intact anti-CEA Mabs. Its immunogenicity after single administration was lower than murine Mabs. These properties make cT84.66 or a lower molecular weight derivative attractive for further evaluation as an imaging agent. These same properties also make it appropriate for future evaluation in Phase I therapy trials. Finally, a wide variation in the rate of antibody clearance was observed, with one patient demonstrating very slow clearance, resulting in the highest estimated marrow dose of the group, and one patient demonstrating unusually rapid clearance, resulting in poor antibody localization to tumor. Data from this study suggest that serum CEA levels, antibody-antigen complex clearance and, therefore, antibody clearance are influenced by both the production and clearance rates of CEA. This underscores the need to further identify, characterize and understand those factors that influence the biodistribution and clearance of radiolabeled anti-CEA antibodies to allow for better selection of patients for therapy and rational planning of radioimmunotherapy.
- Published
- 1997
50. A CT assisted method for absolute quantitation of internal radioactivity.
- Author
-
Liu A, Williams LE, and Raubitschek AA
- Subjects
- Algorithms, Antibodies, Monoclonal, Gamma Cameras, Humans, Indium Radioisotopes, Least-Squares Analysis, Lung diagnostic imaging, Magnetic Resonance Imaging, Phantoms, Imaging, Radiation Dosage, Radioimmunotherapy, Tomography, X-Ray Computed, Radionuclide Imaging methods
- Abstract
A method is described for the determination of radioactivity (microCi or MBq) at an organ site within an object or patient. Using both anatomic image data (CT or MRI scans) and planar gamma camera images, activity at depth is determined using a matrix inversion method based on least squares. The result of the inversion analysis was the unknown set of n linear (uniform) activity densities representative of each organ within the phantom or patient. The problem was overdetermined since the number of unknown activity densities (microCi/cm) was much less than the number of analysis points (N) within the nuclear image. This method, defined as the CT assisted matrix inversion (CAMI) technique, was accurate to within 15% for a three "organ" plastic phantom, wherein the organs were right circular cylinders having activities of 74 to 508 microCi (or 2.74 MBq to 18.8 MBq). This accuracy included image quantitation effects, particularly assumptions concerning attenuation correction. The average absolute percent error of the estimated activity in four distinct radioactive volumes in the phantom was 9.8%. It was found that the background activity within the phantom was estimated to be too high if sampling regions near strong sources were used in the analysis (scatter effect). This was minimized by going at least 2 cm away from such sources. By applying the method to a monoclonal antibody clinical study, activities within the patient's major organs such as liver, spleen, and kidney could be estimated, even in cases where the organ could not be visualized. Here, the CAMI algorithm gave internally consistent results for the patient's left and right lung linear activity concentrations. The CAMI technique resolves the problem of tissue superimposition using depth information from 3-D CT and is applicable in cases where a number of organs overlap in the gamma camera image. Thus, the method should be generally useful to nuclear image quantitation and the estimation of absorbed radiation doses in patients. One particular application is the estimation of radiation doses in radioimmunotherapy (RIT).
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.