Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
MAbs ; 15(1): 2212673, 2023.
Article in English | MEDLINE | ID: mdl-37216961

ABSTRACT

Immune checkpoint inhibitors that overcome T cell suppressive mechanisms in tumors have revolutionized the treatment of cancer but are only efficacious in a small subset of patients. Targeting suppressive mechanisms acting on innate immune cells could significantly improve the incidence of clinical response by facilitating a multi-lineage response against the tumor involving both adaptive and innate immune systems. Here, we show that intra-tumoral interleukin (IL)-38 expression is a feature of a large frequency of head and neck, lung and cervical squamous cancers and correlates with reduced immune cell numbers. We generated IMM20324, an antibody that binds human and mouse IL-38 proteins and inhibits the binding of IL-38 to its putative receptors, interleukin 1 receptor accessory protein-like 1 (IL1RAPL) and IL-36R. In vivo, IMM20324 demonstrated a good safety profile, delayed tumor growth in a subset of mice in an EMT6 syngeneic model of breast cancer, and significantly inhibited tumor expansion in a B16.F10 melanoma model. Notably, IMM20324 treatment resulted in the prevention of tumor growth following re-implantation of tumor cells, indicating the induction of immunological memory. Furthermore, exposure of IMM20324 correlated with decreased tumor volume and increased levels of intra-tumoral chemokines. Together, our data suggest that IL-38 is expressed in a high frequency of cancer patients and allows tumor cells to suppress anti-tumor immunity. Blockade of IL-38 activity using IMM20324 can re-activate immunostimulatory mechanisms in the tumor microenvironment leading to immune infiltration, the generation of tumor-specific memory and abrogation of tumor growth.


Subject(s)
Melanoma, Experimental , T-Lymphocytes , Humans , Mice , Animals , Melanoma, Experimental/drug therapy , Immunologic Memory , Tumor Microenvironment , Cell Line, Tumor , Interleukins
2.
Sci Immunol ; 7(75): eabl9943, 2022 09 09.
Article in English | MEDLINE | ID: mdl-35771946

ABSTRACT

Monoclonal antibodies are an efficacious therapy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, rapid viral mutagenesis led to escape from most of these therapies, outlining the need for an antibody cocktail with a broad neutralizing potency. Using an unbiased interrogation of the memory B cell repertoire of patients with convalescent COVID-19, we identified human antibodies with broad antiviral activity in vitro and efficacy in vivo against all tested SARS-CoV-2 variants of concern, including Delta and Omicron BA.1 and BA.2. Here, we describe an antibody cocktail, IMM-BCP-01, that consists of three patient-derived broadly neutralizing antibodies directed at nonoverlapping surfaces on the SARS-CoV-2 Spike protein. Two antibodies, IMM20184 and IMM20190, directly blocked Spike binding to the ACE2 receptor. Binding of the third antibody, IMM20253, to its cryptic epitope on the outer surface of RBD altered the conformation of the Spike Trimer, promoting the release of Spike monomers. These antibodies decreased Omicron SARS-CoV-2 infection in the lungs of Syrian golden hamsters in vivo and potently induced antiviral effector response in vitro, including phagocytosis, ADCC, and complement pathway activation. Our preclinical data demonstrated that the three-antibody cocktail IMM-BCP-01 could be a promising means for preventing or treating infection of SARS-CoV-2 variants of concern, including Omicron BA.1 and BA.2, in susceptible individuals.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Viral , Cricetinae , Humans , Spike Glycoprotein, Coronavirus/genetics
3.
Vaccine X ; 8: 100098, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33937741

ABSTRACT

Patients who recover from SARS-CoV-2 infections produce antibodies and antigen-specific T cells against multiple viral proteins. Here, an unbiased interrogation of the anti-viral memory B cell repertoire of convalescent patients has been performed by generating large, stable hybridoma libraries and screening thousands of monoclonal antibodies to identify specific, high-affinity immunoglobulins (Igs) directed at distinct viral components. As expected, a significant number of antibodies were directed at the Spike (S) protein, a majority of which recognized the full-length protein. These full-length Spike specific antibodies included a group of somatically hypermutated IgMs. Further, all but one of the six COVID-19 convalescent patients produced class-switched antibodies to a soluble form of the receptor-binding domain (RBD) of S protein. Functional properties of anti-Spike antibodies were confirmed in a pseudovirus neutralization assay. Importantly, more than half of all of the antibodies generated were directed at non-S viral proteins, including structural nucleocapsid (N) and membrane (M) proteins, as well as auxiliary open reading frame-encoded (ORF) proteins. The antibodies were generally characterized as having variable levels of somatic hypermutations (SHM) in all Ig classes and sub-types, and a diversity of VL and VH gene usage. These findings demonstrated that an unbiased, function-based approach towards interrogating the COVID-19 patient memory B cell response may have distinct advantages relative to genomics-based approaches when identifying highly effective anti-viral antibodies directed at SARS-CoV-2.

4.
Mol Ther ; 28(2): 548-560, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31870622

ABSTRACT

The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor ß (TGF-ß) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.


Subject(s)
Genital Neoplasms, Female/immunology , Immunotherapy, Adoptive , Ovarian Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Epitopes/genetics , Epitopes/immunology , Female , Genital Neoplasms, Female/therapy , Humans , Mice , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
5.
Mol Pharm ; 14(2): 415-422, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28048942

ABSTRACT

The targeting of therapeutics specifically to diseased tissue is crucial for the development of successful cancer treatments. The approach here is based on the pH(low) insertion peptide (pHLIP) for the delivery of a potent mitotic inhibitor monomethyl auristatin F (MMAF). We investigated six pHLIP variants conjugated to MMAF to compare their efficacy in vitro against cultured cancer cells. While all pHLIP-MMAF conjugates exhibit potent pH- and concentration-dependent killing, their cytotoxicity profiles are remarkably different. We also show that the lead conjugate exhibits significant therapeutic efficacy in mouse models without overt toxicities. This study confirms pHLIP-monomethyl auristatin conjugates as possible new therapeutic options for cancer treatment and supports their further development.


Subject(s)
Aminobenzoates/pharmacology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Oligopeptides/pharmacology , Peptides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Cell Line , Cell Line, Tumor , Female , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Mice, Nude
6.
J Nucl Med ; 58(1): 175-180, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27539844

ABSTRACT

Antibodies, and engineered antibody fragments, labeled with radioisotopes are being developed as radiotracers for the detection and phenotyping of diseases such as cancer. The development of antibody-based radiotracers requires extensive characterization of their in vitro and in vivo properties, including their ability to target tumors in an antigen-selective manner. In this study, we investigated the use of Cerenkov luminescence imaging (CLI) as compared with PET as a modality for evaluating the in vivo behavior of antibody-based radiotracers. METHODS: The anti-prostate-specific membrane antigen (PSMA) huJ591 antibody (IgG; 150 kDa) and its minibody (Mb; 80 kDa) format were functionalized with the chelator 1,4,7-triazacyclononane-1-glutaric acid-4,7-diacetic acid (NODAGA) and radiolabeled with the positron-emitting radionuclide 64Cu (half-life, 12.7 h). Immunoreactive preparations of the radiolabeled antibodies were injected into NCr nu/nu mice harboring PSMA-positive CWR22Rv1 and PSMA-negative PC-3 tumor xenografts. Tumor targeting was evaluated by both PET and CLI. RESULTS: 64Cu-NODAGA-PSMA-IgG and 64Cu-NODAGA-PSMA-Mb retained the ability to bind cell surface PSMA, and both radiotracers exhibited selective uptake into PSMA-positive tumors. Under the experimental conditions used, PSMA-selective uptake of 64Cu-NODAGA-PSMA-IgG and 64Cu-NODAGA-PSMA-Mb was observed by CLI as early as 3 h after injection, with tumor-to-background ratios peaking at 24 (IgG) and 16 (Mb) h after injection. Targeting data generated by CLI correlated with that generated by PET and necropsy. CONCLUSION: CLI provided a rapid and simple assessment of the targeting specificity and pharmacokinetics of the antibody-based PET radiotracers that correlated well with the behavior observed by standard PET imaging. Moreover, CLI provided clear discrimination between uptake kinetics of an intact IgG and its small-molecular-weight derivative Mb. These data support the use of CLI for the evaluation of radiotracer performance.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Luminescent Measurements/methods , Molecular Imaging/methods , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Reproducibility of Results , Sensitivity and Specificity
7.
MAbs ; 7(6): 1058-71, 2015.
Article in English | MEDLINE | ID: mdl-26337947

ABSTRACT

Phage-display technology facilitates rapid selection of antigen-specific single-chain variable fragment (scFv) antibodies from large recombinant libraries. ScFv antibodies, composed of a VH and VL domain, are readily engineered into multimeric formats for the development of diagnostics and targeted therapies. However, the recombinant nature of the selection strategy can result in VH and VL domains with sub-optimal biophysical properties, such as reduced thermodynamic stability and enhanced aggregation propensity, which lead to poor production and limited application. We found that the C10 anti-epidermal growth factor receptor (EGFR) scFv, and its affinity mutant, P2224, exhibit weak production from E. coli. Interestingly, these scFv contain a fusion of lambda3 and lambda1 V-region (LV3 and LV1) genes, most likely the result of a PCR aberration during library construction. To enhance the biophysical properties of these scFvs, we utilized a structure-based approach to replace and redesign the pre-existing framework of the VL domain to one that best pairs with the existing VH. We describe a method to exchange lambda sequences with a more stable kappa3 framework (KV3) within the VL domain that incorporates the original lambda DE-loop. The resulting scFvs, C10KV3_LV1DE and P2224KV3_LV1DE, are more thermodynamically stable and easier to produce from bacterial culture. Additionally, C10KV3_LV1DE and P2224KV3_LV1DE retain binding affinity to EGFR, suggesting that such a dramatic framework swap does not significantly affect scFv binding. We provide here a novel strategy for redesigning the light chain of problematic scFvs to enhance their stability and therapeutic applicability.


Subject(s)
Drug Design , ErbB Receptors/immunology , Immunoglobulin Light Chains/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Antibody Affinity/immunology , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/genetics , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Immunoglobulin lambda-Chains/chemistry , Immunoglobulin lambda-Chains/genetics , Immunoglobulin lambda-Chains/immunology , Models, Molecular , Molecular Sequence Data , Mutation , Peptide Library , Protein Conformation , Protein Engineering/methods , Sequence Homology, Amino Acid , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Thermodynamics
8.
Expert Opin Biol Ther ; 15(7): 1015-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25936923

ABSTRACT

INTRODUCTION: Use of mAbs to inhibit signaling through the ErbB receptor tyrosine kinase family has proven to be an effective strategy for treating ErbB-driven cancers. Advances in the field of antibody engineering and manufacturing now allow us to more effectively mimic the natural immune response by generating oligoclonal mixtures of antibodies against desired targets of interest. AREAS COVERED: In this review, we examine the literature describing the development of oligoclonal mixtures of antibodies against ErbB family members and the impact of those mixtures on preclinical and clinical efficacy. EXPERT OPINION: Oligoclonal antibodies, facilitated by the improved antibody engineering and manufacturing techniques, hold the promise of improving patient outcomes. Through the use of empirical methods, oligoclonal mixtures with enhanced capacity to block signaling through ErbB family members can be identified. The intrinsic mechanisms associated with each of the component mAbs provide an opportunity to block signaling via multiple mechanisms of action. In addition, combinations of antibodies targeting multiple ErbB family members provide the capacity to down-regulate signaling through multiple components of this critical pathway.


Subject(s)
Antibodies, Monoclonal/therapeutic use , ErbB Receptors/immunology , Neoplasms/drug therapy , Antibodies, Monoclonal/immunology , Humans , Protein Engineering , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Signal Transduction
9.
BMC Cancer ; 15: 436, 2015 May 28.
Article in English | MEDLINE | ID: mdl-26016476

ABSTRACT

BACKGROUND: Overexpression or mutation of the epidermal growth factor receptor (EGFR) potently enhances the growth of many solid tumors. Tumor cells frequently display resistance to mechanistically-distinct EGFR-directed therapeutic agents, making it valuable to develop therapeutics that work by additional mechanisms. Current EGFR-targeting therapeutics include antibodies targeting the extracellular domains, and small molecules inhibiting the intracellular kinase domain. Recent studies have identified a novel prone extracellular tetrameric EGFR configuration, which we identify as a potential target for drug discovery. METHODS: Our focus is on the prone EGFR tetramer, which contains a novel protein-protein interface involving extracellular domain III. This EGFR tetramer is computationally targeted for stabilization by small molecule ligand binding. This study performed virtual screening of a Life Chemicals, Inc. small molecule library of 345,232 drug-like compounds against a molecular dynamics simulation of protein-protein interfaces distinct to the novel tetramer. One hundred nine chemically diverse candidate molecules were selected and evaluated using a cell-based high-content imaging screen that directly assessed induced internalization of the EGFR effector protein Grb2. Positive hits were further evaluated for influence on phosphorylation of EGFR and its effector ERK1/2. RESULTS: Fourteen hit compounds affected internalization of Grb2, an adaptor responsive to EGFR activation. Most hits had limited effect on cell viability, and minimally influenced EGFR and ERK1/2 phosphorylation. Docked hit compound poses generally include Arg270 or neighboring residues, which are also involved in binding the effective therapeutic cetuximab, guiding further chemical optimization. CONCLUSIONS: These data suggest that the EGFR tetrameric configuration offers a novel cancer drug target.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , GRB2 Adaptor Protein/metabolism , Head and Neck Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cetuximab/pharmacology , Drug Evaluation, Preclinical , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Phosphorylation/drug effects , Protein Interaction Domains and Motifs , Signal Transduction
10.
Mol Pharm ; 12(4): 1250-8, 2015 Apr 06.
Article in English | MEDLINE | ID: mdl-25741818

ABSTRACT

Localized delivery is vital for the successful development of novel and effective therapeutics for the treatment of cancer. The targeting and delivery described herein is based on the pH (low) insertion peptide (pHLIP), a unique delivery peptide that can selectively target tumors in mice and translocate and release cargo molecules intracellularly based solely on the low extracellular pH intrinsic to cancer cells. In this study, we investigate the efficacy of pHLIP to target and deliver the highly potent and clinically validated microtubule inhibitor monomethyl auristatin E (MMAE) to cancer cells and breast tumors. We show that pHLIP-MMAE conjugates induce a potent cytotoxic effect (>90% inhibition of cell growth) in a concentration- and pH-dependent manner after only 2 h incubation without any apparent disruption of the plasma membrane. pHLIP-MMAE conjugates exhibit between an 11- and 144-fold higher antiproliferative effect at low pH than that at physiological pH and a pronounced pH-dependent cytotoxicity as compared to that of free drug. Furthermore, we demonstrate that a pHLIP-MMAE drug conjugate effectively targets triple-negative breast tumor xenografts in mice. These results indicate that pHLIP-based auristatin conjugates may have an enhanced therapeutic window as compared to that of free drug, providing a targeting mechanism to attenuate systemic toxicity.


Subject(s)
Membrane Proteins/chemistry , Oligopeptides/chemistry , Triple Negative Breast Neoplasms/drug therapy , Aminobenzoates/chemistry , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Chemistry, Pharmaceutical/methods , Circular Dichroism , Drug Delivery Systems , Female , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Neoplasm Transplantation , Peptides/chemistry , Phosphatidylcholines/chemistry , Spectrometry, Fluorescence , Tryptophan/chemistry
11.
PLoS One ; 9(11): e112376, 2014.
Article in English | MEDLINE | ID: mdl-25386657

ABSTRACT

BACKGROUND: Inappropriate signaling through the epidermal growth factor receptor family (EGFR1/ERBB1, ERBB2/HER2, ERBB3/HER3, and ERBB4/HER4) of receptor tyrosine kinases leads to unregulated activation of multiple downstream signaling pathways that are linked to cancer formation and progression. In particular, ERBB3 plays a critical role in linking ERBB signaling to the phosphoinositide 3-kinase and Akt signaling pathway and increased levels of ERBB3-dependent signaling is also increasingly recognized as a mechanism for acquired resistance to ERBB-targeted therapies. METHODS: We had previously reported the isolation of a panel of anti-ERBB3 single-chain Fv antibodies through use of phage-display technology. In the current study scFv specific for domain I (F4) and domain III (A5) were converted into human IgG1 formats and analyzed for efficacy. RESULTS: Treatment of cells with an oligoclonal mixture of the A5/F4 IgGs appeared more effective at blocking both ligand-induced and ligand-independent signaling through ERBB3 than either single IgG alone. This correlated with improved ability to inhibit the cell growth both as a single agent and in combination with other ERBB-targeted therapies. Treatment of NCI-N87 tumor xenografts with the A5/F4 oligoclonal led to a statistically significant decrease in tumor growth rate that was further enhanced in combination with trastuzumab. CONCLUSION: These results suggest that an oligoclonal antibody mixture may be a more effective approach to downregulate ERBB3-dependent signaling.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Immunoglobulin G/pharmacology , Receptor, ErbB-3/immunology , Signal Transduction/drug effects , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Surface Display Techniques , HEK293 Cells , Heterografts/drug effects , Humans , Immunoglobulin G/chemistry , Immunotherapy/methods , Male , Mice, Nude , Neoplasms/therapy , Receptor, ErbB-3/chemistry , Single-Chain Antibodies/chemistry
12.
FASEB J ; 27(2): 581-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23104982

ABSTRACT

Fibroblast activation protein (FAP) is a serine protease selectively expressed on tumor stromal fibroblasts in epithelial carcinomas and is important in cancer growth, adhesion, and metastases. As FAP enzymatic activity is a potent therapeutic target, we aimed to identify inhibitory antibodies. Using a competitive inhibition strategy, we used phage display techniques to identify 53 single-chain variable fragments (scFvs) after three rounds of panning against FAP. These scFvs were expressed and characterized for binding to FAP by surface plasmon resonance and flow cytometry. Functional assessment of these antibodies yielded an inhibitory scFv antibody, named E3, which could attenuate 35% of FAP cleavage of the fluorescent substrate Ala-Pro-7-amido-4-trifluoromethylcoumarin compared with nonfunctional scFv control. Furthermore, a mutant E3 scFv was identified by yeast affinity maturation. It had higher affinity (4-fold) and enhanced inhibitory effect on FAP enzyme activity (3-fold) than E3. The application of both inhibitory anti-FAP scFvs significantly affected the formation of 3-dimensional FAP-positive cell matrix, as demonstrated by reducing the fibronectin fiber orientation from 41.18% (negative antibody control) to 34.06% (E3) and 36.15% (mutant E3), respectively. Thus, we have identified and affinity-maturated the first scFv antibody capable of inhibiting FAP function. This scFv antibody has the potential to disrupt the role of FAP in tumor invasion and metastasis.


Subject(s)
Gelatinases/antagonists & inhibitors , Gelatinases/immunology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Serine Endopeptidases/immunology , Single-Chain Antibodies/immunology , Animals , Antibody Affinity , Endopeptidases , Flow Cytometry , Gelatinases/genetics , Gelatinases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Targeted Therapy , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/immunology , Neoplasms/drug therapy , Neoplasms/enzymology , Peptide Library , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism , Surface Plasmon Resonance
13.
ACS Nano ; 6(6): 5143-9, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22575126

ABSTRACT

We developed a novel detection method for osteopontin (OPN), a new biomarker for prostate cancer, by attaching a genetically engineered single-chain variable fragment (scFv) protein with high binding affinity for OPN to a carbon nanotube field-effect transistor (NT-FET). Chemical functionalization using diazonium salts is used to covalently attach scFv to NT-FETs, as confirmed by atomic force microscopy, while preserving the activity of the biological binding site for OPN. Electron transport measurements indicate that functionalized NT-FET may be used to detect the binding of OPN to the complementary scFv protein. A concentration-dependent increase in the source-drain current is observed in the regime of clinical significance, with a detection limit of approximately 30 fM. The scFv-NT hybrid devices exhibit selectivity for OPN over other control proteins. These devices respond to the presence of OPN in a background of concentrated bovine serum albumin, without loss of signal. On the basis of these observations, the detection mechanism is attributed to changes in scattering at scFv protein-occupied defect sites on the carbon nanotube sidewall. The functionalization procedure described here is expected to be generalizable to any antibody containing an accessible amine group and to result in biosensors appropriate for detection of corresponding complementary proteins at fM concentrations.


Subject(s)
Biomarkers, Tumor/blood , Immunoassay/instrumentation , Nanotubes, Carbon/chemistry , Osteopontin/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/diagnosis , Transistors, Electronic , Animals , Antibodies/genetics , Cattle , Conductometry/instrumentation , Equipment Design , Equipment Failure Analysis , Humans , Male , Nanotubes, Carbon/ultrastructure , Protein Engineering/methods
14.
Tumour Biol ; 33(3): 617-27, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22383295

ABSTRACT

The ability of engineered antibodies to rapidly and selectively target tumors that express their target antigen makes them well suited for use as radioimaging tracers. The combination of molecular size and bivalent nature makes diabody molecules a particularly promising structure for use as radiotracers for diagnostic imaging. Previous data have demonstrated that the anti-HER2 C6.5 diabody (C6.5db) is an effective radiotracer in preclinical models of HER2-positive cancer. The aim of this study was to evaluate the impact on radiotracer performance, associated with expressing the C6.5db in the Pichia pastoris (P-C6.5db) system as compared to Escherichia coli (E. C6.5db). Glycosylation of P-C6.5db led to faster blood clearance and lower overall tumor uptake than seen with E. coli-produced C6.5db. However, P-C6.5db achieved high tumor/background ratios that are critical for effective imaging. Dosimetry measurements determined in this study for both (124)I-P-C6.5db and (124)I-E-C6.5db suggest that they are equivalent to other radiotracers currently being administered to patients.


Subject(s)
Multimodal Imaging , Neoplasms, Experimental/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals , Receptor, ErbB-2/immunology , Single-Chain Antibodies , Tomography, X-Ray Computed , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Mice, SCID , Radiometry , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacokinetics , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacokinetics , Time Factors , Transplantation, Heterologous , Yeasts/metabolism
15.
Drug Resist Updat ; 14(6): 260-79, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21920801

ABSTRACT

Agents targeting EGFR and related ErbB family proteins are valuable therapies for the treatment of many cancers. For some tumor types, including squamous cell carcinomas of the head and neck (SCCHN), antibodies targeting EGFR were the first protein-directed agents to show clinical benefit, and remain a standard component of clinical strategies for management of the disease. Nevertheless, many patients display either intrinsic or acquired resistance to these drugs; hence, major research goals are to better understand the underlying causes of resistance, and to develop new therapeutic strategies that boost the impact of EGFR/ErbB inhibitors. In this review, we first summarize current standard use of EGFR inhibitors in the context of SCCHN, and described new agents targeting EGFR currently moving through pre-clinical and clinical development. We then discuss how changes in other transmembrane receptors, including IGF1R, c-Met, and TGF-ß, can confer resistance to EGFR-targeted inhibitors, and discuss new agents targeting these proteins. Moving downstream, we discuss critical EGFR-dependent effectors, including PLC-γ; PI3K and PTEN; SHC, GRB2, and RAS and the STAT proteins, as factors in resistance to EGFR-directed inhibitors and as alternative targets of therapeutic inhibition. We summarize alternative sources of resistance among cellular changes that target EGFR itself, through regulation of ligand availability, post-translational modification of EGFR, availability of EGFR partners for hetero-dimerization and control of EGFR intracellular trafficking for recycling versus degradation. Finally, we discuss new strategies to identify effective therapeutic combinations involving EGFR-targeted inhibitors, in the context of new system level data becoming available for analysis of individual tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Head and Neck Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Drug Design , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Head and Neck Neoplasms/pathology , Humans , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
16.
Anal Chem ; 83(9): 3392-7, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21449604

ABSTRACT

Rapid and sensitive detection of serum tumor biomarkers are needed to monitor cancer patients for disease progression. Highly sensitive piezoelectric microcantilever sensors (PEMS) offer an attractive tool for biomarker detection; however, their utility in the complex environment encountered in serum has yet to be determined. As a proof of concept, we have functionalized PEMS with antibodies that specifically bind to HER2, a biomarker (antigen) that is commonly overexpressed in the blood of breast cancer patients. The function and sensitivity of these anti-HER2 PEMS biosensors was initially assessed using recombinant HER2 spiked into human serum. Their ability to detect native HER2 present in the serum of breast cancer patients was then determined. We have found that the anti-HER2 PEMS were able to accurately detect both recombinant and naturally occurring HER2 at clinically relevant levels (>2 ng/mL). This indicates that PEMS-based biosensors provide a potentially effective tool for biomarker detection.


Subject(s)
Blood Chemical Analysis/methods , Breast Neoplasms/blood , Electricity , Extracellular Space , Receptor, ErbB-2/blood , Receptor, ErbB-2/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/blood , Biomarkers, Tumor/chemistry , Case-Control Studies , Humans , Male , Protein Structure, Tertiary , Receptor, ErbB-2/immunology , Trastuzumab
17.
Clin Cancer Res ; 17(6): 1509-20, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21177408

ABSTRACT

PURPOSE: The rapid tumor targeting and pharmacokinetic properties of engineered antibodies make them potentially suitable for use in imaging strategies to predict and monitor response to targeted therapies. This study aims to evaluate C6.5 diabody (C6.5 db), a noncovalent anti-HER2 single-chain Fv dimer, as a radiotracer for predicting response to HER2-targeted therapies such as trastuzumab. EXPERIMENTAL DESIGN: Immunodeficient mice bearing established HER2-positive tumor xenografts were injected with radioiodinated C6.5 db and imaged by PET/CT. Radiotracer biodistribution was quantified by biopsied tumor and normal tissues. Potential competition between trastuzumab and C6.5 db was examined in vitro by flow cytometry and coimmunoprecipitations. RESULTS: Biodistribution analysis of mice bearing xenografts with varying HER2 density revealed that the tumor uptake of (125)I-C6.5 db correlates with HER2 tumor density. In vitro competition experiments suggest that the C6.5 db targets an epitope on HER2 that is distinct from that bound by trastuzumab. Treatment of mice affected with SK-OV-3 tumor with trastuzumab for 3 days caused a 42% (P = 0.002) decrease in tumor uptake of (125)I-C6.5 db. This is consistent with a dramatic decrease in the tumor PET signal of (124)I-C6.5 db after trastuzumab treatment. Furthermore, mice affected with BT-474 tumor showed an approximately 60% decrease (P = 0.0026) in C6.5 db uptake after 6 days of trastuzumab treatment. Immunohistochemistry of excised xenograft sections and in vitro flow cytometry revealed that the decreased C6.5 db uptake on trastuzumab treatment is not associated with HER2 downregulation. CONCLUSIONS: These studies suggest that (124)I-C6.5 db-based imaging can be used to evaluate HER2 levels as a predictor of response to HER2-directed therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Genes, erbB-2 , Positron-Emission Tomography/methods , Receptor, ErbB-2/chemistry , Single-Chain Antibodies/chemistry , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Biopsy , Cell Line, Tumor , Down-Regulation , HEK293 Cells , Humans , Immunoglobulin Fragments/chemistry , Mice , Mice, SCID , Neoplasm Transplantation , Radiopharmaceuticals/pharmacology , Receptor, ErbB-2/immunology , Tomography, X-Ray Computed/methods , Trastuzumab
18.
Semin Nucl Med ; 40(3): 182-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20350627

ABSTRACT

Positron emission tomography (PET) is playing an increasingly important role in the diagnosis, staging, and monitoring response to treatment in a variety of cancers. Recent efforts have focused on immuno-PET, which uses antibody-based radiotracers, to image tumors based on expression of tumor-associated antigens. It is postulated that the specificity afforded by antibody targeting should both improve tumor detection and provide phenotypic information related to primary and metastatic lesions that will guide therapy decisions. Advances in antibody-engineering are providing the tools to develop antibody-based molecules with pharmacokinetic properties optimized for use as immuno-PET radiotracers. Coupled with technical advances in the design of PET scanners, immuno-PET holds promise to improve diagnostic imaging and to guide the use of targeted therapies. An overview of the preclinical immuno-PET studies in cancer models is reviewed here.


Subject(s)
Antibodies, Monoclonal , Disease Models, Animal , Molecular Probe Techniques/trends , Neoplasms/diagnostic imaging , Radioimmunodetection/trends , Radiopharmaceuticals , Tomography, Emission-Computed/trends , Animals , Antibodies, Monoclonal/chemistry , Humans , Image Enhancement/methods , Radiopharmaceuticals/chemistry
19.
Expert Opin Biol Ther ; 10(2): 273-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20053143

ABSTRACT

IMPORTANCE OF THE FIELD: New agents that specifically engage the immune system are being tested in a variety of malignancies. This review provides an overview of naptumomab, an immunotoxin, with encouraging clinical activity in Phase I trials. AREAS COVERED IN THIS REVIEW: This review examines the preclinical and the published clinical data with regards to naptumomab. WHAT THE READER WILL GAIN: This review provides the reader with an understanding of the mechanism of action, immunology, pharmacokinetics and clinical activity of this agent. TAKE HOME MESSAGE: Naptumomab has a unique mechanism of action and appears to be an active agent in the treatment of refractory solid tumors such as renal cell carcinoma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Enterotoxins/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Clinical Trials, Phase I as Topic , Drug Design , Enterotoxins/adverse effects , Enterotoxins/chemistry , Enterotoxins/pharmacokinetics , Enterotoxins/pharmacology , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology
20.
Cancer J ; 14(3): 149-53, 2008.
Article in English | MEDLINE | ID: mdl-18536554

ABSTRACT

Antibodies have emerged as significant agents for the treatment of a number of diseases including cancer and autoimmunity. However, most of the antibodies currently used in clinical practice were developed from humanized or chimeric molecules based on mouse monoclonal antibodies. Recent advances in antibody selection and engineering techniques have led to the development of antibodies specific for highly conserved targets, the creation of novel antibody-based structures, significant improvements in affinity for target antigens, enhanced ability to engage immune effector functions, and the creation of fusion proteins with direct cytotoxic properties. This review provides an overview of the techniques that we expect will have the greatest impact on the field of antibody engineering.


Subject(s)
Antibodies/immunology , Antibodies/pharmacology , Genetic Engineering , Tissue Engineering , Animals , Antibody Formation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...