Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 15(11): 2698-2708, 2016 11.
Article in English | MEDLINE | ID: mdl-27582525

ABSTRACT

Trop-2, also known as TACSTD2, EGP-1, GA733-1, and M1S1, is frequently expressed on a variety of human carcinomas, and its expression is often associated with poor prognosis of the diseases. However, it is also present on the epithelium of several normal tissues. A comprehensively designed Trop-2-targeting antibody-drug conjugate (ADC), balancing both efficacy and toxicity, is therefore necessary to achieve clinical utility. To this end, we developed a cleavable Trop-2 ADC (RN927C) using a site-specific transglutaminase-mediated conjugation method and a proprietary microtubule inhibitor (MTI) linker-payload, PF-06380101. Robust in vitro cytotoxicity of RN927C was observed on a panel of Trop-2-expressing tumor cell lines, with IC50 generally in the subnanomolar range. As expected for an MTI-containing ADC, RN927C readily induced mitotic arrest of treated cells in vitro and in vivo, followed by subsequent cell death. The in vivo efficacy of RN927C was tested in multiple cell line and patient-derived xenograft tumor models, including pancreatic, lung, ovarian, and triple-negative breast tumor types. Single-dose administration of RN927C at 0.75 to 3 mg/kg was generally sufficient to induce sustained regression of Trop-2-expressing tumors and showed superior efficacy over standard treatment with paclitaxel or gemcitabine. Administration of RN927C in nonhuman primate toxicity studies resulted in target-mediated effects in skin and oral mucosa, consistent with Trop-2 expression in these epithelial tissues with minimal, non-dose limiting off-target toxicities. On the basis of the combined efficacy and safety results, RN927C is postulated to have a favorable therapeutic index for treatment of solid tumors. Mol Cancer Ther; 15(11); 2698-708. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Immunoconjugates/pharmacology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Stability , Female , Gene Expression , Humans , Immunoconjugates/chemistry , Lysosomes , Mice , Mitosis/drug effects , Mitosis/genetics , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
J Mol Biol ; 425(10): 1641-54, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23416200

ABSTRACT

The emergence and spread of multi-drug-resistant strains of Staphylococcus aureus in hospitals and in the community emphasize the urgency for the development of novel therapeutic interventions. Our approach was to evaluate the potential of harnessing the human immune system to guide the development of novel therapeutics. We explored the role of preexisting antibodies against S. aureus α-hemolysin in the serum of human individuals by isolating and characterizing one antibody with a remarkably high affinity to α-hemolysin. The antibody provided protection in S. aureus pneumonia, skin, and bacteremia mouse models of infection and also showed therapeutic efficacy when dosed up to 18 h post-infection in the pneumonia model. Additionally, in pneumonia and bacteremia animal models, the therapeutic efficacy of the α-hemolysin antibody appeared additive to the antibiotic linezolid. To better understand the mechanism of action of this isolated antibody, we solved the crystal structure of the α-hemolysin:antibody complex. To our knowledge, this is the first report of the crystal structure of the α-hemolysin monomer. The structure of the complex shows that the antibody binds α-hemolysin between the cap and the rim domains. In combination with biochemical data, the structure suggests that the antibody neutralizes the activity of the toxin by preventing binding to the plasma membrane of susceptible host cells. The data presented here suggest that protective antibodies directed against S. aureus molecules exist in some individuals and that such antibodies have a therapeutic potential either alone or in combination with antibiotics.


Subject(s)
Antibodies, Bacterial/administration & dosage , Antibodies, Bacterial/therapeutic use , Antigen-Antibody Complex/administration & dosage , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/pharmacology , Antibodies, Blocking/blood , Antibodies, Blocking/chemistry , Antibodies, Blocking/therapeutic use , Antigen-Antibody Complex/chemistry , Antigen-Antibody Reactions/immunology , Bacterial Toxins/chemistry , Crystallography, X-Ray , Disease Models, Animal , Female , Hemolysin Proteins/chemistry , Humans , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Staphylococcal Infections/blood , Staphylococcus aureus/pathogenicity
3.
J Mol Biol ; 420(3): 204-19, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22543237

ABSTRACT

Bispecific antibodies and antibody fragments are a new class of therapeutics increasingly utilized in the clinic for T cell recruitment (catumaxomab anti-EpCAM/CD3 and blinatumomab anti-CD19/CD3), increase in the selectivity of targeting, or simultaneous modulation of multiple cellular pathways. While the clinical potential for certain bispecific antibody formats is clear, progress has been hindered because they are often difficult to manufacture, may suffer from suboptimal pharmacokinetic properties, and may be limited due to potential immunogenicity issues. Current state-of-the-art human IgG-like bispecific technologies require co-expression of two heavy chains with a single light chain, use crossover domains to segregate light chains, or utilize scFv (single-chain fragment variable)-Fc fusion. We have engineered both human IgG1 and IgG2 subtypes, with minimal point mutations, to form full-length bispecific human antibodies with high efficiency and in high purity. In our system, the two antibodies of interest can be expressed and purified separately, mixed together under appropriate redox conditions, resulting in a formation of a stable bispecific antibody with high yields. With this approach, it is not necessary to generate new antibodies that share a common light chain, therefore allowing the immediate use of an existing antibody regardless of whether it has been generated via standard hybridoma or display methods. We demonstrate the generality of the approach and show that these bispecific antibodies have properties similar to those of wild-type IgGs, and we further demonstrate the utility of the technology with an example of a CD3/CD20 bispecific antibody that effectively depletes B cells in vitro and in vivo.


Subject(s)
Antibodies, Bispecific/immunology , Immunoglobulin G/metabolism , Protein Engineering/methods , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/isolation & purification , Antibodies, Bispecific/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antigens, CD20/immunology , B-Lymphocytes/immunology , CD3 Complex/immunology , Cetuximab , Cytotoxicity, Immunologic , Female , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Point Mutation , Rats , Rats, Sprague-Dawley , Receptors, Fc/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , T-Lymphocytes/immunology
4.
J Biol Chem ; 287(14): 11090-7, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22294692

ABSTRACT

Target-mediated clearance and high antigen load can hamper the efficacy and dosage of many antibodies. We show for the first time that the mouse, cynomolgus, and human cross-reactive, antagonistic anti-proprotein convertase substilisin kexin type 9 (PCSK9) antibodies J10 and the affinity-matured and humanized J16 exhibit target-mediated clearance, resulting in dose-dependent pharmacokinetic profiles. These antibodies prevent the degradation of low density lipoprotein receptor, thus lowering serum levels of LDL-cholesterol and potently reducing serum cholesterol in mice, and selectively reduce LDL-cholesterol in cynomolgus monkeys. In order to increase the pharmacokinetic and efficacy of this promising therapeutic for hypercholesterolemia, we engineered pH-sensitive binding to mouse, cynomolgus, and human PCSK9 into J16, resulting in J17. This antibody shows prolonged half-life and increased duration of cholesterol lowering in two species in vivo by binding to endogenous PCSK9 in mice and cynomolgus monkeys, respectively. The proposed mechanism of this pH-sensitive antibody is that it binds with high affinity to PCSK9 in the plasma at pH 7.4, whereas the antibody-antigen complex dissociates at the endosomal pH of 5.5-6.0 in order to escape from target-mediated degradation. Additionally, this enables the antibody to bind to another PCSK9 and therefore increase the antigen-binding cycles. Furthermore, we show that this effect is dependent on the neonatal Fc receptor, which rescues the dissociated antibody in the endosome from degradation. Engineered pH-sensitive antibodies may enable less frequent or lower dosing of antibodies hampered by target-mediated clearance and high antigen load.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/pharmacokinetics , Proprotein Convertases/immunology , Protein Engineering , Serine Endopeptidases/immunology , Animals , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacology , Anticholesteremic Agents/blood , Anticholesteremic Agents/immunology , Complementarity Determining Regions/chemistry , Half-Life , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Macaca fascicularis , Male , Mice , Proprotein Convertase 9 , Receptors, Fc/metabolism
5.
Virology ; 375(2): 580-91, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18343477

ABSTRACT

Hepatitis C virus (HCV) envelope glycoprotein co-evolution was studied in 14 genotype 1-infected and treatment-naive subjects, including 7 with mild and 7 with severe liver disease. Cassettes encoding the envelope 1 gene (E1) and hypervariable region (HVR1) of the envelope 2 gene were isolated at 38 different time points over 81 follow-up years. There were no significant differences in age, gender, alcohol use, or viral load between the mild and severe disease groups. Virus from subjects with severe disease had significantly slower evolution in HVR1, and significant divergent evolution of E1 quasispecies, characterized by a preponderance of synonymous mutations, compared to virus from subjects with mild disease. Phylogenetic comparisons indicated higher similarity between amino acid sequences of the E1 and HVR1 regions with mild disease versus severe disease (r=0.44 versus r=0.17, respectively; P=0.01). In summary, HCV envelope quasispecies co-evolution differs during mild versus severe disease.


Subject(s)
Hepacivirus/genetics , Hepatitis C, Chronic/virology , Viral Envelope Proteins/genetics , Amino Acid Sequence , Evolution, Molecular , Female , Humans , Molecular Sequence Data , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...