Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
3.
Mol Cancer Ther ; 17(3): 638-649, 2018 03.
Article in English | MEDLINE | ID: mdl-29282299

ABSTRACT

Luminal A (hormone receptor-positive) breast cancer constitutes 70% of total breast cancer patients. In an attempt to develop a targeted therapeutic for this cancer indication, we have identified and characterized Glial cell line-Derived Neurotrophic Factor (GDNF) Family Receptor Alpha 1 (GFRA1) antibody-drug conjugates (ADC) using a cleavable valine-citrulline-MMAE (vcMMAE) linker-payload. RNAseq and IHC analysis confirmed the abundant expression of GFRA1 in luminal A breast cancer tissues, whereas minimal or no expression was observed in most normal tissues. Anti-GFRA-vcMMAE ADC internalized to the lysosomes and exhibited target-dependent killing of GFRA1-expressing cells both in vitro and in vivo The ADCs using humanized anti-GFRA1 antibodies displayed robust therapeutic activity in clinically relevant cell line-derived (MCF7 and KPL-1) tumor xenograft models. The lead anti-GFRA1 ADC cross-reacts with rodent and cynomolgus monkey GFRA1 antigen and showed optimal pharmacokinetic properties in both species. These properties subsequently enabled a target-dependent toxicity study in rats. Anti-GFRA1 ADC is well tolerated in rats, as seen with other vcMMAE linker-payload based ADCs. Overall, these data suggest that anti-GFRA1-vcMMAE ADC may provide a targeted therapeutic opportunity for luminal A breast cancer patients. Mol Cancer Ther; 17(3); 638-49. ©2017 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Glial Cell Line-Derived Neurotrophic Factor Receptors/antagonists & inhibitors , Immunoconjugates/pharmacology , Xenograft Model Antitumor Assays , Animals , Antibodies/chemistry , Antibodies/immunology , Antibodies/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/immunology , HEK293 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , MCF-7 Cells , Macaca fascicularis , Mice, Nude , Mice, SCID , Rats, Sprague-Dawley , Receptors, Steroid/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
4.
J Med Chem ; 57(19): 7890-9, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25191794

ABSTRACT

Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clinical activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clinical activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clinical activity was demonstrated. Here, we report that through chemical modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chemical synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clinically approved ADC, trastuzumab-MCC-DM1.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/chemical synthesis , Immunoconjugates/pharmacology , Maytansine/analogs & derivatives , Protein Engineering , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Trastuzumab
5.
MAbs ; 6(1): 95-107, 2014.
Article in English | MEDLINE | ID: mdl-24121517

ABSTRACT

Multi-transmembrane proteins are especially difficult targets for antibody generation largely due to the challenge of producing a protein that maintains its native conformation in the absence of a stabilizing membrane. Here, we describe an immunization strategy that successfully resulted in the identification of monoclonal antibodies that bind specifically to extracellular epitopes of a 12 transmembrane protein, multi-drug resistant protein 4 (MRP4). These monoclonal antibodies were developed following hydrodynamic tail vein immunization with a cytomegalovirus (CMV) promoter-based plasmid expressing MRP4 cDNA and were characterized by flow cytometry. As expected, the use of the immune modulators fetal liver tyrosine kinase 3 ligand (Flt3L) and granulocyte-macrophage colony-stimulating factor positively enhanced the immune response against MRP4. Imaging studies using CMV-based plasmids expressing luciferase showed that the in vivo half-life of the target antigen was less than 48 h using CMV-based plasmids, thus necessitating frequent boosting with DNA to achieve an adequate immune response. We also describe a comparison of plasmids, which contained MRP4 cDNA with either the CMV or CAG promoters, used for immunizations. The observed luciferase activity in this comparison demonstrated that the CAG promoter-containing plasmid pCAGGS induced prolonged constitutive expression of MRP4 and an increased anti-MRP4 specific immune response even when the plasmid was injected less frequently. The method described here is one that can be broadly applicable as a general immunization strategy to develop antibodies against multi-transmembrane proteins, as well as target antigens that are difficult to express or purify in native and functionally active conformation.


Subject(s)
Antibodies/immunology , Immunization , Multidrug Resistance-Associated Proteins/immunology , Plasmids , Vaccines, DNA , Animals , Cell Line , DNA, Complementary/immunology , DNA, Complementary/pharmacology , Humans , Mice, Inbred BALB C , Mice, Knockout , Multidrug Resistance-Associated Proteins/biosynthesis , Multidrug Resistance-Associated Proteins/genetics , Plasmids/immunology , Plasmids/pharmacology , Protein Structure, Secondary , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
8.
EMBO J ; 32(5): 713-27, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23386062

ABSTRACT

The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.


Subject(s)
Endosomes/microbiology , Imidazoline Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Salmonella typhimurium/growth & development , Vacuoles/microbiology , rab GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Biological Transport , Blotting, Western , Endosomes/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Imidazoline Receptors/genetics , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/metabolism , Lysosomes/microbiology , Phosphatidylinositol Phosphates/metabolism , Protein Transport , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections/microbiology , Two-Hybrid System Techniques , Vacuoles/metabolism , rab GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/genetics
9.
Nat Biotechnol ; 30(2): 184-9, 2012 Jan 22.
Article in English | MEDLINE | ID: mdl-22267010

ABSTRACT

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Subject(s)
Antibodies/blood , Antibodies/immunology , Binding Sites, Antibody/immunology , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoglobulin G/chemistry , Protein Engineering , Aminobenzoates/chemistry , Aminobenzoates/immunology , Animals , Antibodies/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Cell Line, Tumor , Cell Survival , Cysteine/chemistry , Humans , Immunoconjugates/administration & dosage , Immunoglobulin G/immunology , Macaca fascicularis , Maleimides/chemistry , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Maytansine/chemistry , Maytansine/immunology , Mice , Mice, Nude , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/immunology , Protein Conformation , Rats , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Trastuzumab
10.
Proc Natl Acad Sci U S A ; 108(7): 2759-64, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21273506

ABSTRACT

Sensory and signaling pathways are exquisitely organized in primary cilia. Bardet-Biedl syndrome (BBS) patients have compromised cilia and signaling. BBS proteins form the BBSome, which binds Rabin8, a guanine nucleotide exchange factor (GEF) activating the Rab8 GTPase, required for ciliary assembly. We now describe serum-regulated upstream vesicular transport events leading to centrosomal Rab8 activation and ciliary membrane formation. Using live microscopy imaging, we show that upon serum withdrawal Rab8 is observed to assemble the ciliary membrane in ∼100 min. Rab8-dependent ciliary assembly is initiated by the relocalization of Rabin8 to Rab11-positive vesicles that are transported to the centrosome. After ciliogenesis, Rab8 ciliary transport is strongly reduced, and this reduction appears to be associated with decreased Rabin8 centrosomal accumulation. Rab11-GTP associates with the Rabin8 COOH-terminal region and is required for Rabin8 preciliary membrane trafficking to the centrosome and for ciliogenesis. Using zebrafish as a model organism, we show that Rabin8 and Rab11 are associated with the BBS pathway. Finally, using tandem affinity purification and mass spectrometry, we determined that the transport protein particle (TRAPP) II complex associates with the Rabin8 NH(2)-terminal domain and show that TRAPP II subunits colocalize with centrosomal Rabin8 and are required for Rabin8 preciliary targeting and ciliogenesis.


Subject(s)
Bardet-Biedl Syndrome/physiopathology , Carrier Proteins/metabolism , Centrosome/metabolism , Cilia/physiology , Signal Transduction/physiology , rab GTP-Binding Proteins/metabolism , Analysis of Variance , Animals , Bardet-Biedl Syndrome/metabolism , Fluorescent Antibody Technique , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mass Spectrometry , Membranes/growth & development , Time-Lapse Imaging , Transfection , Two-Hybrid System Techniques , Zebrafish
12.
Clin Cancer Res ; 16(19): 4769-78, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20805300

ABSTRACT

PURPOSE: Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examined the in vitro potency as well as in vivo preclinical efficacy and safety profiles of a heterogeneous preparation of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. EXPERIMENTAL DESIGN AND RESULTS: To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) molecule was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 molecules per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti-cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equivalent doses (µg/m(2)) and retained efficacy at equivalent antibody doses (mg/kg). An improved safety profile of >2-fold was observed in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6,000 µg DM1/m(2)) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6,000 µg DM1/m(2)). CONCLUSIONS: The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clinical development of HER2-positive targeted breast cancer therapies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Maytansine/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Sulfhydryl Compounds/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Macaca fascicularis , Maytansine/chemistry , Mice , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/chemistry , Sulfhydryl Compounds/chemistry , Trastuzumab
15.
Nat Biotechnol ; 26(8): 925-32, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18641636

ABSTRACT

Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogenous conjugates with relatively narrow therapeutic index (maximum tolerated dose/curative dose). Using leads from our previously described phage display-based method to predict suitable conjugation sites, we engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb immunoglobulin folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepared by conventional approaches. The favorable in vivo properties of the near-homogenous composition of this conjugate suggest that our strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Cytotoxins/pharmacology , Immunotoxins/pharmacokinetics , Ovarian Neoplasms/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/genetics , Antibody Specificity , Binding Sites , CA-125 Antigen/immunology , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Cysteine/genetics , Female , Humans , Macaca fascicularis , Membrane Proteins/immunology , Mice , Mutagenesis, Site-Directed , Oligopeptides/pharmacology , Ovarian Neoplasms/drug therapy , Rats , Rats, Sprague-Dawley , Sulfhydryl Compounds/pharmacology
16.
J Immunol Methods ; 332(1-2): 41-52, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18230399

ABSTRACT

Cysteines with reactive thiol groups are attractive tools for site-specific labeling of proteins. Engineering a reactive cysteine residue into proteins with multiple disulfide bonds is often a challenging task as it may interfere with structural and functional properties of the protein. Here we developed a phage display-based biochemical assay, PHESELECTOR (Phage ELISA for Selection of Reactive Thiols) to rapidly screen reactive thiol groups on antibody fragments without interfering with their antigen binding, using trastuzumab-Fab (hu4D5Fab) as a model system. The solvent accessibility values for all the amino acid residues in the hu4D5Fab were calculated using available crystal structure information. Serine, alanine and valine residues with highest solvent accessibility values were selected and tested to compare structure-based design with the PHESELECTOR biochemical method. Cysteine substitutions at partially solvent-accessible alanine or valine residues exhibited better thiol reactivity values than substitutions at serine residues. The poor correlation between fractional solvent accessibility and thiol reactivity of the engineered hu4D5Fab variants indicated the value of PHESELECTOR biochemical assay to identify reactive thiol groups on the antibody-Fab surface. Mass spectrometric analysis of biotinylated ThioFab (Fab with engineered cysteine) variants confirmed that conjugation occurred only at the engineered cysteine thiols of either light or heavy chains. ThioFabs with engineered cysteine residues in the constant domains (CL and CH(1)) should allow universal application for site-specific conjugation of antibody-Fabs.


Subject(s)
Antibodies, Monoclonal/chemistry , Cysteine/chemistry , Immunoglobulin Fab Fragments/chemistry , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Binding Sites/genetics , Binding Sites/immunology , Biotin/chemistry , Cell Line, Tumor , Chromatography, Liquid/methods , Cysteine/genetics , Cysteine/immunology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Gene Expression Regulation/genetics , Genetic Engineering , Genetic Variation , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Maleimides/chemistry , Mass Spectrometry/methods , Models, Molecular , Mutagenesis, Site-Directed , Plasmids/genetics , Sensitivity and Specificity , Staining and Labeling/methods , Surface Properties
18.
Cell ; 129(6): 1201-13, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17574030

ABSTRACT

Primary cilium dysfunction underlies the pathogenesis of Bardet-Biedl syndrome (BBS), a genetic disorder whose symptoms include obesity, retinal degeneration, and nephropathy. However, despite the identification of 12 BBS genes, the molecular basis of BBS remains elusive. Here we identify a complex composed of seven highly conserved BBS proteins. This complex, the BBSome, localizes to nonmembranous centriolar satellites in the cytoplasm but also to the membrane of the cilium. Interestingly, the BBSome is required for ciliogenesis but is dispensable for centriolar satellite function. This ciliogenic function is mediated in part by the Rab8 GDP/GTP exchange factor, which localizes to the basal body and contacts the BBSome. Strikingly, Rab8(GTP) enters the primary cilium and promotes extension of the ciliary membrane. Conversely, preventing Rab8(GTP) production blocks ciliation in cells and yields characteristic BBS phenotypes in zebrafish. Our data reveal that BBS may be caused by defects in vesicular transport to the cilium.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Biological Transport , Cilia/metabolism , rab GTP-Binding Proteins/physiology , Amino Acid Sequence , Animals , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Humans , Microtubules/metabolism , Models, Biological , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Transport , Zebrafish , rab GTP-Binding Proteins/metabolism
19.
Proc Natl Acad Sci U S A ; 104(4): 1236-41, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17229837

ABSTRACT

The Evi5 oncogene has recently been shown to regulate the stability and accumulation of critical G(1) cell cycle factors including Emi1, an inhibitor of the anaphase-promoting complex/cyclosome, and cyclin A. Sequence analysis of the amino terminus of Evi5 reveals a Tre-2, Bub2, Cdc16 domain, which has been shown to be a binding partner and GTPase-activating protein domain for the Rab family of small Ras-like GTPases. Here we describe the identification of Evi5 as a candidate binding protein for Rab11, a GTPase that regulates intracellular transport and has specific roles in endosome recycling and cytokinesis. By yeast two-hybrid analysis, immunoprecipitation, and Biacore analysis, we demonstrate that Evi5 binds Rab11a and Rab11b in a GTP-dependent manner. However, Evi5 displays no activation of Rab11 GTPase activity in vitro. Evi5 colocalizes with Rab11 in vivo, and overexpression of Rab11 perturbs the localization of Evi5, redistributing it into Rab11-positive recycling endosomes. Interestingly, in vitro binding studies show that Rab11 effector proteins including FIP3 compete with Evi5 for binding to Rab11, suggesting a partitioning between Rab11-Evi5 and Rab11 effector complexes. Indeed, ablation of Evi5 by RNA interference causes a mislocalization of FIP3 at the abscission site during cytokinesis. These data demonstrate that Evi5 is a Rab11 binding protein and that Evi5 may cooperate with Rab11 to coordinate vesicular trafficking, cytokinesis, and cell cycle control independent of GTPase-activating protein function.


Subject(s)
GTP Phosphohydrolases/metabolism , Nuclear Proteins/metabolism , Oncogenes , rab GTP-Binding Proteins/metabolism , Cell Cycle , Cell Cycle Proteins , Cell Line , GTPase-Activating Proteins , Humans , I-kappa B Kinase/metabolism , Nuclear Proteins/genetics , Protein Binding , Surface Plasmon Resonance
20.
Nat Rev Mol Cell Biol ; 7(9): 631-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16912714

ABSTRACT

Since the discovery of SNARE proteins in the late 1980s, SNAREs have been recognized as key components of protein complexes that drive membrane fusion. Despite considerable sequence divergence among SNARE proteins, their mechanism seems to be conserved and is adaptable for fusion reactions as diverse as those involved in cell growth, membrane repair, cytokinesis and synaptic transmission. A fascinating picture of these robust nanomachines is emerging.


Subject(s)
Cell Growth Processes/physiology , Membrane Fusion/physiology , Multiprotein Complexes/metabolism , SNARE Proteins/metabolism , Signal Transduction/physiology , Synapses/metabolism , Animals , Humans , SNARE Proteins/genetics , Synapses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...