Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Publication year range
1.
Science ; 381(6659): 794-799, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590355

ABSTRACT

The discovery of small-molecule inhibitors requires suitable binding pockets on protein surfaces. Proteins that lack this feature are considered undruggable and require innovative strategies for therapeutic targeting. KRAS is the most frequently activated oncogene in cancer, and the active state of mutant KRAS is such a recalcitrant target. We designed a natural product-inspired small molecule that remodels the surface of cyclophilin A (CYPA) to create a neomorphic interface with high affinity and selectivity for the active state of KRASG12C (in which glycine-12 is mutated to cysteine). The resulting CYPA:drug:KRASG12C tricomplex inactivated oncogenic signaling and led to tumor regressions in multiple human cancer models. This inhibitory strategy can be used to target additional KRAS mutants and other undruggable cancer drivers. Tricomplex inhibitors that selectively target active KRASG12C or multiple RAS mutants are in clinical trials now (NCT05462717 and NCT05379985).


Subject(s)
Biological Products , Cyclophilin A , Immunophilins , Molecular Chaperones , Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/therapeutic use , Cysteine/chemistry , Cysteine/genetics , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Cyclophilin A/chemistry , Cyclophilin A/metabolism , Immunophilins/chemistry , Immunophilins/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
2.
Nature ; 619(7968): 160-166, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37258666

ABSTRACT

KRAS is one of the most commonly mutated proteins in cancer, and efforts to directly inhibit its function have been continuing for decades. The most successful of these has been the development of covalent allele-specific inhibitors that trap KRAS G12C in its inactive conformation and suppress tumour growth in patients1-7. Whether inactive-state selective inhibition can be used to therapeutically target non-G12C KRAS mutants remains under investigation. Here we report the discovery and characterization of a non-covalent inhibitor that binds preferentially and with high affinity to the inactive state of KRAS while sparing NRAS and HRAS. Although limited to only a few amino acids, the evolutionary divergence in the GTPase domain of RAS isoforms was sufficient to impart orthosteric and allosteric constraints for KRAS selectivity. The inhibitor blocked nucleotide exchange to prevent the activation of wild-type KRAS and a broad range of KRAS mutants, including G12A/C/D/F/V/S, G13C/D, V14I, L19F, Q22K, D33E, Q61H, K117N and A146V/T. Inhibition of downstream signalling and proliferation was restricted to cancer cells harbouring mutant KRAS, and drug treatment suppressed KRAS mutant tumour growth in mice, without having a detrimental effect on animal weight. Our study suggests that most KRAS oncoproteins cycle between an active state and an inactive state in cancer cells and are dependent on nucleotide exchange for activation. Pan-KRAS inhibitors, such as the one described here, have broad therapeutic implications and merit clinical investigation in patients with KRAS-driven cancers.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Animals , Mice , Body Weight , Enzyme Activation , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Nucleotides/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , Cell Division/drug effects , Substrate Specificity
3.
Science ; 374(6564): 197-201, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34618566

ABSTRACT

Recently reported to be effective in patients with lung cancer, KRASG12C inhibitors bind to the inactive, or guanosine diphosphate (GDP)­bound, state of the oncoprotein and require guanosine triphosphate (GTP) hydrolysis for inhibition. However, KRAS mutations prevent the catalytic arginine of GTPase-activating proteins (GAPs) from enhancing an otherwise slow hydrolysis rate. If KRAS mutants are indeed insensitive to GAPs, it is unclear how KRASG12C hydrolyzes sufficient GTP to allow inactive state­selective inhibition. Here, we show that RGS3, a GAP previously known for regulating G protein­coupled receptors, can also enhance the GTPase activity of mutant and wild-type KRAS proteins. Our study reveals an unexpected mechanism that inactivates KRAS and explains the vulnerability to emerging clinically effective therapeutics.


Subject(s)
GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Lung Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , RGS Proteins/metabolism , Animals , Cell Extracts , Cell Line, Tumor , Enzyme Activation , Humans , Hydrolysis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice, Nude , Proto-Oncogene Proteins p21(ras)/genetics , RGS Proteins/genetics , Signal Transduction , Xenograft Model Antitumor Assays
4.
Nature ; 577(7790): 421-425, 2020 01.
Article in English | MEDLINE | ID: mdl-31915379

ABSTRACT

KRAS GTPases are activated in one-third of cancers, and KRAS(G12C) is one of the most common activating alterations in lung adenocarcinoma1,2. KRAS(G12C) inhibitors3,4 are in phase-I clinical trials and early data show partial responses in nearly half of patients with lung cancer. How cancer cells bypass inhibition to prevent maximal response to therapy is not understood. Because KRAS(G12C) cycles between an active and inactive conformation4-6, and the inhibitors bind only to the latter, we tested whether isogenic cell populations respond in a non-uniform manner by studying the effect of treatment at a single-cell resolution. Here we report that, shortly after treatment, some cancer cells are sequestered in a quiescent state with low KRAS activity, whereas others bypass this effect to resume proliferation. This rapid divergent response occurs because some quiescent cells produce new KRAS(G12C) in response to suppressed mitogen-activated protein kinase output. New KRAS(G12C) is maintained in its active, drug-insensitive state by epidermal growth factor receptor and aurora kinase signalling. Cells without these adaptive changes-or cells in which these changes are pharmacologically inhibited-remain sensitive to drug treatment, because new KRAS(G12C) is either not available or exists in its inactive, drug-sensitive state. The direct targeting of KRAS oncoproteins has been a longstanding objective in precision oncology. Our study uncovers a flexible non-uniform fitness mechanism that enables groups of cells within a population to rapidly bypass the effect of treatment. This adaptive process must be overcome if we are to achieve complete and durable responses in the clinic.


Subject(s)
Mutation , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Adaptation, Biological , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects
5.
Univ. med ; 59(4): 1-11, 2018. ilus
Article in Spanish | LILACS, COLNAL | ID: biblio-995609

ABSTRACT

La intoxicación por acetaminofén es una de las causas más frecuentes de falla hepática aguda fulminante y una de las principales causas de atención en los servicios de urgencias, por intoxicación debida a medicamentos. Recientemente, se han registrado avances en la comprensión fisiopatológica, diagnóstico y tratamiento de esta entidad clínica. Los mecanismos de estrés oxidativo y daño del ADN mitocondrial explican los daños producidos en esta intoxicación. En esta revisión se presentan aspectos referentes a la epidemiología, mecanismos fisiopatológicos, diagnóstico y tratamiento de la intoxicación por acetaminofén.


Acetaminophen poisoning is one of the most frequent causes of acute fulminant hepatic failure and one of the main causes of care in emergency services due to drug intoxication. Recent progress has been made in the pathophysiological understanding, diagnosis and treatment of this clinical entity. The mechanisms of oxidative stress and mitochondrial DNA damage explain the damage caused by this intoxication. This review presents aspects related to the epidemiology, pathophysiological mechanisms, diagnosis and treatment of acetaminophen intoxication.


Subject(s)
Humans , Drug Overdose , Chemical and Drug Induced Liver Injury , Acetaminophen
6.
Nat Med ; 23(8): 929-937, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28714990

ABSTRACT

The principles that govern the evolution of tumors exposed to targeted therapy are poorly understood. Here we modeled the selection and propagation of an amplification in the BRAF oncogene (BRAFamp) in patient-derived tumor xenografts (PDXs) that were treated with a direct inhibitor of the kinase ERK, either alone or in combination with other ERK signaling inhibitors. Single-cell sequencing and multiplex fluorescence in situ hybridization analyses mapped the emergence of extra-chromosomal amplification in parallel evolutionary trajectories that arose in the same tumor shortly after treatment. The evolutionary selection of BRAFamp was determined by the fitness threshold, the barrier that subclonal populations need to overcome to regain fitness in the presence of therapy. This differed for inhibitors of ERK signaling, suggesting that sequential monotherapy is ineffective and selects for a progressively higher BRAF copy number. Concurrent targeting of the RAF, MEK and ERK kinases, however, imposed a sufficiently high fitness threshold to prevent the propagation of subclones with high-level BRAFamp. When administered on an intermittent schedule, this treatment inhibited tumor growth in 11/11 PDXs of lung cancer or melanoma without apparent toxicity in mice. Thus, gene amplification can be acquired and expanded through parallel evolution, enabling tumors to adapt while maintaining their intratumoral heterogeneity. Treatments that impose the highest fitness threshold will likely prevent the evolution of resistance-causing alterations and, thus, merit testing in patients.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Adult , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/administration & dosage , Carboplatin/administration & dosage , Cisplatin/administration & dosage , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Male , Melanoma/genetics , Melanoma/secondary , Mice , Middle Aged , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Pemetrexed/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Single-Cell Analysis , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays , raf Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...