Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38712232

ABSTRACT

Many disease-causing proteins have multiple pathogenic mechanisms, and conventional inhibitors struggle to reliably disrupt more than one. Targeted protein degradation (TPD) can eliminate the protein, and thus all its functions, by directing a cell's protein turnover machinery towards it. Two established strategies either engage catalytic E3 ligases or drive uptake towards the endolysosomal pathway. Here we describe CYpHER (CatalYtic pH-dependent Endolysosomal delivery with Recycling) technology with potency and durability from a novel catalytic mechanism that shares the specificity and straightforward modular design of endolysosomal uptake. By bestowing pH-dependent release on the target engager and using the rapid-cycling transferrin receptor as the uptake receptor, CYpHER induces endolysosomal target delivery while re-using drug, potentially yielding increased potency and reduced off-target tissue exposure risks. The TfR-based approach allows targeting to tumors that overexpress this receptor and offers the potential for transport to the CNS. CYpHER function was demonstrated in vitro with EGFR and PD-L1, and in vivo with EGFR in a model of EGFR-driven non-small cell lung cancer.

2.
J Pharmacol Exp Ther ; 353(1): 119-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25653417

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as an attractive therapeutic target for cardiovascular disease. Monoclonal antibodies (mAbs) that bind PCSK9 and prevent PCSK9:low-density lipoprotein receptor complex formation reduce serum low-density lipoprotein-cholesterol (LDL-C) in vivo. PCSK9-mediated lysosomal degradation of bound mAb, however, dramatically reduces mAb exposure and limits duration of effect. Administration of high-affinity mAb1:PCSK9 complex (1:2) to mice resulted in significantly lower mAb1 exposure compared with mAb1 dosed alone in normal mice or in PCSK9 knockout mice lacking antigen. To identify mAb-binding characteristics that minimize lysosomal disposition, the pharmacokinetic behavior of four mAbs representing a diverse range of PCSK9-binding affinities at neutral (serum) and acidic (endosomal) pH was evaluated in cynomolgus monkeys. Results revealed an inverse correlation between affinity and both mAb exposure and duration of LDL-C lowering. High-affinity mAb1 exhibited the lowest exposure and shortest duration of action (6 days), whereas mAb2 displayed prolonged exposure and LDL-C reduction (51 days) as a consequence of lower affinity and pH-sensitive PCSK9 binding. mAbs with shorter endosomal PCSK9:mAb complex dissociation half-lives (<20 seconds) produced optimal exposure-response profiles. Interestingly, incorporation of previously reported Fc-region amino acid substitutions or novel loop-insertion peptides that enhance in vitro neonatal Fc receptor binding, led to only modest pharmacokinetic improvements for mAbs with pH-dependent PCSK9 binding, with only limited augmentation of pharmacodynamic activity relative to native mAbs. A pivotal role for PCSK9 in mAb clearance was demonstrated, more broadly suggesting that therapeutic mAb-binding characteristics require optimization based on target pharmacology.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cholesterol, LDL/blood , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Immunoglobulin Fc Fragments/genetics , Macaca mulatta , Male , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 9 , Proprotein Convertases/genetics , Proprotein Convertases/immunology , Protein Binding , Receptors, Fc/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology
3.
J Biol Chem ; 290(12): 7535-62, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25583986

ABSTRACT

Producing pure and well behaved bispecific antibodies (bsAbs) on a large scale for preclinical and clinical testing is a challenging task. Here, we describe a new strategy for making monovalent bispecific heterodimeric IgG antibodies in mammalian cells. We applied an electrostatic steering mechanism to engineer antibody light chain-heavy chain (LC-HC) interface residues in such a way that each LC strongly favors its cognate HC when two different HCs and two different LCs are co-expressed in the same cell to assemble a functional bispecific antibody. We produced heterodimeric IgGs from transiently and stably transfected mammalian cells. The engineered heterodimeric IgG molecules maintain the overall IgG structure with correct LC-HC pairings, bind to two different antigens with comparable affinity when compared with their parental antibodies, and retain the functionality of parental antibodies in biological assays. In addition, the bispecific heterodimeric IgG derived from anti-HER2 and anti-EGF receptor (EGFR) antibody was shown to induce a higher level of receptor internalization than the combination of two parental antibodies. Mouse xenograft BxPC-3, Panc-1, and Calu-3 human tumor models showed that the heterodimeric IgGs strongly inhibited tumor growth. The described approach can be used to generate tools from two pre-existent antibodies and explore the potential of bispecific antibodies. The asymmetrically engineered Fc variants for antibody-dependent cellular cytotoxicity enhancement could be embedded in monovalent bispecific heterodimeric IgG to make best-in-class therapeutic antibodies.


Subject(s)
Immunoglobulin G/chemistry , Static Electricity , Amino Acids/chemistry , Animals , Antibody-Dependent Cell Cytotoxicity , CHO Cells , Cell Line , Cricetulus , Dimerization , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology , Mice , Protein Engineering , Surface Plasmon Resonance
4.
J Biol Chem ; 289(6): 3571-90, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24311787

ABSTRACT

Antibody-dependent cellular cytotoxicity (ADCC) is mediated through the engagement of the Fc segment of antibodies with Fcγ receptors (FcγRs) on immune cells upon binding of tumor or viral antigen. The co-crystal structure of FcγRIII in complex with Fc revealed that Fc binds to FcγRIII asymmetrically with two Fc chains contacting separate regions of the FcγRIII by utilizing different residues. To fully explore this asymmetrical nature of the Fc-FcγR interaction, we screened more than 9,000 individual clones in Fc heterodimer format in which different mutations were introduced at the same position of two Fc chains using a high throughput competition AlphaLISA® assay. To this end, we have identified a panel of novel Fc variants with significant binding improvement to FcγRIIIA (both Phe-158 and Val-158 allotypes), increased ADCC activity in vitro, and strong tumor growth inhibition in mice xenograft human tumor models. Compared with previously identified Fc variants in conventional IgG format, Fc heterodimers with asymmetrical mutations can achieve similar or superior potency in ADCC-mediated tumor cell killing and demonstrate improved stability in the CH2 domain. Fc heterodimers also allow more selectivity toward activating FcγRIIA than inhibitory FcγRIIB. Afucosylation of Fc variants further increases the affinity of Fc to FcγRIIIA, leading to much higher ADCC activity. The discovery of these Fc variants will potentially open up new opportunities of building the next generation of therapeutic antibodies with enhanced ADCC effector function for the treatment of cancers and infectious diseases.


Subject(s)
Antibodies, Neoplasm , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G , Protein Engineering , Receptors, IgG/immunology , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Cell Line, Tumor , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Mice, SCID , Mutation , Neoplasms , Receptors, IgG/genetics , Xenograft Model Antitumor Assays
5.
Neurobiol Dis ; 45(3): 913-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198572

ABSTRACT

Insufficiency of the transcriptional regulator GTF2IRD1 has become a strong potential explanation for some of the major characteristic features of the neurodevelopmental disorder Williams-Beuren syndrome (WBS). Genotype/phenotype correlations in humans indicate that the hemizygous loss of the GTF2IRD1 gene and an adjacent paralogue, GTF2I, play crucial roles in the neurocognitive and craniofacial aspects of the disease. In order to explore this genetic relationship in greater detail, we have generated a targeted Gtf2ird1 mutation in mice that blocks normal GTF2IRD1 protein production. Detailed analyses of homozygous null Gtf2ird1 mice have revealed a series of phenotypes that share some intriguing parallels with WBS. These include reduced body weight, a facial deformity resulting from localised epidermal hyperplasia, a motor coordination deficit, alterations in exploratory activity and, in response to specific stress-inducing stimuli; a novel audible vocalisation and increased serum corticosterone. Analysis of Gtf2ird1 expression patterns in the brain using a knock-in LacZ reporter and c-fos activity mapping illustrates the regions where these neurological abnormalities may originate. These data provide new mechanistic insight into the clinical genetic findings in WBS patients and indicate that insufficiency of GTF2IRD1 protein contributes to abnormalities of facial development, motor function and specific behavioural disorders that accompany this disease.


Subject(s)
Focal Epithelial Hyperplasia/etiology , Motor Skills Disorders/etiology , Muscle Proteins/genetics , Mutation/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Vocalization, Animal/physiology , Williams Syndrome/complications , Analysis of Variance , Animals , Animals, Newborn/blood , Body Temperature/genetics , Body Weight/genetics , Brain/metabolism , Circadian Rhythm/genetics , Corticosterone/blood , Disease Models, Animal , Exploratory Behavior/physiology , Fats , Female , Focal Epithelial Hyperplasia/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Skills Disorders/genetics , Muscle Strength , Muscle, Skeletal/pathology , Phenotype , Sex Factors , Sleep/genetics , Sound Spectrography , Stress, Psychological/genetics , Swimming/psychology , Williams Syndrome/genetics , Williams Syndrome/pathology
6.
Comb Chem High Throughput Screen ; 13(2): 170-87, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20053162

ABSTRACT

This review describes methods for quantifying the binding of small molecule drug candidates to plasma proteins and the application of these methods in drug discovery and development. Particular attention is devoted to methods amenable to medium-to-high throughput analysis and those well suited for measurement of compounds that are highly protein bound. The methods reviewed herein include the conventional techniques of equilibrium dialysis, ultrafiltration and ultracentrifugation, as well as some more novel approaches utilizing micropartitioning and biosensor-based analysis. Additional concepts that are discussed include plasma protein structure, enantioselective protein binding, drug displacement, the effect of patient demographics and disease states on free (unbound) drug levels, and the influence of protein binding on drug candidate pharmacokinetics and pharmacodynamics. Practical considerations pertaining to the evaluation of highly protein bound drug candidates are also highlighted.


Subject(s)
Blood Proteins/metabolism , Drug Discovery/methods , Pharmaceutical Preparations/metabolism , Biosensing Techniques/methods , High-Throughput Screening Assays/methods , Humans , Protein Binding
7.
Hum Mol Genet ; 18(4): 753-66, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19008301

ABSTRACT

Dysregulation of the serotonergic signaling system has been implicated in the pathology of mood disorders including depression, and various rodent models of disrupted serotonergic signaling display depression-related behavioral phenotypes. Depression is a common neuropsychiatric feature of preclinical Huntington's disease (HD) but the underlying changes in the HD brain contributing to the development of depression are unknown. Using the R6/1 transgenic mouse model of HD, we show that pre-motor symptomatic HD mice display sex-specific depressive-related behaviors on the forced-swim (FST), tail-suspension (TST) and novelty-suppressed feeding (NSFT) tests while having muted responses to acute anti-depressant administration. The baseline behaviors of HD mice were similar to the behavioral phenotypes of serotonin (5-HT) receptor and transporter null mutants, and gene expression of specific serotonin receptors were subsequently found to be reduced in the hippocampus and cortex of HD mice. Female HD mice had an additional deficit in cortical expression of serotonin transporter (SerT). Environmental enrichment normalized the FST behavioral response of female HD mice corresponding with increased gene expression of specific 5-HT receptors in the hippocampus and cortex. Our findings implicate altered serotonergic signaling as the basis for the development of depression during the preclinical stages of HD.


Subject(s)
Depression/physiopathology , Gene Expression , Huntington Disease/psychology , Receptor, Serotonin, 5-HT1B/metabolism , Receptors, Serotonin/metabolism , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal , Body Weight , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Female , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/physiopathology , Male , Mice , Mice, Transgenic , Motor Activity , Receptor, Serotonin, 5-HT1B/genetics , Receptors, Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Species Specificity
8.
Eur J Neurosci ; 28(4): 717-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18702691

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterised by motor neuron degeneration, muscle wasting and paralysis. While twin studies support a role for both genetic and environmental factors in ALS, the nature of environmental modifiers is unknown. We therefore compared onset and progression of disease symptoms in female and male transgenic ALS mice (expressing the human SOD1(G93A) gene mutation) and their wild-type littermates, housed in environmentally enriched versus standard conditions. Environmental enrichment significantly improved motor performance, as measured using the accelerating rotarod, in particular for female mice. This enhanced motor coordination was observed for both SOD1(G93A) and wild-type mice, suggesting this effect is independent of genotype. Female SOD1(G93A) mice housed with environmental enrichment were found to reach overt end-stage disease sooner than their standard-housed littermates. However, male SOD1(G93A) mice did not show significantly accelerated disease progression. This evidence for environmental modulation of ALS pathogenesis in transgenic mice provides insights into activity-dependent aspects of the disease process, and may help identify molecular targets for pharmacological modulators as future therapeutics.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Behavior, Animal/physiology , Disease Models, Animal , Environment , Housing, Animal , Mice, Transgenic , Motor Activity/physiology , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Progression , Female , Humans , Male , Mice , Sex Factors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Survival Rate
9.
Hippocampus ; 18(8): 824-34, 2008.
Article in English | MEDLINE | ID: mdl-18493969

ABSTRACT

The complexity of the genetics underlying schizophrenia is highlighted by the multitude of molecular pathways that have been reported to be disrupted in the disorder including muscarinic, serotonergic, and glutamatergic signaling systems. It is of interest, therefore, that phospholipase C-beta1 (PLC-beta1) acts as a point of convergence for these pathways during cortical development and plasticity. These signaling pathways, furthermore, are susceptible to modulation by RGS4, one of the more promising candidate genes for schizophrenia. PLC-beta1 knockout mice were behaviorally assessed on tests including fear conditioning, elevated plus maze, and the Y maze. In situ hybridization was used to assess RGS4 expression. We found that PLC-beta1 knockout mice display abnormal anxiety profiles on some, but not all measures assessed, including decreased anxiety on the elevated plus maze. We also show memory impairment and a complete absence of acquisition of hippocampal-dependent fear conditioning. Furthermore, at a molecular level, we demonstrate dramatic changes in expression of RGS4 mRNA in selective regions of the PLC-beta1 knockout mouse brain, particularly the CA1 region of the hippocampus. These results validate the utility of the PLC-beta1 knockout mouse as a model of schizophrenia, including molecular and cellular evidence for disrupted cortical maturation and associated behavioral endophenotypes.


Subject(s)
Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Phospholipase C beta/deficiency , RGS Proteins/genetics , Animals , Anxiety/physiopathology , Base Sequence , Behavior, Animal/physiology , Conditioning, Psychological/physiology , DNA Primers/genetics , Fear/physiology , Female , Gene Expression Regulation , Hippocampus/physiology , Humans , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phospholipase C beta/genetics , Phospholipase C beta/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizophrenia/etiology , Schizophrenia/genetics , Schizophrenia/physiopathology , Signal Detection, Psychological
10.
Eur J Neurosci ; 22(8): 2081-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16262645

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG trinucleotide repeat encoding an extended polyglutamine tract in the huntingtin protein. Affected individuals display progressive motor, cognitive and psychiatric symptoms (including depression), leading to terminal decline. Given that transgenic HD mice have decreased hippocampal cell proliferation and that a deficit in neurogenesis has been postulated as an underlying cause of depression, we hypothesized that decreased hippocampal neurogenesis contributes to depressive symptoms and cognitive decline in HD. Fluoxetine, a serotonin-reuptake inhibitor commonly prescribed for the treatment of depression, is known to increase neurogenesis in the dentate gyrus of wild-type mouse hippocampus. Here we show that hippocampal-dependent cognitive and depressive-like behavioural symptoms occur in HD mice, and that the administration of fluoxetine produces a marked improvement in these deficits. Furthermore, fluoxetine was found to rescue deficits of neurogenesis and volume loss in the dentate gyrus of HD mice.


Subject(s)
Cell Proliferation/drug effects , Cognition Disorders/etiology , Fluoxetine/therapeutic use , Huntington Disease/complications , Neurons/drug effects , Selective Serotonin Reuptake Inhibitors/therapeutic use , Age Factors , Animals , Behavior, Animal , Cells, Cultured , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Dentate Gyrus/cytology , Disease Models, Animal , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Humans , Huntingtin Protein , Huntington Disease/drug therapy , Huntington Disease/pathology , Immunohistochemistry/methods , Mice , Mice, Transgenic , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Rotarod Performance Test/methods , Trinucleotide Repeat Expansion
11.
Clin Exp Metastasis ; 22(8): 643-52, 2005.
Article in English | MEDLINE | ID: mdl-16649073

ABSTRACT

The uPA/uPAR system is involved in tumour progression and metastasis of a variety of cancers. Previously, we have shown that increased expression of urokinase plasminogen activator (uPA) correlated with malignancy grade in certain sarcomas. A study looking at in vivo inhibition of this system has not been done to date for osteosarcoma. More recently, this laboratory developed a clinically relevant mouse model where intratibial injection of UMR106-01 cells resulted in the development of osteosarcoma and lung metastases. Expression of uPA and its receptor (uPAR) were localised to the invading front of the tumours. Pulmonary metastasis is a predominant feature of the disease and is the major cause of death in patients. In the present study, the effects of down-regulating uPAR were observed in vitro and in vivo. UMR106-01 cells were transfected with either antisense-uPAR or vector control plasmids. Two antisense clones, exhibiting uPAR downregulation, demonstrated decreased adhesion, migration and invasion in cell-based assays in vitro (P<0.05). Cellular proliferation was not affected by uPAR downregulation. In vivo, a marked reduction of 80% in tibial tumour volumes (P<0.05), and total inhibition of pulmonary metastases were observed in mice injected with the more potent of the antisense clones. This study proves seminally the usefulness of uPAR antisense in curbing the growth and spread of osteosarcoma.


Subject(s)
Cell Division/physiology , Down-Regulation , Lung Neoplasms/secondary , Receptors, Cell Surface/physiology , Animals , Blotting, Western , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Receptors, Urokinase Plasminogen Activator
12.
Cell Tissue Res ; 307(3): 337-45, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11904770

ABSTRACT

Prostatic carcinoma affects 1 in 11 men and targets bone with sclerotic metastases. The study of prostate carcinoma growth in bone has been hampered by the lack of suitable animal models. We have developed an in vivo model of prostate carcinoma growth in bone by inoculating three human prostate carcinoma cell lines (PC-3, DU-145, and LNCaP) into the tibia of congenitally athymic mice. Developing tumors were analyzed by radiographic, histologic, immunohistochemical, and in situ hybridization examination. Seven of the nine PC-3 inoculated mice and all (9/9) of the DU-145 inoculated mice developed tumors in the injected limb. In contrast, inoculation with LNCaP cells failed to produce tumors (0/9). Radiologically, the tumors had a mixed sclerotic/lytic appearance with extracortical extension. All the PC-3 tumors invaded the bone marrow cavity, cortical bone, and surrounding soft tissue. The DU-145 tumors were confined to the bone marrow cavity in 7/9 animals. CK18 and Ki67 localization identified the human tumor cells and their proliferative activity, respectively. The PC-3- and DU-145-induced tibial tumors expressed alpha(1)I procollagen and osteopontin mRNA, to varying degrees. All the tumors demonstrated an up-regulation of osteoclasts at the bone/tumor interface compared with the control limbs. Thus, this is a reliable and reproducible in vivo model of prostate carcinoma growth in bone enabling the study of the interactions that occur between prostate cancer cells and bone at an important part of the metastatic cascade, namely, growth and invasion at a distant site.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Disease Models, Animal , Prostatic Neoplasms/pathology , Animals , Bone Marrow/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/physiopathology , Cell Transplantation , Collagen Type I/genetics , Collagen Type I/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteopontin , Prostatic Neoplasms/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Tibia , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...