Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Anal Chem ; 90(1): 866-871, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29206445

ABSTRACT

Large molecule quantitation by LC-MS/MS commonly relies on bottom-up or so-called surrogate peptide measurements to infer the whole-molecule concentration. This can lead to questions about what is actually being measured in the assay (intact drug and/or other drug related material). An intact sequential affinity capture (ISAC) assay was developed utilizing two different immunoaffinity (IA) reagents. The reagents were selective for the heavy and light chain of a monoclonal antibody, which when used consecutively, ensures that only the intact form of the antibody is represented by the surrogate peptide. The approach provided comparable results to a traditional sandwich IA assay indicating similar capture populations. The use of an initial ISAC assessment of affinity capture purification, should add a degree of confidence in the use of a single IA-LC-MS/MS quantitation assay.


Subject(s)
Antibodies, Monoclonal/blood , Biological Assay/methods , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Animals , Antibodies, Monoclonal/chemistry , Macaca mulatta , Proteolysis , Trypsin/chemistry
2.
Infect Immun ; 83(2): 822-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25486992

ABSTRACT

Clostridium difficile infection (CDI) represents the most prevalent cause of antibiotic-associated gastrointestinal infections in health care facilities in the developed world. Disease symptoms are caused by the two homologous exotoxins, TcdA and TcdB. Standard therapy for CDI involves administration of antibiotics that are associated with a high rate of disease recurrence, highlighting the need for novel treatment paradigms that target the toxins rather than the organism itself. A combination of human monoclonal antibodies, actoxumab and bezlotoxumab, directed against TcdA and TcdB, respectively, has been shown to decrease the rate of recurrence in patients treated with standard-of-care antibiotics. However, the exact mechanism of antibody-mediated protection is poorly understood. In this study, we show that the antitoxin antibodies are protective in multiple murine models of CDI, including systemic and local (gut) toxin challenge models, as well as primary and recurrent models of infection in mice. Systemically administered actoxumab-bezlotoxumab prevents both the damage to the gut wall and the inflammatory response, which are associated with C. difficile in these models, including in mice challenged with a strain of the hypervirulent ribotype 027. Furthermore, mutant antibodies (N297Q) that do not bind to Fcγ receptors provide a level of protection similar to that of wild-type antibodies, demonstrating that the mechanism of protection is through direct neutralization of the toxins and does not involve host effector functions. These data provide a mechanistic basis for the prevention of recurrent disease observed in CDI patients in clinical trials.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antitoxins/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/prevention & control , Enterotoxins/immunology , Animals , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antitoxins/therapeutic use , Chlorocebus aethiops , Disease Models, Animal , Enterocolitis, Pseudomembranous/immunology , Mice , Mice, Inbred C57BL , Mutation , Receptors, IgG/immunology , Recurrence , Vero Cells
3.
Drug Metab Dispos ; 43(5): 774-85, 2015 May.
Article in English | MEDLINE | ID: mdl-25739975

ABSTRACT

Elevated levels of proinflammatory cytokines associated with infection and inflammation can modulate cytochrome P450 enzymes, leading to potential disease-drug interactions and altered small-molecule drug disposition. We established a human-derived hepatocyte-Kupffer cell (Hep:KC) coculture model to assess the indirect cytokine impact on hepatocytes through stimulation of KC-mediated cytokine release and compared this model with hepatocytes alone. Characterization of Hep:KC cocultures showed an inflammation response after treatment with lipopolysaccharide and interleukin (IL)-6 (indicated by secretion of various cytokines). Additionally, IL-6 exposure upregulated acute-phase proteins (C-reactive protein, alpha-1-acid glycoprotein, and serum amyloid A2) and downregulated CYP3A4. Compared with hepatocytes alone, Hep:KC cocultures showed enhanced IL-1ß-mediated effects but less impact from both IL-2 and IL-23. Hep:KC cocultures treated with IL-1ß exhibited a higher release of proinflammatory cytokines, an increased upregulation of acute-phase proteins, and a larger extent of metabolic enzyme and transporter suppression. IC50 values for IL-1ß-mediated CYP3A4 suppression were lower in Hep:KC cocultures (98.0-144 pg/ml) compared with hepatocytes alone (IC50 > 5000 pg/ml). Cytochrome suppression was preventable by blocking IL-1ß interaction with IL-1R1 using an antagonist cytokine or an anti-IL-1ß antibody. Unlike IL-1ß, IL-6-mediated effects were comparable between hepatocyte monocultures and Hep:KC cocultures. IL-2 and IL-23 caused a negligible inflammation response and a minimal inhibition of CYP3A4. In both hepatocyte monocultures and Hep:KC cocultures, IL-2RB and IL-23R were undetectable, whereas IL-6R and IL-1R1 levels were higher in Hep:KC cocultures. In summary, compared with hepatocyte monocultures, the Hep:KC coculture system is a more robust in vitro model for studying the impact of proinflammatory cytokines on metabolic enzymes.


Subject(s)
Carrier Proteins/metabolism , Hepatocytes/metabolism , Inflammation/metabolism , Interleukins/metabolism , Kupffer Cells/metabolism , 3T3 Cells , Adult , Animals , Biological Transport/physiology , C-Reactive Protein/metabolism , Cell Line , Coculture Techniques/methods , Cytochrome P-450 CYP3A/metabolism , Down-Regulation/physiology , Glycoproteins/metabolism , Humans , Male , Mice , Middle Aged , Serum Amyloid A Protein/metabolism , Up-Regulation/physiology
4.
Anal Chem ; 86(17): 8776-84, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25010922

ABSTRACT

Quantitation of therapeutic monoclonal antibodies (mAb) using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for pharmacokinetic (PK) studies is becoming an essential complement to traditional antibody-based ligand binding assays (LBA). Here we show an automated method to perform LC-MS/MS-based quantitation, with IgG1 conserved peptides, a heavy isotope labeled mAb internal standard, and anti-human Fc enrichment. All reagents in the method are commercially available with no requirement to develop novel assay-specific reagents. The method met traditional quantitative LC-MS/MS assay analytical characteristics in terms of precision, accuracy, and specificity. The method was applied to the pharmacokinetic study of a mAb dosed in cynomolgus monkey, and the results were compared with the immunoassay data. This methodology has the potential to benefit and accelerate the early biopharmaceutical development process, particularly by enabling PK analysis across species and candidate molecules with minimal method development.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Blood Chemical Analysis/instrumentation , Peptides/analysis , Tandem Mass Spectrometry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/blood , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Half-Life , Immunoglobulin G/metabolism , Immunoprecipitation , Isotope Labeling , Macaca fascicularis , Molecular Sequence Data , Peptides/chemistry
5.
Bioorg Med Chem ; 22(7): 2303-10, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24588962

ABSTRACT

The ribonucleotide reductase (RNR) enzyme is a heteromer of RRM1 and RRM2 subunits. The active enzyme catalyzes de novo reduction of ribonucleotides to generate deoxyribonucleotides (dNTPs), which are required for DNA replication and DNA repair processes. Complexity in the generation of physiologically relevant, active RRM1/RRM2 heterodimers was perceived as limiting to the identification of selective RRM1 inhibitors by high-throughput screening of compound libraries and led us to seek alternative methods to identify lead series. In short, we found that gemcitabine, as its diphosphate metabolite, represents one of the few described active site inhibitors of RRM1. We herein describe the identification of novel 5'-amino gemcitabine analogs as potent RRM1 inhibitors through in-cell phenotypic screening.


Subject(s)
Deoxycytidine/analogs & derivatives , Tumor Suppressor Proteins/antagonists & inhibitors , Cell Line, Tumor , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Ribonucleoside Diphosphate Reductase , Structure-Activity Relationship , Gemcitabine
6.
Bioorg Med Chem Lett ; 23(9): 2590-4, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23535330

ABSTRACT

Drug design efforts in the emerging 2-aminothiazole-4-carboxamide class of CHK1 inhibitors have uncovered specific combinations of key substructures within the molecule; resulting in significant improvements in cell-based activity while retaining a greater than one hundred-fold selectivity against CDK2. The X-ray crystal structure of a complex between compound 39 and the CHK1 protein detailing a 'U-shaped' topology and key interactions with the protein surface at the ATP site is also reported.


Subject(s)
Amides/chemistry , Drug Design , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Thiazoles/chemistry , Amides/chemical synthesis , Amides/metabolism , Binding Sites , Checkpoint Kinase 1 , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinases/metabolism , Structure-Activity Relationship
7.
J Immunol Methods ; 522: 113553, 2023 11.
Article in English | MEDLINE | ID: mdl-37661047

ABSTRACT

Targeted immune agonist (TIA) comprising a TLR7 agonist conjugated to tumor-targeting antibodies have been shown to induce potent anti-tumor responses in various preclinical models. However, the clinical proof-of-concept of a TIA has been hampered by systemic dose-limiting immune-related toxicities, including rapid induction of anti-drug antibodies in patients. We have developed ELISPOT-based assay to measure activation of antibody-secreting cells (ASCs), intended to simulate the interaction between TIA and peripheral B cells as a tool to pre-clinically de-risk tumor target-independent peripheral B-cell activation by TIA. This method has proven to be robust and has fast turn-around time to evaluate the induction of spontaneous B-cell activation by TIA in a tumor target- and FcγR-independent manner. This novel ASC assay platform may serve as a preclinical tool to de-risk TIAs that can potentially induce immune-related adverse effects in the clinic.


Subject(s)
Neoplasms , Toll-Like Receptor 7 , Humans , Adjuvants, Immunologic , Antibodies , Lymphocyte Activation
8.
Bioorg Med Chem Lett ; 21(1): 471-4, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21094607

ABSTRACT

Previous efforts by our group have established pyrazolo[1,5-a]pyrimidine as a viable core for the development of potent and selective CDK inhibitors. As part of an effort to utilize the pyrazolo[1,5-a]pyrimidine core as a template for the design and synthesis of potent and selective kinase inhibitors, we focused on a key regulator in the cell cycle progression, CHK1. Continued SAR development of the pyrazolo[1,5-a]pyrimidine core at the C5 and C6 positions, in conjunction with previously disclosed SAR at the C3 and C7 positions, led to the discovery of potent and selective CHK1 inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Binding Sites , Catalytic Domain , Checkpoint Kinase 1 , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Drug Evaluation, Preclinical , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship
10.
J Immunol Methods ; 494: 113047, 2021 07.
Article in English | MEDLINE | ID: mdl-33775673

ABSTRACT

Peggy Sue is a capillary-based western/immunoassay platform that can separate and characterize proteins by size or charge. A quick and automated immunogenicity assay was developed on Peggy Sue based on charge separation and compared with a traditional bridging method using preclinical samples from non-human primate studies. The results generated with the Peggy Sue assay were comparable to those of the bridging assays. The Peggy Sue platform has several advantages, including time efficiency, low sample consumption, and easy automation. The platform is especially ideal for further characterization of anti-drug antibody (ADA) specificity against complex biologics such as bispecific or multi-specific biotherapeutics as it is easy to conduct domain specificity assessment of observed ADA responses. Our evaluation suggests that the Peggy Sue platform is a promising tool for preclinical ADA analysis.


Subject(s)
Antibodies/isolation & purification , Biological Products/therapeutic use , Drug-Related Side Effects and Adverse Reactions/diagnosis , Immunoassay/methods , Single-Domain Antibodies/isolation & purification , Animals , Automation , Camelids, New World , Electrophoresis, Capillary , Humans , Macaca mulatta
11.
Mol Imaging Biol ; 23(2): 250-259, 2021 04.
Article in English | MEDLINE | ID: mdl-33104972

ABSTRACT

PURPOSE: Programmed cell death-1 receptor (PD-1) and its ligand (PD-L1) are the targets for immunotherapy in many cancer types. Although PD-1 blockade has therapeutic effects, the efficacy differs between patients. Factors contributing to this variability are PD-L1 expression levels and immune cells present in tumors. However, it is not well understood how PD-1 expression in the tumor microenvironment impacts immunotherapy response. Thus, imaging of PD-1-expressing immune cells is of interest. This study aims to evaluate the biodistribution of Zirconium-89 (89Zr)-labeled pembrolizumab, a humanized IgG4 kappa monoclonal antibody targeting PD-1, in healthy cynomolgus monkeys as a translational model of tracking PD-1-positive immune cells. PROCEDURES: Pembrolizumab was conjugated with the tetrafluorophenol-N-succinyl desferal-Fe(III) ester (TFP-N-sucDf) and subsequently radiolabeled with 89Zr. Four cynomolgus monkeys with no previous exposure to humanized monoclonal antibodies received tracer only or tracer co-injected with pembrolizumab intravenously over 5 min. Thereafter, a static whole-body positron emission tomography (PET) scan was acquired with 10 min per bed position on days 0, 2, 5, and 7. Image-derived standardized uptake values (SUVmean) were quantified by region of interest (ROI) analysis. RESULTS: 89Zr-N-sucDf-pembrolizumab was synthesized with high radiochemical purity (> 99 %) and acceptable molar activity (> 7 MBq/nmol). In animals dosed with tracer only, 89Zr-N-sucDf-pembrolizumab distribution in lymphoid tissues such as mesenteric lymph nodes, spleen, and tonsils increased over time. Except for the liver, low radiotracer distribution was observed in all non-lymphoid tissue including the lung, muscle, brain, heart, and kidney. When a large excess of pembrolizumab was co-administered with a radiotracer, accumulation in the lymph nodes, spleen, and tonsils was reduced, suggestive of target-mediated accumulation. CONCLUSIONS: 89Zr-N-sucDf-pembrolizumab shows preferential uptake in the lymphoid tissues including the lymph nodes, spleen, and tonsils. 89Zr-N-sucDf-pembrolizumab may be useful in tracking the distribution of a subset of immune cells in non-human primates and humans. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02760225.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Molecular Imaging/methods , Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Programmed Cell Death 1 Receptor/metabolism , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacokinetics , Female , Immunotherapy/methods , Macaca fascicularis , Male , Models, Animal , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/immunology , Radioisotopes , Tissue Distribution , Zirconium
12.
Biochemistry ; 49(38): 8350-8, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20718440

ABSTRACT

Affinity selection-mass spectrometry (AS-MS) screening of kinesin spindle protein (KSP) followed by enzyme inhibition studies and temperature-dependent circular dichroism (TdCD) characterization was utilized to identify a series of benzimidazole compounds. This series also binds in the presence of Ispinesib, a known anticancer KSP inhibitor in phase I/II clinical trials for breast cancer. TdCD and AS-MS analyses support simultaneous binding implying existence of a novel non-Ispinesib binding pocket within KSP. Additional TdCD analyses demonstrate direct binding of these compounds to Ispinesib-resistant mutants (D130V, A133D, and A133D + D130V double mutant), further strengthening the hypothesis that the compounds bind to a distinct binding pocket. Also importantly, binding to this pocket causes uncompetitive inhibition of KSP ATPase activity. The uncompetitive inhibition with respect to ATP is also confirmed by the requirement of nucleotide for binding of the compounds. After preliminary affinity optimization, the benzimidazole series exhibited distinctive antimitotic activity as evidenced by blockade of bipolar spindle formation and appearance of monoasters. Cancer cell growth inhibition was also demonstrated either as a single agent or in combination with Ispinesib. The combination was additive as predicted by the binding studies using TdCD and AS-MS analyses. The available data support the existence of a KSP inhibitory site hitherto unknown in the literature. The data also suggest that targeting this novel site could be a productive strategy for eluding Ispinesib-resistant tumors. Finally, AS-MS and TdCD techniques are general in scope and may enable screening other targets in the presence of known drugs, clinical candidates, or tool compounds that bind to the protein of interest in an effort to identify potency-enhancing small molecules that increase efficacy and impede resistance in combination therapy.


Subject(s)
Benzimidazoles/pharmacology , Kinesins/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides/metabolism , Benzimidazoles/antagonists & inhibitors , Binding Sites , Circular Dichroism , Humans , Kinesins/antagonists & inhibitors , Kinesins/chemistry , Mass Spectrometry , Nucleotides/antagonists & inhibitors , Nucleotides/chemistry , Protein Structure, Tertiary , Quinazolines/metabolism
13.
Bioorg Med Chem Lett ; 20(24): 7216-21, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21074424

ABSTRACT

A novel series of CHK1 inhibitors based on thienopyridine template has been designed and synthesized. These inhibitors maintain critical hydrogen bonding with the hinge and conserved water in the ATP binding site. Several compounds show single digit nanomolar CHK1 activities. Compound 70 shows excellent enzymatic activity of 1 nM.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/chemistry , Pyridazines/chemical synthesis , Thienopyridines/chemistry , Thiophenes/chemical synthesis , Adenosine Triphosphate/chemistry , Binding Sites , Checkpoint Kinase 1 , Crystallography, X-Ray , Drug Design , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyridazines/chemistry , Pyridazines/pharmacology , Structure-Activity Relationship , Thienopyridines/chemical synthesis , Thienopyridines/pharmacology , Thiophenes/chemistry , Thiophenes/pharmacology
15.
Front Immunol ; 11: 573405, 2020.
Article in English | MEDLINE | ID: mdl-33117369

ABSTRACT

The molecule "T cell immunoreceptor with immunoglobulin and ITIM domain," or TIGIT, has recently received much attention as a promising target in the treatment of various malignancies. In spite of the quick progression of anti-TIGIT antibodies into clinical testing both as monotherapy and in combination with programmed cell death-1 (PD-1)-directed immune checkpoint blockade, the molecular mechanism behind the observed therapeutic benefits remains poorly understood. Here we demonstrate, using mouse tumor models, that TIGIT blocking antibodies with functional Fc binding potential induce effective anti-tumor response. Our observations reveal that the anti-TIGIT therapeutic effect is not achieved by depletion of intratumoral regulatory T cells (Treg) or any cell population expressing TIGIT, but instead is mediated by possible "reverse activating signals" through FcγRs on myeloid cells, inducing expression of various mediators such as cytokines and chemokines. Furthermore, we discovered an induction of a robust and persistent granzyme B and perforin response, distinct from a predominantly interferon-γ (IFN-γ)-driven anti-PD-1 blockade. Our observations, for the first time, provide the basis for a mechanistic hypothesis involving the requirement of a functional Fc domain of anti-TIGIT monoclonal antibodies, of which various isotypes are currently under intense clinical investigation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Colonic Neoplasms/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Myeloid Cells/drug effects , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Granzymes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Signal Transduction , Tumor Burden/drug effects , Tumor Microenvironment
16.
Bioorg Med Chem Lett ; 19(13): 3405-9, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19481450

ABSTRACT

Substituted benzimidazoles were profiled as inhibitors of kinesin spindle protein (KSP), an increasingly important target for the development of anticancer drugs. This series demonstrated the monoastral phenotypic response and was found to be active in both enzymatic and cellular-based assays.


Subject(s)
Benzimidazoles/chemistry , Enzyme Inhibitors/chemistry , Kinesins/antagonists & inhibitors , Animals , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Caco-2 Cells , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Kinesins/metabolism , Rats , Structure-Activity Relationship
17.
AAPS J ; 21(3): 39, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30868312

ABSTRACT

Assessment of the factors that regulate antibody exposure-response relationships in the relevant animal models is critical for the design of successful translational strategies from discovery to the clinic. Depending on the specific clinical indication, preclinical development paradigms may require that the efficacy or dosing-related attributes for the existing antibody be assessed in various species when cross-reactivity of the lead antibody to the intended species is justified. Additionally, with the success of monoclonal antibodies for management of various human conditions, a parallel interest in therapeutic use of these novel modalities in various veterinary species has followed. The protective role of neonatal Fc receptor (FcRn) in regulation of IgG homeostasis and clearance is now well recognized and the "nonspecific clearance" of antibodies through bone marrow-derived phagocytic and vascular endothelial cells (via lysosomal processes) is modulated by interactions with FcRn receptors. In this study, we have attempted to examine the PK properties of human IgG antibodies in dog and monkey. These studies establish a translational framework for evaluation of IgG antibody PK properties across species.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Immunoglobulin G/pharmacology , Administration, Intravenous , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Macaca mulatta , Mice , Models, Animal , Respiratory Syncytial Viruses/immunology , Species Specificity
18.
J Immunol Methods ; 456: 7-14, 2018 05.
Article in English | MEDLINE | ID: mdl-29427592

ABSTRACT

LAG3 is an important regulator of T cell homeostasis and studies in mouse tumor models have demonstrated that simultaneously antagonizing LAG3 and PD1 can augment tumor-specific T cell responses and induce tumor rejection. The combined use of LAG3 antagonist antibodies with established anti-PD1 therapies is currently being evaluated in human clinical trials. A functional assay for human LAG3 was developed by co-culture of a Jurkat T-cell lymphoma line overexpressing LAG3 with a Raji B-cell lymphoma line in the presence of staphylococcal enterotoxins. Reversal of LAG3 repression was measured as an increase in IL-2 production or NFAT activation in response to treatment with MK-4280, an anti-human LAG3 antagonist antibody. Changes in cytokines, chemokines, and other mRNA transcripts were in agreement with published in vitro and in vivo models for LAG3 biology which highlights the physiological relevance of the Jurkat functional assay. Additional engineering of PD1 and PDL1 components into the LAG3 assay resulted in a bi-functional assay that is capable of inducing a 10-fold response to individual antibodies blocking either PD1 or LAG3. Importantly, when MK-4280 and pembrolizumab were combined to block both pathways, a synergistic 50-fold increase in response was observed.


Subject(s)
Antibodies, Blocking/analysis , Antigens, CD/immunology , Cell Engineering , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Antibodies, Blocking/immunology , Antigens, CD/genetics , Flow Cytometry , Gene Expression Profiling , Humans , Jurkat Cells , Programmed Cell Death 1 Receptor/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , T-Lymphocytes/immunology , Tumor Cells, Cultured , Lymphocyte Activation Gene 3 Protein
19.
Diabetes ; 67(2): 299-308, 2018 02.
Article in English | MEDLINE | ID: mdl-29097375

ABSTRACT

Insulin has a narrow therapeutic index, reflected in a small margin between a dose that achieves good glycemic control and one that causes hypoglycemia. Once injected, the clearance of exogenous insulin is invariant regardless of blood glucose, aggravating the potential to cause hypoglycemia. We sought to create a "smart" insulin, one that can alter insulin clearance and hence insulin action in response to blood glucose, mitigating risk for hypoglycemia. The approach added saccharide units to insulin to create insulin analogs with affinity for both the insulin receptor (IR) and mannose receptor C-type 1 (MR), which functions to clear endogenous mannosylated proteins, a principle used to endow insulin analogs with glucose responsivity. Iteration of these efforts culminated in the discovery of MK-2640, and its in vitro and in vivo preclinical properties are detailed in this report. In glucose clamp experiments conducted in healthy dogs, as plasma glucose was lowered stepwise from 280 mg/dL to 80 mg/dL, progressively more MK-2640 was cleared via MR, reducing by ∼30% its availability for binding to the IR. In dose escalations studies in diabetic minipigs, a higher therapeutic index for MK-2640 (threefold) was observed versus regular insulin (1.3-fold).


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Drug Design , Hypoglycemic Agents/therapeutic use , Insulin, Regular, Human/analogs & derivatives , Lectins, C-Type/agonists , Mannose-Binding Lectins/agonists , Receptor, Insulin/agonists , Receptors, Cell Surface/agonists , Animals , Animals, Inbred Strains , Binding, Competitive , CHO Cells , Cricetulus , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Half-Life , Humans , Hyperglycemia/prevention & control , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Insulin, Regular, Human/adverse effects , Insulin, Regular, Human/pharmacokinetics , Insulin, Regular, Human/therapeutic use , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Male , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Metabolic Clearance Rate , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Swine , Swine, Miniature
20.
Oncogene ; 22(51): 8293-301, 2003 Nov 13.
Article in English | MEDLINE | ID: mdl-14614453

ABSTRACT

AuroraA, a mitotic kinase, is reported to be amplified and overexpressed in a variety of human tumors. Active mutants of AuroraA can transform mouse fibroblasts and form tumors in nude mice. However, the mechanism behind this oncogenic potential remains elusive. In this study, we investigated the consequences of AuroraA overexpression and showed that increased AuroraA levels compromise the mitotic spindle checkpoint triggered by nocodazole, a microtubule polymerization inhibitor. This is accomplished by disrupting the proper assembly of the mitotic checkpoint complex at the level of the Cdc20-BubR1 interaction. As a result, the spindle checkpoint complex fails to form and cells progress through mitosis without proper arrest in response to nocodazole. This ability to override the mitotic spindle checkpoint was found to be independent of AuroraA kinase activity. We conclude that maintenance of a functional balance between AuroraA and mitotic checkpoint proteins is essential for the proper progression through mitosis. This study therefore offers a possible explanation of how deregulation of AuroraA can contribute to genetic instability and tumorigenesis.


Subject(s)
Microtubules/drug effects , Nocodazole/pharmacology , Protein Kinases/metabolism , Spindle Apparatus , Aurora Kinases , Cell Cycle Proteins , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Protein Serine-Threonine Kinases , Xenopus Proteins
SELECTION OF CITATIONS
SEARCH DETAIL