Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 693
Filter
2.
Bioorg Chem ; 147: 107316, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38583246

ABSTRACT

Ras GTPases and other CaaX proteins undergo multiple post-translational modifications at their carboxyl-terminus. These events initiate with prenylation of a cysteine and are followed by endoproteolytic removal of the 'aaX' tripeptide and carboxylmethylation. Some CaaX proteins are only subject to prenylation, however, due to the presence of an uncleavable sequence. In this study, uncleavable sequences were used to stage Ras isoforms in a farnesylated and uncleaved state to address the impact of CaaX proteolysis on protein localization and function. This targeted strategy is more specific than those that chemically inhibit the Rce1 CaaX protease or delete the RCE1 gene because global abrogation of CaaX proteolysis impacts the entire CaaX protein proteome and effects cannot be attributed to any specific CaaX protein of the many concurrently affected. With this targeted strategy, clear mislocalization and reduced activity of farnesylated and uncleaved Ras isoforms was observed. In addition, new peptidomimetics based on cleavable Ras CaaX sequences and the uncleavable CAHQ sequence were synthesized and tested as Rce1 inhibitors using in vitro and cell-based assays. Consistently, these non-hydrolyzable peptidomimetic Rce1 inhibitors recapitulate Ras mislocalization effects when modeled on cleavable but not uncleavable CaaX sequences. These findings indicate that a prenylated and uncleavable CaaX sequence, which can be easily applied to a wide range of mammalian CaaX proteins, can be used to probe the specific impact of CaaX proteolysis on CaaX protein properties under conditions of an otherwise normally processed CaaX protein proteome.


Subject(s)
ras Proteins , Humans , ras Proteins/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Proteolysis/drug effects , Molecular Structure , Peptidomimetics/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/chemical synthesis , Endopeptidases
3.
Nat Rev Cancer ; 24(5): 316-337, 2024 May.
Article in English | MEDLINE | ID: mdl-38627557

ABSTRACT

Although RAS was formerly considered undruggable, various agents that inhibit RAS or specific RAS oncoproteins have now been developed. Indeed, the importance of directly targeting RAS has recently been illustrated by the clinical success of mutant-selective KRAS inhibitors. Nevertheless, responses to these agents are typically incomplete and restricted to a subset of patients, highlighting the need to develop more effective treatments, which will likely require a combinatorial approach. Vertical strategies that target multiple nodes within the RAS pathway to achieve deeper suppression are being investigated and have precedence in other contexts. However, alternative strategies that co-target RAS and other therapeutic vulnerabilities have been identified, which may mitigate the requirement for profound pathway suppression. Regardless, the efficacy of any given approach will likely be dictated by genetic, epigenetic and tumour-specific variables. Here we discuss various combinatorial strategies to treat KRAS-driven cancers, highlighting mechanistic concepts that may extend to tumours harbouring other RAS mutations. Although many promising combinations have been identified, clinical responses will ultimately depend on whether a therapeutic window can be achieved and our ability to prospectively select responsive patients. Therefore, we must continue to develop and understand biologically diverse strategies to maximize our likelihood of success.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Mutation , ras Proteins/metabolism , ras Proteins/genetics , ras Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Signal Transduction , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology
4.
Cancer Cell ; 41(1): 88-105.e8, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36525973

ABSTRACT

Lung squamous cell carcinoma (LUSC) represents a major subtype of lung cancer with limited treatment options. KMT2D is one of the most frequently mutated genes in LUSC (>20%), and yet its role in LUSC oncogenesis remains unknown. Here, we identify KMT2D as a key regulator of LUSC tumorigenesis wherein Kmt2d deletion transforms lung basal cell organoids to LUSC. Kmt2d loss increases activation of receptor tyrosine kinases (RTKs), EGFR and ERBB2, partly through reprogramming the chromatin landscape to repress the expression of protein tyrosine phosphatases. These events provoke a robust elevation in the oncogenic RTK-RAS signaling. Combining SHP2 inhibitor SHP099 and pan-ERBB inhibitor afatinib inhibits lung tumor growth in Kmt2d-deficient LUSC murine models and in patient-derived xenografts (PDXs) harboring KMT2D mutations. Our study identifies KMT2D as a pivotal epigenetic modulator for LUSC oncogenesis and suggests that KMT2D loss renders LUSC therapeutically vulnerable to RTK-RAS inhibition.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Animals , Humans , Mice , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic , Lung/metabolism , Lung Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
5.
J Mol Biol ; 434(17): 167626, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35595166

ABSTRACT

Allosteric mechanisms are pervasive in nature, but human-designed allosteric perturbagens are rare. The history of KRASG12C inhibitor development suggests that covalent chemistry may be a key to expanding the armamentarium of allosteric inhibitors. In that effort, irreversible targeting of a cysteine converted a non-deal allosteric binding pocket and low affinity ligands into a tractable drugging strategy. Here we examine the feasibility of expanding this approach to other allosteric pockets of RAS and kinase family members, given that both protein families are regulators of vital cellular processes that are often dysregulated in cancer and other human diseases. Moreover, these heavily studied families are the subject of numerous drug development campaigns that have resulted, sometimes serendipitously, in the discovery of allosteric inhibitors. We consequently conducted a comprehensive search for cysteines, a commonly targeted amino acid for covalent drugs, using AlphaFold-generated structures of those families. This new analysis presents potential opportunities for allosteric targeting of validated and understudied drug targets, with an emphasis on cancer therapy.


Subject(s)
Antineoplastic Agents , Molecular Targeted Therapy , Neoplasms , Protein Kinase Inhibitors , Protein Kinases , ras Proteins , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cysteine/metabolism , Humans , Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry
6.
Adv Cancer Res ; 153: 131-168, 2022.
Article in English | MEDLINE | ID: mdl-35101229

ABSTRACT

Mutations in the three RAS oncogenes are present in approximately 30% of all human cancers that drive tumor growth and metastasis by aberrant activation of RAS-mediated signaling. Despite the well-established role of RAS in tumorigenesis, past efforts to develop small molecule inhibitors have failed for various reasons leading many to consider RAS as "undruggable." Advances over the past decade with KRAS(G12C) mutation-specific inhibitors have culminated in the first FDA-approved RAS drug, sotorasib. However, the patient population that stands to benefit from KRAS(G12C) inhibitors is inherently limited to those patients harboring KRAS(G12C) mutations. Additionally, both intrinsic and acquired mechanisms of resistance have been reported that indicate allele-specificity may afford disadvantages. For example, the compensatory activation of uninhibited wild-type (WT) NRAS and HRAS isozymes can rescue cancer cells harboring KRAS(G12C) mutations from allele-specific inhibition or the occurrence of other mutations in KRAS. It is therefore prudent to consider alternative drug discovery strategies that may overcome these potential limitations. One such approach is pan-RAS inhibition, whereby all RAS isozymes co-expressed in the tumor cell population are targeted by a single inhibitor to block constitutively activated RAS regardless of the underlying mutation. This chapter provides a review of past and ongoing strategies to develop pan-RAS inhibitors in detail and seeks to outline the trajectory of this promising strategy of RAS inhibition.


Subject(s)
Antineoplastic Agents , Neoplasms , ras Proteins , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Isoenzymes , Mutation , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
7.
Anal Cell Pathol (Amst) ; 2021: 5228713, 2021.
Article in English | MEDLINE | ID: mdl-34926131

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a malignant tumour originating from the mucosal lining of the oral cavity. Its characteristics include hidden onset, high recurrence, and distant metastasis after operation. At present, clinical treatment usually includes surgery, chemotherapy, radiotherapy, or the joint use of these modalities. Unfortunately, multidrug resistant is one of the important obstacles that causes cancer chemotherapy failure. Anlotinib, which has recently been proven to have good antitumour effects, is a novel multitargeted tyrosine kinase inhibitor. However, there are few studies of the anlotinib-associated mechanism in OSCC and its underlying molecular mechanism. In our study, in vitro models of human oral squamous cell carcinoma HSC-3 cells were used to determine the efficacy of anlotinib. On the one hand, we showed that anlotinib treatment significantly reduced the viability and proliferation of HSC-3 cells and decreased cell migration by inhibiting the activation of the Akt phosphorylation pathway. On the other side, anlotinib inhibited PI3K/Akt/Bad phosphorylation and promoted apoptosis of HSC-3 cells by activating RAS protein expression. In brief, these results indicated that anlotinib had prominent antitumour activity in OSCC, mainly by inhibiting the PI3K/Akt phosphorylation pathway. This work provides evidences and a basic principle for using anlotinib to treat patients with OSCC for clinical research.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Indoles/pharmacology , Mouth Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/pharmacology , Signal Transduction/drug effects , ras Proteins/antagonists & inhibitors , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Mouth Neoplasms/pathology , ras Proteins/metabolism
8.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830283

ABSTRACT

RAS (rat sarcoma virus) mutant cancers remain difficult to treat despite the advances in targeted therapy and immunotherapy. Targeted therapies against the components of mitogen-activated protein kinase (MAPK) pathways, including RAS, RAF, MEK, and ERK, have demonstrated activity in BRAF mutant and, in limited cases, RAS mutant cancer. RAS mutant cancers have been found to activate adaptive resistance mechanisms such as autophagy during MAPK inhibition. Here, we review the recent clinically relevant advances in the development of the MAPK pathway and autophagy inhibitors and focus on their application to RAS mutant cancers. We provide analysis of the preclinical rationale for combining the MAPK pathway and autophagy and highlight the most recent clinical trials that have been launched to capitalize on this potentially synthetic lethal approach to cancer therapy.


Subject(s)
Autophagy/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Therapy, Combination/methods , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Treatment Outcome , ras Proteins/metabolism
9.
Bioorg Med Chem Lett ; 53: 128414, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34666187

ABSTRACT

S-Palmitoylation is a reversible post-translational lipid modification that regulates protein trafficking and signaling. The enzymatic depalmitoylation of proteins is inhibited by the beta-lactones Palmostatin M and B, which have been found to target several serine hydrolases. In efforts to better understand the mechanism of action of Palmostatin M, we describe herein the synthesis, chemical proteomic analysis, and functional characterization of analogs of this compound. We identify Palmostatin M analogs that maintain inhibitory activity in N-Ras depalmitoylation assays while displaying complementary reactivity across the serine hydrolase class as measured by activity-based protein profiling. Active Palmostatin M analogs inhibit the recently characterized ABHD17 subfamily of depalmitoylating enzymes, while sparing other candidate depalmitoylases such as LYPLA1 and LYPLA2. These findings improve our understanding of the structure-activity relationship of Palmostatin M and refine the set of serine hydrolase targets relevant to the compound's effects on N-Ras palmitoylation dynamics.


Subject(s)
Lactones/analysis , Propiolactone/analogs & derivatives , Proteomics , Sulfones/analysis , ras Proteins/metabolism , Humans , Lactones/metabolism , Lactones/pharmacology , Molecular Structure , Propiolactone/analysis , Propiolactone/metabolism , Propiolactone/pharmacology , Sulfones/metabolism , Sulfones/pharmacology , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry
10.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34675073

ABSTRACT

Neutrophils sense and migrate through an enormous range of chemoattractant gradients through adaptation. Here, we reveal that in human neutrophils, calcium-promoted Ras inactivator (CAPRI) locally controls the GPCR-stimulated Ras adaptation. Human neutrophils lacking CAPRI (caprikd ) exhibit chemoattractant-induced, nonadaptive Ras activation; significantly increased phosphorylation of AKT, GSK-3α/3ß, and cofilin; and excessive actin polymerization. caprikd cells display defective chemotaxis in response to high-concentration gradients but exhibit improved chemotaxis in low- or subsensitive-concentration gradients of various chemoattractants, as a result of their enhanced sensitivity. Taken together, our data reveal that CAPRI controls GPCR activation-mediated Ras adaptation and lowers the sensitivity of human neutrophils so that they are able to chemotax through a higher-concentration range of chemoattractant gradients.


Subject(s)
Chemotaxis, Leukocyte/immunology , Neutrophils/immunology , ras GTPase-Activating Proteins/immunology , ras Proteins/antagonists & inhibitors , Actins/immunology , Cell Movement , Cell Polarity , Gene Knockdown Techniques , HL-60 Cells , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/drug effects , Neutrophils/physiology , Receptors, G-Protein-Coupled/immunology , Shelterin Complex/immunology , Signal Transduction , Telomere-Binding Proteins/immunology , ras GTPase-Activating Proteins/deficiency , ras GTPase-Activating Proteins/genetics , ras Proteins/immunology
11.
Cancer Sci ; 112(10): 4026-4036, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34382720

ABSTRACT

Disialoganglioside (GD2)-specific chimeric antigen receptor (CAR)-T cells (GD2-CAR-T cells) have been developed and tested in early clinical trials in patients with relapsed/refractory neuroblastoma. However, the effectiveness of immunotherapy using these cells is limited, and requires improvement. Combined therapy with CAR-T cells and molecular targeted drugs could be a promising strategy to enhance the antitumor efficacy of CAR T cell immunotherapy. Here, we generated GD2-CAR-T cells through piggyBac transposon (PB)-based gene transfer (PB-GD2-CAR-T cells), and analyzed the combined effect of these cells and a MEK inhibitor in vitro and in vivo on neuroblastoma. Trametinib, a MEK inhibitor, ameliorated the killing efficacy of PB-GD2-CAR-T cells in vitro, whereas a combined treatment of the two showed superior antitumor efficacy in a murine xenograft model compared to that of PB-GD2-CAR-T cell monotherapy, regardless of the mutation status of the MAPK pathway in tumor cells. The results presented here provide new insights into the feasibility of combined treatment with CAR-T cells and MEK inhibitors in patients with neuroblastoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Gangliosides/therapeutic use , Immunotherapy, Adoptive/methods , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neuroblastoma/therapy , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Animals , Cell Line, Tumor , Combined Modality Therapy/methods , Coumarins/therapeutic use , DNA Transposable Elements , Drug Resistance, Neoplasm , Female , Genetic Therapy/methods , Humans , Mice , Mice, SCID , Mutation , Neoplasm Recurrence, Local/therapy , Protein Kinase Inhibitors/therapeutic use , T-Lymphocytes , Xenograft Model Antitumor Assays , ras Proteins/antagonists & inhibitors
12.
Nat Commun ; 12(1): 4688, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344896

ABSTRACT

Internalization and intracellular trafficking of G protein-coupled receptors (GPCRs) play pivotal roles in cell responsiveness. Dysregulation in receptor trafficking can lead to aberrant signaling and cell behavior. Here, using an endosomal BRET-based assay in a high-throughput screen with the prototypical GPCR angiotensin II type 1 receptor (AT1R), we sought to identify receptor trafficking inhibitors from a library of ~115,000 small molecules. We identified a novel dual Ras and ARF6 inhibitor, which we named Rasarfin, that blocks agonist-mediated internalization of AT1R and other GPCRs. Rasarfin also potently inhibits agonist-induced ERK1/2 signaling by GPCRs, and MAPK and Akt signaling by EGFR, as well as prevents cancer cell proliferation. In silico modeling and in vitro studies reveal a unique binding modality of Rasarfin within the SOS-binding domain of Ras. Our findings unveil a class of dual small G protein inhibitors for receptor trafficking and signaling, useful for the inhibition of oncogenic cellular responses.


Subject(s)
ADP-Ribosylation Factors/antagonists & inhibitors , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , ras Proteins/antagonists & inhibitors , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Binding Sites , Bioluminescence Resonance Energy Transfer Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Molecular Dynamics Simulation , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , ras Proteins/chemistry , ras Proteins/metabolism
13.
J Med Chem ; 64(17): 13038-13053, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34415745

ABSTRACT

The Ras subfamily of small GTPases is mutated in ∼30% of human cancers and represents compelling yet challenging anticancer drug targets owing to their flat protein surface. We previously reported a bicyclic peptidyl inhibitor, cyclorasin B3, which binds selectively to Ras-GTP with modest affinity and blocks its interaction with downstream effector proteins in vitro but lacks cell permeability or biological activity. In this study, optimization of B3 yielded a potent pan-Ras inhibitor, cyclorasin B4-27, which binds selectively to the GTP-bound forms of wild-type and mutant Ras isoforms (KD = 21 nM for KRasG12V-GppNHp) and is highly cell-permeable and metabolically stable (serum t1/2 > 24 h). B4-27 inhibits Ras signaling in vitro and in vivo by blocking Ras from interacting with downstream effector proteins and induces apoptosis of Ras-mutant cancer cells. When administered systemically (i.v.), B4-27 suppressed tumor growth in two different mouse xenograft models at 1-5 mg/kg of daily doses.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Discovery , Humans , Male , Mice , Mice, Nude , Protein Isoforms , Xenograft Model Antitumor Assays
14.
Drugs ; 81(13): 1573-1579, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34357500

ABSTRACT

Sotorasib (LUMAKRAS™) is a RAS GTPase family inhibitor being developed by Amgen for the treatment of solid tumours with KRAS mutations, including non-small cell lung cancer (NSCLC) and colorectal cancer. In May 2021, sotorasib was granted accelerated approval by the US FDA for the treatment of adult patients with KRAS G12C-mutated locally advanced or metastatic NSCLC, as determined by an FDA-approved test, who have received at least one prior systemic therapy. This article summarizes the milestones in the development of sotorasib leading to this first approval for KRAS G12C-mutated NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Piperazines/pharmacology , Piperazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials as Topic , Drug Approval , Humans , Lung Neoplasms/pathology , Piperazines/adverse effects , Piperazines/pharmacokinetics , Pyridines/adverse effects , Pyridines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , United States , United States Food and Drug Administration , ras Proteins/antagonists & inhibitors
15.
Pharmacol Res ; 172: 105806, 2021 10.
Article in English | MEDLINE | ID: mdl-34450320

ABSTRACT

RAS proteins (HRAS, KRAS, NRAS) participate in many physiological signal transduction processes related to cell growth, division, and survival. The RAS proteins are small (188/189 amino acid residues) and they function as GTPases. These proteins toggle between inactive and functional forms; the conversion of inactive RAS-GDP to active RAS-GTP as mediated by guanine nucleotide exchange factors (GEFs) turns the switch on and the intrinsic RAS-GTPase activity stimulated by the GTPase activating proteins (GAPs) turns the switch off. RAS is upstream to the RAS-RAF-MEK-ERK and the PI3-kinase-AKT signaling modules. Importantly, the overall incidence of RAS mutations in all cancers is about 19% and RAS mutants have been a pharmacological target for more than three decades. About 84% of all RAS mutations involve KRAS. Except for the GTP/GDP binding site, the RAS proteins lack other deep surface pockets thereby hindering efforts to identify high-affinity antagonists; thus, they have been considered to be undruggable. KRAS mutations frequently occur in lung, colorectal, and pancreatic cancers, the three most deadly cancers in the United States. Studies within the last decade demonstrated that the covalent modification of KRAS C12, which accounts for about 10% of all RAS mutations, led to the discovery of an adjacent pocket (called the switch II pocket) that accommodated a portion of the drug. This led to the development of sotorasib as a second-line treatment of KRASG12C-mutant non-small cell lung cancer. Considerable effort also has been expended to develop MAP kinase and PI3-kinase pathway inhibitors as indirect RAS antagonists.


Subject(s)
ras Proteins , Animals , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , ras Proteins/antagonists & inhibitors , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism
16.
Nat Commun ; 12(1): 4045, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34193876

ABSTRACT

RAS mutations are the most common oncogenic drivers across human cancers, but there remains a paucity of clinically-validated pharmacological inhibitors of RAS, as druggable pockets have proven difficult to identify. Here, we identify two RAS-binding Affimer proteins, K3 and K6, that inhibit nucleotide exchange and downstream signaling pathways with distinct isoform and mutant profiles. Affimer K6 binds in the SI/SII pocket, whilst Affimer K3 is a non-covalent inhibitor of the SII region that reveals a conformer of wild-type RAS with a large, druggable SII/α3 pocket. Competitive NanoBRET between the RAS-binding Affimers and known RAS binding small-molecules demonstrates the potential to use Affimers as tools to identify pharmacophores. This work highlights the potential of using biologics with small interface surfaces to select unseen, druggable conformations in conjunction with pharmacophore identification for hard-to-drug proteins.


Subject(s)
Biological Products/pharmacology , Cell Surface Display Techniques/methods , Drug Discovery/methods , Neoplasms/drug therapy , ras Proteins/antagonists & inhibitors , Allosteric Site , Biological Products/chemistry , Humans , Neoplasms/chemistry , Neoplasms/enzymology , Signal Transduction , ras Proteins/metabolism
17.
Genes (Basel) ; 12(6)2021 06 10.
Article in English | MEDLINE | ID: mdl-34200676

ABSTRACT

Around 20% of all malignancies harbour activating mutations in RAS isoforms. Despite this, there is a deficiency of RAS-targeting agents licensed for therapeutic use. The picomolar affinity of RAS for GTP, and the lack of suitable pockets for high-affinity small-molecule binding, precluded effective therapies despite decades of research. Recently, characterisation of the biochemical properties of KRAS-G12C along with discovery of its 'switch-II pocket' have allowed development of effective mutant-specific inhibitors. Currently seven KRAS-G12C inhibitors are in clinical trials and sotorasib has become the first one to be granted FDA approval. Here, we discuss historical efforts to target RAS directly and approaches to target RAS effector signalling, including combinations that overcome limitations of single-agent targeting. We also review pre-clinical and clinical evidence for the efficacy of KRAS-G12C inhibitor monotherapy followed by an illustration of combination therapies designed to overcome primary resistance and extend durability of response. Finally, we briefly discuss novel approaches to targeting non-G12C mutant isoforms.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Humans , Mutation , Neoplasms/genetics , ras Proteins/genetics , ras Proteins/metabolism
18.
Sci Rep ; 11(1): 12590, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131238

ABSTRACT

Colorectal cancer (CRC) is one of the most well-known malignancies with high prevalence and poor 5-year survival. Previous studies have demonstrated that a high-fat diet (HFD) is capable of increasing the odds of developing CRC. Acrolein, an IARC group 2A carcinogen, can be formed from carbohydrates, vegetable oils, animal fats, and amino acids through the Maillard reaction during the preparation of foods. Consequently, humans are at risk of acrolein exposure through the consumption of foods rich in fat. However, whether acrolein contributes to HFD-induced CRC has not been determined. In this study, we found that acrolein induced oncogenic transformation, including faster cell cycling, proliferation, soft agar formation, sphere formation and cell migration, in NIH/3T3 cells. Using xenograft tumorigenicity assays, the acrolein-transformed NIH/3T3 clone formed tumors. In addition, cDNA microarray and bioinformatics studies by Ingenuity Pathway Analysis pointed to the fact that RAS/MAPK pathway was activated in acrolein-transformed clones that contributed to colon tumorigenesis. Furthermore, acrolein-induced DNA damages (Acr-dG adducts) were higher in CRC tumor tissues than in normal epithelial cells in CRC patients. Notably, CRC patients with higher levels of Acr-dG adducts appeared to have better prognosis. The results of this study demonstrate for the first time that acrolein is important in oncogenic transformation through activation of the RAS/MAPK signaling pathway, contributing to colon tumorigenesis.


Subject(s)
Acrolein/toxicity , Carcinogenesis/drug effects , Carcinogens/toxicity , Colorectal Neoplasms/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , DNA Adducts/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Maillard Reaction , Mice , NIH 3T3 Cells , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
19.
J Nanobiotechnology ; 19(1): 177, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-34118951

ABSTRACT

BACKGROUND: Ras activation is a frequent event in hepatocellular carcinoma (HCC). Combining a RAS inhibitor with traditional clinical therapeutics might be hampered by a variety of side effects, thus hindering further clinical translation. Herein, we report on integrating an IR820 nanocapsule-augmented sonodynamic therapy (SDT) with the RAS inhibitor farnesyl-thiosalicylic acid (FTS). Using cellular and tumor models, we demonstrate that combined nanocapsule-augmented SDT with FTS induces an anti-tumor effect, which not only inhibits tumor progression, and enables fluorescence imaging. To dissect the mechanism of a combined tumoricidal therapeutic strategy, we investigated the scRNA-seq transcriptional profiles of an HCC xenograft following treatment. RESULTS: Integrative single-cell analysis identified several clusters that defined many corresponding differentially expressed genes, which provided a global view of cellular heterogeneity in HCC after combined SDT/FTS treatment. We conclude that the combination treatment suppressed HCC, and did so by inhibiting endothelial cells and a modulated immunity. Moreover, hepatic stellate secretes hepatocyte growth factor, which plays a key role in treating SDT combined FTS. By contrast, enrichment analysis estimated the functional roles of differentially expressed genes. The Gene Ontology terms "cadherin binding" and "cell adhesion molecule binding" and KEGG pathway "pathway in cancer" were significantly enriched by differentially expressed genes after combined SDT/FTS therapy. CONCLUSIONS: Thus, some undefined mechanisms were revealed by scRNA-seq analysis. This report provides a novel proof-of-concept for combinatorial HCC-targeted therapeutics that is based on a non-invasive anti-tumor therapeutic strategy and a RAS inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Diathermy/methods , Liver Neoplasms/drug therapy , Sequence Analysis, RNA , ras Proteins/antagonists & inhibitors , Animals , Carcinoma, Hepatocellular/radiotherapy , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Endothelial Cells , Farnesol/analogs & derivatives , Farnesol/pharmacology , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/radiotherapy , Mice, Inbred BALB C , Mice, Nude , Salicylates
20.
Mol Cancer Ther ; 20(9): 1743-1754, 2021 09.
Article in English | MEDLINE | ID: mdl-34158349

ABSTRACT

Activating mutations in RAS are found in approximately 30% of human cancers, resulting in the delivery of a persistent signal to critical downstream effectors that drive tumorigenesis. RAS-driven malignancies respond poorly to conventional cancer treatments and inhibitors that target RAS directly are limited; therefore, the identification of new strategies and/or drugs to disrupt RAS signaling in tumor cells remains a pressing therapeutic need. Taking advantage of the live-cell bioluminescence resonance energy transfer (BRET) methodology, we describe the development of a NanoBRET screening platform to identify compounds that modulate binding between activated KRAS and the CRAF kinase, an essential effector of RAS that initiates ERK cascade signaling. Using this strategy, libraries containing synthetic compounds, targeted inhibitors, purified natural products, and natural product extracts were evaluated. These efforts resulted in the identification of compounds that inhibit RAS/RAF binding and in turn suppress RAS-driven ERK activation, but also compounds that have the deleterious effect of enhancing the interaction to upregulate pathway signaling. Among the inhibitor hits identified, the majority were compounds derived from natural products, including ones reported to alter KRAS nanoclustering (ophiobolin A), to impact RAF function (HSP90 inhibitors and ROS inducers) as well as some with unknown targets and activities. These findings demonstrate the potential for this screening platform in natural product drug discovery and in the development of new therapeutic agents to target dysregulated RAS signaling in human disease states such as cancer.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Fibroblasts/drug effects , High-Throughput Screening Assays/methods , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , ras Proteins/agonists , ras Proteins/antagonists & inhibitors , Animals , Fibroblasts/metabolism , Humans , Ligands , Nanotechnology/methods , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...