Fact-checked by Grok 2 weeks ago

HRAS

HRAS is a proto-oncogene located on 11p15.5 that encodes a 21-kDa protein belonging to the family of small . This protein functions as a in pathways, cycling between an active GTP-bound state and an inactive GDP-bound state to regulate cellular processes such as , , and survival. The HRAS product is ubiquitously expressed, with particularly high levels in skin and brain tissues, and it localizes primarily to the plasma membrane and Golgi apparatus through post-translational modifications like palmitoylation. As part of the , HRAS plays a central role in the (MAPK) signaling cascade, transmitting signals from extracellular receptors to the nucleus to control gene expression and cellular responses. It interacts with downstream effectors like RAF kinases to activate pathways involved in and division, and its activity is tightly regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Dysregulation of HRAS, often through point mutations that impair GTP hydrolysis, leads to constitutive activation and promotes oncogenesis. Germline mutations in HRAS cause , a rare autosomal dominant disorder characterized by developmental delay, coarse facial features, cardiac abnormalities like , , and an elevated risk of tumors. Common germline variants include G12S, G12A, and G13D, which are typically and result in hyperactivation of the /MAPK pathway. HRAS mutations occur in approximately 1-3% of human cancers, with higher prevalence in specific types such as (up to 13%), head and neck (3-4%), and thyroid carcinoma; these mutations, often at codons 12, 13, or 61, drive tumor progression by enhancing and survival. Despite the lower frequency compared to KRAS and NRAS mutations, which together account for the majority of alterations (approximately 17% of all cancers), HRAS alterations define distinct clinical subgroups with implications for targeted therapies like MEK inhibitors. As of 2025, emerging therapies such as farnesyltransferase inhibitors show promise for HRAS-mutant cancers.

Discovery and History

Gene Identification

The Harvey rat sarcoma (Ha-MuSV), identified by Jennifer Harvey in 1964 during studies of rat models, was one of the first retroviruses linked to rapid tumor induction in , carrying a transforming later designated v-H-ras. In the 1970s, researchers including Edward Scolnick and colleagues at the (NIH) characterized the viral oncogene's product, a 21-kDa protein (p21) with GTP-binding properties, establishing its role as a proto-oncogene derived from cellular sequences transduced by the . In 1982, independent groups, including one led by Robert Weinberg, identified the human homolog of v-H-ras (c-H-ras, now ) through analysis of transforming DNA from the EJ human bladder carcinoma cell line, demonstrating via Southern blotting of human genomic DNA hybridized with v-H-ras probes. This discovery revealed that the EJ shared a conserved 3.0-kilobase region with the viral gene, confirming HRAS as the cellular counterpart responsible for neoplastic transformation in human tumors. The full cloning of the human HRAS gene was achieved that same year by Edward Scolnick's group at NIH, including Mariano Barbacid and colleagues, who isolated and sequenced the proto-oncogene from normal human DNA using screening and assays in NIH/3T3 mouse fibroblasts to verify transforming activity. They distinguished it as a proto-oncogene activated by point mutations, such as the glycine-to-valine substitution at codon 12 observed in the T24 bladder carcinoma variant.

Oncogenic Discovery

The oncogenic potential of HRAS was first demonstrated through DNA transfection experiments in the early 1980s, which showed that mutated forms of the gene could transform normal cells. In 1982, researchers led by Robert A. Weinberg transfected DNA from the human bladder carcinoma cell line EJ into NIH 3T3 mouse fibroblasts, resulting in the formation of transformed foci—piled-up, anchorage-independent cell clusters indicative of oncogenic activity. Analysis of the transforming DNA identified HRAS as the responsible gene, harboring a specific point mutation at codon 12 (G12V), which substitutes glycine with valine and locks the protein in an active GTP-bound state. Independent studies by other groups confirmed this finding, sequencing the mutated HRAS from EJ cells and linking the single amino acid change to its transforming capability. These assays established HRAS mutations as sufficient to induce cellular transformation in vitro, marking a pivotal experimental validation of its role as an oncogene. Extending these transfection approaches, Weinberg's team in 1983 applied the NIH 3T3 formation assay to DNA from a cell line, identifying another activated HRAS variant capable of oncogenic . This work reinforced that mutations in HRAS, detected via -induced phenotypic changes, were not limited to but extended to other tumor types. Parallel efforts sequenced HRAS from primary tumors, revealing point mutations such as G12V in and , directly associating these alterations with cancer development. These discoveries highlighted the assay's power in isolating dominant oncogenes from tumor genomes, shifting from viral to cellular origins of cancer. The identification of HRAS as an built on foundational insights into proto-oncogenes, exemplified by the 1989 in Physiology or Medicine awarded to J. Michael Bishop and for elucidating the cellular origin of retroviral oncogenes. Their work demonstrated that viral oncogenes like v-src derive from normal cellular genes, providing the theoretical basis for recognizing mutated cellular HRAS as an activated counterpart. By the late 1980s, epidemiological surveys indicated that mutations in the family of genes (, , ), mainly via codon 12, 13, or 61 mutations, occurred in approximately 10-20% of human tumors across various sites, with HRAS contributing a smaller proportion, underscoring its broad relevance in . These early links emphasized HRAS's role in driving uncontrolled proliferation through gain-of-function changes.

Molecular Biology

Genomic Structure

The HRAS gene is located on the short arm of human at cytogenetic band p15.5. The genomic locus spans approximately 3.3 , extending from base pair 532242 to 535576 on the complement strand according to the GRCh38.p14 assembly. The gene consists of 7 s, with the canonical transcript (NM_005343.4) comprising 6 s: 1 is entirely non-coding (part of the ), and s 2 through 5 encode the full 189-amino acid H-Ras protein, while 6 contains the 3' . The coding sequence is 570 long (including the ), initiating in 2 and terminating in 5. The promoter region immediately upstream of the transcription start site is GC-rich and lacks a , relying instead on multiple Sp1-binding GC box motifs (at least four to six) for basal transcriptional initiation at multiple start sites. Although the primary transcript is highly conserved, alternative splicing produces four variants: NM_005343.4 and NM_001130442.3 (canonical isoform 1, 1,070 bp, 189 aa), NM_176795.5 (isoform 2, shorter 5' UTR, 171 aa), and NM_001318054.2 (isoform 3, shorter ). The HRAS coding exhibits high evolutionary across mammals, with orthologs identified in over 290 sharing greater than 90% identity in key functional domains. The complete genomic and cDNA sequences were first determined in 1983 through and sequencing efforts on normal and oncogenically activated alleles from the T24 bladder carcinoma cell line.

Expression and Regulation

HRAS exhibits ubiquitous expression across tissues, with particularly elevated levels in epithelial tissues such as skin and , as well as in and muscle. This pattern reflects its role as a fundamental regulator of , though tissue-specific variations influence baseline activity in contexts like epithelial and . At the transcriptional level, HRAS expression is modulated by regulatory elements including enhancers that respond to feedback from the MAPK pathway. Deregulated Ras-ERK signaling alters the enhancer landscape and associated modifications, thereby influencing HRAS-dependent in a context-specific manner. Growth factors such as (EGF) can upregulate HRAS transcription indirectly through EGFR-mediated activation of downstream transcription factors like AP-1, contributing to enhanced expression in responsive cells. Post-transcriptional regulation of HRAS involves microRNAs and alternative . For instance, miR-143-3p targets the 3' of HRAS mRNA, suppressing its expression and thereby inhibiting oncogenic signaling in cancers like . Additionally, alternative generates HRAS mRNA isoforms with varying 3' UTR lengths, which affect mRNA stability and translation efficiency; oncogenic mutations can promote usage of proximal sites, leading to more stable transcripts that enhance tumor progression. Epigenetic mechanisms further fine-tune HRAS expression. Oncogenic RAS signaling stimulates acetylation at promoters of target genes, promoting transcriptional . In certain cancers, aberrant patterns in non-coding regions of HRAS exhibit tissue-specific variations, potentially contributing to modulated expression levels, though hypermethylation-mediated silencing is less common compared to events.

Protein Function

Structural Features

HRAS is a small monomeric protein with a molecular weight of approximately 21 , comprising 189 residues. The core structure consists of two main domains: the G-domain (residues 1-166), which encompasses the nucleotide-binding and catalytic sites, and the C-terminal (HVR; residues 167-189), an unstructured segment that facilitates membrane association through post-translational lipid modifications. The G-domain fold is characteristic of the , featuring a six-stranded β-sheet flanked by five α-helices, enabling the protein to function as a conformational switch. The G-domain structure is highly conserved among family members (, , ). Key structural motifs within the G-domain are essential for nucleotide interactions and conformational dynamics. The P-loop (G1 motif, residues 10-17, sequence GAGGVGKS) forms a flexible phosphate-binding loop that coordinates the β- and γ-phosphates of GTP via bonds and magnesium ion stabilization. Adjacent to this, Switch I (G2 motif, residues 30-40) and Switch II (G3 motif, residues 60-76) are intrinsically disordered loops in the apo form. Switch I adopts an α-helical conformation in the GTP-bound state and is extended in the GDP-bound state; Switch II adopts a β-strand conformation in the GTP-bound state and an α-helical conformation in the GDP-bound state, thereby transmitting signals to downstream effectors. These regions, conserved across isoforms, underscore HRAS's role in . Post-translational lipid modifications at the are critical for HRAS's subcellular localization. The CAAX box (residues 186-189, CVIM) directs farnesylation of Cys186 by protein farnesyltransferase, attaching a 15-carbon farnesyl isoprenoid that promotes initial insertion. HRAS further undergoes reversible palmitoylation at Cys181 and Cys184 via linkages, which increases affinity by approximately 100-fold compared to farnesylation alone and facilitates dynamic trafficking between compartments. The atomic-level of HRAS was first resolved in the early 1990s through , providing foundational insights into its mechanism. Seminal studies, such as Milburn et al. (1990) for the GTP analog-bound form (PDB 5P21) and Pai et al. (1990) for the GDP-bound form (PDB 4Q21), revealed how binding stabilizes distinct Switch conformations. Subsequent structures, including the GTP-bound form in PDB 1QRA (resolved at 1.6 ), have refined these models, confirming the protein's compact, globular architecture and flexibility in the switch regions.

Biochemical Mechanism

HRAS operates as a molecular switch in cellular signaling, cycling between an inactive GDP-bound conformation and an active GTP-bound state. In the inactive state, HRAS tightly binds guanosine diphosphate (GDP), which maintains a closed conformation of its switch I and switch II regions, preventing effector interactions. Activation occurs through guanine nucleotide exchange factors (GEFs), such as son of sevenless 1 (SOS1), which catalyze the dissociation of GDP and facilitate binding of guanosine triphosphate (GTP). This exchange is driven by SOS1's catalytic domain, which stabilizes a nucleotide-free intermediate of HRAS, promoting the high-affinity binding of abundant cellular GTP over GDP. The resulting HRAS-GTP complex undergoes conformational changes in its switch regions, enabling interactions with downstream effectors. Inactivation is achieved via GTP hydrolysis to GDP, reverting HRAS to its inactive form; this intrinsic GTPase activity is inherently slow, with a rate constant of approximately $10^{-4} s^{-1} at physiological temperatures. GTPase-activating proteins (GAPs), such as (NF1), dramatically accelerate this by up to $10^5-fold through insertion of an "arginine finger" (e.g., in p120GAP homologs) into the , which stabilizes the and positions a nucleophilic for attack on the \gamma-. The is catalyzed by 61 (Gln61) of HRAS, which orients the for inline : \text{HRAS-GTP} + \text{H}_2\text{O} \rightarrow \text{HRAS-GDP} + \text{P}_\text{i} Mutations at Gln61 impair this positioning, reducing GAP-stimulated rates by orders of magnitude. The helical domain of HRAS contributes to autoinhibition in the GDP-bound state by interacting with the G domain to stabilize the closed nucleotide-binding pocket and restrict conformational flexibility, thereby hindering spontaneous activation.

Signaling Pathways

RAF-MEK-ERK Cascade

Upon activation, GTP-bound HRAS interacts directly with RAF kinases (ARAF, BRAF, and ), binding to their to relieve autoinhibition of the kinase domain and recruit RAF to the plasma membrane where HRAS is localized. This interaction disrupts intramolecular inhibitory contacts within RAF, enabling dimerization and subsequent autophosphorylation for full activation. Activated RAF then phosphorylates and activates mitogen-activated protein kinase kinases MEK1 and MEK2 at specific serine residues. MEK1/2 in turn phosphorylate extracellular signal-regulated kinases ERK1 and ERK2 on threonine and tyrosine residues within the TEY motif, leading to their activation. Phosphorylated ERK1/2 translocate to the nucleus, where they phosphorylate transcription factors such as Elk-1 and c-Fos, inducing expression of genes including cyclin D1 and c-Myc, which promote G1 to S phase progression by activating cyclin-dependent kinases and driving cell proliferation. To prevent excessive signaling, activated ERK provides negative feedback by phosphorylating son of sevenless 1 (SOS1), the guanine nucleotide exchange factor (GEF) for HRAS, at multiple sites including serine 1132, which inhibits SOS1's GEF activity and reduces further HRAS activation. In fibroblasts, HRAS activation typically results in a 5- to 10-fold increase in ERK phosphorylation, underscoring the pathway's amplification of mitogenic signals for proliferation.

PI3K-AKT Pathway

The GTP-bound form of HRAS directly interacts with the Ras-binding domain (RBD) of the PI3K catalytic subunit p110α, thereby activating the lipid kinase activity of class IA PI3K. This binding, which is dependent on the effector region of HRAS, promotes the of phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), a key second messenger that accumulates at the plasma membrane. The interaction enhances PI3K activity through conformational changes and facilitates membrane recruitment, essential for downstream signaling in and survival. PIP3 generated by HRAS-activated PI3K recruits the serine/ AKT (also known as PKB) to the plasma membrane, where it undergoes activation via at Thr308 by PDK1 and at Ser473 by mTORC2. Activated AKT then multiple targets to promote cell survival and metabolism; for instance, of BAD sequesters it to 14-3-3 proteins, inhibiting its pro-apoptotic function. Similarly, AKT-mediated of TSC2 inactivates the TSC1/TSC2 complex, relieving inhibition of Rheb and thereby activating to drive protein through targets like p70 S6 . These effects collectively enhance anti-apoptotic signaling and anabolic processes critical for cellular . HRAS signaling through the PI3K-AKT pathway integrates with the pathway to enable full oncogenic transformation, as evidenced by synergistic tumor regression in mouse models upon combined inhibition of both cascades. This crosstalk ensures coordinated regulation of survival and growth signals, with PI3K-AKT providing metabolic and anti-apoptotic support alongside -driven proliferation. In certain cellular contexts, such as fibroblasts and epithelial cells, HRAS exhibits higher potency in activating PI3K compared to , attributed to differences in their ability to stimulate PIP3 production and downstream AKT signaling. This isoform-specific affinity contributes to distinct oncogenic potentials, with HRAS more effectively promoting PI3K-dependent transformation in some tissues.

Pathogenic Variants

Germline Mutations

Germline mutations in the HRAS gene are rare, accounting for less than 1% of all cases among the , a group of developmental disorders caused by variants in the RAS/MAPK pathway. These mutations are predominantly activating and cluster at specific hotspots, including codons 12, 13, and 61, which alter the GTP-binding domain of the HRAS protein. For instance, the p.G12S variant is identified in approximately 80% of individuals with , the primary RASopathy associated with HRAS germline changes. These variants follow an autosomal dominant pattern, with the vast majority arising in affected individuals rather than being inherited from parents. In rare instances, or direct transmission has been documented, but occurrence predominates due to the mutations' high and the selective pressure against their propagation in the . Functionally, HRAS mutations impair the protein's intrinsic activity and its responsiveness to GTPase-activating proteins (GAPs), resulting in reduced and a 10- to 100-fold prolongation of the GTP-bound active state compared to wild-type HRAS. This sustained activation hyperdrives downstream /MAPK signaling, contributing to the developmental perturbations characteristic of associated syndromes. In , common variants include p.G12A, p.G12S, and p.G12V, while p.G13C and p.G13R have been reported in cases of milder phenotypes. These effects parallel somatic HRAS alterations in certain cancers, though variants exert whole-body influences during .

Somatic Mutations

Somatic mutations in HRAS are acquired genetic alterations that occur in tumor cells and contribute to oncogenesis by locking the protein in its active GTP-bound state. These mutations are less common than those in or NRAS but are recurrent in specific cancer types, driving uncontrolled through dysregulated signaling. Unlike variants, somatic HRAS mutations are not inherited and arise sporadically during tumorigenesis. The vast majority of oncogenic HRAS somatic mutations cluster at three hotspot codons: G12, G13, and Q61, accounting for approximately 90-95% of all detected variants across cancers. At these sites, missense substitutions such as G12V, G12S, G13R, and Q61R predominate, with codon 12 mutations comprising about 50% and codon 61 around 40% of HRAS alterations. For instance, in head and neck squamous cell carcinoma (HNSCC), the G12S variant is the most frequent among HRAS mutants, occurring in roughly 3-4% of cases overall, though specific subtypes may show higher enrichment. These s are evolutionarily conserved and critical for GTP hydrolysis. Mechanistically, these missense at G12, G13, and Q61 impair the of GTPase-activating proteins (GAPs), such as NF1, which normally accelerate GTP to inactivate HRAS. This results in prolonged GTP and constitutive activation of downstream effectors like RAF and PI3K, promoting tumor growth and survival. In addition to point , HRAS occurs in 5-10% of certain tumors, leading to overexpression of wild-type protein and enhanced signaling, though this is less frequent than mutational activation. Next-generation sequencing (NGS) has facilitated the detection of HRAS , revealing a pan-cancer of 3-5%, with higher rates in specific histologies such as (around 10%) and salivary gland tumors (up to 20% in mucoepidermoid carcinomas). In bladder urothelial carcinoma, HRAS often co-occur with FGFR3 alterations in low-grade tumors, while in salivary gland neoplasms, they are enriched in subsets like epithelial-myoepithelial . These tools enable precise identification in clinical settings, informing potential targeted approaches. Recent analyses from 2023-2025, leveraging TCGA datasets, confirm HRAS mutations as predominant in distinct HNSCC subgroups, occurring in about 6% of cases overall and associating with unique immunologic profiles, such as increased expression and pre-exhausted T cells. A 2025 study further revealed that wild-type HRAS overexpression promotes chemoresistance in HNSCC by enhancing , suggesting broader roles beyond mutations. These findings highlight HRAS-altered HNSCC as a molecularly defined entity with prognostic implications, underscoring the value of ongoing genomic profiling in refining tumor classification.

Clinical Associations

Rasopathies

Rasopathies are a group of developmental disorders caused by mutations in genes of the /MAPK signaling pathway, with mutations primarily associated with , a rare multisystem condition characterized by distinctive facial features, cardiac abnormalities, and growth issues. In , affected individuals typically exhibit coarse facial features including full lips, large mouth, and low-set ears, along with congenital heart defects such as or , and severe due to feeding difficulties and metabolic demands. Approximately 80% of cases result from the specific HRAS p.Gly12Ser (G12S) variant, a gain-of-function that constitutively activates the protein. Diagnosis is confirmed through identifying heterozygous HRAS mutations, often performed via targeted sequencing of gene panels when clinical features suggest the condition. Costello syndrome shares overlapping phenotypic features with other RASopathies, such as cardiofaciocutaneous (CFC) syndrome, which presents with similar cardiac and facial anomalies but is predominantly caused by mutations in BRAF or other pathway genes; however, HRAS variants at codon 13, such as p.Gly13Asp or p.Gly13Val, account for about 5-10% of Costello syndrome cases and contribute to milder or variable expressions within the spectrum. The pathophysiology stems from hyperactive HRAS signaling, where these germline mutations impair GTPase activity, leading to persistent activation of downstream effectors like RAF-MEK-ERK, which promotes excessive prenatal and postnatal cell proliferation, differentiation, and survival, ultimately disrupting normal organ development. This dysregulated growth signaling explains the multisystem involvement, including skeletal, cutaneous, and neurological manifestations observed from infancy. Management of Costello syndrome is symptomatic and multidisciplinary, focusing on supportive care to address complications like feeding tubes for , cardiac monitoring and medications for arrhythmias, and physical/ for developmental delays. Regular screening for associated issues, such as orthopedic evaluations and dermatologic interventions for skin laxity, is recommended, with considered in select cases to mitigate after weighing risks. The estimated incidence is approximately 1 in 300,000 live births, underscoring its rarity and the need for specialized .

Cancers

Somatic mutations in HRAS are oncogenic drivers in a subset of human malignancies, occurring in approximately 3-5% of all tumors across cancer types, with higher frequencies in specific epithelial cancers. These mutations constitutively activate HRAS, promoting uncontrolled and survival through dysregulated downstream signaling. Notably, HRAS alterations often with mutations in the PI3K-AKT pathway, such as PIK3CA gain-of-function variants, which synergistically enhance tumor aggressiveness by amplifying both MAPK and PI3K signaling axes. This is particularly evident in salivary duct carcinoma, where HRAS and PIK3CA mutations are found in up to 23% and 23% of cases, respectively, contributing to a more invasive . In , HRAS mutations are present in up to 13-15% of cases, predominantly affecting non-muscle-invasive urothelial carcinomas. The G12V hotspot mutation is associated with increased risk of tumor recurrence and progression to muscle-invasive disease, leading to worse overall prognosis compared to wild-type tumors. Patients with HRAS-mutant exhibit higher rates of local relapse post-resection, with studies indicating that activated HRAS drives resistance to standard therapies and . HRAS mutations occur in 3-5% of head and neck squamous cell carcinomas (HNSCC), less frequently than KRAS or EGFR alterations, but define a distinct molecular subgroup with aggressive behavior. The G12S variant predominates, correlating with poorer survival outcomes in the absence of targeted interventions. Recent analyses from 2023 highlight that HRAS-mutant HNSCC tumors exhibit an immunosuppressive tumor microenvironment, marked by elevated PD-L1 expression and reduced T-cell infiltration, resulting in diminished response to immune checkpoint inhibitors like pembrolizumab. Beyond these, HRAS mutations contribute to in about 5-10% of follicular and subtypes, often alongside NRAS variants, and are linked to higher malignancy risk in indeterminate nodules. In , mutation rates reach 20-23%, frequently with androgen receptor overexpression, underscoring a hormone-driven oncogenic . Epithelial-myoepithelial shows even higher , with HRAS mutations in up to 85% of cases, serving as a diagnostic hallmark across histologic variants.

Therapeutic Targeting

Historical Inhibitors

Early efforts to therapeutically target HRAS focused on farnesyltransferase inhibitors (FTIs), a class of small molecules developed in the late and extensively tested in phase II clinical trials during the 2000s. Exemplars include tipifarnib (R115777) and (SCH 66336), which competitively inhibit farnesyltransferase, the enzyme responsible for attaching a 15-carbon farnesyl isoprenoid to the residue in HRAS's C-terminal CAAX motif. This is crucial for HRAS's anchoring to the inner plasma membrane, enabling its interaction with upstream activators like receptor tyrosine kinases and downstream effectors in signaling cascades. By blocking farnesylation, FTIs prevent HRAS membrane localization, thereby attenuating its oncogenic activity specifically in HRAS-dependent cells. Clinical evaluation of FTIs in the in unselected advanced solid tumors showed modest antitumor efficacy, with partial responses reported in approximately 10-20% of patients across indications like and head and cancers. For instance, a 2003 phase II study evaluated tipifarnib in 76 patients with , demonstrating partial responses in 14% (5/35) of the intermittent dosing cohort, highlighting limited but observable activity in hormone-resistant disease. Later HRAS-selected trials, such as a 2015 phase II in head and squamous cell carcinoma (HNSCC), reported higher objective response rates of 50% in small cohorts (n=18). These results underscored FTIs' potential in HRAS-driven contexts but fell short of expectations for broad approval in . Despite HRAS specificity, FTIs largely failed to deliver transformative outcomes in cancer due to compensatory mechanisms, particularly alternative via geranylgeranylation for and NRAS isoforms, which allowed these mutants to evade inhibition and sustain membrane association. This bypass pathway rendered FTIs ineffective in the majority of RAS-mutant cancers, where predominates, limiting their potency to HRAS-enriched subsets like certain head and neck tumors. Lonafarnib, while unsuccessful in oncology trials, found a non-cancer application; it received FDA approval in 2020 for treating Hutchinson-Gilford progeria syndrome, where it inhibits farnesylation of the mutant lamin A precursor to improve survival by about 2.5 years in affected children.

Emerging Therapies

Recent advances in HRAS-targeted therapies have focused on mutation-specific inhibitors and strategies to overcome limitations of earlier approaches. Tipifarnib, a farnesyltransferase , has shown promising results in HRAS-mutant head and neck (HNSCC). In the phase II AIM-HN study, tipifarnib achieved an objective response rate (ORR) of 30% by investigator assessment (95% CI: 18.1-44.9) in 59 patients with recurrent or metastatic HRAS-mutant HNSCC, including one complete response, with a disease control rate of 48%. These findings, reported in 2023, support tipifarnib as a potential first for this rare subset, comprising 4-8% of HNSCC cases; the (NDA) was accepted by the FDA on June 1, 2025, with a (PDUFA) target action date of November 30, 2025. Direct inhibitors targeting HRAS have emerged, leveraging strategies from KRAS-targeted agents. , originally developed as a G12C inhibitor, demonstrates potent inhibition of HRAS G12C mutants with comparable efficacy to G12C, binding irreversibly to the switch pocket and suppressing downstream ERK signaling in preclinical models. Although HRAS G12C mutations are rare (occurring in <1% of cancers), this pan-RAS G12C activity expands its potential utility. Additionally, SOS1 degraders, such as bifunctional CRBN-based PROTACs, have shown preclinical efficacy by reducing active HRAS levels alongside and NRAS in RAS-mutant cell lines and xenografts, leading to >90% target degradation and significant tumor growth inhibition, including up to 50% reduction in some models resistant to direct inhibitors. These degraders address reactivation of wild-type RAS isoforms, a common resistance mechanism. Immunotherapies face challenges in HRAS-mutant tumors, where mutations correlate with reduced response to PD-1 blockade due to immunosuppressive microenvironments and lower . Combination approaches are under investigation, including MEK inhibitors with PD-1 inhibitors to enhance antitumor immunity; for instance, trametinib pretreatment sensitizes /HRAS-driven lung adenocarcinoma models to PD-1 blockade, prolonging survival by modulating T-cell infiltration. Ongoing trials, such as those evaluating MEK/PD-1 combinations in RAS-mutant solid tumors (e.g., NCT05440942), report preliminary immune activation without excessive toxicity as of 2025. Looking ahead, functional screens of rare HRAS variants presented at the 2025 AACR meeting classified 57 HRAS variants from COSMIC, identifying 15 novel oncogenic drivers and guiding variant-specific therapies through enhanced understanding of signaling potency. Pan-RAS inhibitors like RMC-6236, a RAS(ON) tri-complex agent targeting mutant and wild-type HRAS, NRAS, and , have advanced to phase III trials by 2025, demonstrating broad antitumor activity in preclinical HRAS models and potential to circumvent isoform-specific resistance.

References

  1. [1]
    Gene ResultHRAS HRas proto-oncogene, GTPase [ (human)] - NCBI
    Sep 27, 2025 · This gene belongs to the Ras oncogene family, whose members are related to the transforming genes of mammalian sarcoma retroviruses.
  2. [2]
    Human RAS Superfamily Proteins and Related GTPases - PMC
    For KRAS (67, 68) and HRAS (69), the primary function of alternate splicing may be to generate isoforms with distinct subcellular localizations.
  3. [3]
    The frequency of Ras mutations in cancer - PMC - PubMed Central
    We find that ~19% of cancer patients harbor Ras mutations; equivalent to ~3.4 million new cases per year worldwide.
  4. [4]
  5. [5]
    Worldwide Prevalence and Clinical Characteristics of RAS ...
    The estimated mutation rate is highest for HRAS (7%), followed by KRAS (2.89%) and NRAS (2.20%). HRAS prevalence in South Asia (15.28%) is twice as high as the ...
  6. [6]
    HRAS Mutations Define a Distinct Subgroup in Head and Neck ... - NIH
    Jan 5, 2023 · Mutations in oncogenic HRAS are rare (3%) in patients with HNSCC and occur more frequently in human papillomavirus (HPV)–negative tumors, with ...
  7. [7]
    Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene - Nature
    - **Abstract**: Examination of homologies between retroviral oncogenes and transforming sequences reveals that the human bladder carcinoma (EJ) oncogene is homologous to the Harvey sarcoma virus oncogene (ras). Structural analysis limits the region of homology to a 3.0-kilobase SacI fragment of the EJ oncogene. Both EJ and ras DNA probes detect similar transcripts in transfectants derived from bladder carcinoma cell lines.
  8. [8]
    A point mutation is responsible for the acquisition of transforming ...
    Nov 11, 1982 · Mariano Barbacid. Show authors. Nature volume 300, pages 149–152 (1982)Cite this article. 3427 Accesses. 1227 Citations. 28 Altmetric. Metrics ...Missing: cloning | Show results with:cloning
  9. [9]
    Entry - *190020 - HRAS PROTOONCOGENE, GTPase - OMIM
    HGNC Approved Gene Symbol: HRAS. Cytogenetic location: 11p15.5 ... Chromosomal localization of three human ras genes by in situ molecular hybridization.
  10. [10]
    Activation of the T24 bladder carcinoma transforming gene is linked ...
    Dec 23, 1982 · The transforming gene found in a human bladder carcinoma cell line, T24, is H-ras-1, the human homologue of the Harvey sarcoma virus oncogene (vH-ras).
  11. [11]
    The Nobel Prize in Physiology or Medicine 1989 - NobelPrize.org
    Michael Bishop and Harold E. Varmus "for their discovery of the cellular origin of retroviral oncogenes". To cite this section. MLA style: The Nobel Prize in ...Missing: 2000 HRAS<|control11|><|separator|>
  12. [12]
  13. [13]
    Start Site Selection by Sp1 in the TATA-less Human Ha-ras Promoter
    Feb 18, 1994 · The promoter of the human Ha-ras gene is very GC-rich, has four Sp1-binding sites and no TATA element. Transcription initiates at multiple start sites.Missing: AP- | Show results with:AP-
  14. [14]
    Binding of the Sp1 transcription factor by the human Harvey ras1 ...
    Footprinting analysis with deoxyribonuclease I was used to show that Sp1 binds to six GC box sequences within the c-Ha-ras1 promoter.Missing: TATA AP- 1
  15. [15]
    Tissue expression of HRAS - Summary - The Human Protein Atlas
    Tau specificity score is a numerical indicator of the specificity of the gene expression across cells or tissues. The value ranges from 0 and 1, where 0 ...
  16. [16]
    Functional Specificity of Ras Isoforms: So Similar but So Different
    In mice, the level of H-ras transcripts is highest in the brain, muscle, and skin and lowest in the liver; K-ras transcripts are more abundant in the gut, lung, ...
  17. [17]
    Deregulation of the Ras-Erk Signaling Axis Modulates the Enhancer ...
    Aug 25, 2015 · Unrestrained Ras-Erk signaling deregulates histone marking at active enhancers. Sprouty loss and oncogenic HRas G12V alter distinct enhancers and target genes.
  18. [18]
    Gene expression signatures modulated by epidermal growth factor ...
    May 1, 2012 · Isogenic HaCaT variants overexpressing either wild-type EGFR or the transcription factor NF-kappa-B p65 subunit (p65), or mutant HRAS have ...
  19. [19]
    miR-143-3p: A Critical Regulator of KRAS and HRAS with Potential ...
    Aug 10, 2025 · therapeutic target for improving outcomes in aggressive breast cancer subtypes. Keywords: Breast cancer, Gene regulation, HRAS, KRAS, MicroRNA, ...
  20. [20]
    Alternative polyadenylation is a determinant of oncogenic Ras function
    Dec 17, 2021 · Alternative polyadenylation of mRNA has important but poorly understood roles in development and cancer. Activating mutations in the Ras ...
  21. [21]
    Oncogenic N-Ras Stimulates SRF-Mediated Transactivation via H3 ...
    In addition, oncogenic H-Ras regulates CBP and Tip60, histone acetyltransferases, which modulate histone acetylation at local and global levels [18], and ...
  22. [22]
    DNA methylation of coding and non-coding regions of the human H ...
    The majority of the tissues showed an intermediate level of methylation, and a certain tissue specificity emerged by comparing tissues of the same and different ...
  23. [23]
    GTPase HRas - Homo sapiens (Human) | UniProtKB | UniProt
    GTPase HRas. EC number. EC:3.6.5.2 (UniProtKB | ENZYME | Rhea ) 1 publication. Alternative names. H-Ras-1; Ha-Ras; Transforming protein p21; c-H-ras; p21ras.
  24. [24]
    A Structure is Worth a Thousand Words: New Insights for RAS and ...
    Although all four RAS isoforms share high sequence and structural homology in the GTPase domain (G-domain; 1–166), they differ significantly in the last 20 ...
  25. [25]
    Structural Insights into the Regulation Mechanism of Small GTPases ...
    The G domain consists of a six-stranded β-sheet and 5 α-helices and has conserved sequence motifs called the G1, G2, G3, G4, and G5 motifs (G boxes), which are ...<|control11|><|separator|>
  26. [26]
    Posttranslational Modifications of RAS Proteins - PMC
    Upon palmitoylation, RAS acquires a 100-fold greater affinity for membranes than that of the prenylated-only protein (Shahinian and Silvius 1995; Schroeder et ...
  27. [27]
    Targeting the Ras palmitoylation/depalmitoylation cycle in cancer
    The remaining three proteins, H-Ras, N-Ras, and K-Ras4A, are modified by palmitoylation of cysteines proximal to the C-terminal cysteinyl site of farnesylation ...
  28. [28]
    Three-dimensional structures of H-ras p21 mutants: Molecular basis ...
    Aug 10, 1990 · Article. Three-dimensional structures of H-ras p21 mutants: Molecular basis for their inability to function as signal switch molecules.
  29. [29]
    1QRA: STRUCTURE OF P21RAS IN COMPLEX WITH GTP AT 100 K
    Nov 5, 1999 · Structure 1QRA is the structure of P21RAS in complex with GTP at 100K, determined by X-ray diffraction at 1.60 Å resolution.
  30. [30]
    Regulation of Small GTPases by GEFs, GAPs, and GDIs
    The GTPase switch is turned on by GEFs, which stimulate dissociation of the tightly bound GDP, and turned off by GAPs, which accelerate the intrinsically ...
  31. [31]
    Dynamic regulation of RAS and RAS signaling | Biochemical Journal
    Jan 6, 2023 · The signaling dynamics of RAS proteins is thus regulated by three types of rates: (i) GTP hydrolysis, (ii) nucleotide binding/unbinding and (iii) ...
  32. [32]
    The arginine finger of RasGAP helps Gln-61 align the nucleophilic ...
    Mutations of Gln-61 have been found that reduce the rate of GAP-stimulated hydrolysis by as much as 106-fold (17, 18), which corresponds to an increase in the ...Missing: HRAS enhancement
  33. [33]
    Ras and GTPase-activating protein (GAP) drive GTP into a ... - PNAS
    Sep 4, 2012 · Ras proteins accelerate GTP hydrolysis by a factor of 10 5 compared to GTP in water. GTPase-activating proteins (GAPs) accelerate hydrolysis by another factor ...
  34. [34]
    Plasma membrane regulates Ras signaling networks - PMC - NIH
    Interaction with membrane-anchored GTP-bound Ras relieves the autoinhibition. RBD at the N-terminal of Raf interacts with Ras, swinging the CRD away, and ...
  35. [35]
    RAF inhibitors promote RAS-RAF interaction by allosterically ... - NIH
    Oct 31, 2017 · Under physiological conditions, RAS-GTP binding is the primary trigger implementing the relief of autoinhibition, which subsequently leads to ...
  36. [36]
    Ras-Mediated Activation of the Raf Family Kinases - PMC
    The Raf kinases interact directly with active, membrane-localized Ras, and this interaction is often the first step in the Raf activation process.
  37. [37]
    Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR ... - PMC - NIH
    The Ras/Raf/MEK/ERK and Ras/PI3K/PTEN/Akt/mTOR pathways can be activated by mutations/amplifications of upstream growth factor receptors. The abnormal ...
  38. [38]
    ROLES OF THE RAF/MEK/ERK PATHWAY IN CELL GROWTH ...
    Growth factors and mitogens use the Ras/Raf/MEK/ERK signaling cascade to transmit signals from their receptors to regulate gene expression and prevent ...
  39. [39]
    ERK MAP kinase in G1 cell cycle progression and cancer - PMC - NIH
    Activation of the Ras/ERK signaling pathway in cell cycle progression from G0/G1 to S phase has been linked to cyclin D induction and consequent retinoblastoma ...
  40. [40]
    Down-regulation of c-Myc following MEK/ERK inhibition halts the ...
    Both the c-Myc and Ras/MEK/ERK pathways play an important role in the progression of the G1-cell cycle phase by enhancing cyclins expression [13,14] and CDK/ ...
  41. [41]
    Extracellular Regulated Kinases: Signaling from Ras to ERK ... - NIH
    Further upstream on the pathway, ERK phosphorylates the Ras GEF Sos1. Initial studies had shown that Sos1 became phosphorylated in response to growth factor ...
  42. [42]
    RSK phosphorylates SOS1 creating 14-3-3 docking sites and ... - NIH
    SOS1 phosphorylation by RSK inhibits EGF-induced MAPK activation. We next ... In stage one of the negative feedback loop on SOS1, ERK phosphorylates ...The Ras-Mapk Pathway... · Rsk Phosphorylates Sos1 At... · Sos1 Phosphorylation By Rsk...
  43. [43]
    Progress on Ras/MAPK Signaling Research and Targeting in Blood ...
    Della Rocca et al. found that stimulation of α1B or α2A adrenergic receptors triggered these pathways to cause a rapid 5–10-fold increase in ERK phosphorylation ...2. Activation Of Ras And Raf... · 3. Ras-Raf Pathway Mutations · 3.1. Ras Mutations<|control11|><|separator|>
  44. [44]
    RAS Interaction with PI3K: More Than Just Another Effector Pathway
    RAS proteins interact with PI3K, a main effector pathway, regulating cell growth, cell cycle, and survival. This interaction is direct and essential for RAS- ...
  45. [45]
  46. [46]
    The RASopathies: from pathogenetics to therapeutics
    Feb 18, 2022 · The RASopathies are a group of disorders caused by a germline mutation in one of the genes encoding a component of the RAS/MAPK pathway.
  47. [47]
    The RASopathies - PMC - PubMed Central - NIH
    The RASopathies are a clinically defined group of medical genetic syndromes caused by germline mutations in genes that encode components or regulators of the ...
  48. [48]
  49. [49]
    Germline and sporadic cancers driven by the RAS pathway
    This review examines the parallels in pathological and genetic details of RASopathies and somatic RAS-driven cancers.
  50. [50]
    Worldwide Prevalence and Clinical Characteristics of RAS ...
    Mutations in RAS family members are well-established drivers of cancer. Gain-of-function mutations in RAS genes are found in ∼19% of human cancers, most ...
  51. [51]
    A comprehensive survey of Ras mutations in cancer - PMC
    The COSMIC dataset confirms that K-Ras is the most frequently mutated isoform present in 22% of all tumours analysed compared to 8% for N-Ras and 3% for H-Ras ( ...
  52. [52]
    RAS mutational frequency and hotspots in human cancer. A The ...
    G12, G13, and Q61 occupy 96-98% of all mutations in KRAS and NRAS isoforms. The data are taken from the Catalogue of Somatic Mutations in Cancer (COSMIC v94).<|separator|>
  53. [53]
    HRAS Mutations Define a Distinct Subgroup in Head and Neck ...
    Oncogenic mutations in HRAS occur in 3%-4% of HNSCC, with G12S being the most frequent. Without targeted therapy, patients with HRAS-mutant HNSCC had poor ...Missing: prevalence | Show results with:prevalence
  54. [54]
    Emerging strategies to target RAS signaling in human cancer therapy
    Jul 23, 2021 · Here we fully review RAS mutations in human cancer and especially focus on emerging strategies that have been recently developed for RAS-targeting therapy.
  55. [55]
    Duplication of Glu37 in the switch I region of HRAS impairs effector ...
    Mar 1, 2010 · Duplication of Glu37 in the switch I region of HRAS impairs effector/GAP binding and underlies Costello syndrome by promoting enhanced growth ...
  56. [56]
    The Genomic, Transcriptomic, and Immunologic Landscape of ...
    The frequency of somatic HRAS mutations in solid tumors has been estimated to be between 1.0 and 6.9% [9,10] depending on the cohort of patients studied.<|separator|>
  57. [57]
    FGFR3, HRAS, KRAS, NRAS and PIK3CA Mutations in Bladder ...
    KRAS and HRAS mutations occurred with equal frequency, whereas NRAS mutations were not frequent in bladder cancer.
  58. [58]
    H-ras gene mutations in salivary gland mucoepidermoid carcinomas
    Results: Mutated H-ras genes were detected in 9 patients, for an overall incidence of 18% (9 of 50 patients). All but 1 of the mutations occurred at codon 12: a ...Missing: bladder | Show results with:bladder
  59. [59]
    The immunologic landscape of HRAS-mutant head and neck ...
    Aug 12, 2025 · Based on TCGA HNSCC mutational and mRNA expression data, we found that 6% of HNSCC cases harbor HRAS mutations. · Transcriptome analysis showed ...
  60. [60]
    Regulation of nucleotide excision repair by wild-type HRAS ... - Nature
    Apr 12, 2025 · Our findings indicate that in a specific cluster of head and neck tumors, ERK/cJun signaling activation is strongly reliant on HRAS activity.
  61. [61]
    HRAS-Related Costello Syndrome - GeneReviews® - NCBI Bookshelf
    Aug 29, 2006 · Kratz et al [2011] reviewed published cases and confirmed the 15% cumulative incidence of cancer in individuals with Costello syndrome by age ...Diagnosis · Clinical Characteristics · Management · Genetic Counseling
  62. [62]
    Costello syndrome: Clinical phenotype, genotype, and management ...
    HRAS mutations associated with CS result in a gain-of-function, which causes constitutive activation of the RAS protein (Aoki et al., 2005) or more complex ...
  63. [63]
    HRAS mutation analysis in Costello syndrome: Genotype and ...
    Dec 2, 2005 · Our results confirm that germline HRAS mutations cause Costello syndrome in most patients [Aoki et al., 2005]. All mutations occurred de novo ...
  64. [64]
    RASopathies: unraveling mechanisms with animal models
    Aug 1, 2015 · RASopathies are developmental disorders caused by germline mutations in the Ras-MAPK pathway, and are characterized by a broad spectrum of ...
  65. [65]
    Multidisciplinary Management of Costello Syndrome - PubMed Central
    Jun 2, 2022 · Antihistaminic oral therapy and topical treatment with moisturizing to improve patient' discomfort due to continuous itching are often necessary ...
  66. [66]
    Costello Syndrome - Symptoms, Causes, Treatment | NORD
    Jan 4, 2017 · Costello syndrome is an extremely rare disorder that affects multiple organ systems of the body. This condition is characterized by growth delays after birth.
  67. [67]
    Tipifarnib as a Precision Therapy for HRAS-Mutant Head and Neck ...
    Tipifarnib is a potent and highly selective inhibitor of farnesyltransferase (FTase). FTase catalyzes the posttranslational attachment of farnesyl groups to ...Missing: lonafarnib | Show results with:lonafarnib
  68. [68]
    Tipifarnib Shows Potential in HRAS-Mutant Head and Neck Cancers
    Nov 4, 2020 · Tipifarnib is an FTI initially studied extensively in the late 1990s and early 2000s but did not achieve response rates robust enough to support ...Missing: II | Show results with:II
  69. [69]
    Dual farnesyl and geranylgeranyl transferase inhibitor thwarts ... - NIH
    However, KRAS becomes geranylgeranylated and active when cancer cells are treated with FTIs. To overcome this geranylgeranylation-dependent resistance to FTIs ...
  70. [70]
    FDA approval summary for lonafarnib (Zokinvy) for the treatment of ...
    The U.S. Food and Drug Administration recently approved lonafarnib as the first treatment for Hutchinson-Gilford progeria syndrome (HGPS) and processing- ...
  71. [71]
  72. [72]
    Kura Oncology Announces Positive Results from Registration ...
    Oct 17, 2023 · As of the data cutoff on June 15, 2023 , 59 patients with R/M HRAS mutant HNSCC were enrolled, of whom 50 had high HRAS mutant variant allele ...
  73. [73]
    Sotorasib Is a Pan-RAS G12C Inhibitor Capable of Driving Clinical ...
    May 1, 2024 · Our data also demonstrate that sotorasib can inhibit HRASG12C with similar potency to KRASG12C. Although HRASG12C mutations are rare and no ...Missing: analogous | Show results with:analogous
  74. [74]
    Development of bifunctional CRBN-SOS1 degraders for treatment of ...
    May 31, 2023 · SOS1 degradation resulted in the reduction of active KRAS, HRAS and MRAS levels when assayed by RAF-RBD pulldowns in MIA PaCa-2 (KRAS G12C) ...
  75. [75]
    Targeted Degradation of SOS1 Exhibits Potent Anticancer Activity ...
    Jan 2, 2025 · Targeted degradation of SOS1 exhibits potent anticancer activity and overcomes resistance in KRAS-mutant tumors and BCR–ABL–positive leukemia.
  76. [76]
    Trametinib sensitizes KRAS-mutant lung adenocarcinoma tumors to ...
    Apr 20, 2024 · We found that trametinib synergizes with PD-1/PD-L1 blockade to hamper lung cancer progression and increase survival.Missing: NCT04592211 trial
  77. [77]
  78. [78]
    Evolution of direct RAS inhibitors: from undruggable target to clinical ...
    Sep 24, 2025 · HRAS mutations are less common but appear in cancers such as bladder cancer and head and neck squamous cell carcinomas (HNSCCs), while NRAS ...Evolution Of Kras Inhibitors... · Targeted Protein Degraders... · Targeting Kras Mutation