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










Publication year range
1.
Nat Commun ; 8: 14837, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332616

ABSTRACT

Protein drugs that neutralize vascular endothelial growth factor (VEGF), such as aflibercept or ranibizumab, rescue vision in patients with retinal vascular diseases. Nonetheless, optimal visual outcomes require intraocular injections as frequently as every month. Here we report a method to extend the intravitreal half-life of protein drugs as an alternative to either encapsulation or chemical modifications with polymers. We combine a 97-amino-acid peptide of human origin that binds hyaluronan, a major macromolecular component of the eye's vitreous, with therapeutic antibodies and proteins. When administered to rabbit and monkey eyes, the half-life of the modified proteins is increased ∼3-4-fold relative to unmodified proteins. We further show that prototype long-acting anti-VEGF drugs (LAVAs) that include this peptide attenuate VEGF-induced retinal changes in animal models of neovascular retinal disease ∼3-4-fold longer than unmodified drugs. This approach has the potential to reduce the dosing frequency associated with retinal disease treatments.


Subject(s)
Bevacizumab/administration & dosage , Ranibizumab/administration & dosage , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Retinal Diseases/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Animals , Bevacizumab/chemistry , Bevacizumab/pharmacokinetics , Disease Models, Animal , Female , Half-Life , Humans , Hyaluronic Acid/chemistry , Intravitreal Injections , Macaca fascicularis , Male , Rabbits , Ranibizumab/chemistry , Ranibizumab/pharmacokinetics , Receptors, Vascular Endothelial Growth Factor/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Retinal Diseases/metabolism
2.
Cancer Biother Radiopharm ; 29(3): 91-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24443889

ABSTRACT

PURPOSE: Single-step iodination is often performed in preference to use of a radiometal label for initial animal biodistributions. Yet loss of iodine occurs in vivo so that it is important to measure uptake (%ID/g) differences between radiolabels. METHODS: Murine biodistributions of four radioiodinated and (111)In-labeled cognate anti-carcinoembryonic antigen antibodies were compared. Uptakes were obtained in athymic mice out to 96 hours for diabody, minibody, scFv-Fc, and intact humanized (M5A) versions of the T84.66 antibody. Tissues included liver, spleen, kidneys, lungs, and human LS174T colorectal xenografts. Ratios (R) of iodine uptake to indium uptake were calculated. RESULTS: For all cognates, no significant differences were found in the blood uptakes between the two labels. In normal solid organs, iodine was generally cleared more rapidly with decreasing molecular weight (MW). In liver and spleen by 24 hours, the R value was 5% for diabody and minibody, whereas a value of 50% was seen for the intact mAb. Renal differences were even more marked for the two lower MW species. Tumor losses, however, were found to be essentially independent of MW and were modest; 50% by 48 hours. To test the generality of the results, comparisons were then made for normal organs and tumors of the R values found above for M5A and MN-14 intact antibody literature results. Good agreement, within estimated errors in R, was seen between these two cases, although (111)In and (88)Y were the respective radiometals. CONCLUSIONS: Loss of iodine from labeled antibodies can be marked in normal organs and that correction (by 1/R) of the iodine biodistribution to estimate the associated radiometal result may be possible. Explicit differences between the two types of biodistribution also imply that separate uptake results need to be considered when evaluating the impact on imaging and therapy using various possible radiolabels.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoembryonic Antigen/immunology , Indium Radioisotopes , Iodine Radioisotopes , Animals , Colonic Neoplasms/metabolism , Humans , Kidney/metabolism , Lung/metabolism , Mice , Spleen/metabolism , Tissue Distribution
3.
J Biol Chem ; 287(27): 22927-37, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22570488

ABSTRACT

Serum half-life of IgG is controlled by the neonatal Fc receptor (FcRn) that interacts with the IgG Fc region and may be increased or decreased as a function of altered FcRn binding. Preclinical evaluations of modified IgGs are frequently carried out in mice, but such IgGs may bind differently to mouse and human FcRn (mFcRn and hFcRn). Here, we report a detailed characterization of a matched set of mouse-human chimeric T84.66 scFv-Fc variants with specificity for the tumor carcinoembryonic antigen and mutations in the FcRn-binding site. Binding to soluble mFcRn and hFcRn was measured using in vitro assays, and the results were compared with blood clearance in vivo in normal (mFcRn bearing) and hFcRn transgenic mice. All variants bound better to mFcRn than to hFcRn. The loss of affinity varied among the mutants, however, and also the hierarchy of binding differed depending on the receptor. The mutations had no major impact on binding to the classical Fcγ receptors. Importantly, the trend of blood clearance in both strains of mice correlated with the hierarchy of binding obtained using soluble FcRn. Consequently, in vitro interaction analysis of engineered IgGs regarding their cross-species FcRn binding ability provides information for prediction of in vivo pharmacokinetics.


Subject(s)
Carcinoembryonic Antigen/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Protein Engineering/methods , Receptors, Fc/immunology , Receptors, IgG/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Specificity , Cross Reactions/immunology , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/blood , Iodine Radioisotopes , Mice , Mice, Inbred BALB C , Mice, Transgenic , Single-Chain Antibodies/pharmacokinetics , Species Specificity
4.
Int J Mol Imaging ; 2011: 834515, 2011.
Article in English | MEDLINE | ID: mdl-21912743

ABSTRACT

Introduction. Sensitive and specific imaging of pancreas cancer are necessary for accurate diagnosis, staging, and treatment. The vast majority of pancreas cancers express the carbohydrate tumor antigen CA19-9. The goal of this study was to determine the potential to target CA19-9 with a radiolabeled anti-CA19-9 antibody for imaging pancreas cancer. Methods. CA19-9 was quantified using flow cytometry on human pancreas cancer cell lines. An intact murine anti-CA19-9 monoclonal antibody was labeled with a positron emitting radionuclide (Iodine-124) and injected into mice harboring antigen positive and negative xenografts. MicroPET/CT were performed at successive time intervals (72 hours, 96 hours, 120 hours) after injection. Radioactivity was measured in blood and tumor to provide objective confirmation of the images. Results. Antigen expression by flow cytometry revealed approximately 1.3 × 10(6) CA19-9 antigens for the positive cell line and no expression in the negative cell line. Pancreas xenograft imaging with Iodine-124-labeled anti-CA19-9 mAb demonstrated an average tumor to blood ratio of 5 and positive to negative tumor ratio of 20. Conclusion. We show in vivo targeting of our antigen positive xenograft with a radiolabeled anti-CA19-9 antibody. These data demonstrate the potential to achieve anti-CA19-9 antibody based positron emission tomography of pancreas cancer.

5.
J Surg Res ; 170(2): 169-78, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21601881

ABSTRACT

BACKGROUND: Intact antibodies are poor imaging agents due to a long serum half-life (10-20 d) preventing adequate contrast between the tumor and surrounding blood. Smaller engineered antibody fragments overcome this problem by exhibiting shorter serum half-lives (4-20 h).The diabody (55 kDa) is the smallest antibody fragment, which retains the bivalency of the intact antibody. Our goal was to develop and characterize the anti-CA19-9 diabody fragment and determine its ability to provide antigen specific imaging of pancreas cancer. METHODS: The diabody DNA construct was created by isolation of the variable region genes of the intact anti-CA19-9 antibody. Diabody expression was carried out in NS0 cells and purified using HPLC from supernatant. Specific antigen binding was confirmed with flow cytometry and immunofluorescence. Radiolabeled diabody was injected into mice harboring an antigen positive xenograft (BxPC3 or Capan-2) and a negative xenograft (MiaPaca-2). MicroCT and MicroPET were performed at successive time intervals after injection. Radioactivity was measured in blood and tumor to provide objective confirmation of the microPET images. RESULTS: Immunofluorescence and flow cytometry showed specific binding of the anti-CA19-9 diabody. Pancreas xenograft imaging of BxPC3/MiaPaca-2 and Capan-2/MiaPaca-2 models with the anti-CA19-9 diabody demonstrated an average tumor:blood ratio of 5.0 and 2.0, respectively, and an average positive:negative tumor ratio of 11 and 6, respectively. With respect to the tumor:blood ratio, these data indicate five times and two times more radioactivity in the tumor than in the blood yielding adequate contrast between tumor tissue and background (i.e., blood) to create the representative microPET images. CONCLUSIONS: We successfully engineered a functional diabody against CA19-9, a tumor antigen present on the vast majority of pancreas cancers. Additionally, we demonstrate high contrast antigen specific microPET imaging of pancreas cancer in xenograft models.


Subject(s)
Antibodies, Monoclonal , CA-19-9 Antigen/immunology , Immunoglobulin Fragments , Pancreatic Neoplasms/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , CA-19-9 Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Humans , Hybridomas , Immunoglobulin Fragments/blood , Immunoglobulin Fragments/pharmacology , Iodine Radioisotopes/pharmacokinetics , Mice , Multiple Myeloma , Pancreatic Neoplasms/immunology , Protein Engineering/methods , Tissue Distribution , Transplantation, Heterologous
6.
Mol Imaging Biol ; 13(3): 526-535, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20628903

ABSTRACT

PURPOSE: The objective of this article is to develop internalizing positron emission tomography (PET) reporter genes for tracking genetically modified T cells in vivo. PROCEDURES: The transmembrane and cytoplasmic domains of the human transferrin receptor (TfR) and CD5 were each fused to the carcinoembryonic (CEA) minigene N-A3 and expressed in Jurkat T cells. Internalization was evaluated by confocal microscopy or by intracellular uptake of ¹²5I-labeled anti-CEA scFv-Fc. Reporter gene-transfected Jurkat xenografts in mice were analyzed by immunohistochemistry (IHC) and imaged by PET using ¹²4I- or 64Cu-scFv-Fc as tracers. RESULTS: Surface expression of TR(1-99)-NA3 was lower than that of NA3-CD5. Both reporter genes were internalized following binding of the anti-CEA antibody fragment. IHC of tumors showed strong staining of NA3-CD5, whereas TR(1-99)-NA3 stained weakly. Specific targeting of TR(1-99)-NA3 or NA3-CD5 was shown by PET in xenografted mice. CONCLUSIONS: The in vivo imaging studies suggest a potential application of the internalizing form of CEA (N-A3) as a PET reporter gene.


Subject(s)
Carcinoembryonic Antigen/genetics , Endocytosis , Genes, Reporter/genetics , Molecular Imaging/methods , Animals , Carcinoembryonic Antigen/metabolism , Female , Flow Cytometry , Humans , Immunoglobulin Fragments , Immunohistochemistry , Jurkat Cells , Mice , Mice, Nude , Positron-Emission Tomography , Recombinant Proteins/metabolism , Transfection , Xenograft Model Antitumor Assays
7.
EJNMMI Res ; 1(1): 24, 2011 Nov 07.
Article in English | MEDLINE | ID: mdl-22214289

ABSTRACT

BACKGROUND: Sensitive antibody-based tumor targeting has the potential not only to image metastatic and micrometastatic disease, but also to be the basis of targeted therapy. The vast majority of pancreas cancers express carcinoembryonic antigen (CEA). Thus, we sought to evaluate the potential of CEA as a pancreatic cancer target utilizing a rapidly clearing engineered anti-CEA scFv-Fc antibody fragment with a mutation in the Fc region [anti-CEA scFv-Fc H310A]. METHODS: Immunohistochemistry (IHC) with the antibody fragment was used to confirm expression of CEA on human pancreas cancer specimens. In vivo tumor targeting was evaluated by tail vein injection of I124-labeled anti-CEA scFv-Fc(H310A) into mice harboring CEA-positive and -negative xenografts. MicroPET/CT imaging was performed at successive time intervals. Radioactivity in blood and tumor was measured after the last time point. Additionally, unlabeled anti-CEA scFv-Fc(H310A) was injected into CEA-positive tumor bearing mice and ex vivo IHC was performed to identify the presence of the antibody to define the microscopic intratumoral pattern of targeting. RESULTS: Moderate to strong staining by IHC was noted on 84% of our human pancreatic cancer specimens and was comparable to staining of our xenografts. Pancreas xenograft imaging with the radiolabeled anti-CEA scFv-Fc(H310A) antibody demonstrated average tumor/blood ratios of 4.0. Immunolocalization demonstrated peripheral antibody fragment penetration of one to five cell diameters (0.75 to 1.5 µm). CONCLUSIONS: We characterized a preclinical xenograft model with respect to CEA expression that was comparable to human cases. We demonstrated that the anti-CEA scFv-Fc(H310A) antibody exhibited antigen-specific tumor targeting and shows promise as an imaging and potentially therapeutic agent.

8.
J Biol Chem ; 285(50): 39239-48, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20889497

ABSTRACT

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), a GPI-anchored endothelial cell protein, binds lipoprotein lipase (LPL) and transports it into the lumen of capillaries where it hydrolyzes triglycerides in lipoproteins. GPIHBP1 is assumed to be expressed mainly within the heart, skeletal muscle, and adipose tissue, the sites where most lipolysis occurs, but the tissue pattern of GPIHBP1 expression has never been evaluated systematically. Because GPIHBP1 is found on the luminal face of capillaries, we predicted that it would be possible to define GPIHBP1 expression patterns with radiolabeled GPIHBP1-specific antibodies and positron emission tomography (PET) scanning. In Gpihbp1(-/-) mice, GPIHBP1-specific antibodies were cleared slowly from the blood, and PET imaging showed retention of the antibodies in the blood pools (heart and great vessels). In Gpihbp1(+/+) mice, the antibodies were cleared extremely rapidly from the blood and, to our surprise, were taken up mainly by lung and liver. Immunofluorescence microscopy confirmed the presence of GPIHBP1 in the capillary endothelium of both lung and liver. In most tissues with high levels of Gpihbp1 expression, Lpl expression was also high, but the lung was an exception (very high Gpihbp1 expression and extremely low Lpl expression). Despite low Lpl transcript levels, however, LPL protein was readily detectable in the lung, suggesting that some of that LPL originates elsewhere and then is captured by GPIHBP1 in the lung. In support of this concept, lung LPL levels were significantly lower in Gpihbp1(-/-) mice than in Gpihbp1(+/+) mice. In addition, Lpl(-/-) mice expressing human LPL exclusively in muscle contained high levels of human LPL in the lung.


Subject(s)
Gene Expression Regulation , Glycosylphosphatidylinositols/metabolism , Receptors, Lipoprotein/chemistry , Animals , Binding Sites , Capillaries/metabolism , Cell Membrane/metabolism , Endothelium/metabolism , Kinetics , Lung/metabolism , Mice , Mice, Transgenic , Models, Biological , Positron-Emission Tomography/methods
9.
Protein Eng Des Sel ; 23(10): 789-98, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20802234

ABSTRACT

The long circulation persistence of human serum albumin (HSA) is enabled by its domain III (DIII) interaction with the neonatal Fc receptor (FcRn). A protein scaffold based on HSA DIII was designed. To modify the serum half life of the scaffold, residues H535, H510, and H464 were individually mutated to alanine. HSA DIII wild type (WT) and variants were fused to the anti-carcinoembryonic antigen (CEA) T84.66 diabody (Db), radiolabeled with (124)I and injected into xenografted athymic mice for serial PET/CT imaging. All proteins targeted the CEA-positive tumor. The mean residence times (MRT) of the proteins, calculated by quantifying blood activity from the PET images, were: Db-DIII WT (56.7 h), H535A (25 h), H510A (20 h), H464A (17 h), compared with Db (2.9 h). Biodistribution confirmed the order of blood clearance from slow to fast: Db-DIII WT > H535A > H510A > H464A > Db with 4.0, 2.0, 1.8, 1.6 and 0.08 %ID/g of remaining blood activity at 51 h, respectively. This study demonstrates that attenuating the DIII-FcRn interaction provides a way of controlling the pharmacokinetics of the entire Db-DIII fusion protein without compromising tumor targeting. H464 appears to be most crucial for FcRn binding (greatest reduction in MRT), followed by H510 and H535. By mutating the DIII scaffold, we can dial serum kinetics for imaging or therapy applications.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Serum Albumin/metabolism , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Animals , Base Sequence , Carcinoembryonic Antigen/blood , Carcinoembryonic Antigen/metabolism , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/chemistry , Humans , Iodine Radioisotopes , Isotope Labeling , Kinetics , Mice , Mice, Nude , Molecular Sequence Data , Mutation , Positron-Emission Tomography , Receptors, Fc/blood , Receptors, Fc/chemistry , Recombinant Fusion Proteins/blood , Serum Albumin/chemistry , Serum Albumin/classification , Serum Albumin/genetics , Tomography, X-Ray Computed , Transplantation, Heterologous
10.
Anal Biochem ; 401(2): 173-81, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20178770

ABSTRACT

Many biological and biomedical laboratory assays require the use of antibodies and antibody fragments that strongly bind to their cell surface targets. Conventional binding assays, such as the enzyme-linked immunosorbent assay (ELISA) and flow cytometry, have many challenges, including capital equipment requirements, labor intensiveness, and large reagent and sample consumption. Although these techniques are successful in mainstream biology, there is an unmet need for a tool to quickly ascertain the relative binding capabilities of antibodies/antibody fragments to cell surface targets on the benchtop at low cost. We describe a novel cell capture assay that enables several candidate antibodies to be evaluated quickly as to their relative binding efficacies to their cell surface targets. We used chimeric rituximab and murine anti-CD20 monoclonal antibodies as cell capture agents on a functionalized microscope slide surface to assess their relative binding affinities based on how well they capture CD20-expressing mammalian cells. We found that these antibodies' concentration-dependent cell capture profiles correlate with their relative binding affinities. A key observation of this assay involved understanding how differences in capture surfaces affect the assay results. This approach can find utility when an antibody or antibody fragment against a known cell line needs to be selected for targeting studies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Affinity , Antigens, CD20/immunology , Lymphocytes/immunology , Microscopy, Fluorescence/methods , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20/genetics , Biotinylation , Cell Line , Flow Cytometry , Gene Expression , Humans , Jurkat Cells , Lymphocytes/cytology , Rituximab
11.
Protein Eng Des Sel ; 23(4): 243-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20053640

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.


Subject(s)
Antibodies, Monoclonal/chemistry , Lymphoma, B-Cell/diagnostic imaging , Positron-Emission Tomography , Single-Chain Antibodies/chemistry , 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
12.
J Nucl Med ; 50(9): 1500-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19690034

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.


Subject(s)
Antibodies, Monoclonal , Lymphoma, B-Cell/diagnostic imaging , Positron-Emission Tomography/methods , Positron-Emission Tomography/veterinary , Radiopharmaceuticals , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , Disease Models, Animal , Female , Humans , Immunoglobulin Fragments/immunology , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred C3H , Radiopharmaceuticals/immunology , Reproducibility of Results , Rituximab , Sensitivity and Specificity
13.
Eur J Nucl Med Mol Imaging ; 36(1): 104-14, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18719907

ABSTRACT

PURPOSE: Reporter genes can provide a way of noninvasively assessing gene activity in vivo. However, current reporter gene strategies may be limited by the immunogenicity of foreign reporter proteins, endogenous expression, or unwanted biological activity. We have developed a reporter gene based on carcinoembryonic antigen (CEA), a human protein with limited normal tissue expression. METHODS: To construct a CEA reporter gene for PET, a CEA minigene (N-A3) was fused to the extracellular and transmembrane domains of the human Fc gamma RIIb receptor. The NA3-Fc gamma RIIb recombinant gene, driven by a CMV promoter, was transfected in Jurkat (human T cell leukemia) cells. Expression was analyzed by flow cytometry, immunohistochemistry (IHC), and microPET imaging. RESULTS: Flow cytometry identified Jurkat clones stably expressing NA3-Fc gamma RIIb at low, medium, and high levels. High and medium NA3-Fc gamma RIIb expression could also be detected by Western blot. Reporter gene positive and negative Jurkat cells were used to establish xenografts in athymic mice. IHC showed staining of the tumor with high reporter gene expression; medium and low N-A3 expression was not detected. MicroPET imaging, using an anti-CEA (124)I-labeled single-chain Fv-Fc antibody fragment, demonstrated that only high N-A3 expression could be detected. Specific accumulation of activity was visualized at the N-A3 positive tumor as early as 4 h. MicroPET image quantitation showed tumor activity of 1.8 +/- 0.2, 15.2 +/- 1.3, and 4.6 +/- 1.2 percent injected dose per gram (%ID/g) at 4, 20, and 48 h, respectively. Biodistribution at 48 h demonstrated tumor uptake of 4.8 +/- 0.8%ID/g. CONCLUSION: The CEA N-A3 minigene has the potential to be used as a reporter gene for imaging cells in vivo.


Subject(s)
Carcinoembryonic Antigen/analysis , Carcinoembryonic Antigen/genetics , Genes, Reporter , Recombinant Proteins/genetics , Animals , Carcinoembryonic Antigen/immunology , Cell Line , GPI-Linked Proteins , Humans , Immunoglobulin Fragments/immunology , Isoantigens/genetics , Mice , Neoplasms/diagnostic imaging , Neoplasms/genetics , Neoplasms/metabolism , Positron-Emission Tomography , Receptors, IgG/genetics , Recombinant Proteins/analysis , Recombinant Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tomography, X-Ray Computed
14.
FEBS J ; 275(16): 4097-110, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18637944

ABSTRACT

The neonatal Fc receptor (FcRn) is a major histocompatibility complex class I-related molecule that regulates the half-life of IgG and albumin. In addition, FcRn directs the transport of IgG across both mucosal epithelium and placenta and also enhances phagocytosis in neutrophils. This new knowledge gives incentives for the design of IgG and albumin-based diagnostics and therapeutics. To study FcRn in vitro and to select and characterize FcRn binders, large quantities of soluble human FcRn are needed. In this report, we explored the impact of two free cysteine residues (C48 and C251) of the FcRn heavy chain on the overall structure and function of soluble human FcRn and described an improved bacterial production strategy based on removal of these residues, yielding approximately 70 mg.L(-1) of fermentation of refolded soluble human FcRn. The structural and functional integrity was proved by CD, surface plasmon resonance and MALDI-TOF peptide mapping analyses. The strategy may generally be translated to the large-scale production of other major histocompatibility complex class I-related molecules with nonfunctional unpaired cysteine residues. Furthermore, the anti-FcRn response in goats immunized with the FcRn heavy chain alone was analyzed following affinity purification on heavy chain-coupled Sepharose. Importantly, purified antibodies blocked the binding of both ligands to soluble human FcRn and were thus directed to both binding sites. This implies that the FcRn heavy chain, without prior assembly with human beta2-microglobulin, contains the relevant epitopes found in soluble human FcRn, and is therefore sufficient to obtain binders to either ligand-binding site. This finding will greatly facilitate the selection and characterization of such binders.


Subject(s)
Cysteine/chemistry , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Receptors, Fc/chemistry , Receptors, Fc/immunology , Amino Acid Sequence , Cell Line , Cysteine/genetics , Disulfides/chemistry , Escherichia coli/genetics , Histocompatibility Antigens Class I/genetics , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Fc/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Temperature
15.
Cancer Res ; 67(2): 718-26, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234783

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.


Subject(s)
Carcinoembryonic Antigen/immunology , Immunoconjugates/pharmacokinetics , Immunoglobulin Fragments/metabolism , Indium Radioisotopes/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , 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
16.
Mol Cancer Ther ; 5(6): 1550-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818514

ABSTRACT

Monoclonal antibodies (mAb) are being used at an increasing rate in the treatment of cancer, with current efforts focused on developing engineered antibodies that exhibit optimal biodistribution profiles for imaging and/or radioimmunotherapy. We recently developed the single-chain Fv-Fc (scFv-Fc) mAb, which consists of a single-chain antibody Fv fragment (light-chain and heavy-chain variable domains) coupled to the IgG1 Fc region. Point mutations that attenuate binding affinity to FcRn were introduced into the Fc region of the wild-type scFv-Fc mAb, resulting in several new antibodies, each with a different half-life. Here, we describe the construction of a two-tiered physiologically based pharmacokinetic model capable of simulating the apparent biodistribution of both (111)In- and (125)I-labeled scFv-Fc mAbs, where (111)In-labeled metabolites from degraded (111)In-labeled mAbs tend to become trapped within the lysosomal compartment, whereas free (125)I from degraded (125)I-labeled mAbs is quickly eliminated via the urinary pathway. The different concentration-time profiles of (111)In- and (125)I-labeled mAbs permits estimation of the degradation capacity of each organ and elucidates the dependence of cumulative degradation in liver, muscle, and skin on FcRn affinity and tumor mass. Liver is estimated to account for approximately 50% of all degraded mAb when tumor is small (approximately 0.1 g) and drops to about 35% when tumor mass is larger (approximately 0.3 g). mAb degradation in residual carcass (primarily skin and muscle) decreases from approximately 45% to 16% as FcRn affinity of the three mAb variants under consideration increases. In addition, elimination of a small amount of mAb in the kidneys is shown to be required for a successful fit of model to data.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Immunoconjugates/pharmacokinetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fragments/metabolism , Models, Biological , Animals , Female , Indium Radioisotopes/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Tissue Distribution
17.
Expert Opin Drug Deliv ; 3(1): 53-70, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16370940

ABSTRACT

Therapeutic antibodies are well established as an important class of drugs in modern medicine. The exquisite specificity and affinity for a specific target offered by antibodies has also encouraged their development as delivery vehicles for agents such as radionuclides to target tissues, for radioimmunoimaging and radioimmunotherapy. Specifically, in nuclear medicine, radionuclide-conjugated antibody molecules make it possible to image diseased loci with greater sensitivity than other imaging modalities such as magnetic resonance imaging. Furthermore, two radionuclide-conjugated antibodies have recently been approved for the therapy of non-Hodgkin's lymphoma. However, optimal implementation of antibodies has been limited by the extended circulation persistence that is characteristic of native antibodies, which is responsible for increased background activity in radioimmunoimaging applications and dose-related normal organ toxicities in radioimmunotherapy. In this article the current status of radiolabelled intact antibodies is reviewed, focusing on strategies to improve their pharmacokinetic properties to suit a desired application. Examples from the literature that represent different approaches to accomplishing this task in terms of their successes as well as limitations, and perspectives for the future are discussed.


Subject(s)
Antibodies/therapeutic use , Immunoglobulin Fragments/therapeutic use , Neoplasms/radiotherapy , Animals , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/diagnostic imaging , Neoplasms/immunology , Radioimmunodetection/methods , Radioimmunotherapy/methods , Radioisotopes/therapeutic use
18.
Nat Protoc ; 1(4): 2048-60, 2006.
Article in English | MEDLINE | ID: mdl-17487194

ABSTRACT

Immunoglobulins (Igs) are large proteins of 150 kDa with prolonged residence time in blood. Their half-life is controlled by their ability to interact with the protective neonatal Fc receptor (FcRn, Brambell receptor) present on endothelial cells. Here, we describe a protocol using site-specific mutagenesis of individual residues responsible for this interaction, resulting in engineered antibodies with distinct half-lives. The method is a powerful tool that enables manipulation of half-lives and is applicable to all antibodies and Fc fusion proteins for the development of agents with controlled pharmacokinetic properties. Moreover, the protocol is applicable to any situation where the structure and/or function of engineered proteins are to be studied. The protocol begins with the mutagenesis reaction at the DNA level and proceeds to describe mammalian expression and purification of recombinant proteins, radiolabeling and evaluation in vivo. The time frame for completing the procedure is about 6 months, provided that no complications are encountered.


Subject(s)
Antibodies/genetics , Mutagenesis, Site-Directed/methods , Receptors, Fc/metabolism , Animals , Antibodies/metabolism , Half-Life , Isotope Labeling/methods , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Cancer Res ; 65(13): 5907-16, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15994969

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.


Subject(s)
Immunoconjugates/pharmacokinetics , Immunoglobulin Fragments/metabolism , Neoplasms/diagnostic imaging , Receptor, ErbB-2/immunology , 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
20.
Cancer Res ; 65(2): 622-31, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15695407

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.


Subject(s)
Carcinoembryonic Antigen/immunology , Immunoconjugates/pharmacokinetics , Immunoglobulin Fragments/metabolism , Iodine Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...