Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348885

ABSTRACT

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Subject(s)
Benzamides/metabolism , Bridged Bicyclo Compounds/pharmacology , Heptanes/pharmacology , Lysosomes/drug effects , Vesicular Transport Proteins/metabolism , Activating Transcription Factor 6/metabolism , Animals , Benzamides/chemistry , Benzamides/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Frameshift Mutation , Heptanes/therapeutic use , Humans , Imidazoline Receptors/antagonists & inhibitors , Imidazoline Receptors/genetics , Imidazoline Receptors/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lysosomes/metabolism , Male , Mice , Mice, Transgenic , Mucin-1/chemistry , Mucin-1/genetics , Mucin-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Unfolded Protein Response/drug effects , Vesicular Transport Proteins/chemistry
3.
Nature ; 547(7662): 217-221, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678778

ABSTRACT

Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Melanoma/immunology , Melanoma/therapy , Precision Medicine/methods , Amino Acid Sequence , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/chemistry , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class II/immunology , Humans , Machine Learning , Melanoma/genetics , Mutation , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Patient Safety , Programmed Cell Death 1 Receptor/antagonists & inhibitors
5.
Nat Biotechnol ; 23(3): 329-36, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711537

ABSTRACT

Kinase inhibitors show great promise as a new class of therapeutics. Here we describe an efficient way to determine kinase inhibitor specificity by measuring binding of small molecules to the ATP site of kinases. We have profiled 20 kinase inhibitors, including 16 that are approved drugs or in clinical development, against a panel of 119 protein kinases. We find that specificity varies widely and is not strongly correlated with chemical structure or the identity of the intended target. Many novel interactions were identified, including tight binding of the p38 inhibitor BIRB-796 to an imatinib-resistant variant of the ABL kinase, and binding of imatinib to the SRC-family kinase LCK. We also show that mutations in the epidermal growth factor receptor (EGFR) found in gefitinib-responsive patients do not affect the binding affinity of gefitinib or erlotinib. Our results represent a systematic small molecule-protein interaction map for clinical compounds across a large number of related proteins.


Subject(s)
Drug Design , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Pharmaceutical Preparations/metabolism , Piperazines/metabolism , Protein Interaction Mapping/methods , Protein Kinase Inhibitors/metabolism , Pyrimidines/metabolism , Benzamides , Imatinib Mesylate , Microchemistry/methods , Protein Binding
6.
Nucleic Acids Res ; 31(10): e57, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12736323

ABSTRACT

A common method for generating mice with subtle genetic manipulations uses homologous recombination (HR) in embryonic stem (ES) cells to replace a wild-type gene with a slightly modified one. Generally, a drug resistance gene is inserted with the modified gene to select correctly targeted clones. Often, however, the presence of this drug resistance gene interferes with the normal locus and creates a null or hypomorphic allele. Flanking of the selectable marker by loxP sites followed by Cre-mediated deletion after drug selection can overcome this problem. The simplest method used to remove a loxP-flanked selectable marker is to breed an animal carrying a loxP-flanked drug resistance gene to an animal that expresses Cre recombinase in the germline. To date only outbred transgenic mice are available for this purpose. This can be problematic for phenotypic analysis in many organ systems, including the brain, and for the analysis of behavior. While attempting to make 129S6/SvEvTac inbred background (isogenic to our ES cells) mice that express Cre under the control of several tissue-specific promoters, we serendipitously generated a line that excises loxP-flanked drug resistance genes in all tissues, including the germline. This reagent allows deletion of loxP-flanked sequences while maintaining the mutation on an inbred background.


Subject(s)
Gene Deletion , Integrases/genetics , Viral Proteins/genetics , Animals , Binding Sites/genetics , Drug Resistance/genetics , Female , Genome , Green Fluorescent Proteins , Homozygote , Hypoxanthine Phosphoribosyltransferase/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Confocal , Mutagenesis, Insertional , Mutation , Recombination, Genetic , Viral Proteins/metabolism
7.
J Mol Diagn ; 18(4): 566-71, 2016 07.
Article in English | MEDLINE | ID: mdl-27157321

ABSTRACT

Mucin-1 kidney disease, previously described as medullary cystic kidney disease type 1 (MCKD1, OMIM 174000), is an autosomal dominant tubulointerstitial kidney disease recently shown to be caused by a single-base insertion within the variable number tandem repeat region of the MUC1 gene. Because of variable age of disease onset and often subtle signs and symptoms, clinical diagnosis of mucin-1 kidney disease and differentiation from other forms of hereditary kidney disease have been difficult. The causal insertion resides in a variable number tandem repeat region with high GC content, which has made detection by standard next-generation sequencing impossible to date. The inherently difficult nature of this mutation required an alternative method for routine detection and clinical diagnosis of the disease. We therefore developed and validated a mass spectrometry-based probe extension assay with a series of internal controls to detect the insertion event using 24 previously characterized positive samples from patients with mucin-1 kidney disease and 24 control samples known to be wild type for the variant. Validation results indicate an accurate and reliable test for clinically establishing the molecular diagnosis of mucin-1 kidney disease with 100% sensitivity and specificity across 275 tests called.


Subject(s)
Mass Spectrometry/methods , Molecular Diagnostic Techniques , Mucin-1/genetics , Mutation , Polycystic Kidney, Autosomal Dominant/diagnosis , Polycystic Kidney, Autosomal Dominant/genetics , Genotype , Humans , Mass Spectrometry/standards , Reproducibility of Results , Sensitivity and Specificity , Workflow
8.
Cancer Immunol Res ; 1(1): 11-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24777245

ABSTRACT

Despite years of preclinical efforts and hundreds of clinical studies, therapeutic cancer vaccines with the routine ability to limit or eliminate tumor growth in humans have been elusive. With advances in genome sequencing, it is now possible to identify a new class of tumor-specific antigens derived from mutated proteins that are present only in the tumor. These "neoantigens" should provide highly specific targets for antitumor immunity. Although many challenges remain in producing and testing neoantigen-based vaccines customized for each patient, a neoantigen vaccine offers a promising new approach to induce highly focused antitumor T cells aimed at eradicating cancer cells.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Neoplasms/immunology , Neoplasms/therapy , Humans , Precision Medicine/methods
9.
J Psychiatr Res ; 46(12): 1553-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22998742

ABSTRACT

We used in vitro neurogenesis-based human neural stem cell (hNSCs) assays and rodent in vivo behavioral assays to identify potential novel antidepressants. A combination of buspirone and melatonin displayed antidepressant activity in these assays whereas neither buspirone nor melatonin alone showed any antidepressant-like profile. After evaluating numerous combination ratios, we determined that low dose buspirone 15 mg combined with melatonin-SR 3 mg yielded optimal antidepressant efficacy in our pre-clinical platform. The low dose of buspirone suggested that antidepressant efficacy might be achieved with only minimal adverse event liability. Based on these data, we conducted an exploratory 6-week, multi-center, double-blind, randomized, placebo- and comparator-controlled study of the combination of buspirone and melatonin in subjects with acute Major Depressive Disorder (MDD). The combination treatment revealed a significant antidepressant response in subjects with MDD on several measures (Clinical Global Impression of Severity and Improvement, Inventory of Depressive Symptomatology) compared to either placebo or buspirone 15 mg monotherapy. These preliminary findings have clinical implications and suggest that a platform of pre-clinical neurogenesis matched with confirmatory behavioral assays may be useful as a drug discovery strategy.


Subject(s)
Behavior, Animal/drug effects , Buspirone/administration & dosage , Depressive Disorder, Major/drug therapy , Melatonin/administration & dosage , Neurogenesis/drug effects , Adult , Animals , Buspirone/adverse effects , Buspirone/pharmacology , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/pharmacology , Double-Blind Method , Drug Synergism , Drug Therapy, Combination , Female , Humans , Male , Melatonin/adverse effects , Melatonin/pharmacology , Middle Aged , Placebos , Rats , Rats, Inbred F344 , Treatment Outcome
10.
J Med Chem ; 52(23): 7808-16, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19754199

ABSTRACT

Treatment of AML patients with small molecule inhibitors of FLT3 kinase has been explored as a viable therapy. However, these agents are found to be less than optimal for the treatment of AML because of lack of sufficient potency or suboptimal oral pharmacokinetics (PK) or lack of adequate tolerability at efficacious doses. We have developed a series of extremely potent and highly selective FLT3 inhibitors with good oral PK properties. The first series of compounds represented by 1 (AB530) was found to be a potent and selective FLT3 kinase inhibitor with good PK properties. The aqueous solubility and oral PK properties at higher doses in rodents were found to be less than optimal for clinical development. A novel series of compounds were designed lacking the carboxamide group of 1 with an added water solubilizing group. Compound 7 (AC220) was identified from this series to be the most potent and selective FLT3 inhibitor with good pharmaceutical properties, excellent PK profile, and superior efficacy and tolerability in tumor xenograft models. Compound 7 has demonstrated a desirable safety and PK profile in humans and is currently in phase II clinical trials.


Subject(s)
Benzothiazoles/pharmacology , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Rats , Solubility , Substrate Specificity , Xenograft Model Antitumor Assays
11.
Genome Biol ; 6(6): R48, 2005.
Article in English | MEDLINE | ID: mdl-15960800

ABSTRACT

BACKGROUND: Progressive neurological dysfunction is a key aspect of human aging. Because of underlying differences in the aging of mice and humans, useful mouse models have been difficult to obtain and study. We have used gene-expression analysis and polymorphism screening to study molecular senescence of the retina and hippocampus in two rare inbred mouse models of accelerated neurological senescence (SAMP8 and SAMP10) that closely mimic human neurological aging, and in a related normal strain (SAMR1) and an unrelated normal strain (C57BL/6J). RESULTS: The majority of age-related gene expression changes were strain-specific, with only a few common pathways found for normal and accelerated neurological aging. Polymorphism screening led to the identification of mutations that could have a direct impact on important disease processes, including a mutation in a fibroblast growth factor gene, Fgf1, and a mutation in and ectopic expression of the gene for the chemokine CCL19, which is involved in the inflammatory response. CONCLUSION: We show that combining the study of inbred mouse strains with interesting traits and gene-expression profiling can lead to the discovery of genes important for complex phenotypes. Furthermore, full-genome polymorphism detection, sequencing and gene-expression profiling of inbred mouse strains with interesting phenotypic differences may provide unique insights into the molecular genetics of late-manifesting complex diseases.


Subject(s)
Aging/genetics , Aging/physiology , Amino Acid Sequence , Animals , Brain/cytology , Chemokine CCL19 , Chemokines, CC/chemistry , Chemokines, CC/genetics , Chromosomes, Mammalian/genetics , Fibroblast Growth Factor 1/genetics , Gene Expression Profiling , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutation/genetics , Nervous System/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Species Specificity
12.
Proc Natl Acad Sci U S A ; 102(31): 11011-6, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16046538

ABSTRACT

To realize the full potential of targeted protein kinase inhibitors for the treatment of cancer, it is important to address the emergence of drug resistance in treated patients. Mutant forms of BCR-ABL, KIT, and the EGF receptor (EGFR) have been found that confer resistance to the drugs imatinib, gefitinib, and erlotinib. The mutations weaken or prevent drug binding, and interestingly, one of the most common sites of mutation in all three kinases is a highly conserved "gatekeeper" threonine residue near the kinase active site. We have identified existing clinical compounds that bind and inhibit drug-resistant mutant variants of ABL, KIT, and EGFR. We found that the Aurora kinase inhibitor VX-680 and the p38 inhibitor BIRB-796 inhibit the imatinib- and BMS-354825-resistant ABL(T315I) kinase. The KIT/FLT3 inhibitor SU-11248 potently inhibits the imatinib-resistant KIT(V559D/T670I) kinase, consistent with the clinical efficacy of SU-11248 against imatinib-resistant gastrointestinal tumors, and the EGFR inhibitors EKB-569 and CI-1033, but not GW-572016 and ZD-6474, potently inhibit the gefitinib- and erlotinib-resistant EGFR(L858R/T790M) kinase. EKB-569 and CI-1033 are already in clinical trials, and our results suggest that they should be considered for testing in the treatment of gefitinib/erlotinib-resistant non-small cell lung cancer. The results highlight the strategy of screening existing clinical compounds against newly identified drug-resistant mutant variants to find compounds that may serve as starting points for the development of next-generation drugs, or that could be used directly to treat patients that have acquired resistance to first-generation targeted therapy.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Aminoquinolines , Aniline Compounds , Cell Line , Drug Resistance, Neoplasm/genetics , Humans , Indoles/pharmacology , Kinetics , Morpholines/pharmacology , Mutation , Naphthalenes/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Organic Chemicals/pharmacology , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrroles/pharmacology , Sunitinib
SELECTION OF CITATIONS
SEARCH DETAIL