Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (38)

Search Parameters:
Keywords = oncogenic Hras

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
39 pages, 3100 KiB  
Review
RESEARCH CHALLENGES IN STAGE III AND IV RAS-ASSOCIATED CANCERS: A Narrative Review of the Complexities and Functions of the Family of RAS Genes and Ras Proteins in Housekeeping and Tumorigenesis
by Richard A. McDonald, Armando Varela-Ramirez and Amanda K. Ashley
Biology 2025, 14(8), 936; https://doi.org/10.3390/biology14080936 - 25 Jul 2025
Viewed by 498
Abstract
Proto-oncogenes in the RAS superfamily play dual roles in maintaining cellular homeostasis, such as regulating growth signals and contributing to cancer development through proliferation and deregulation. Activating proto-oncogenes in vitro transforms cells, underscoring their centrality in gene regulation and cellular networks. Despite decades [...] Read more.
Proto-oncogenes in the RAS superfamily play dual roles in maintaining cellular homeostasis, such as regulating growth signals and contributing to cancer development through proliferation and deregulation. Activating proto-oncogenes in vitro transforms cells, underscoring their centrality in gene regulation and cellular networks. Despite decades of research, poor outcomes in advanced cancers reveal gaps in understanding Ras-driven mechanisms or therapeutic strategies. This narrative review examines RAS genes and Ras proteins in both housekeeping functions, such as cell growth, apoptosis, and protein trafficking, as well as in tumorigenesis, integrating insights from human (HRAS, KRAS, NRAS), mouse (Hras, Kras, Nras), and Drosophila melanogaster (ras) models. While RAS mutations are tightly linked to human tumors, the interplay between their standard and oncogenic functions remains complex. Even within the same tissue, distinct cancer pathways—such as the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) pathways—can drive varied disease courses, complicating treatment. Advanced-stage cancers add further challenges, including heterogeneity, protective microenvironments, drug resistance, and adaptive progression. This synthesis organizes current knowledge of RAS gene regulation and Ras protein function from genomic alterations and intracellular signaling to membrane dynamics and extracellular interactions, offering a layered perspective on the Ras pathway’s role in both housekeeping and tumorigenic contexts. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

16 pages, 831 KiB  
Article
Mutational Profiling of Medullary Thyroid Carcinoma via a Large-Scale Genomic Repository
by Beau Hsia, Elijah Torbenson, Nigel Lang and Peter T. Silberstein
DNA 2025, 5(3), 35; https://doi.org/10.3390/dna5030035 - 17 Jul 2025
Viewed by 260
Abstract
Background: Medullary thyroid cancer (MTC), a neuroendocrine tumor originating from thyroid parafollicular C-cells, presents therapeutic challenges, particularly in advanced stages. While RET proto-oncogene mutations are known drivers, a comprehensive understanding of the broader somatic mutation landscape is needed to identify novel therapeutic targets [...] Read more.
Background: Medullary thyroid cancer (MTC), a neuroendocrine tumor originating from thyroid parafollicular C-cells, presents therapeutic challenges, particularly in advanced stages. While RET proto-oncogene mutations are known drivers, a comprehensive understanding of the broader somatic mutation landscape is needed to identify novel therapeutic targets and improve prognostication. This study leveraged the extensive AACR Project GENIE dataset to characterize MTC genomics. Methods: A retrospective analysis of MTC samples from GENIE examined recurrent somatic mutations, demographic/survival correlations, and copy number variations using targeted sequencing data (significance: p < 0.05). Results: Among 341 samples, RET mutations predominated (75.7%, mostly M918T), followed by HRAS (10.0%) and KRAS (5.6%), with mutual exclusivity between RET and RAS alterations. Recurrent mutations included KMT2D (5.3%), CDH11 (5.3%), ATM (5.0%), and TP53 (4.1%). NOTCH1 mutations were enriched in metastatic cases (p = 0.023). Preliminary associations included sex-linked mutations (BRAF/BRCA1/KIT in females, p = 0.028), and survival (ATM associated with longer survival, p = 0.016; BARD1/BLM/UBR5/MYH11 with shorter survival, p < 0.05), though limited subgroup sizes warrant caution. Conclusions: This large-scale genomic analysis confirms the centrality of RET and RAS pathway alterations in MTC and their mutual exclusivity. The association of NOTCH1 mutations with metastasis suggests a potential role in disease progression. While findings regarding demographic and survival correlations are preliminary, they generate hypotheses for future validation. This study enhances the genomic foundation for understanding MTC and underscores the need for integrated clinico-genomic datasets to refine therapeutic approaches. Full article
Show Figures

Figure 1

14 pages, 1562 KiB  
Article
A Cross-Sectional Exploratory Study of Rat Sarcoid (Ras) Activation in Women with and Without Polycystic Ovary Syndrome
by Sara Anjum Niinuma, Haniya Habib, Ashleigh Suzu-Nishio Takemoto, Priya Das, Thozhukat Sathyapalan, Stephen L. Atkin and Alexandra E. Butler
Cells 2025, 14(5), 377; https://doi.org/10.3390/cells14050377 - 5 Mar 2025
Viewed by 1222
Abstract
Objective: Rat sarcoma (Ras) proteins, Kirsten, Harvey, and Neuroblastoma rat sarcoma viral oncogene homolog (KRAS, HRAS, and NRAS, respectively), are a family of GTPases, which are key regulators of cellular growth, differentiation, and apoptosis through signal transduction pathways modulated by growth factors [...] Read more.
Objective: Rat sarcoma (Ras) proteins, Kirsten, Harvey, and Neuroblastoma rat sarcoma viral oncogene homolog (KRAS, HRAS, and NRAS, respectively), are a family of GTPases, which are key regulators of cellular growth, differentiation, and apoptosis through signal transduction pathways modulated by growth factors that have been recognized to be dysregulated in PCOS. This study explores Ras signaling proteins and growth factor-related proteins in polycystic ovary syndrome (PCOS). Methods: In a well-validated PCOS database of 147 PCOS and 97 control women, plasma was batch analyzed using Somascan proteomic analysis for circulating KRas, Ras GTPase-activating protein-1 (RASA1), and 45 growth factor-related proteins. The cohort was subsequently stratified for BMI (body mass index), testosterone, and insulin resistance (HOMA-IR) for subset analysis. Results: Circulating KRas, and RASA1 did not differ between PCOS and control women (p > 0.05). EGF1, EGFR, and EGFRvIII were decreased in PCOS (p = 0.04, p = 0.04 and p < 0.001, respectively). FGF8, FGF9, and FGF17 were increased in PCOS (p = 0.02, p = 0.03 and p = 0.04, respectively), and FGFR1 was decreased in PCOS (p < 0.001). VEGF-D (p < 0.001), IGF1 (p < 0.001), IGF-1sR (p = 0.02), and PDGFRA (p < 0.001) were decreased in PCOS compared to controls. After stratifying for BMI ≤ 29.9 kg/m2, EGFR FGF8, FGFR1 VEGF-D, IGF1, and IGF-1sR differed (p < 0.05) though EGF1, EGFRvIII, FGF8, FGFR1, and VEGF-D no longer differed; after subsequently stratifying for HOMA-IR, only FGFR1, VEGF-D, IGF1, and IGF-1sR differed between groups (p < 0.05). Conclusions: Several growth factors that activate Ras differ between women with and without PCOS, and when stratified for BMI and HOMA-IR, only FGFR1, VEGF-D, IGF1, and IGF-1sR differed; these appear to be inherent features of the pathophysiology of PCOS. Full article
(This article belongs to the Special Issue Ras Family of Genes and Proteins: Structure, Function and Regulation)
Show Figures

Graphical abstract

10 pages, 15870 KiB  
Case Report
Association of Schimmelpenning Syndrome with Astrocytoma (WHO Grade 3): Case Report
by Aija Tumova, Kaspars Auslands, Andrejs Millers, Zanda Priede, Māris Buks, Agnese Ozola, Elīna Ozoliņa, Kārlis Bicāns and Rūdolfs Ulmanis
Medicina 2024, 60(10), 1688; https://doi.org/10.3390/medicina60101688 - 14 Oct 2024
Viewed by 1640
Abstract
Schimmelpenning syndrome, or epidermal nevus syndrome, is a rare, neurocutaneous disorder characterized by skin abnormalities, such as epidermal nevi, and involvement of the central nervous system, including intracranial tumors. There are only a few reported cases of intracranial tumors associated with Schimmelpenning syndrome. [...] Read more.
Schimmelpenning syndrome, or epidermal nevus syndrome, is a rare, neurocutaneous disorder characterized by skin abnormalities, such as epidermal nevi, and involvement of the central nervous system, including intracranial tumors. There are only a few reported cases of intracranial tumors associated with Schimmelpenning syndrome. In most cases, a single nucleotide mutation in the RAS family proto-oncogenes, like HRAS or KRAS genes, can result in the genetic mosaicism that is responsible for the clinical manifestations of this syndrome. The authors present a case report of a woman with Schimmelpenning syndrome who sought medical help with complaints of progressive headache and dizziness. The radiological and histopathological findings indicated an astrocytoma, IDH-mutant (WHO grade 3). The molecular analysis revealed pathogenic changes in the oncogenic HRAS gene with a prevalence of 31%. The patient underwent surgical treatment and had no neurological sequelae. By presenting such a clinical case, attention is paid to the interrelationship between genetic syndromes and intracranial tumors. Full article
(This article belongs to the Section Neurology)
Show Figures

Figure 1

15 pages, 7928 KiB  
Article
GTP-Bound N-Ras Conformational States and Substates Are Modulated by Membrane and Point Mutation
by Alexandra Farcas and Lorant Janosi
Int. J. Mol. Sci. 2024, 25(3), 1430; https://doi.org/10.3390/ijms25031430 - 24 Jan 2024
Viewed by 2180
Abstract
Oncogenic Ras proteins are known to present multiple conformational states, as reported by the great variety of crystallographic structures. The GTP-bound states are grouped into two main states: the “inactive” state 1 and the “active” state 2. Recent reports on H-Ras have shown [...] Read more.
Oncogenic Ras proteins are known to present multiple conformational states, as reported by the great variety of crystallographic structures. The GTP-bound states are grouped into two main states: the “inactive” state 1 and the “active” state 2. Recent reports on H-Ras have shown that state 2 exhibits two substates, directly related to the orientation of Tyr32: toward the GTP-bound pocket and outwards. In this paper, we show that N-Ras exhibits another substate of state 2, related to a third orientation of Tyr32, toward Ala18 and parallel to the GTP-bound pocket. We also show that this substate is highly sampled in the G12V mutation of N-Ras and barely present in its wild-type form, and that the G12V mutation prohibits the sampling of the GTPase-activating protein (GAP) binding substate, rendering this mutation oncogenic. Furthermore, using molecular dynamics simulations, we explore the importance of the membrane on N-Ras’ conformational state dynamics and its strong influence on Ras protein stability. Moreover, the membrane has a significant influence on the conformational (sub)states sampling of Ras. This, in turn, is of crucial importance in the activation/deactivation cycle of Ras, due to the binding of guanine nucleotide exchange factor proteins (GEFs)/GTPase-activating proteins (GAPs). Full article
Show Figures

Figure 1

16 pages, 2960 KiB  
Article
Oncogene-Induced Senescence Is a Crucial Antitumor Defense Mechanism of Human Endometrial Stromal Cells
by Artem L. Toropov, Pavel I. Deryabin, Alla N. Shatrova and Aleksandra V. Borodkina
Int. J. Mol. Sci. 2023, 24(18), 14089; https://doi.org/10.3390/ijms241814089 - 14 Sep 2023
Cited by 6 | Viewed by 2649
Abstract
Being the major cellular component of highly dynamic tissue, endometrial stromal cells (EnSCs) are exposed to cycles of proliferation upon hormonal stimulation, which might pose risks for the accumulation of mutations and malignization. However, endometrial stromal tumors are rare and uncommon. The present [...] Read more.
Being the major cellular component of highly dynamic tissue, endometrial stromal cells (EnSCs) are exposed to cycles of proliferation upon hormonal stimulation, which might pose risks for the accumulation of mutations and malignization. However, endometrial stromal tumors are rare and uncommon. The present study uncovered defense mechanisms that might underlie the resistance of EnSCs against oncogenic transformation. All experiments were performed in vitro using the following methods: FACS, WB, RT-PCR, IF, molecular cloning, lentiviral transduction, and CRISPR/Cas9 genome editing. We revealed that the expression of the mutant HRASG12V leads to EnSC senescence. We experimentally confirmed the inability of HRASG12V-expressing EnSCs to bypass senescence and resume proliferation, even upon estrogen stimulation. At the molecular level, the induction of oncogene-induced senescence (OIS) was accompanied by activation of the MEK/ERK, PI3K/AKT, p53/p21WAF/CIP/Rb, and p38/p16INK4a/Rb pathways; however, inhibiting either pathway did not prevent cell cycle arrest. PTEN loss was established as an additional feature of HRASG12V-induced senescence in EnSCs. Using CRISPR-Cas9-mediated PTEN knockout, we identified PTEN loss-induced senescence as a reserve molecular mechanism to prevent the transformation of HRASG12V-expressing EnSCs. The present study highlights oncogene-induced senescence as an antitumor defense mechanism of EnSCs controlled by multiple backup molecular pathways. Full article
(This article belongs to the Collection Feature Papers in “Molecular Biology”)
Show Figures

Figure 1

2 pages, 185 KiB  
Editorial
Advances in Molecular Research of Oncogenes
by Fernando C. Baltanas and Eugenio Santos
Int. J. Mol. Sci. 2023, 24(8), 7222; https://doi.org/10.3390/ijms24087222 - 13 Apr 2023
Cited by 1 | Viewed by 1624
Abstract
The isolation of the first human oncogene (HRAS), a critical breakthrough in cancer research, has occurred over forty years ago, and the identification of new pathogenic oncogenes has continuously grown since [...] Full article
(This article belongs to the Special Issue Advances in Molecular Research of Oncogenes)
1 pages, 172 KiB  
Abstract
The Effect of RAS2 Gene Mutation in Single Cell Yeast Model
by Nazela Ahmed Chowdhury, Fatema Akter Sadia, Ismat Jahan Anee and Ashfaqul Muid Khandaker
Biol. Life Sci. Forum 2023, 21(1), 9; https://doi.org/10.3390/blsf2023021009 - 20 Mar 2023
Viewed by 1313
Abstract
More than 30% of all human malignancies are brought about by mutations in RAS proto-oncogenes (HRAS, KRAS, and NRAS) that are greatly conserved in yeast RAS1 and RAS2. This makes yeast (Saccharomyces cerevisiae) an efficient single-celled eukaryotic model organism to [...] Read more.
More than 30% of all human malignancies are brought about by mutations in RAS proto-oncogenes (HRAS, KRAS, and NRAS) that are greatly conserved in yeast RAS1 and RAS2. This makes yeast (Saccharomyces cerevisiae) an efficient single-celled eukaryotic model organism to study their functions. In the current investigation, the null mutation of the RAS2 gene was analyzed to find out its deleterious consequences in yeast cells based on their ability to utilize glycerol as a respiratory substrate, mtDNA mutation rate, mtDNA abundance, and distribution pattern. Mutant cells grown in YPEG plates demonstrated slight respiratory deficiency compared to the wild type. An erythromycin-resistant assay was carried out to analyze the spontaneous mitochondrial DNA mutation rate in the Δras2 mutant and it was found to be greater than that of wild type. In addition, the mitochondrial DNAs of both strains were also visualized under a fluorescence microscope using DAPI fluorescent stain. It was observed that mtDNA abundance was much lower compared to wild type cells. Thus, the present investigation revealed that deletion of the RAS2 gene resulted in mtDNA mutation and depletion. Full article
17 pages, 3487 KiB  
Article
Gain-of-Function p53N236S Mutation Drives the Bypassing of HRasV12-Induced Cellular Senescence via PGC–1α
by Hao Yang, Ke Zhang, Yusheng Guo, Xin Guo, Kailong Hou, Jing Hou, Ying Luo, Jing Liu and Shuting Jia
Int. J. Mol. Sci. 2023, 24(4), 3790; https://doi.org/10.3390/ijms24043790 - 14 Feb 2023
Viewed by 2679
Abstract
One of the key steps in tumorigenic transformation is immortalization in which cells bypass cancer-initiating barriers such as senescence. Senescence can be triggered by either telomere erosion or oncogenic stress (oncogene-induced senescence, OIS) and undergo p53- or Rb-dependent cell cycle arrest. The tumor [...] Read more.
One of the key steps in tumorigenic transformation is immortalization in which cells bypass cancer-initiating barriers such as senescence. Senescence can be triggered by either telomere erosion or oncogenic stress (oncogene-induced senescence, OIS) and undergo p53- or Rb-dependent cell cycle arrest. The tumor suppressor p53 is mutated in 50% of human cancers. In this study, we generated p53N236S (p53S) mutant knock-in mice and observed that p53S heterozygous mouse embryonic fibroblasts (p53S/+) escaped HRasV12-induced senescence after subculture in vitro and formed tumors after subcutaneous injection into severe combined immune deficiency (SCID) mice. We found that p53S increased the level and nuclear translocation of PGC–1α in late-stage p53S/++Ras cells (LS cells, which bypassed the OIS). The increase in PGC–1α promoted the biosynthesis and function of mitochondria in LS cells by inhibiting senescence-associated reactive oxygen species (ROS) and ROS-induced autophagy. In addition, p53S regulated the interaction between PGC–1α and PPARγ and promoted lipid synthesis, which may indicate an auxiliary pathway for facilitating cell escape from aging. Our results illuminate the mechanisms underlying p53S mutant-regulated senescence bypass and demonstrate the role played by PGC–1α in this process. Full article
(This article belongs to the Special Issue Targeted Cancer Therapies and Programmed Cell Death)
Show Figures

Figure 1

19 pages, 4128 KiB  
Article
Comprehensive Analysis of the Expression and Clinical Significance of RAS Family Members in Non-Small Cell Lung Cancer Based on Bioinformatics Data and the A549 Cell Line Model
by Milena Pązik, Marta Żebrowska-Nawrocka, Agnieszka Wosiak, Jacek Pietrzak and Ewa Balcerczak
Appl. Sci. 2023, 13(1), 166; https://doi.org/10.3390/app13010166 - 23 Dec 2022
Viewed by 4100
Abstract
Lung cancer is the second most frequent worldwide diagnosed cancer. Mutations in the RAS genes family are among the most common oncogenic alterations occurring in non-small cell lung cancer (NSCLC). Many treatment options against KRAS mutations have been developed for NSCLC; however, they [...] Read more.
Lung cancer is the second most frequent worldwide diagnosed cancer. Mutations in the RAS genes family are among the most common oncogenic alterations occurring in non-small cell lung cancer (NSCLC). Many treatment options against KRAS mutations have been developed for NSCLC; however, they remain insufficient. Moreover, the role of KRAS and HRAS gene expression in lung cancer remains unclear. However, inhibitors of RAS genes expression seem to be a good candidate for new drugs agents in NSCLC. This study used bioinformatical analysis to determine KRAS and HRAS gene expression and its clinical significance, and then examined the influence of three different RAS inhibitors (farnesythiosalicylic acid (FTS), deltarasin and Kobe0065) on cell growth and the KRAS and HRAS gene expression (by RT-qPCR) in human NSCLC A549 cells. KRAS and HRAS were shown to be overexpressed in NSCLC compared to non-tumor lung tissues of healthy individuals (from databases) and significantly associated with different clinicopathological features. It was also found that FTS, in a dose-dependent manner, suppressed proliferation of human A549 cells, while deltarasin reduced expression of HRAS in the lung cancer cells. To sum up, the results obtained from analyses based on bioinformatics databases indicate that the studied genes are potential risk factors for the development of lung cancer. On the other hand, studies of their expression on cell lines indicated that they may also be potentially important in the response to treatment using RAS inhibitors. Full article
(This article belongs to the Section Biomedical Engineering)
Show Figures

Figure 1

23 pages, 808 KiB  
Review
Senolytic Therapy: A Potential Approach for the Elimination of Oncogene-Induced Senescent HPV-Positive Cells
by Tareq Saleh, Ashraf I. Khasawneh, Nisreen Himsawi, Jumana Abu-Raideh, Vera Ejeilat, Ahmed M. Elshazly and David A. Gewirtz
Int. J. Mol. Sci. 2022, 23(24), 15512; https://doi.org/10.3390/ijms232415512 - 8 Dec 2022
Cited by 16 | Viewed by 4128
Abstract
Senescence represents a unique cellular stress response characterized by a stable growth arrest, macromolecular alterations, and wide spectrum changes in gene expression. Classically, senescence is the end-product of progressive telomeric attrition resulting from the repetitive division of somatic cells. In addition, senescent cells [...] Read more.
Senescence represents a unique cellular stress response characterized by a stable growth arrest, macromolecular alterations, and wide spectrum changes in gene expression. Classically, senescence is the end-product of progressive telomeric attrition resulting from the repetitive division of somatic cells. In addition, senescent cells accumulate in premalignant lesions, in part, as a product of oncogene hyperactivation, reflecting one element of the tumor suppressive function of senescence. Oncogenic processes that induce senescence include overexpression/hyperactivation of H-Ras, B-Raf, and cyclin E as well as inactivation of PTEN. Oncogenic viruses, such as Human Papilloma Virus (HPV), have also been shown to induce senescence. High-risk strains of HPV drive the immortalization, and hence transformation, of cervical epithelial cells via several mechanisms, but primarily via deregulation of the cell cycle, and possibly, by facilitating escape from senescence. Despite the wide and successful utilization of HPV vaccines in reducing the incidence of cervical cancer, this measure is not effective in preventing cancer development in individuals already positive for HPV. Accordingly, in this commentary, we focus on the potential contribution of oncogene and HPV-induced senescence (OIS) in cervical cancer. We further consider the potential utility of senolytic agents for the elimination of HPV-harboring senescent cells as a strategy for reducing HPV-driven transformation and the risk of cervical cancer development. Full article
Show Figures

Figure 1

16 pages, 2294 KiB  
Article
Predicting Deleterious Non-Synonymous Single Nucleotide Polymorphisms (nsSNPs) of HRAS Gene and In Silico Evaluation of Their Structural and Functional Consequences towards Diagnosis and Prognosis of Cancer
by Chuan-Yu Chai, Sathiya Maran, Hin-Yee Thew, Yong-Chiang Tan, Nik Mohd Afizan Nik Abd Rahman, Wan-Hee Cheng, Kok-Song Lai, Jiun-Yan Loh and Wai-Sum Yap
Biology 2022, 11(11), 1604; https://doi.org/10.3390/biology11111604 - 2 Nov 2022
Cited by 11 | Viewed by 3912
Abstract
The Harvey rat sarcoma (HRAS) proto-oncogene belongs to the RAS family and is one of the pathogenic genes that cause cancer. Deleterious nsSNPs might have adverse consequences at the protein level. This study aimed to investigate deleterious nsSNPs in the HRAS gene in [...] Read more.
The Harvey rat sarcoma (HRAS) proto-oncogene belongs to the RAS family and is one of the pathogenic genes that cause cancer. Deleterious nsSNPs might have adverse consequences at the protein level. This study aimed to investigate deleterious nsSNPs in the HRAS gene in predicting structural alterations associated with mutants that disrupt normal protein–protein interactions. Functional and structural analysis was employed in analyzing the HRAS nsSNPs. Putative post-translational modification sites and the changes in protein–protein interactions, which included a variety of signal cascades, were also investigated. Five different bioinformatics tools predicted 33 nsSNPs as “pathogenic” or “harmful”. Stability analysis predicted rs1554885139, rs770492627, rs1589792804, rs730880460, rs104894227, rs104894227, and rs121917759 as unstable. Protein–protein interaction analysis revealed that HRAS has a hub connecting three clusters consisting of 11 proteins, and changes in HRAS might cause signal cascades to dissociate. Furthermore, Kaplan–Meier bioinformatics analyses indicated that the HRAS gene deregulation affected the overall survival rate of patients with breast cancer, leading to prognostic significance. Thus, based on these analyses, our study suggests that the reported nsSNPs of HRAS may serve as potential targets for different proteomic studies, diagnoses, and therapeutic interventions focusing on cancer. Full article
(This article belongs to the Special Issue Cancer Signaling Pathways, Crosstalk and Therapeutics)
Show Figures

Figure 1

23 pages, 2667 KiB  
Review
Computer-Aided Drug Design Boosts RAS Inhibitor Discovery
by Ge Wang, Yuhao Bai, Jiarui Cui, Zirui Zong, Yuan Gao and Zhen Zheng
Molecules 2022, 27(17), 5710; https://doi.org/10.3390/molecules27175710 - 5 Sep 2022
Cited by 18 | Viewed by 4935
Abstract
The Rat Sarcoma (RAS) family (NRAS, HRAS, and KRAS) is endowed with GTPase activity to regulate various signaling pathways in ubiquitous animal cells. As proto-oncogenes, RAS mutations can maintain activation, leading to the growth and proliferation of abnormal cells and the development of [...] Read more.
The Rat Sarcoma (RAS) family (NRAS, HRAS, and KRAS) is endowed with GTPase activity to regulate various signaling pathways in ubiquitous animal cells. As proto-oncogenes, RAS mutations can maintain activation, leading to the growth and proliferation of abnormal cells and the development of a variety of human cancers. For the fight against tumors, the discovery of RAS-targeted drugs is of high significance. On the one hand, the structural properties of the RAS protein make it difficult to find inhibitors specifically targeted to it. On the other hand, targeting other molecules in the RAS signaling pathway often leads to severe tissue toxicities due to the lack of disease specificity. However, computer-aided drug design (CADD) can help solve the above problems. As an interdisciplinary approach that combines computational biology with medicinal chemistry, CADD has brought a variety of advances and numerous benefits to drug design, such as the rapid identification of new targets and discovery of new drugs. Based on an overview of RAS features and the history of inhibitor discovery, this review provides insight into the application of mainstream CADD methods to RAS drug design. Full article
(This article belongs to the Special Issue Computational Strategy for Drug Design)
Show Figures

Figure 1

15 pages, 423 KiB  
Article
Treatment of RET-Positive Advanced Medullary Thyroid Cancer with Multi-Tyrosine Kinase Inhibitors—A Retrospective Multi-Center Registry Analysis
by Viktoria Florentine Koehler, Pia Adam, Carmina Teresa Fuss, Linmiao Jiang, Elke Berg, Karin Frank-Raue, Friedhelm Raue, Eva Hoster, Thomas Knösel, Hans-Ulrich Schildhaus, Thomas Negele, Udo Siebolts, Kerstin Lorenz, Stephanie Allelein, Matthias Schott, Christine Spitzweg and Matthias Kroiss
Cancers 2022, 14(14), 3405; https://doi.org/10.3390/cancers14143405 - 13 Jul 2022
Cited by 7 | Viewed by 3373
Abstract
Background: RET (rearranged during transfection) variants are the most prevalent oncogenic events in medullary thyroid cancer (MTC). In advanced disease, multi-tyrosine kinase inhibitors (MKIs) cabozantinib and vandetanib are the approved standard treatment irrespective of RET status. The actual outcome of patients with RET [...] Read more.
Background: RET (rearranged during transfection) variants are the most prevalent oncogenic events in medullary thyroid cancer (MTC). In advanced disease, multi-tyrosine kinase inhibitors (MKIs) cabozantinib and vandetanib are the approved standard treatment irrespective of RET status. The actual outcome of patients with RET-positive MTC treated with MKIs is ill described. Methods: We here retrospectively determined the RET oncogene variant status with a targeted DNA Custom Panel in a prospectively collected cohort of 48 patients with advanced MTC treated with vandetanib and/or cabozantinib at four German referral centers. Progression-free survival (PFS) and overall survival (OS) probabilities were estimated using the Kaplan-Meier method. Results: In total, 44/48 (92%) patients had germline or somatic RET variants. The M918T variant was found in 29/44 (66%) cases. In total, 2/32 (6%) patients with a somatic RET variant had further somatic variants, while in 1/32 (3%) patient with a germline RET variant, additional variants were found. Only 1/48 (2%) patient had a pathogenic HRAS variant, and no variants were found in 3 cases. In first-line treatment, the median OS was 53 (95% CI (95% confidence interval), 32–NR (not reached); n = 36), and the median PFS was 21 months (12–39; n = 33) in RET-positive MTC patients. In second-line treatment, the median OS was 18 (13–79; n = 22), and the median PFS was 3.5 months (2–14; n = 22) in RET-positive cases. Conclusions: RET variants were highly prevalent in patients with advanced MTC. The treatment results in RET-positive cases were similar to those reported in unselected cohorts. Full article
(This article belongs to the Special Issue Advances in Thyroid Cancer)
Show Figures

Figure 1

11 pages, 976 KiB  
Case Report
HRAS Q61L Mutation as a Possible Target for Non-Small Cell Lung Cancer: Case Series and Review of Literature
by Laurent Mathiot, Guillaume Herbreteau, Siméon Robin, Charlotte Fenat, Jaafar Bennouna, Christophe Blanquart, Marc Denis and Elvire Pons-Tostivint
Curr. Oncol. 2022, 29(5), 3748-3758; https://doi.org/10.3390/curroncol29050300 - 20 May 2022
Cited by 9 | Viewed by 4541
Abstract
Introduction: Assessment of actionable gene mutations and oncogene fusions have made a paradigm shift in treatment strategies of non-small cell lung cancer (NSCLC). HRAS mutations involved around 0.2–0.8% of NSCLC patients, mostly on codon 61. For these patients, few data are available regarding [...] Read more.
Introduction: Assessment of actionable gene mutations and oncogene fusions have made a paradigm shift in treatment strategies of non-small cell lung cancer (NSCLC). HRAS mutations involved around 0.2–0.8% of NSCLC patients, mostly on codon 61. For these patients, few data are available regarding clinical characteristics and response to therapies. Methods: Next-Generation Sequencing (NGS) done routinely at Nantes University Hospital was used to identify HRAS molecular alterations in NSCLC patients. We identified and described four HRAS p.GlnQ61Leu mutated patients. Literature of previously HRAS-mutant NSCLC cases was reviewed, and available data in solid tumour with the most advanced H-Ras specific inhibitor, tipifarnib, were presented. Results: Of 1614 patients diagnosed with advanced NSCLC from January 2018 to December 2020, four (0.25%) had HRAS p.Gln61Leu mutation. Three of them died during the first-line systemic therapy. Furthermore, three additional cases were identified in literature. All cases were current or former smokers, most of them had pleural or pericardial effusion at diagnosis. Conclusions: The clinical course of patients with HRAS-mutant NSCLC remains unclear. Furthers cases should be identified in order to clarify prognosis and response to therapies. Tipifarnib, a farnesyl transferase inhibitor, is a promising candidate to target HRAS-mutant tumours and should be explored in NSCLC patients. Full article
Show Figures

Figure 1

Back to TopTop