Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
J Med Chem ; 67(9): 7130-7145, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38630077

ABSTRACT

Multitarget medications represent an appealing therapy against the disease with multifactorial abnormalities─cancer. Therefore, simultaneously targeting son of sevenless 1 (SOS1) and epidermal growth factor receptor (EGFR), two aberrantly expressed proteins crucial for the oncogenesis and progression of prostate cancer, may achieve active antitumor effects. Here, we discovered dual SOS1/EGFR-targeting compounds via pharmacophore-based docking screening. The most prominent compound SE-9 exhibited nanomolar inhibition activity against both SOS1 and EGFR and efficiently suppressed the phosphorylation of ERK and AKT in prostate cancer cells PC-3. Cellular assays also revealed that SE-9 displayed strong antiproliferative activities through diverse mechanisms, such as induction of cell apoptosis and G1 phase cell cycle arrest, as well as reduction of angiogenesis and migration. Further in vivo findings showed that SE-9 potently inhibited tumor growth in PC-3 xenografts without obvious toxicity. Overall, SE-9 is a novel dual-targeting SOS1/EGFR inhibitor that represents a promising treatment strategy for prostate cancer.


Subject(s)
Antineoplastic Agents , Cell Proliferation , ErbB Receptors , Prostatic Neoplasms , SOS1 Protein , Male , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , SOS1 Protein/antagonists & inhibitors , SOS1 Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Cell Line, Tumor , Mice , Apoptosis/drug effects , Drug Discovery , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/therapeutic use , Mice, Nude , Structure-Activity Relationship , Xenograft Model Antitumor Assays , Mice, Inbred BALB C
2.
J Med Chem ; 67(1): 774-781, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38156904

ABSTRACT

SOS1 and SOS2 are guanine nucleotide exchange factors that mediate RTK-stimulated RAS activation. Selective SOS1:KRAS PPI inhibitors are currently under clinical investigation, whereas there are no reports to date of SOS2:KRAS PPI inhibitors. SOS2 activity is implicated in MAPK rebound when divergent SOS1 mutant cell lines are treated with the SOS1 inhibitor BI-3406; therefore, SOS2:KRAS inhibitors are of therapeutic interest. In this report, we detail a fragment-based screening strategy to identify X-ray cocrystal structures of five diverse fragment hits bound to SOS2.


Subject(s)
Furans , Guanine Nucleotide Exchange Factors , Proto-Oncogene Proteins p21(ras) , Quinazolines , X-Rays , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Cell Line , SOS1 Protein/metabolism
3.
Eur J Med Chem ; 261: 115828, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37778239

ABSTRACT

Kristen rat sarcoma (KRAS) is one of the most common oncogenes in human cancers. As a guanine nucleotide exchange factor, Son of Sevenless Homologue 1 (SOS1) represents a potential therapeutic concept for the treatment of KRAS-mutant cancers because of its activation on KRAS and downstream signaling pathways. In this review, we provide a comprehensive overview of the structure, biological function, and regulation of SOS1. We also focus on the recent advances in SOS1 inhibitors and emphasize their binding modes, structure-activity relationships and pharmacological activities. We hope that this publication can provide a comprehensive compendium on the rational design of SOS1 inhibitors.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/metabolism , Nuclear Family , Signal Transduction
4.
Bioorg Chem ; 136: 106536, 2023 07.
Article in English | MEDLINE | ID: mdl-37054529

ABSTRACT

KRAS mutations (G12C, G12D, etc.) are implicated in the oncogenesis and progression of many deadliest cancers. Son of sevenless homolog 1 (SOS1) is a crucial regulator of KRAS to modulate KRAS from inactive to active states. We previously discovered tetra-cyclic quinazolines as an improved scaffold for inhibiting SOS1-KRAS interaction. In this work, we report the design of tetra-cyclic phthalazine derivatives for selectively inhibiting SOS1 against EGFR. The lead compound 6c displayed remarkable activity to inhibit the proliferation of KRAS(G12C)-mutant pancreas cells. 6c showed a favorable pharmacokinetic profile in vivo, with a bioavailability of 65.8% and exhibited potent tumor suppression in pancreas tumor xenograft models. These intriguing results suggested that 6c has the potential to be developed as a drug candidate for KRAS-driven tumors.


Subject(s)
Proto-Oncogene Proteins p21(ras) , SOS1 Protein , Humans , SOS1 Protein/genetics , SOS1 Protein/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Mutation , Quinazolines/pharmacology , ErbB Receptors/genetics
5.
Bioorg Chem ; 135: 106500, 2023 06.
Article in English | MEDLINE | ID: mdl-37003134

ABSTRACT

Blocking the interaction between Ras and Son of Sevenless homolog 1 (SOS1) has been an attractive therapeutic strategy for treating cancers involving oncogenic Ras mutations. K-Ras mutation is the most common in Ras-driven cancers, accounting for 86%, with N-Ras mutation and H-Ras mutation accounting for 11% and 3%, respectively. Here, we report the design and synthesis of a series of hydrocarbon-stapled peptides to mimic the alpha-helix of SOS1 as pan-Ras inhibitors. Among these stapled peptides, SSOSH-5 was identified to maintain a well-constrained alpha-helical structure and bind to H-Ras with high affinity. SSOSH-5 was furthermore validated to bind with Ras similarly to the parent linear peptide through structural modeling analysis. This optimized stapled peptide was proven to be capable of effectively inhibiting the proliferation of pan-Ras-mutated cancer cells and inducing apoptosis in a dose-dependent manner by modulating downstream kinase signaling. Of note, SSOSH-5 exhibited a high capability of crossing cell membranes and strong proteolytic resistance. We demonstrated that the peptide stapling strategy is a feasible approach for developing peptide-based pan-Ras inhibitors. Furthermore, we expect that SSOSH-5 can be further characterized and optimized for the treatment of Ras-driven cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , SOS1 Protein/chemistry , SOS1 Protein/genetics , SOS1 Protein/metabolism , Peptides/pharmacology , Signal Transduction , Mutation , Antineoplastic Agents/pharmacology
6.
Curr Pharm Des ; 29(22): 1741-1746, 2023.
Article in English | MEDLINE | ID: mdl-37073657

ABSTRACT

The most frequent mutated oncogene KRAS in lung cancer is targeted by KRAS G12C-directed drugs, such as Sotorasib and Adagrasib. Still, other alleles frequently expressed in pancreatic and colon cancer may be attacked indirectly by hitting the guanine nucleotide exchange factor (GEF) SOS1 that loads and activates KRAS. The first modulators of SOS1 were found to act as agonists and defined a hydrophobic pocket at the catalytic site. High throughput screenings resulted in the detection of SOS1 inhibitors Bay-293 and BI-3406 comprising amino quinazoline scaffolds optimized for binding to the pocket by various substituents. The first inhibitor, BI-1701963, is in clinical studies alone or in combination with a KRAS inhibitor, a MAPK inhibitor or chemotherapeutics. An optimized agonist, VUBI-1, shows activity against tumor cells by destructive overactivation of cellular signaling. This agonist was used to formulate a proteolysis targeting chimera (PROTAC), that labels SOS1 for degradation by proteasomal degradation through a linked VHL E3 ligase ligand. This PROTAC exhibited the highest SOS1-directed activity due to target destruction, recycling and removal of SOS1 as a scaffolding protein. Although other first PROTACs have entered clinical trials, each conjugate must be meticulously adapted as an efficient clinical drug.


Subject(s)
Proteolysis Targeting Chimera , Proto-Oncogene Proteins p21(ras) , SOS1 Protein , Humans , High-Throughput Screening Assays , Proteolysis , Ubiquitin-Protein Ligases/metabolism , SOS1 Protein/agonists , SOS1 Protein/metabolism
7.
Mol Carcinog ; 62(7): 1025-1037, 2023 07.
Article in English | MEDLINE | ID: mdl-37042566

ABSTRACT

It has been challenging to target mutant KRAS (mKRAS) in colorectal cancer (CRC) and other malignancies. Recent efforts have focused on developing inhibitors blocking molecules essential for KRAS activity. In this regard, SOS1 inhibition has arisen as an attractive approach for mKRAS CRC given its essential role as a guanine nucleotide exchange factor for this GTPase. Here, we demonstrated the translational value of SOS1 blockade in mKRAS CRC. We used CRC patient-derived organoids (PDOs) as preclinical models to evaluate their sensitivity to SOS1 inhibitor BI3406. A combination of in silico analyses and wet lab techniques was utilized to define potential predictive markers for SOS1 sensitivity and potential mechanisms of resistance in CRC. RNA-seq analysis of CRC PDOs revealed two groups of CRC PDOs with differential sensitivities to SOS1 inhibitor BI3406. The resistant group was enriched in gene sets involving cholesterol homeostasis, epithelial-mesenchymal transition, and TNF-α/NFκB signaling. Expression analysis identified a significant correlation between SOS1 and SOS2 mRNA levels (Spearman's ρ 0.56, p < 0.001). SOS1/2 protein expression was universally present with heterogeneous patterns in CRC cells but only minimal to none in surrounding nonmalignant cells. Only SOS1 protein expression was associated with worse survival in patients with RAS/RAF mutant CRC (p = 0.04). We also found that SOS1/SOS2 protein expression ratio >1 by immunohistochemistry (p = 0.03) instead of KRAS mutation (p = 1) was a better predictive marker to BI3406 sensitivity of CRC PDOs, concordant with the significant positive correlation between SOS1/SOS2 protein expression ratio and SOS1 dependency. Finally, we showed that GTP-bound RAS level underwent rebound even in BI3406-sensitive PDOs with no change of KRAS downstream effector genes, thus suggesting upregulation of guanine nucleotide exchange factor as potential cellular adaptation mechanisms to SOS1 inhibition. Taken together, our results show that high SOS1/SOS2 protein expression ratio predicts sensitivity to SOS1 inhibition and support further clinical development of SOS1-targeting agents in CRC.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , SOS1 Protein/genetics , SOS1 Protein/metabolism , Guanine Nucleotide Exchange Factors/genetics , Mutation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics
8.
J Med Chem ; 66(7): 4324-4341, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36987571

ABSTRACT

Son of sevenless homologue 1 (SOS1) protein is universally expressed in cells and plays an important role in the RAS signaling pathway. Specifically, this protein interacts with RAS in response to upstream stimuli to promote guanine nucleotide exchange in RAS and activates the downstream signaling pathways. Thus, targeting SOS1 is a new approach for treating RAS-driven cancers. In this Perspective, we briefly summarize the structural and functional aspects of SOS1 and focus on recent advances in the discovery of activators, inhibitors, and PROTACs that target SOS1. This review aims to provide a timely and updated overview on the strategies for targeting SOS1 in cancer therapy.


Subject(s)
Neoplasms , Nuclear Family , Humans , SOS1 Protein/metabolism , Signal Transduction , Neoplasms/drug therapy
9.
J Pharmacol Exp Ther ; 385(2): 106-116, 2023 05.
Article in English | MEDLINE | ID: mdl-36849412

ABSTRACT

Individuals with neurofibromatosis type 1 develop rat sarcoma virus (RAS)-mitogen-activated protein kinase-mitogen-activated and extracellular signal-regulated kinase (RAS-MAPK-MEK)-driven nerve tumors called neurofibromas. Although MEK inhibitors transiently reduce volumes of most plexiform neurofibromas in mouse models and in neurofibromatosis type 1 (NF1) patients, therapies that increase the efficacy of MEK inhibitors are needed. BI-3406 is a small molecule that prevents Son of Sevenless (SOS)1 interaction with Kirsten rat sarcoma viral oncoprotein (KRAS)-GDP, interfering with the RAS-MAPK cascade upstream of MEK. Single agent SOS1 inhibition had no significant effect in the DhhCre;Nf1 fl/fl mouse model of plexiform neurofibroma, but pharmacokinetics (PK)-driven combination of selumetinib with BI-3406 significantly improved tumor parameters. Tumor volumes and neurofibroma cell proliferation, reduced by MEK inhibition, were further reduced by the combination. Neurofibromas are rich in ionized calcium binding adaptor molecule 1 (Iba1)+ macrophages; combination treatment resulted in small and round macrophages, with altered cytokine expression indicative of altered activation. The significant effects of MEK inhibitor plus SOS1 inhibition in this preclinical study suggest potential clinical benefit of dual targeting of the RAS-MAPK pathway in neurofibromas. SIGNIFICANCE STATEMENT: Interfering with the RAS-mitogen-activated protein kinase (RAS-MAPK) cascade upstream of mitogen activated protein kinase kinase (MEK), together with MEK inhibition, augment effects of MEK inhibition on neurofibroma volume and tumor macrophages in a preclinical model system. This study emphasizes the critical role of the RAS-MAPK pathway in controlling tumor cell proliferation and the tumor microenvironment in benign neurofibromas.


Subject(s)
Neurofibroma, Plexiform , Neurofibroma , Neurofibromatosis 1 , Animals , Mice , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases , Neurofibroma/drug therapy , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/therapeutic use , Tumor Microenvironment , SOS1 Protein/metabolism
10.
J Med Chem ; 65(19): 13158-13171, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36173339

ABSTRACT

The interaction between son of sevenless 1 (SOS1) gene and Kirsten rat sarcoma viral oncogene (KRAS) is crucial for activating signals of proliferation and survival in a range of cancers. We previously discovered compound 40a with a tetracyclic quinazoline pharmacophore as a potent orally bioavailable SOS1 inhibitor. Herein, we disclosed the discovery of compound 13c, which substituted the third ring with the seven-membered ring, as a clinical drug candidate for suppressing KRAS-driven tumors. 13c strongly disrupted the protein-protein interaction between SOS1 and KRAS with low IC50 values of 3.9 nM (biochemical) and 21 nM (cellular). 13c showed a favorable pharmacokinetic profile with a bioavailability of 86.8% in beagles and exhibited 83.0% tumor suppression in Mia-paca-2 pancreas xenograft mice tumor models. 13c exhibited a weak time-dependent CY3A4P inhibition than BI-3406, thereby reducing the risk of drug-drug interaction in drug combination. Toxicological investigations revealed that 13c had a lower risk of sudden cardiac death than BI-3406. Overall, 13c has been under evaluation in preclinical trials.


Subject(s)
Carcinoma , Pancreatic Neoplasms , Animals , Dogs , Humans , Mice , Carbon Isotopes , Nuclear Family , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/genetics , Quinazolines/therapeutic use , SOS1 Protein/metabolism
11.
J Med Chem ; 65(14): 9678-9690, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35833726

ABSTRACT

SOS1 is one of the major guanine nucleotide exchange factors that regulates the ability of KRAS to cycle through its "on" and "off" states. Disrupting the SOS1:KRASG12C protein-protein interaction (PPI) can increase the proportion of GDP-loaded KRASG12C, providing a strong mechanistic rationale for combining inhibitors of the SOS1:KRAS complex with inhibitors like MRTX849 that target GDP-loaded KRASG12C. In this report, we detail the design and discovery of MRTX0902─a potent, selective, brain-penetrant, and orally bioavailable SOS1 binder that disrupts the SOS1:KRASG12C PPI. Oral administration of MRTX0902 in combination with MRTX849 results in a significant increase in antitumor activity relative to that of either single agent, including tumor regressions in a subset of animals in the MIA PaCa-2 tumor mouse xenograft model.


Subject(s)
Brain , Proto-Oncogene Proteins p21(ras) , Acetonitriles , Animals , Cell Line, Tumor , Humans , Mice , Mutation , Piperazines , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines , SOS1 Protein/metabolism
12.
Adv Cancer Res ; 153: 169-203, 2022.
Article in English | MEDLINE | ID: mdl-35101230

ABSTRACT

RAS proteins play major roles in many human cancers, but programs to develop direct RAS inhibitors so far have only been successful for the oncogenic KRAS mutant G12C. As an alternative approach, inhibitors for the RAS guanine nucleotide exchange factor SOS1 have been investigated by several academic groups and companies, and major progress has been achieved in recent years in the optimization of small molecule activators and inhibitors of SOS1. Here, we review the discovery and development of small molecule modulators of SOS1 and their molecular binding modes and modes of action. As targeting the RAS pathway is expected to result in the development of resistance mechanisms, SOS1 inhibitors will most likely be best applied in vertical combination approaches where two nodes of the RAS signaling pathway are hit simultaneously. We summarize the current understanding of which combination partners may be most beneficial for patients with RAS driven tumors.


Subject(s)
Neoplasms , SOS1 Protein , ras Proteins , Carcinogenesis , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oncogenes , SOS1 Protein/antagonists & inhibitors , SOS1 Protein/chemistry , SOS1 Protein/metabolism , ras Proteins/genetics , ras Proteins/metabolism
13.
J Mol Histol ; 52(5): 991-1006, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34387789

ABSTRACT

Emerging evidence reveals that long noncoding RNAs (lncRNAs) contribute to human tumorigenesis. Nevertheless, the function of HOXC cluster antisense RNA 3 (HOXC-AS3) in human cervical cancer (CC) remains largely unknown. The levels of HOXC-AS3, miR-105-5p and SOS1 in CC tissues and cells were monitored by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Gain- and loss-of-function experiments were conducted to verify the function of HOXC-AS3 and miR-105-5p in CC cells. Meanwhile, cell proliferation, apoptosis, migration and invasion were examined by the cell counting kit-8 (CCK8) experiment, colony formation assay, flow cytometry and Transwell assay. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were carried out to test the regulatory interaction of HOXC-AS3, miR-105-5p and SOS1. In addition, in vivo experiment was performed to certain the role of HOXC-AS3 in tumorigenesis of CC. HOXC-AS3 was overexpressed in CC tissues (vs. adjacent normal tissues) and CC cells. Besides, the higher HOXC-AS3 profile was associated with the poorer clinical prognosis of CC patients. Overexpression of HOXC-AS3 promoted cell growth, migration and invasion, hampered apoptosis, whereas knocking down HOXC-AS3 exhibited the reverse effects. MiR-105-5p was a downstream target of HOXC-AS3, and it mediated the HOXC-AS3-induced oncogenic effects. Mechanistically, the bioinformatic analysis illustrated that SOS1 was targeted by miR-105-5p. Up-regulating SOS1 heightened the growth, migration and invasion of CC cells by enhancing the ErbB signaling pathway, which was reversed by miR-105-5p. Up-regulated HOXC-AS3 aggravates CC by promoting SOS1 expression via targeting miR-105-5p.


Subject(s)
Disease Progression , ErbB Receptors/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Animals , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Neoplasm Metastasis , RNA, Long Noncoding/genetics , SOS1 Protein/metabolism
14.
Biomolecules ; 11(8)2021 07 30.
Article in English | MEDLINE | ID: mdl-34439794

ABSTRACT

Recent breakthroughs have reignited interest in RAS GEFs as direct therapeutic targets. To search for new inhibitors of SOS GEF activity, a repository of known/approved compounds (NIH-NACTS) and a library of new marine compounds (Biomar Microbial Technologies) were screened by means of in vitro RAS-GEF assays using purified, bacterially expressed SOS and RAS constructs. Interestingly, all inhibitors identified in our screenings (two per library) shared related chemical structures belonging to the anthraquinone family of compounds. All our anthraquinone SOS inhibitors were active against the three canonical RAS isoforms when tested in our SOS GEF assays, inhibited RAS activation in mouse embryonic fibroblasts, and were also able to inhibit the growth of different cancer cell lines harboring WT or mutant RAS genes. In contrast to the commercially available anthraquinone inhibitors, our new marine anthraquinone inhibitors did not show in vivo cardiotoxicity, thus providing a lead for future discovery of stronger, clinically useful anthraquinone SOS GEF blockers.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , GTP Phosphohydrolases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Cardiotoxicity/prevention & control , Cell Line, Transformed , Cell Line, Tumor , Doxorubicin/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Idarubicin/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/genetics , SOS1 Protein/metabolism , Son of Sevenless Proteins/deficiency , Son of Sevenless Proteins/genetics
15.
Int J Med Sci ; 18(14): 3097-3105, 2021.
Article in English | MEDLINE | ID: mdl-34400880

ABSTRACT

Emerging evidence revealed that UHRF2 was implicated in a variety of human diseases, especially in cancer. However, the biological function, clinical significance and underly mechanisms of UHRF2 in hepatocellular carcinoma (HCC) is largely unknown. We analyzed the expression of UHRF2 in 371 HCC tissues and 50 para-cancerous tissues of TCGA database. We found that UHRF2 was significantly upregulated in HCC tissues, which was further confirmed in HCC cells and tissues by western blot. More importantly, the level of UHRF2 was correlated with pathological grade and clinical stage, and the patients with high level of UHRF2 had lower overall survival, disease-free survival and higher recurrence rate than those with low UHRF2 level. Univariate and multivariate Cox regression analysis revealed that high level of UHRF2 might be an independent prognostic factor for HCC patients. Functional investigations suggested that ectopic expression of UHRF2 could promote the proliferation, migration and invasion of HCC cell lines, whereas knock down of UHRF2 exhibited an opposite effect. Additionally, gene set enrichment analysis indicated that ERBB signaling pathway was upregulated in patients with high level of UHRF2. Pearson correlation analysis indicated that the expression of UHRF2 was positively correlated with ErbB3 and its downstream targets SOS1, Ras and Raf-1. Furthermore, we found that overexpression of UHRF2 could upregulate the expression of ErbB3, SOS1, Ras and Raf-1. Our findings suggested that UHRF2 might accelerate HCC progression by upregulating ErbB3/Ras/Raf signaling pathway and it might serve as a diagnostic marker and therapeutic target for HCC patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Receptor, ErbB-3/genetics , Ubiquitin-Protein Ligases/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hep G2 Cells , Hepatectomy , Humans , Liver/pathology , Liver/surgery , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Proto-Oncogene Proteins c-raf/metabolism , Receptor, ErbB-3/metabolism , SOS1 Protein/metabolism , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Up-Regulation , ras Proteins/metabolism
16.
Expert Opin Ther Pat ; 31(12): 1189-1204, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34253125

ABSTRACT

Introduction: Up to 30% of all human cancers are driven by the overactivation of RAS signaling. Son of Sevenless 1 (SOS1) is a central node in RAS signaling pathways and modulation of SOS1-mediated RAS activation represents a unique opportunity for treating RAS-addicted cancers. Several recent publications and patent documents have demonstrated the ability of small molecules to affect the activation of RAS by SOS1 and have shown their potential for the treatment of cancers driven by RAS mutants.Areas covered: Documents focusing on both small-molecule inhibitors and activators of the SOS1:RAS interaction and their potential use as cancer therapeutics are covered. A total of 10 documents from 4 applicants are evaluated with discussion focusing on structural modifications of these compounds as well as relevant preclinical data.Expert opinion: The last decade has seen a significant increase in research and disclosures in the development of small-molecule SOS1 inhibitors. Considering the promising data that have been disclosed, interest in this area of research will likely remain strong for the foreseeable future. With the first SOS1 inhibitor currently in phase I clinical trials, the outcome of these trials will likely influence future development of SOS1 inhibitors for treatment of RAS-driven cancers.


Subject(s)
Neoplasms/drug therapy , SOS1 Protein/antagonists & inhibitors , ras Proteins/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Development , Humans , Mutation , Neoplasms/genetics , Patents as Topic , SOS1 Protein/metabolism , Signal Transduction
17.
Biochem J ; 478(14): 2793-2809, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34232285

ABSTRACT

Growth factor receptor-bound protein 2 (GRB2) is a trivalent adaptor protein and a key element in signal transduction. It interacts via its flanking nSH3 and cSH3 domains with the proline-rich domain (PRD) of the RAS activator SOS1 and via its central SH2 domain with phosphorylated tyrosine residues of receptor tyrosine kinases (RTKs; e.g. HER2). The elucidation of structural organization and mechanistic insights into GRB2 interactions, however, remain challenging due to their inherent flexibility. This study represents an important advance in our mechanistic understanding of how GRB2 links RTKs to SOS1. Accordingly, it can be proposed that (1) HER2 pYP-bound SH2 potentiates GRB2 SH3 domain interactions with SOS1 (an allosteric mechanism); (2) the SH2 domain blocks cSH3, enabling nSH3 to bind SOS1 first before cSH3 follows (an avidity-based mechanism); and (3) the allosteric behavior of cSH3 to other domains appears to be unidirectional, although there is an allosteric effect between the SH2 and SH3 domains.


Subject(s)
GRB2 Adaptor Protein/chemistry , Phosphotyrosine/chemistry , Protein Domains , SOS1 Protein/chemistry , src Homology Domains , Amino Acid Sequence , Binding Sites/genetics , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Kinetics , Ligands , Models, Molecular , Phosphotyrosine/metabolism , Protein Binding , SOS1 Protein/genetics , SOS1 Protein/metabolism
19.
Curr Opin Chem Biol ; 62: 109-118, 2021 06.
Article in English | MEDLINE | ID: mdl-33848766

ABSTRACT

Son of Sevenless (SOS) is a guanine nucleotide exchange factor that activates the important cell signaling switch KRAS. SOS acts as a pacemaker for KRAS, the beating heart of cancer, by catalyzing the "beating" from the KRAS(off) to the KRAS(on) conformation. Activating mutations in SOS1 are common in Noonan syndrome and oncogenic alterations in KRAS drive 1 in seven human cancers. Promising clinical efficacy has been observed for selective KRASG12C inhibitors, but the vast majority of oncogenic KRAS alterations remain undrugged. The discovery of a druggable pocket on SOS1 has led to potent SOS1 inhibitors such as BI-3406. SOS1 inhibition leads to antiproliferative effects against all major KRAS mutants. The first SOS1 inhibitor has entered clinical trials for KRAS-mutated cancers. In this review, we provide an overview of SOS1 function, its association with cancer and RASopathies, known SOS1 activators and inhibitors, and a future perspective is provided.


Subject(s)
Antineoplastic Agents/chemistry , Mutant Proteins/chemistry , Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/antagonists & inhibitors , Acetonitriles/pharmacology , Antineoplastic Agents/pharmacology , Gene Expression Regulation , Humans , Mutation , Pacemaker, Artificial , Piperazines/pharmacology , Protein Conformation , Pyridines/pharmacology , Pyrimidines/pharmacology , SOS1 Protein/metabolism , Signal Transduction , Structure-Activity Relationship
20.
Cell ; 184(10): 2649-2664.e18, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33848463

ABSTRACT

Receptor tyrosine kinase (RTK)-mediated activation of downstream effector pathways such as the RAS GTPase/MAP kinase (MAPK) signaling cascade is thought to occur exclusively from lipid membrane compartments in mammalian cells. Here, we uncover a membraneless, protein granule-based subcellular structure that can organize RTK/RAS/MAPK signaling in cancer. Chimeric (fusion) oncoproteins involving certain RTKs including ALK and RET undergo de novo higher-order assembly into membraneless cytoplasmic protein granules that actively signal. These pathogenic biomolecular condensates locally concentrate the RAS activating complex GRB2/SOS1 and activate RAS in a lipid membrane-independent manner. RTK protein granule formation is critical for oncogenic RAS/MAPK signaling output in these cells. We identify a set of protein granule components and establish structural rules that define the formation of membraneless protein granules by RTK oncoproteins. Our findings reveal membraneless, higher-order cytoplasmic protein assembly as a distinct subcellular platform for organizing oncogenic RTK and RAS signaling.


Subject(s)
Biomolecular Condensates/metabolism , Cytoplasmic Granules/metabolism , Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , ras Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Enzyme Activation , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HEK293 Cells , Humans , SOS1 Protein/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...