Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 10(6)2022 06.
Article in English | MEDLINE | ID: mdl-35728872

ABSTRACT

BACKGROUND: Despite clinical success with T cell engagers (TCEs) targeting hematological malignancies, achieving a safe and efficacious dose in patients with solid tumors remains challenging. Due to potency, low levels of target antigen expression on normal tissues may not be tolerated. To overcome this, we engineered a novel conditionally active TCE design called COBRA (Conditional Bispecific Redirected Activation). Administered as prodrugs, COBRAs bind to cell surface antigens on both normal and tumor tissues but are preferentially activated within the tumor microenvironment. METHODS: A COBRA was engineered to target EGFR, TAK-186. The potency of precleaved TAK-186 relative to a non-cleavable control was assessed in vitro. Mice bearing established solid tumors expressing a range of EGFR levels were administered a single bolus of human T cells, and concurrently treated with TAK-186 and associated controls intravenously. We assessed the plasma and tumor exposure of intact and cleaved TAK-186. RESULTS: TAK-186 shows potent redirected T cell killing of antigen expressing tumor cells. In vivo efficacy studies demonstrate regressions of established solid tumors, dependent on intratumoral COBRA cleavage. Pharmacokinetic studies reveal TAK-186 is stable in circulation, but once activated is rapidly cleared due to loss of its albumin-binding half-life extension domain. CONCLUSIONS: The studies shown support the advancement of TAK-186, and the pursuit of additional COBRA TCEs for the treatment of solid tumors.


Subject(s)
ErbB Receptors , Neoplasms , T-Lymphocytes , Animals , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Immunotherapy , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment
2.
Antib Ther ; 5(1): 1-10, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35005430

ABSTRACT

BACKGROUND: COBRA™ (COnditional Bispecific Redirected Activation) T-cell engagers are designed to target solid tumors as a single polypeptide chain prodrug that becomes activated by proteolysis in the tumor microenvironment. One COBRA molecule comprises seven Ig domains: three single-domain antibodies (sdAbs) recognizing a tumor target or human serum albumin (HSA), and CD3ε-binding variable fragment heavy chain (VH) and variable fragment light chain (VL) and their inactivated counterparts, VHi and VLi. Pairing of VH and VL, and VLi and VHi into single-chain variable fragments (Fv) is prevented by shortened inter-domain linkers. Instead, VH and VL are expected to interact with VLi and VHi, respectively, thus making a diabody whose binding to CD3ε on the T-cells is impaired. METHODS: We analyzed the structure of an epidermal growth factor receptor (EGFR) COBRA in solution using negative stain electron microscopy (EM) and small-angle X-ray scattering (SAXS). RESULTS: We found that this EGFR COBRA forms stable monomers with a very dynamic interdomain arrangement. At most, only five domains at a time appeared ordered, and only one VH-VL pair was found in the Fv orientation. Nonenzymatic posttranslational modifications suggest that the CDR3 loops in the VL-VHi pair are exposed but are buried in the VH-VLi pair. The MMP9 cleavage rate of the prodrug when bound to recombinant EGFR or HSA is not affected, indicating positioning of the MMP9-cleavable linker away from the EGFR and HSA binding sites. CONCLUSION: Here, we propose a model for EGFR COBRA where VH and VLi form an Fv, and VL and VHi do not, possibly interacting with other Ig domains. SAXS and MMP9 cleavage analyses suggest that all COBRA molecules tested have a similar structural architecture.

3.
Biochem Biophys Rep ; 29: 101205, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35071801

ABSTRACT

CRISPR/Cas9 gene-editing technology allows researchers to study protein function by specifically introducing double-stranded breaks in the gene of interest then analyze its subsequent loss in sensitive biological assays. To help characterize one of a series of highly potent, conditionally active, T cell engaging bispecific molecules called COBRA™, the human EpCAM gene was disrupted in HT29 cells using CRISPR/Cas9 and guide RNA targeting its Exon 2. Although a commercially available antibody indicated loss of cell-surface expression, the EpCAM targeting bispecific COBRA was still able to lyse these cells in a T cell dependent cellular cytotoxicity assay. RT-PCR sequence analysis of these cells showed a major alternative transcript generated after CRISPR/Cas9, with Exon 1 and 3 spliced together in-frame, skipping Exon 2 completely, to express a truncated cell-surface receptor recognized by the EpCAM-COBRA. Researchers who use CRISPR/Cas9 must be cognizant of this potential to express alternative versions of their proteins and use sensitive orthogonal detection methods to ensure complete gene disruption.

4.
Mol Cancer Ther ; 20(1): 109-120, 2021 01.
Article in English | MEDLINE | ID: mdl-33203731

ABSTRACT

T cells have a unique capability to eliminate cancer cells and fight malignancies. Cancer cells have adopted multiple immune evasion mechanisms aimed at inhibiting T cells. Dramatically improved patient outcomes have been achieved with therapies genetically reprogramming T cells, blocking T-cell inhibition by cancer cells, or transiently connecting T cells with cancer cells for redirected lysis. This last modality is based on antibody constructs that bind a surface antigen on cancer cells and an invariant component of the T-cell receptor. Although high response rates were observed with T-cell engagers specific for CD19, CD20, or BCMA in patients with hematologic cancers, the treatment of solid tumors has been less successful. Here, we developed and characterized a novel T-cell engager format, called TriTAC (for Trispecific T-cell Activating Construct). TriTACs are engineered with features to improve patient safety and solid tumor activity, including high stability, small size, flexible linkers, long serum half-life, and highly specific and potent redirected lysis. The present study establishes the structure/activity relationship of TriTACs and describes the development of HPN424, a PSMA- (FOLH1-) targeting TriTAC in clinical development for patients with metastatic castration-resistant prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , T-Lymphocytes/metabolism , Albumins/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , CD3 Complex/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Half-Life , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca fascicularis , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Prostate-Specific Antigen/metabolism , T-Lymphocytes/drug effects
5.
MAbs ; 12(1): 1792130, 2020.
Article in English | MEDLINE | ID: mdl-32684124

ABSTRACT

Conditionally active COBRA™ (COnditional Bispecific Redirected Activation) T cell engagers are engineered to overcome the limitations of inherently active first-generation T cell engagers, which are unable to discern between tumor and healthy tissues. Designed to be administered as prodrugs, COBRAs target cell surface antigens upon administration, but engage T cells only after they are activated within the tumor microenvironment (TME). This allows COBRAs to be preferentially turned on in tumors while safely remaining inactive in healthy tissue. Here, we describe the development of the COBRA design and the characterization of these conditionally active T cell engagers. Upon administration COBRAs are engineered to bind to tumor-associated antigens (TAAs) and serum albumin (to extend their half-life in circulation), but are inhibited from interacting with the T cell receptor complex signaling molecule CD3. In the TME, a matrix metalloproteinase (MMP)-mediated linker cleavage event occurs within the COBRA construct, which rearranges the molecule, allowing it to co-engage TAAs and CD3, thereby activating T cells against the tumor. COBRAs are conditionally activated through cleavage with MMP9, and once active are highly potent, displaying sub-pM EC50s in T cell killing assays. Studies in tumor-bearing mice demonstrate COBRA administration completely regresses established solid tumor xenografts. These results strongly support the further characterization of the novel COBRA design in preclinical development studies.


Subject(s)
Antibodies, Bispecific , Antigens, Neoplasm , Antineoplastic Agents, Immunological , Immunotherapy , Lymphocyte Activation , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , HT29 Cells , Humans , Jurkat Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/immunology , Protein Engineering , Xenograft Model Antitumor Assays
6.
MAbs ; 5(4): 523-32, 2013.
Article in English | MEDLINE | ID: mdl-23765106

ABSTRACT

We developed a method for deep mutational scanning of antibody complementarity-determining regions (CDRs) that can determine in parallel the effect of every possible single amino acid CDR substitution on antigen binding. The method uses libraries of full length IgGs containing more than 1000 CDR point mutations displayed on mammalian cells, sorted by flow cytometry into subpopulations based on antigen affinity and analyzed by massively parallel pyrosequencing. Higher, lower and neutral affinity mutations are identified by their enrichment or depletion in the FACS subpopulations. We applied this method to a humanized version of the anti-epidermal growth factor receptor antibody cetuximab, generated a near comprehensive data set for 1060 point mutations that recapitulates previously determined structural and mutational data for these CDRs and identified 67 point mutations that increase affinity. The large-scale, comprehensive sequence-function data sets generated by this method should have broad utility for engineering properties such as antibody affinity and specificity and may advance theoretical understanding of antibody-antigen recognition.


Subject(s)
Amino Acid Substitution , Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Complementarity Determining Regions , ErbB Receptors/immunology , Mutation, Missense , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cetuximab , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , ErbB Receptors/genetics , HEK293 Cells , Humans
7.
J Immunol ; 189(9): 4470-7, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018459

ABSTRACT

CTLA4-Ig is an Fc fusion protein containing the extracellular domain of CTLA-4, a receptor known to deliver a negative signal to T cells. CTLA4-Ig modulates T cell costimulatory signals by blocking the CD80 and CD86 ligands from binding to CD28, which delivers a positive T cell costimulatory signal. To engineer CTLA4-Ig variants with altered binding affinity to CD80 and CD86, we employed a high-throughput protein engineering method to map the ligand binding surface of CTLA-4. The resulting mutagenesis map identified positions critical for the recognition of each ligand on the three CDR-like loops of CTLA-4, consistent with the published site-directed mutagenesis and x-ray crystal structures of the CTLA-4/CD80 and CTLA-4/CD86 complexes. A number of single amino acid substitutions were identified that equally affected the binding affinity of CTLA4-Ig for both ligands as well as those that differentially affected binding. All of the high-affinity variants showed improved off-rates, with the best one being a 17.5-fold improved off-rate over parental CTLA4-Ig binding to CD86. Allostimulation of human CD4(+) T cells showed that improvement of CD80 and CD86 binding activity augmented inhibition of naive and primed T cell activation. In general, increased affinity for CD86 resulted in more potent inhibition of T cell response than did increased affinity for CD80. Optimization of the affinity balance to CD80 and CD86 to particular disease settings may lead to development of a CTLA4-Ig molecule with improved efficacy and safety profiles.


Subject(s)
Immunoconjugates/genetics , Immunoconjugates/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Abatacept , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , B7-1 Antigen/antagonists & inhibitors , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/biosynthesis , B7-2 Antigen/genetics , CHO Cells , Cricetinae , Cricetulus , Cross Reactions/genetics , Cross Reactions/immunology , Genes, Synthetic/immunology , HEK293 Cells , Humans , Immunoconjugates/therapeutic use , Jurkat Cells , Peptide Library , Plasmids/genetics , Plasmids/immunology , Protein Binding/genetics , Protein Binding/immunology
8.
Proteomics Clin Appl ; 4(8-9): 726-38, 2010 Sep.
Article in English | MEDLINE | ID: mdl-21137090

ABSTRACT

PURPOSE: We investigated the ability to perform a clinical proteomic study using samples collected at different times from two independent clinical sites. EXPERIMENTAL DESIGN: Label-free 2-D-LC-MS proteomic analysis was used to differentially quantify tens of thousands of peptides from human plasma. We have asked whether samples collected from two sites, when analyzed by this type of peptide profiling, reproducibly contain detectable peptide markers that are differentially expressed in the plasma of disease (advanced renal cancer) patients relative to healthy normals. RESULTS: We have demonstrated that plasma proteins enriched in disease patients are indeed detected reproducibly in both clinical collections. Regression analysis, unsupervised hierarchical clustering and PCA detected no systematic bias in the data related to site of sample collection and processing. Using a genetic algorithm, support vector machine classification method, we were able to correctly classify disease samples at 88% sensitivity and 94% specificity using the second site as an independent validation set. CONCLUSIONS AND CLINICAL RELEVANCE: We conclude that multiple site collection, when analyzed by label-free 2-D-LC-MS, generates data that are sufficiently reproducible to guide reliable biomarker discovery.


Subject(s)
Biomarkers/analysis , Blood Proteins/analysis , Proteomics/methods , Specimen Handling/methods , Biomarkers/metabolism , Blood Proteins/metabolism , Chromatography, Liquid/methods , Humans , Mass Spectrometry/methods , Principal Component Analysis , Regression Analysis , Reproducibility of Results , Sensitivity and Specificity , Time Factors
9.
MAbs ; 2(3): 256-65, 2010.
Article in English | MEDLINE | ID: mdl-20400861

ABSTRACT

Monoclonal antibodies represent an attractive therapeutic tool as they are highly specific for their targets, convey effector functions and enjoy robust manufacturing procedures. Humanization of murine monoclonal antibodies has vastly improved their in vivo tolerability. Humanization, the replacement of mouse constant regions and V framework regions for human sequences, results in a significantly less immunogenic product. However, some humanized and even fully human sequence-derived antibody molecules still carry immunological risk. To more fully understand the immunologic potential of humanized and human antibodies, we analyzed CD4(+) helper T cell epitopes in a set of eight humanized antibodies. The antibodies studied represented a number of different VH and VL family members carrying unique CDR regions. In spite of these differences, CD4(+) T cell epitopes were found only in CDR-sequence containing regions. We were able to incorporate up to two amino acid modifications in a single epitope that reduced the immunogenic potential while retaining full biologic function. We propose that immunogenicity will always be present in some antibody molecules due to the nature of the antigen-specific combining sites. A consequence of this result is modifications to reduce immunogenicity will be centered on the affinity-determining regions. Modifications to CDR regions can be designed that reduce the immunogenic potential while maintaining the bioactivity of the antibody molecule.


Subject(s)
Antibodies, Monoclonal/immunology , Complementarity Determining Regions/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibody Affinity , Antibody Specificity , Cells, Cultured , Complementarity Determining Regions/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Mice , Protein Engineering , T-Lymphocytes, Helper-Inducer/immunology
10.
Clin Cancer Res ; 16(2): 497-508, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068083

ABSTRACT

PURPOSE: Targeted therapeutics have significantly changed the outcome for patients diagnosed with cancer. Still, effective therapeutic intervention does not exist for many cancers and much remains to be done. The objective of this study was to identify novel genes that potentially regulate tumor growth, to target these gene products with monoclonal antibodies, and to examine the therapeutic potential of these antibodies. EXPERIMENTAL DESIGN: Using cDNA microarray analysis, we identified genes overexpressed in several solid malignancies. We generated a mouse monoclonal antibody, 19.2.1, and its humanized counterpart, PDL192, to one such target, TweakR (TWEAK receptor, Fn14, TNFRSF12A, CD266), and characterized the antitumor activities in vitro and in mouse xenograft models. RESULTS: Both 19.2.1 (mouse IgG2a) and PDL192 (human IgG1), like TWEAK, the natural ligand of TweakR, inhibited the growth of several TweakR-expressing cancer cell lines in anchorage-dependent and anchorage-independent assays in vitro. Both antibodies showed significant antitumor activity in multiple mouse xenograft models. PDL192 and 19.2.1 also induced antibody-dependent cellular cytotoxicity (ADCC) of cancer cell lines in vitro. A chimeric version of 19.2.1 containing the mouse IgG1 Fc region (19.2.1 x G1) exhibited significantly less ADCC than 19.2.1. However, 19.2. 1x G1 showed differential activity in vivo, with activity equivalent to 19.2.1 in one model, but significantly less efficacy than 19.2.1 in a second model. These results indicate that PDL192 and 19.2.1 mediate their antitumor effects by signaling through TweakR, resulting in reduced tumor cell proliferation, and by ADCC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cell Proliferation/drug effects , Neoplasms/pathology , Tumor Necrosis Factors/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Cytokine TWEAK , Dose-Response Relationship, Drug , Humans , Immunotherapy/methods , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Neoplasm Metastasis , Neoplasms/therapy , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Invest New Drugs ; 26(1): 7-12, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17786386

ABSTRACT

Angiogenesis, the process by which new blood vessels form from existing vasculature, is critical for tumor growth and invasion. Growth factors, such as VEGF, initiate signaling cascades resulting in the proliferation of resting endothelial cells. Blockade of growth factor pathways has proven effective in inhibiting angiogenesis and tumor growth in vivo. Integrins, including the integrin alpha5beta1, are also important mediators of angiogenesis and these adhesion molecules also regulate cancer cell growth and migration in vitro. Volociximab is a high affinity, function-blocking antibody against integrin alpha5beta1 that is currently in multiple Phase II oncology clinical trials. Volociximab displays potent anti-angiogenic activity in a monkey model of choroidal neovascularization. In this study, we explored the consequences of integrin alpha5beta1 blockade on tumorigenesis. Because volociximab does not cross-react with rodent alpha5beta1, the syngeneic rabbit VX2 carcinoma model was utilized as an alternative to standard mouse xenograft models for the assessment of anti-tumor activity of volociximab. Volociximab administered intravenously to rabbits bearing VX2 tumors is detectable on tumor cells and vasculature 45 min post-administration. Volociximab was found to significantly inhibit the growth of tumors growing subcutaneously or intramuscularly, despite a 20-fold lower affinity for rabbit integrin, relative to human. This effect was found to correlate with decreased blood vessel density within these tumors. These results support the use of volociximab in the intervention of malignant disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Integrin alpha5beta1/antagonists & inhibitors , Neoplasms, Experimental/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Antibody Specificity , Clinical Trials, Phase III as Topic , Dose-Response Relationship, Drug , Fibronectins/metabolism , Humans , Immunochemistry , Injections, Intravenous , Integrin alpha5beta1/immunology , Integrin alpha5beta1/metabolism , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Binding , Rabbits , Species Specificity , Surface Plasmon Resonance/methods , Tumor Burden/drug effects
12.
J Transl Med ; 5: 61, 2007 Nov 27.
Article in English | MEDLINE | ID: mdl-18042290

ABSTRACT

BACKGROUND: Integrins are important adhesion molecules that regulate tumor and endothelial cell survival, proliferation and migration. The integrin alpha5beta1 has been shown to play a critical role during angiogenesis. An inhibitor of this integrin, volociximab (M200), inhibits endothelial cell growth and movement in vitro, independent of the growth factor milieu, and inhibits tumor growth in vivo in the rabbit VX2 carcinoma model. Although volociximab has already been tested in open label, pilot phase II clinical trials in melanoma, pancreatic and renal cell cancer, evaluation of the mechanism of action of volociximab has been limited because this antibody does not cross-react with murine alpha5beta1, precluding its use in standard mouse xenograft models. METHODS: We generated a panel of rat-anti-mouse alpha5beta1 antibodies, with the intent of identifying an antibody that recapitulated the properties of volociximab. Hybridoma clones were screened for analogous function to volociximab, including specificity for alpha5beta1 heterodimer and blocking of integrin binding to fibronectin. A subset of antibodies that met these criteria were further characterized for their capacities to bind to mouse endothelial cells, inhibit cell migration and block angiogenesis in vitro. One antibody that encompassed all of these attributes, 339.1, was selected from this panel and tested in xenograft models. RESULTS: A panel of antibodies was characterized for specificity and potency. The affinity of antibody 339.1 for mouse integrin alpha5beta1 was determined to be 0.59 nM, as measured by BIAcore. This antibody does not significantly cross-react with human integrin, however 339.1 inhibits murine endothelial cell migration and tube formation and elicits cell death in these cells (EC50 = 5.3 nM). In multiple xenograft models, 339.1 inhibited the growth of established tumors by 40-60% (p < 0.05) and this inhibition correlates with a concomitant decrease in vessel density. CONCLUSION: The results herein demonstrate that 339.1, like volociximab, exhibits potent anti-alpha5beta1 activity and confirms that inhibition of integrin alpha5beta1 impedes angiogenesis and slows tumor growth in vivo.


Subject(s)
Angiogenesis Inhibitors/immunology , Antibodies, Monoclonal/therapeutic use , Integrin alpha5beta1/immunology , Animals , Annexin A5/immunology , Antineoplastic Agents/therapeutic use , Cell Death , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Female , Fibronectins/antagonists & inhibitors , Fibronectins/immunology , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/immunology , Integrin alpha5beta1/genetics , Integrin alpha5beta1/therapeutic use , Mice , Mice, SCID , Placenta/immunology , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/immunology , Transplantation, Heterologous
13.
J Exp Ther Oncol ; 5(4): 273-86, 2006.
Article in English | MEDLINE | ID: mdl-17024968

ABSTRACT

Integrin alpha5beta1, the principal fibronectin receptor, is an important survival factor, playing a key role in angiogenesis. Angiogenesis is critical for tumor growth, and anti-angiogenic therapies have met clinical success. To validate the therapeutic potential of an anti-alpha5beta1 strategy, we generated volociximab (M200) a chimeric human IgG4 version of the alpha5beta1 function-blocking murine antibody IIA1; and F200, the Fab derivative. Volociximab, F200 and IIA1 showed similar activity by ELISA (EC50= 0.2nM), Biacore (Kd= 0.1-0.4nM) and inhibition of fibronectin binding (IC50= 2-3nM). The inhibitory potential of alpha5beta1 antibodies was compared to HuMV833, an anti-VEGF antibody. Both volociximab and HuMV833 inhibited HUVEC proliferation (IC50 of volociximab = 0.2-0.5nM; IC50 of HuMV833 = 45nM). However, IIA1, volociximab and F200 were also potent inhibitors of an in vitro model of angiogenesis (HUVEC tube formation assay), unlike HuMV833. Additionally, volociximab inhibited in vitro tube formation induced by VEGF and/or bFGF, suggesting a mechanism of action independent of growth factor stimulus. In fact, inhibition of alpha5beta1 function by volociximab induced apoptosis of actively proliferating, but not resting, endothelial cells. Volociximab does not cross-react with rodent alpha5beta1, therefore in vivo validation of an anti-alpha5beta1 approach was conducted in a cynomolgus model of choroidal revascularization. Volociximab and F200 were potent inhibitors of neovessel formation in this model. These data demonstrate that volociximab has therapeutic potential in diseases in which new vessel formation is a component of the pathology.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies/therapeutic use , Drug Evaluation, Preclinical , Integrin alpha5beta1/immunology , Animals , Antibodies, Monoclonal, Murine-Derived , COS Cells , Chlorocebus aethiops , Extracellular Matrix/metabolism , Humans , Inhibitory Concentration 50 , Integrin alpha5beta1/chemistry , Kinetics , Macaca fascicularis , Macular Degeneration/drug therapy , Neovascularization, Pathologic , Rituximab
14.
Mol Cancer Ther ; 3(8): 921-32, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15299075

ABSTRACT

Current treatments for advanced stage, hormone-resistant prostate cancer are largely ineffective, leading to high patient mortality and morbidity. To fulfill this unmet medical need, we used global gene expression profiling to identify new potential antibody-drug conjugate (ADC) targets that showed maximal prostate cancer-specific expression. TMEFF2, a gene encoding a plasma membrane protein with two follistatin-like domains and one epidermal growth factor-like domain, had limited normal tissue distribution and was highly overexpressed in prostate cancer. Immunohistochemistry analysis using a specific monoclonal antibody (mAb) to human TMEFF2 showed significant protein expression in 74% of primary prostate cancers and 42% of metastatic lesions from lymph nodes and bone that represented both hormone-naïve and hormone-resistant disease. To evaluate anti-TMEFF2 mAbs as potential ADCs, one mAb was conjugated to the cytotoxic agent auristatin E via a cathepsin B-sensitive valine-citrulline linker. This ADC, Pr1-vcMMAE, was used to treat male severe combined immunodeficient mice bearing xenografted LNCaP and CWR22 prostate cancers expressing TMEFF2. Doses of 3 to 10 mg/kg of this specific ADC resulted in significant and sustained tumor growth inhibition, whereas an isotype control ADC had no significant effect. Similar efficacy and specificity was shown with huPr1-vcMMAE, a humanized anti-TMEFF2 ADC. No overt in vivo toxicity was observed with either murine or human ADC, despite significant cross-reactivity of anti-TMEFF2 mAb with the murine TMEFF2 protein, implying minimal toxicity to other body tissues. These data support the further evaluation and clinical testing of huPr1-vcMMAE as a novel therapeutic for the treatment of metastatic and hormone-resistant prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Membrane Proteins/chemistry , Membrane Proteins/immunology , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Oligopeptides/therapeutic use , Prostatic Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Brain/metabolism , Cell Membrane/metabolism , Cell Proliferation , Cloning, Molecular , DNA, Complementary/metabolism , Flow Cytometry , Follistatin/chemistry , Humans , Hybridomas/chemistry , Immunohistochemistry , Kinetics , Lymphatic Metastasis , Male , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Oligopeptides/chemistry , Prostate/metabolism , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Time Factors , Transfection
15.
Cancer Res ; 63(19): 6387-94, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14559828

ABSTRACT

We have used the Eos Hu03 GeneChip array, which represents over 92% of the transcribed human genome, to measure gene expression in a panel of normal and diseased human tissues. This analysis revealed that E-selectin mRNA is selectively overexpressed in prostate cancer epithelium, a finding that correlated strongly with E-selectin protein expression as assessed by immunohistochemistry. Antibodies against E-selectin that blocked function failed to impede cancer cell growth, suggesting that overexpression of E-selectin was not essential for cell growth. However, a novel auristatin E-based antibody drug conjugate (ADC), E-selectin antibody valine-citrulline monomethyl-auristatin E, was a potent and selective agent against E-selectin-expressing cancer cell lines in vitro, with the degree of cytotoxicity varying with surface antigen density. Interestingly, sensitivity to the ADC differed among cell lines from different tissues expressing similar amounts of E-selectin and was found to correlate with sensitivity to free auristatin E. Furthermore, E-selectin-expressing tumors grown as xenografts in severe combined immunodeficient mice were responsive to treatment with E-selectin antibody valine-citrulline monomethyl-auristatin E in vivo, with more than 85% inhibition of tumor growth observed in treated mice. These findings demonstrate that an E-selectin-targeting ADC has potential as a prostate cancer therapy and validates a genomics-based paradigm for the identification of cancer-specific antigens suitable for targeted therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , E-Selectin/biosynthesis , Immunotoxins/metabolism , Oligopeptides/administration & dosage , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , E-Selectin/genetics , E-Selectin/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunotoxins/immunology , Immunotoxins/pharmacology , Male , Mice , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...