Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Oncotarget ; 6(31): 31104-18, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26418751

ABSTRACT

Serous Ovarian Cancers (SOC) are frequently resistant to programmed cell death. However, here we describe that these programmed death-resistant cells are nonetheless sensitive to agents that modulate autophagy. Cytotoxicity is not dependent upon apoptosis, necroptosis, or autophagy resolution. A screen of NCBI yielded more than one dozen FDA-approved agents displaying perturbed autophagy in ovarian cancer. The effects were maximized via combinatorial use of the agents that impinged upon distinct points of autophagy regulation. Autophagosome formation correlated with efficacy in vitro and the most cytotoxic two agents gave similar effects to a pentadrug combination that impinged upon five distinct modulators of autophagy. However, in a complex in vivo SOC system, the pentadrug combination outperformed the best two, leaving trace or no disease and with no evidence of systemic toxicity. Targeting the autophagy pathway in a multi-modal fashion might therefore offer a clinical option for treating recalcitrant SOC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Molecular Targeted Therapy , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Ovarian Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Mice, Inbred C57BL , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
2.
PLoS One ; 10(5): e0120815, 2015.
Article in English | MEDLINE | ID: mdl-25973900

ABSTRACT

High-risk neuroblastoma is associated with an overall survival rate of 30-50%. Neuroblastoma-expressed cell adhesion receptors of the integrin family impact cell adhesion, migration, proliferation and survival. Integrin α4 is essential for neural crest cell motility during development, is highly expressed on leukocytes, and is critical for transendothelial migration. Thus, cancer cells that express this receptor may exhibit increased metastatic potential. We show that α4 expression in human and murine neuroblastoma cell lines selectively enhances in vitro interaction with the alternatively spliced connecting segment 1 of fibronectin, as well as vascular cell adhesion molecule-1 and increases migration. Integrin α4 expression enhanced experimental metastasis in a syngeneic tumor model, reconstituting a pattern of organ involvement similar to that seen in patients. Accordingly, antagonism of integrin α4 blocked metastasis, suggesting adhesive function of the integrin is required. However, adhesive function was not sufficient, as mutants of integrin α4 that conserved the matrix-adhesive and promigratory function in vitro were compromised in their metastatic capacity in vivo. Clinically, integrin α4 is more frequently expressed in non-MYNC amplified tumors, and is selectively associated with poor prognosis in this subset of disease. These results reveal an unexpected role for integrin α4 in neuroblastoma dissemination and identify α4 as a potential prognostic indicator and therapeutic target.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin alpha4/genetics , Liver Neoplasms/genetics , Nervous System Neoplasms/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Humans , Integrin alpha4/metabolism , Intercellular Signaling Peptides and Proteins , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis , Mice , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Nervous System Neoplasms/metabolism , Nervous System Neoplasms/pathology , Neuroblastoma/metabolism , Neuroblastoma/secondary , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Prognosis , Signal Transduction , Survival Analysis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
3.
Protein Eng Des Sel ; 27(10): 317-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25095796

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) is an immunoglobulin superfamily cell adhesion molecule that is aberrantly expressed in a wide variety of human tumors, including melanoma, prostate cancer, breast cancer, colorectal carcinoma, bladder cancer and pancreatic adenocarcinoma. This wide spectrum of human malignancies makes ALCAM a prospective pan-cancer immunoPET target to aid in detection and diagnosis in multiple malignancies. In this study, we assess site-specific versus non-site-specific conjugation strategies for (64)Cu-DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) immunoPET imaging of a fully human ALCAM cys-diabody (cDb) with a reduced linker length that retains its bivalent binding ability. ALCAM constructs with linker lengths of eight, five and three amino acids were produced to make true non-covalent site-specifically modified cDbs. Characterization by gel electrophoresis, size exclusion chromatography, flow cytometry and mass spectrometry of the various constructs was performed. To demonstrate the increased utility of targeting multiple malignancies expressing ALCAM, we compare the targeting of the site-specific versus non-site-specific conjugated cDbs to the human colorectal cancer xenograft LS174T. Interestingly, the conjugation strategy not only affects tumor targeting but also hepatic and renal uptake/clearance.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/chemistry , Colorectal Neoplasms/diagnostic imaging , Copper , Heterocyclic Compounds, 1-Ring , Immunohistochemistry/methods , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/metabolism , Humans , Molecular Imaging/methods , Rats , Tissue Distribution
4.
J Surg Res ; 185(1): 45-55, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23827791

ABSTRACT

BACKGROUND: Antibody-based therapeutics is a rapidly growing field. Small engineered antibody fragments demonstrate similar antigen affinity compared with the parental antibody but have a shorter serum half-life and possess the ability to be conjugated to nanoparticles. The goal of this study was to engineer an anti-carbohydrate antigen 19-9 (CA19-9) cys-diabody fragment in hopes of targeting nanoparticles to pancreatic cancer. METHODS: The anti-CA19-9 cys-diabody was created by engineering a C-terminal cysteine residue into the DNA single-chain Fv construct of the anti-CA19-9 diabody and expressed in NS0 cells. Maleimide chemistry was used to conjugate the cys-diabody to polymerized liposomal nanoparticles (PLNs) through the cysteine residues. Flow cytometry was used to evaluate targeting of cys-diabody and cys-diabody-PLN conjugate to human pancreatic cancer cell lines. The cys-diabody was radiolabeled with a positron emitter ((124)I) and evaluated in a mouse model of CA19-9-positive and CA19-9-negative xenografts with micro-positron emission tomography/micro-computed tomography at successive time intervals after injection. Percentage of injected dose per gram of radioactivity was measured in blood and tumor to provide objective confirmation of the micro-positron emission tomographic images. RESULTS: Tumor xenograft imaging of the anti-CA19-9 cys-diabody demonstrated an average tumor-to-blood ratio of 3.0 and positive-to-negative tumor ratio of 7.4. Successful conjugation of the cys-diabody to PLNs was indicated by flow cytometry showing specific binding of cys-diabody-PLN conjugate to human pancreatic cancer cells in vitro. CONCLUSIONS: Our results show that the anti-CA19-9 cys-diabody targets pancreatic cancer providing specific molecular imaging in tumor xenograft models. Furthermore, the cys-diabody-PLN conjugate demonstrates target-specific binding of human pancreatic cancer cells with the potential to deliver targeted treatment.


Subject(s)
Antibodies, Bispecific/pharmacology , CA-19-9 Antigen/immunology , Nanoparticles/therapeutic use , Pancreatic Neoplasms/therapy , Positron-Emission Tomography/methods , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Bispecific/chemistry , Cell Line, Tumor , Cystine/chemistry , Cystine/pharmacology , Female , Humans , Immunotherapy/methods , Liposomes/pharmacology , Mice , Mice, Nude , Multiple Myeloma , Pancreatic Neoplasms/diagnostic imaging , Protein Engineering , Single-Chain Antibodies/chemistry , Xenograft Model Antitumor Assays
5.
Mol Imaging Biol ; 14(3): 336-47, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21630083

ABSTRACT

PURPOSE: The purpose of this study was to generate and evaluate a positron emission tomography (PET) radiotracer targeting activated leukocyte cell adhesion molecule (ALCAM). PROCEDURES: A human anti-ALCAM single chain variable fragment was reformatted to produce a covalent dimer, termed a cys-diabody (CysDb). Purified CysDb was characterized by gel electrophoresis and size exclusion chromatography, and immunoreactivity was assessed by flow cytometry and immunofluorescence. Targeting and imaging of ALCAM-positive tumors using (64)Cu-DOTA-CysDb were evaluated in mice bearing human pancreatic adenocarcinoma xenografts (HPAF-II or BxPC-3). RESULTS: CysDb binds specifically to ALCAM-positive cells in vitro with an apparent affinity in the range of 1-3 nM. MicroPET images at 4 h showed specific targeting of positive tumors in vivo, a finding confirmed by biodistribution analysis, with positive/negative tumor ratios of 1.9 ± 0.6 and 2.4 ± 0.6, and positive tumor/blood ratios of 2.5 ± 0.9 and 2.9 ± 0.6 (HPAF-II and BxPC-3, respectively). CONCLUSIONS: Successful imaging with (64)Cu-DOTA-CysDb in animal models suggests further investigation of ALCAM as an imaging biomarker is warranted.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Biomarkers, Tumor/metabolism , Cysteine/metabolism , Positron-Emission Tomography/methods , Protein Engineering/methods , Single-Chain Antibodies/metabolism , Activated-Leukocyte Cell Adhesion Molecule/analysis , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Copper Radioisotopes/chemistry , Cysteine/chemistry , Cysteine/genetics , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/chemistry , Neoplasms, Experimental/metabolism , Organometallic Compounds/chemistry , Pancreatic Neoplasms/metabolism , Rats , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
6.
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.

7.
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
8.
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.

9.
Cancer Biother Radiopharm ; 25(3): 253-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20578830

ABSTRACT

The identification of tumor tissue biomarkers has led to the production, validation, and Food and Drug Administration-approval of a number of antibody-based targeted therapeutics in the past two decades. As a result of the significant role that these immunotherapeutics play in the management of cancer, and the potential utility of complementary imaging agents, immunoPET imaging has generated considerable interest. This update discusses the important factors to consider when designing a PET (positron emission tomography) imaging agent from the molecular target to the biological targeting molecule and radionuclide combination and also reviews recent preclinical and clinical findings in the immunoPET field. Although there are a variety of radionuclides that are currently utilized in PET studies, this update focuses on four of the positron emitters commonly used in labeling proteins: iodine-124, zirconium-89, copper-64, and fluorine-18. Notable advances in the preclinical setting include the continued development of immunoPET probes to predict the biodistribution of related radioimmunotherapeutics, the success of nontraditional radionuclide and antibody fragment combinations, the broader use of zirconium-89, and the recent emergence of (18)F-labeled diabodies for same-day imaging. Antibody-based PET probes constitute a valuable class of molecular imaging agents, and the progress made preclinically should expedite the transition of these targeted diagnostics to clinical applications.


Subject(s)
Immunoconjugates , Neoplasms/diagnosis , Positron-Emission Tomography/trends , Radiopharmaceuticals , Animals , Antibodies , Clinical Trials as Topic , Copper Radioisotopes , Drug Evaluation, Preclinical , Fluorine Radioisotopes , Humans , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunoglobulin Fragments , Iodine Radioisotopes , Neoplasms/therapy , Radioisotopes , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Recombinant Fusion Proteins , Tissue Distribution , Zirconium
10.
Eur J Nucl Med Mol Imaging ; 37(8): 1529-38, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20354850

ABSTRACT

PURPOSE: Prostate stem cell antigen (PSCA), a cell surface glycoprotein expressed in normal human prostate and bladder, is over-expressed in the majority of localized prostate cancer and most bone metastases. We have previously shown that the hu1G8 minibody, a humanized anti-PSCA antibody fragment (single-chain Fv-C(H)3 dimer, 80 kDa), can localize specifically and image PSCA-expressing xenografts at 21 h post-injection. However, the humanization and antibody fragment reformatting decreased its apparent affinity. Here, we sought to evaluate PET imaging contrast with affinity matured minibodies. METHODS: Yeast scFv display, involving four rounds of selection, was used to generate the three affinity matured antibody fragments (A2, A11, and C5) that were reformatted into minibodies. These three affinity matured anti-PSCA minibodies were characterized in vitro, and following radiolabeling with (124)I were evaluated in vivo for microPET imaging of PSCA-expressing tumors. RESULTS: The A2, A11, and C5 minibody variants all demonstrated improved affinity compared to the parental (P) minibody and were ranked as follows: A2 > A11 > C5 > P. The (124)I-labeled A11 minibody demonstrated higher immunoreactivity than the parental minibody and also achieved the best microPET imaging contrast in two xenograft models, LAPC-9 (prostate cancer) and Capan-1 (pancreatic cancer), when evaluated in vivo. CONCLUSION: Of the affinity variant minibodies tested, the A11 minibody that ranked second in affinity was selected as the best immunoPET tracer to image PSCA-expressing xenografts. This candidate is currently under development for evaluation in a pilot clinical imaging study.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , Immunoglobulin Fragments/immunology , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Amino Acid Sequence , Animals , Antibody Affinity , Cell Line, Tumor , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Male , Mice , Molecular Sequence Data , Mutation , Prostatic Neoplasms/pathology , Radioactive Tracers
11.
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
12.
Bioconjug Chem ; 20(8): 1474-81, 2009 Aug 19.
Article in English | MEDLINE | ID: mdl-19642689

ABSTRACT

The present work demonstrates the use of small bivalent engineered antibody fragments, cys-diabodies, for biological modification of nanoscale particles such as quantum dots (Qdots) for detection of target antigens. Novel bioconjugated quantum dots known as immunoQdots (iQdots) were developed by thiol-specific oriented coupling of tumor specific cys-diabodies, at a position away from the antigen binding site to amino PEG CdSe/ZnS Qdots. Initially, amino PEG Qdot 655 were coupled with reduced anti-HER2 cys-diabody by amine-sulfhydryl-reactive linker [N-ε-maleimidocaproyloxy] succinimide ester (EMCS) to produce anti-HER2 iQdot 655. Spectral characterization of the conjugate revealed that the spectrum was symmetrical and essentially identical to unconjugated Qdot. Specific receptor binding activity of anti-HER2 iQdot 655 was confirmed by flow cytometry on HER2 positive and negative cells. Immunofluorescence results showed homogeneous surface labeling of the cell membrane with Qdot 655 conjugate. In addition, cys-diabodies specific for HER2, as well as prostate stem cell antigen (PSCA), were conjugated successfully with amino PEG Qdot 800. All of these iQdots retain the photoluminescence properties of the unconjugated Qdot 800 as well as the antigen binding specificity of the cys-diabody as demonstrated by flow cytometry. Simultaneous detection of two tumor antigens on LNCaP/PSCA prostate cancer cells (which express PSCA and HER2) in culture was possible using two iQdots, anti-HER2 iQdot 655 and anti-PSCA iQdot 800. Thus, these iQdots are potentially useful as optical probes for sensitive, multiplexed detection of surface markers on tumor cells. The present thiol-specific conjugation method demonstrates a general approach for site-specific oriented coupling of cys-diabodies to a wide variety of nanoparticles without disturbing the antigen binding site and maintaining small size compared to intact antibody.


Subject(s)
Antibodies/chemistry , Biomarkers, Tumor/analysis , Immunoconjugates/chemistry , Neoplasms/diagnosis , Quantum Dots , Animals , Antibodies/immunology , Antibodies/isolation & purification , Cadmium Compounds/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/immunology , Luminescence , Mice , Polyethylene Glycols/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Selenium Compounds/chemistry , Sulfides/chemistry , Zinc Compounds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...