Fact-checked by Grok 2 weeks ago

Insulin-like growth factor 1 receptor

The insulin-like growth factor 1 receptor (IGF1R) is a transmembrane receptor tyrosine kinase that plays a central role in mediating the biological effects of insulin-like growth factors (IGFs), particularly IGF-1 and IGF-2, by binding them with high affinity and activating downstream signaling pathways essential for cell growth, proliferation, differentiation, survival, and metabolism. Encoded by the IGF1R gene on human chromosome 15q26.3, it forms a heterotetrameric structure consisting of two extracellular α-subunits responsible for ligand binding and two transmembrane β-subunits containing the intracellular tyrosine kinase domain. Expressed ubiquitously from the oocyte stage through adulthood, with notably high levels in tissues such as the kidney and ovary, IGF1R is critical for embryonic development and postnatal growth, as evidenced by severe growth retardation and developmental defects in mice with disrupted Igf1r genes. Upon ligand binding, IGF1R undergoes dimerization and autophosphorylation of its β-subunits, triggering multiple intracellular signaling cascades, including the phosphatidylinositol 3-kinase (PI3K)/AKT pathway for anti-apoptotic and metabolic effects, the /mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway for and differentiation, and the ()/ () pathway. IGF1R can also form hybrid receptors with the (), which bind IGFs and insulin with altered affinities, further modulating signaling outputs in tissues like muscle and fat. In some contexts, activated IGF1R translocates to the , where it influences gene transcription independently of cytoplasmic pathways. These mechanisms underscore IGF1R's role in maintaining , as its knockout in animal models leads to and perinatal lethality. Dysregulation of IGF1R, most commonly through overexpression or hyperactivation, is strongly associated with oncogenesis and tumor progression in multiple malignancies, including , , , and colorectal cancers, where it promotes cell survival, , and resistance to by suppressing . Germline mutations in IGF1R cause rare growth disorders such as Desbuquois dysplasia type 2 and severe intrauterine growth retardation due to IGF-1 resistance, highlighting its non-redundant function in linear growth. Therapeutically, IGF1R has been targeted with monoclonal antibodies (e.g., figitumumab, ganitumab) and small-molecule inhibitors in clinical trials, showing promise in combination regimens for advanced solid tumors despite challenges like pathway crosstalk and feedback activation.

Structure

Subunit Composition and Assembly

The insulin-like growth factor 1 receptor (IGF1R) is a heterotetrameric transmembrane receptor tyrosine kinase composed of two extracellular α-subunits linked by disulfide bonds and two β-subunits that span the plasma membrane. The α-subunits are entirely extracellular and primarily responsible for ligand recognition, while the β-subunits contain the intracellular tyrosine kinase domains essential for signal transduction. IGF1R is synthesized as a single-chain 1367-amino acid precursor pro-receptor, which undergoes post-translational processing including removal and in the . Furin-mediated proteolytic at a specific site (between residues 740 and 741 of the precursor) generates the mature α-subunit (approximately 710 residues, encompassing residues 31-740 of the precursor) and β-subunit (approximately 627 residues, residues 741-1367). This occurs after and is necessary for the receptor to achieve its functional configuration. The functional receptor assembles into an α₂β₂ heterotetramer through a combination of disulfide bridges and non-covalent interactions, with intra-monomer s linking each α to its paired β-subunit and inter-monomer s connecting the two α-subunits. This assembly begins with dimerization of the pro-receptor monomers in the , followed by cleavage and maturation in the Golgi, ensuring proper binding and activation. The heterotetrameric structure is critical for high-affinity interaction and downstream signaling. Structural studies of the IGF1R ectodomain, such as the crystal structure of its first three domains (PDB entry 1IGR), reveal a modular that contributes to the overall V-shaped conformation of the assembled ectodomain in higher-resolution models. This V-shaped arrangement positions the ligand-binding sites at the apex, facilitating dimerization and activation upon engagement.

Domain Organization and Ligand Binding

The insulin-like growth factor 1 receptor (IGF1R) is a transmembrane composed of distinct domains that contribute to its structural integrity and function. The extracellular region, spanning approximately the first 900 residues, is organized into several modules: two domains (L1 and L2), a (CR), and three type III domains (FnIII-1, FnIII-2, and FnIII-3). The L1 domain (residues 1–150) forms a compact β-helix structure with that participates in recognition, while the L2 domain (residues 300–460) adopts a similar fold but plays a lesser direct role in binding. The CR (residues 152–298) consists of 11 disulfide-linked modules stabilized by 22 residues, bridging L1 and L2 and contributing critically to the receptor's specificity. The FnIII domains (residues 461–901) facilitate interdomain interactions and include the site for proteolytic cleavage into α- and β-subunits, with FnIII-2 containing an insert domain. The (residues 906–929) comprises a single α-helical span that anchors the receptor in the plasma membrane and supports preformed dimerization essential for its activity. Intracellularly, the β-subunit features a juxtamembrane region, a domain (TK; residues 973–1229), and a C-terminal tail. The TK domain exhibits a bilobal typical of receptor kinases, with an activation loop containing key residues (Y1161, Y1165, and Y1166) that undergo autophosphorylation to regulate kinase activity. The C-terminal tail (beyond residue 1229) includes additional autophosphorylation sites, such as Y1250 and Y1251, which serve as docking platforms for downstream effectors. Ligand binding occurs primarily through the extracellular α-subunits, with high-affinity interaction sites formed by the L1, , and domains of one protomer in conjunction with the α-C-terminal segment and FnIII-1 of the opposing protomer. IGF-1 binds with high affinity (Kd ≈ 0.1 ), engaging residues in the L1- groove and inducing structural rearrangements that separate the β-subunits. IGF-2 exhibits lower affinity (approximately 10–50-fold reduced compared to IGF-1), while insulin binds with even lower affinity (about 100-fold less than IGF-1), underscoring the receptor's specificity despite among the ligands. These binding events rely on the dimeric assembly of the receptor, with the and L1 domains providing the primary contacts for ligand specificity.

Receptor Family

Insulin Receptor Superfamily

The insulin-like growth factor 1 receptor (IGF1R) is a receptor tyrosine kinase (RTK) that belongs to the insulin receptor (IR) superfamily, a distinct group within the broader RTK family characterized by their unique preformed dimeric architecture. Encoded by the IGF1R gene located on chromosome 15q26.3 in humans, IGF1R shares this classification with other members due to its role in transducing signals for growth and metabolic regulation through tyrosine kinase activity. This superfamily is often grouped under RTK class II, emphasizing their evolutionary divergence from other RTK classes that typically dimerize only upon ligand binding. The core members of the IR superfamily include the (IR), encoded by the INSR gene on 19p13.2; IGF1R itself; and the insulin receptor-related receptor (IRR), which has more restricted expression and an unidentified physiological . Additionally, hybrid receptors form through heterodimerization between these members, such as IR/IGF1R hybrids, which combine α and β subunits from different receptors and exhibit altered ligand affinities that influence both metabolic and mitogenic signaling. These hybrids are prevalent in tissues where both IR and IGF1R are co-expressed, contributing to the superfamily's functional versatility. The IR superfamily arose from ancient gene duplications during early evolution, approximately 500 million years ago, coinciding with the two rounds of whole-genome duplication (2R ) at the base of s. A primary duplication event generated the ancestral IR lineage, followed by a secondary duplication that separated the progenitors of IGF1R and IRR, leading to their diversification while preserving high sequence conservation. For instance, IGF1R shares over 50% overall sequence identity with IR, rising to 84-85% in the intracellular domain, reflecting their common evolutionary origin. All superfamily members exhibit a conserved heterotetrameric structure composed of two extracellular α-subunits for binding and two transmembrane β-subunits containing the domain, activated via ligand-induced trans-autophosphorylation to mediate overlapping roles in cellular growth and .

Structural and Functional Homologs

The insulin-like growth factor 1 receptor (IGF1R) has structural and functional homologs in various species that underscore its evolutionary conservation within the receptor tyrosine kinase family, particularly in regulating growth and metabolism. In Drosophila melanogaster, the insulin-like receptor (dIR, also known as InR) serves as a key homolog, sharing approximately 40-50% sequence identity in the kinase domain with human IGF1R and playing a central role in nutrient sensing, growth control, and lifespan regulation through binding to Drosophila insulin-like peptides (DILPs). Similarly, in Caenorhabditis elegans, the daf-2 gene encodes a receptor tyrosine kinase that is the ortholog of IGF1R, exhibiting sequence homology in the kinase domain and mediating insulin/IGF-1-like signaling to influence dauer formation, reproductive development, and longevity, with reduced daf-2 activity extending lifespan by up to twofold. A notable non-tyrosine kinase relative is the insulin-like growth factor 2 receptor (IGF2R), also known as the cation-independent mannose-6-phosphate receptor (CI-MPR), which evolved from an ancestral mannose-6-phosphate receptor and binds IGF-2 with high affinity but lacks an intracellular domain, thereby functioning primarily as a scavenger receptor to internalize and degrade IGF-2, preventing its interaction with signaling receptors like IGF1R. This contrasts with IGF1R's role in , as IGF2R does not propagate growth-promoting signals and instead modulates IGF-2 to maintain tissue . Structural comparisons across these homologs reveal conserved features in ligand-induced conformational changes, with cryo-EM and crystal structures of apo (unliganded) and holo (-bound) forms showing similar transitions from an autoinhibited, T-shaped dimer to an active, λ-shaped conformation upon engagement, as observed in human IGF1R and dIR ectodomains. However, differences in ligand specificity are evident; for instance, dIR accommodates multiple DILPs with varying affinities due to its broader binding pocket in the L1 and cysteine-rich domains, unlike the more selective IGF-1 binding of human IGF1R. These shared structural motifs, including the leucine-rich repeats and type III domains, highlight evolutionary divergence while preserving core activation mechanisms. Functional divergences among homologs illustrate specialized adaptations beyond growth regulation. The insulin receptor-related receptor (IRR), a close paralog of IGF1R in mammals, primarily functions as an extracellular alkali sensor, activated by mildly alkaline (above 7.4) rather than peptide ligands, with pH-induced proton dissociation in the L1 domain triggering autophosphorylation and downstream signaling in tissues like the and that interface with external environments. This pH-sensing role in IRR contrasts sharply with IGF1R's focus on IGF-1-mediated anabolic processes, emphasizing how can lead to distinct physiological contexts within the superfamily.

Activation and Signaling

Ligand Binding and Receptor Activation

The insulin-like growth factor 1 receptor (IGF1R) is primarily activated by its cognate ligands, (IGF-1) and (IGF-2). IGF-1 serves as the main , exhibiting high binding affinity (Kd ≈ 0.1–1 nM) and produced predominantly by the liver as an endocrine , with additional local synthesis in various tissues such as muscle and . In contrast, IGF-2 predominates during fetal development, with circulating levels 3–10 times higher than IGF-1 in the , and displays lower affinity for IGF1R (Kd ≈ 10–100 nM) compared to IGF-1, though it binds with high affinity to the isoform A (IR-A). Structures reveal polymorphic ligand binding stoichiometries for IGF1R, including asymmetric 1:1 and symmetric 2:1 :receptor ratios under saturating conditions, where ligands interact with the α-subunits to induce activation. Ligand engagement with the extracellular α-subunits triggers a series of conformational changes that relieve autoinhibition and activate the intracellular domains of the β-subunits. In the inactive state, the α-subunits are tethered in close proximity, constraining the β-subunits and preventing activity; upon binding, the α-subunits separate, allowing the transmembrane and intracellular regions to extend into an active configuration. This separation facilitates trans-autophosphorylation between the β-subunits, primarily at three conserved residues in the activation loop (Tyr1131, Tyr1135, and Tyr1136 in the human sequence), located in the juxtamembrane and domains. at these sites stabilizes the active conformation, enhancing catalytic activity by up to 100-fold. Cryo-electron (cryo-EM) have elucidated these , revealing a transition from a compact, autoinhibited "tethered" state to an extended, asymmetric "Γ-shaped" active form upon IGF-1 . For instance, the of the full-length IGF1R in complex with IGF-1 (PDB: 6PYH) at 4.3 shows asymmetric 1:1 , with the occupying a single formed by the L1 and cysteine-rich of one α-subunit and the C-terminal of the other, promoting α-subunit dissociation and β-subunit dimerization for trans-phosphorylation. A 2022 of IGF1R (PDB: 7YRR) further demonstrates symmetric 2:1 at site 2 epitopes. This asymmetric in 1:1 complexes reflects negative , where initial occupancy reduces for a second , though 2:1 indicates context-dependent activation. IGF1R can also form hybrid receptors with the structurally related (IR), influencing ligand specificity and activation profiles. These hybrids, composed of one IR αβ and one IGF1R αβ , include IR-A/IGF1R and IR-B/IGF1R variants, where IR-A is the exon-11-minus fetal isoform and IR-B is the exon-11-plus metabolic isoform. IR-A/IGF1R hybrids exhibit higher affinity for insulin (Kd ≈ 0.3 nM) compared to homotypic IGF1R (Kd > 100 nM), conferring greater insulin sensitivity, while both hybrid types respond preferentially to IGF-1 (approximately 20- to 50-fold higher potency than insulin) over IGF-2. In contrast, IR-B/IGF1R hybrids show reduced insulin binding, altering tissue-specific signaling in contexts like fetal growth and cancer.

Downstream Signaling Pathways

Upon ligand-induced dimerization and autophosphorylation of the insulin-like growth factor 1 receptor (IGF1R), primarily at residues Tyr1131, Tyr1135, and Tyr1136 in the activation loop, the receptor recruits key adaptor proteins to initiate intracellular signaling. These include the insulin receptor substrates IRS-1 and IRS-2, which bind via their phosphotyrosine-binding domains to the phosphorylated juxtamembrane region (notably Tyr950), and the adaptor protein Shc, which also docks at Tyr950. Additionally, binds to phosphorylated IRS or Shc, facilitating downstream activation. The primary downstream pathways activated by IGF1R include the PI3K/Akt/mTOR cascade, which promotes cell survival and metabolism, and the MAPK/ERK pathway, which drives proliferation. In the PI3K/Akt/mTOR pathway, phosphorylated IRS-1/2 recruits the p85 regulatory subunit of PI3K, leading to activation of its p110 catalytic subunit and generation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3) at the plasma membrane. PIP3 then recruits and activates phosphoinositide-dependent kinase 1 (PDK1) and Akt (also known as PKB), with PDK1 phosphorylating Akt at Thr308 and mTORC2 at Ser473 for full activation; activated Akt subsequently stimulates mTORC1 to enhance protein synthesis and inhibit apoptosis. In parallel, the MAPK/ERK pathway is engaged through binding to phosphorylated Shc or IRS, recruiting the SOS to activate , which in turn stimulates the Raf-MEK-ERK kinase cascade, culminating in ERK1/2 and nuclear translocation to regulate for and division. IGF1R also activates additional pathways, such as via (JAK) , contributing to and anti-apoptotic effects, and Cγ (PLCγ), which hydrolyzes PIP2 to produce (IP3) and diacylglycerol (DAG), modulating calcium release and activity. IGF1R signaling exhibits crosstalk with the (IR), sharing adaptor proteins like IRS-1/2 and Shc, but IGF1R preferentially activates mitogenic pathways such as MAPK/ERK, whereas IR biases toward metabolic responses via enhanced PI3K/Akt signaling. This distinction arises from differential affinities for ligands and substrates, with hybrid IGF1R/IR receptors integrating signals in a context-dependent manner. To prevent excessive signaling, mechanisms attenuate pathway activity, including serine of IRS-1/2 by downstream effectors such as S6K1 (activated by ) or components of the MAPK cascade, which promotes IRS ubiquitination and proteasomal degradation, thereby dampening PI3K recruitment and Akt activation.

Physiological Functions

Role in Embryonic and Postnatal Development

The insulin-like growth factor 1 receptor (IGF1R) is indispensable for embryonic development, as demonstrated by studies in models. Homozygous Igf1r-null mice display severe intrauterine growth retardation, with birth weights reduced to approximately 45% of wild-type littermates (a 55% reduction), and die shortly after birth due to stemming from muscle , underdeveloped diaphragms, and immaturity. This phenotype highlights the non-redundancy of IGF1R with the (IR), as IR-null mice exhibit embryonic lethality from hyperglycemia and organ atrophy but lack the profound muscle and skeletal defects seen in IGF1R deficiency. The absence of IGF1R signaling disrupts , survival, and across multiple tissues, underscoring its central role in coordinating embryonic . In development, IGF1R promotes the and of myoblasts, which are critical precursors for muscle fiber formation. Activation of IGF1R by IGF-1 stimulates myoblast expansion during early and subsequently drives their withdrawal from the to fuse into multinucleated myotubes, enhancing muscle mass and function in the . Similarly, in , IGF1R signaling in chondrocytes of the growth plate facilitates , the terminal step where cells enlarge and secrete to enable longitudinal elongation. IGF-1 exerts insulin-like effects to augment this hypertrophic response, independent of , thereby supporting the structural integrity of developing bones. Postnatally, IGF1R sustains linear growth through the (GH)-IGF1 axis, in which pituitary-derived GH induces hepatic IGF-1 synthesis, and circulating IGF-1 binds IGF1R in peripheral tissues to promote activity in growth plates. This endocrine mechanism accounts for a substantial portion of postnatal body size increase, with IGF1R mediating local paracrine effects in proliferative zones. IGF1R expression is elevated in rapidly growing tissues such as the liver and during early postnatal life, correlating with heightened anabolic demands and facilitating the transition from embryonic to juvenile growth phases. In humans, IGF1R mutations are linked to (IUGR), characterized by and head circumference, followed by severe postnatal growth failure. Heterozygous loss-of-function mutations, such as those impairing receptor kinase activity, reduce IGF-1 responsiveness and result in persistent , often with birth weights below the third percentile and minimal catch-up growth. Compound heterozygous variants, including missense changes in the extracellular and domains, have been documented in cases of extreme pre- and postnatal growth impairment, emphasizing IGF1R's non-redundant contributions to human developmental growth.

Involvement in Metabolism and Lactation

The insulin-like growth factor 1 receptor (IGF1R) plays a key role in metabolic by facilitating and in and tissues. Upon ligand binding, IGF1R activates the IRS-1/PI3K/AKT signaling pathway, which promotes translocation of glucose transporters (such as ) to the , thereby enhancing insulin-stimulated in muscle cells. In , this pathway similarly supports glucose transport and drives de novo by upregulating enzymes like and , contributing to fat storage and energy balance. While IGF1R signaling overlaps with that of the (IR) in these metabolic functions, it is distinguished by a stronger emphasis on proliferative effects rather than purely anabolic ones, as evidenced by tissue-specific differences in receptor activation and downstream . In the context of lactation, IGF1R is essential for mammary epithelial cell function, particularly in driving alveolar and the synthesis of milk proteins such as β-casein. During and early , IGF1R expression is upregulated in the , where it synergizes with signaling to induce β-casein transcription via STAT5 activation and enhanced IGF-1 bioavailability. This interaction promotes alveolar differentiation and secretory capacity, ensuring efficient production; disruption of IGF1R signaling impairs these processes, leading to reduced β-casein expression and structural defects in alveolar architecture. Studies using conditional models in mice highlight IGF1R's integration of metabolic and lactational roles. Mammary-specific dominant-negative IGF1R transgenics exhibit reduced alveolar during , culminating in diminished milk protein synthesis and lower yield during , which correlates with altered maternal glucose tolerance due to impaired partitioning toward the . These findings link IGF1R-mediated signaling to overall maternal metabolic adaptations, such as enhanced in mammary tissue to support synthesis and energy demands of . In humans, parallels emerge in gestational physiology, where increased IGF1R is observed in placentas from pregnancies with gestational diabetes mellitus and , potentially promoting fetal growth through enhanced nutrient supply (particularly glucose and ). This upregulation enhances IGF1R in cells, promoting fetal nutrient supply but potentially exacerbating maternal if unchecked.

Regulation of Body Size and Aging

The insulin-like growth factor 1 receptor (IGF1R) plays a central role in regulating body size through the (GH)-IGF1 axis, where GH stimulates the liver and other tissues to produce IGF1, which then binds to IGF1R to promote growth and linear during and adulthood. Expression levels of IGF1R in various tissues directly correlate with overall body size, as demonstrated in mouse models where altered receptor abundance influences proportional growth. Genetic polymorphisms in the IGF1R gene, particularly in promoter regions, contribute to variation in populations, with specific variants associated with modest differences in stature across diverse cohorts. In aging research, reduced IGF1R signaling has been shown to extend lifespan in model organisms by activating downstream pathways that enhance resistance and cellular maintenance. In , mutations in daf-2, the homolog of IGF1R, approximately double the lifespan through activation of the DAF-16/FoxO , which promotes by upregulating genes involved in response and . This insulin/IGF-1 signaling pathway highlights IGF1R's evolutionarily conserved role in modulating aging processes. In mammals, diminished IGF1R activity similarly promotes , as seen in heterozygous Igf1r mice (Igf1r^{+/-}), which exhibit extended lifespan compared to wild-type controls, accompanied by improved healthspan markers such as reduced oxidative damage. Dwarf mouse models with disrupted GH-IGF1 axis signaling, including low IGF1R activation, display approximately 40% lifespan extension, mimicking the effects of caloric restriction by boosting and resistance to metabolic stress without compromising . Late-life reduction in IGF1R signaling further enhances these benefits, delaying age-related decline in physical function. Human studies link lower IGF1R signaling to exceptional , with rare variants in the IGF1R identified in centenarians that impair receptor function and attenuate IGF1-mediated pathways, potentially contributing to prolonged lifespan. Offspring of long-lived individuals often show signs of reduced IGF1R sensitivity, such as elevated IGF1 levels relative to body size, supporting a balanced role for moderated signaling in maintaining vitality into advanced age. However, optimal IGF1R activity is necessary for preserving muscle mass and overall physical , underscoring the importance of signaling equilibrium for healthy aging.

Pathological Roles

Association with Craniosynostosis and Growth Disorders

Overactive signaling through the insulin-like growth factor 1 receptor (IGF1R) contributes to by accelerating the fusion of calvarial sutures through enhanced and . In calvarial s derived from patients with single-suture (SSC), activation of the IGF1 pathway increases cellular contractility and motility, promoting premature suture closure. This pathway's dysregulation correlates with craniofacial defects in mouse models, where IGF1 overexpression leads to skeletal abnormalities including premature suture fusion via heightened activity. Rare genetic variants in IGF1R have been identified in families with isolated , particularly affecting single sutures such as the sagittal or coronal. A resequencing study of 130 SSC cases identified five rare missense variants in IGF1R, including three novel ones, that were significantly enriched compared to controls and segregated with the in affected families. These variants likely enhance IGF1R signaling, contributing to suture pathogenesis, and in some instances present with features resembling Saethre-Chotzen syndrome but without mutations in the TWIST1 gene. IGF1R mutations are also linked to various growth disorders, with inactivating variants causing syndromes characterized by intrauterine and postnatal growth failure. Heterozygous loss-of-function , such as nonsense or frameshift changes in the , impair IGF1 signaling, resulting in and often elevated serum IGF-1 levels. Recent studies as of 2025 have identified novel IGF1R variants associated with , further delineating phenotypic variability. Conversely, gain-of-function alterations, including increased IGF1R copy number from 15q terminal duplications that encompass IGF1R, have been observed in overgrowth syndromes featuring tall stature and , though the specific contribution of IGF1R remains uncertain. In conditions like Beckwith-Wiedemann syndrome, upregulation of IGF2 can activate IGF1R signaling, contributing to overgrowth.

Effects of Gene Inactivation or Deletion

Inactivation or deletion of the IGF1R gene in model organisms reveals profound disruptions in growth and development, underscoring its essential role in prenatal and postnatal . Homozygous null of Igf1r in mice results in perinatal , with embryos displaying a 45% reduction in body weight at birth compared to wild-type littermates, accompanied by severe muscle hypotrophy, delayed , and abnormalities in development such as a thin and impaired dermal proliferation. These defects arise from impaired IGF-1 signaling, leading to shortly after birth. In contrast, heterozygous Igf1r null mice are viable but exhibit a consistent 20% reduction in adult body size and weight, with no overt or major organ malformations, highlighting a effect on somatic growth. Tissue-specific deletions of Igf1r further delineate its localized functions while revealing organ-specific vulnerabilities. Liver-specific knockout of Igf1r in mice does not substantially alter circulating IGF-1 levels, as hepatic IGF-1 production is primarily regulated by via the rather than autocrine IGF1R signaling. Neuronal-specific deletion of Igf1r, particularly in the and , disrupts and expression, resulting in impaired spatial learning, memory deficits, and increased anxiety-like behaviors in behavioral assays such as the and Morris water maze. These effects demonstrate IGF1R's critical involvement in maturation and function, with deficits emerging from reduced PI3K/Akt pathway activation in affected neurons. In humans, biallelic inactivating mutations in IGF1R, including homozygous nonsense and missense variants, cause severe intrauterine growth retardation (IUGR), , and postnatal , often with additional features like developmental delay and dysmorphic facial traits. Such cases, first systematically reported in cohorts during the , typically result in birth weights below the 3rd and head circumferences reduced by 2-3 standard deviations, reflecting profound impairment of IGF-1-mediated mitogenic signaling during embryogenesis. For instance, homozygous missense mutations in IGF1R disrupt activity, leading to extreme and metabolic dysregulation without complete lethality. Compensatory mechanisms partially mitigate IGF1R loss in certain contexts, particularly through upregulation of insulin receptor (IR)/IGF1R hybrid receptors, which form when IR and IGF1R monomers dimerize and retain partial affinity for IGF-1 ligands. In tissues like muscle and liver, these hybrids sustain some downstream signaling via shared PI3K/Akt and MAPK pathways, attenuating growth and metabolic defects in heterozygous or tissue-specific knockouts, though they cannot fully replicate homodimeric IGF1R functions.

Clinical Significance

Role in Cancer

The insulin-like growth factor 1 receptor (IGF1R) is frequently overexpressed in various human cancers, contributing to enhanced tumor cell proliferation and survival. In , IGF1R upregulation occurs in approximately 50-90% of cases, particularly in estrogen receptor-positive subtypes, where it correlates with aggressive disease and reduced patient survival. Similarly, elevated IGF1R expression is observed in 60-80% of cancers, promoting androgen-independent growth, while in and colorectal cancers, it is detected in over 70% of tumors, associating with advanced stages and poor prognosis. IGF1R exerts its oncogenic effects through autocrine and loops involving IGF-1 and IGF-2 ligands, which activate the receptor to drive downstream pathways that inhibit and enhance cell survival. For instance, IGF1R activation stimulates the PI3K/Akt pathway, conferring resistance to by upregulating anti-apoptotic proteins such as Bcl-2. Additionally, IGF1R promotes tumor by inducing epithelial-mesenchymal transition (), a process that increases cell motility and invasiveness through transcriptional changes in E-cadherin and expression. These mechanisms underscore IGF1R's role in sustaining tumor progression beyond initial proliferation signals. In specific malignancies, IGF1R plays distinct roles that amplify cancer hallmarks. In , IGF1R signaling drives by upregulating (VEGF) expression in tumor cells, facilitating nutrient supply and metastatic spread. In , IGF1R supports the survival of malignant plasma cells within the niche, where it interacts with stromal cells to provide protective signals against and chemotherapy-induced death. As a prognostic , high IGF1R expression predicts increased risk across several cancers. In , particularly ERBB2 (HER2)-enriched subtypes, co-expression of IGF1R and ERBB2 amplifies signaling crosstalk, correlating with worse overall survival and resistance to targeted therapies. The insulin-like growth factor 1 receptor (IGF1R) plays a significant role in the pathogenesis of and . Activation of IGF1R signaling contributes to compensatory β-cell in response to chronic , as evidenced by studies showing that IGF1R-mediated pathways, particularly through IRS-2, promote β-cell to offset insulin demand in insulin-resistant states. However, formation of hybrid receptors between IGF1R and the (IR) disrupts normal insulin signaling, as these hybrids exhibit high affinity for IGF-1 but low affinity for insulin, leading to reduced and exacerbated . Increased prevalence of such hybrids in adipose and tissues of individuals with correlates with diminished insulin sensitivity and contributes to systemic . In age-related neurodegenerative diseases, dysregulated IGF1R signaling is implicated in pathological processes. Elevated IGF1R activity has been linked to progression through promotion of tau hyperphosphorylation, a hallmark of formation, as IGF-1 stimulation activates downstream kinases like GSK-3β that phosphorylate . In , reduced IGF1R/IGF-1 signaling is associated with disease progression, as partial IGF-1 deficiency exacerbates oxidative damage, inflammation, and neuronal in toxin-induced models like , highlighting the neuroprotective role of adequate IGF1R activity in preventing microglial activation and loss. IGF1R influences cardiovascular health, particularly in , by regulating vascular cell (VSMC) behavior. IGF1R signaling promotes VSMC proliferation and migration, which stabilizes atherosclerotic plaques by increasing content and fibrous cap thickness, thereby reducing rupture risk; deficiency in VSMC-specific IGF1R leads to plaque instability, larger necrotic cores, and accelerated progression in hyperlipidemic models. Additionally, low IGF1R activity, reflected by reduced circulating IGF-1 levels, correlates with increased frailty in the elderly, independently associating with higher odds of frailty (adjusted OR 1.54 for lowest vs. highest IGF-1 quartile) due to impaired muscle maintenance and physical resilience. Recent genome-wide association studies (GWAS) post-2020 have identified IGF1R variants influencing metabolic and outcomes. Rare loss-of-function variants in IGF1R, such as those reducing receptor signaling, are enriched in centenarians and linked to extended lifespan by mimicking caloric restriction effects on insulin/IGF-1 pathways. Similarly, damaging missense variants in IGF1R confer risk for and in diverse populations, highlighting its role in glucose and age-related metabolic decline. Emerging as of 2025 also associates IGF1R dysregulation with increased risk of and metabolic complications in survivors, expanding its clinical implications in age-related and post-viral conditions.

Regulation and Interactions

Post-Translational Regulation

The insulin-like growth factor 1 receptor (IGF1R) is subject to multiple post-translational modifications that fine-tune its , , trafficking, and subcellular localization following protein synthesis. Among these, plays a central role in modulating receptor beyond initial ligand-induced autophosphorylation on key residues in the activation loop. Inhibitory phosphorylation occurs on serine residues within the C-terminal tail, such as Ser1280–1283, mediated by kinases including (PKC), which attenuates kinase activity and downstream signaling. by protein tyrosine phosphatase 1B (PTP1B) reverses activating tyrosine phosphorylations on IGF1R, while phosphatase and tensin homolog (PTEN) dephosphorylates PIP3 to attenuate downstream signaling, thereby dampening signaling intensity and duration in response to stimuli like or nutrient deprivation. Ubiquitination serves as a critical for controlling IGF1R turnover, particularly during prolonged stimulation. The E3 ubiquitin ligase c-Cbl promotes K48-linked ubiquitination of the receptor, facilitating clathrin-mediated , trafficking to lysosomes, and subsequent proteolytic degradation, which prevents sustained signaling and maintains cellular . This process is kinase-dependent, requiring initial receptor autophosphorylation, and involves adaptors like β-arrestin to recruit c-Cbl to the endocytic machinery. Glycosylation is essential for IGF1R maturation and function, with N-linked glycans on the α-subunit playing a pivotal role in proper folding, proteolytic processing of the proreceptor, and high-affinity ligand binding. Defects in N-glycosylation, as seen in congenital disorders, lead to retention, reduced surface expression, and impaired signaling. Additionally, O-GlcNAcylation, a dynamic nutrient-sensitive modification, modulates IGF1R activity by promoting autophosphorylation, likely through inhibition of PTP1B, thereby enhancing PI3K/Akt pathway activation under hyperglycemic conditions. Other modifications include SUMOylation and palmitoylation, which influence IGF1R localization. SUMO-1 conjugation on conserved residues (Lys1025, Lys1100, Lys1120) in the β-subunit, induced by IGF-1, enables translocation of the receptor, where it acts as a transcriptional co-activator of its canonical signaling. Palmitoylation, indirectly regulating IGF1R via the scaffold protein flotillin-1, controls receptor trafficking from the to the plasma membrane; depalmitoylation-repalmitoylation cycles sustain activation upon binding.

Protein-Protein Interactions

The insulin-like growth factor 1 receptor (IGF1R) participates in a diverse array of non-covalent protein-protein interactions that modulate its activation, localization, and downstream signaling without involving post-translational modifications. These interactions typically occur at the phosphorylated residues within the receptor's intracellular , serving as docking sites for adaptor proteins and coregulators that fine-tune IGF1R function in various cellular contexts. Key adaptor proteins include insulin receptor substrates 1 and 2 (IRS-1 and IRS-2), which bind directly to autophosphorylated IGF1R through their phosphotyrosine-binding (PTB) domains. This interaction recruits the p85 regulatory subunit of 3-kinase (PI3K) to IRS proteins, thereby initiating the PI3K-Akt pathway essential for metabolic and proliferative responses. In parallel, the adaptor protein Shc docks to tyrosine-phosphorylated IGF1R and associates with receptor-bound protein 2 (), which recruits son of sevenless (Sos) to activate the Ras-mitogen-activated protein kinase (MAPK) cascade, promoting and . Among coregulators, caveolin-1 binds IGF1R within plasma membrane lipid rafts, sequestering the receptor in these cholesterol-rich microdomains to inhibit its activation and downstream signaling, such as in differentiation processes. Similarly, β-arrestin interacts with IGF1R after G-protein-coupled receptor kinase (GRK)-mediated of the receptor's C-terminal tail, facilitating rapid desensitization, , and signal attenuation to prevent prolonged activation. IGF1R also forms functional hybrids through dimerization with the (IR), creating heterotetrameric complexes composed of one IGF1R αβ and one IR αβ ; these hybrids exhibit biased ligand affinity, with insulin preferentially activating hybrid signaling toward metabolic outcomes rather than mitogenic ones. In cancer contexts, IGF1R heterodimerizes with the (EGFR), enabling that stabilizes IGF1R expression, enhances PI3K-Akt and MAPK pathway activation, and contributes to resistance against EGFR-targeted therapies. Interaction databases highlight the breadth of IGF1R's binding partners, underscoring its role as a signaling hub. The BioGRID database catalogs 526 unique interactors for human IGF1R, derived from experimental evidence including affinity capture and co-immunoprecipitation assays. Prominent among these are PIK3R1 (the p85 subunit of PI3K) and MAPK1 (Erk2), which rank highly due to frequent co-occurrence in signaling complexes, as evidenced by high-confidence networks in resources like .

Therapeutic Targeting

Inhibitors and Modulators

The insulin-like growth factor 1 receptor (IGF1R) has been targeted by various pharmacological agents, primarily to disrupt its role in and survival signaling. Monoclonal antibodies represent a key class of inhibitors, with figitumumab (CP-751,871) being a fully IgG2 that selectively binds to the α-subunit of IGF1R, thereby blocking ligand access by IGF-1 and IGF-2 and promoting receptor internalization and degradation. This mechanism inhibits IGF1R autophosphorylation and downstream signaling, leading to antiproliferative effects in preclinical models. However, development of figitumumab was discontinued following III trials due to lack of in improving outcomes, such as in non-small cell . Tyrosine kinase inhibitors (TKIs) constitute another major category, exemplified by linsitinib (OSI-906), a small-molecule inhibitor of and the (). Linsitinib competitively binds to the ATP-binding of the kinase domain, preventing autophosphorylation and activation of downstream pathways like PI3K/AKT. This agent has been evaluated in clinical trials, including for , where it demonstrated preliminary antitumor activity despite challenges in broader application and subsequent termination of development. Other modulators include antisense oligonucleotides (ASOs) that target IGF1R mRNA to reduce receptor expression. For instance, 2′-MOE-modified ASOs bind to IGF1R mRNA sequences, inducing RNase H-mediated degradation and thereby decreasing IGF1R protein levels, which inhibits tumor cell proliferation in vitro and in vivo models. Similarly, natural compounds like genistein, a soy , act as partial antagonists by inactivating IGF1R and suppressing of downstream effectors such as AKT, contributing to antiproliferative and pro-apoptotic effects in cancer cells. Development of IGF1R inhibitors faces significant challenges, including toxicity from cross-inhibition of , which disrupts glucose and causes . Additionally, resistance often arises through pathway , where IGF1R inhibition leads to compensatory activation of alternative signaling networks, such as or MAPK pathways, thereby limiting therapeutic efficacy.

Clinical Applications and Challenges

The clinical translation of IGF1R-targeted therapies has shown mixed results, with notable successes in specific pediatric sarcomas but broader challenges in solid tumors. In a phase I/II trial of figitumumab monotherapy in patients with refractory , the objective response rate was 2%, with stable disease in 29% of patients. However, trials in non-small cell (NSCLC) have largely failed to demonstrate significant efficacy, attributed to intratumoral heterogeneity in IGF1R expression and signaling, which complicates patient selection and leads to inconsistent responses across cohorts. These outcomes highlight the potential of IGF1R inhibition in IGF1R-overexpressing tumors like while underscoring the need for refined trial designs in heterogeneous cancers such as NSCLC. Combination strategies have emerged to enhance IGF1R-targeted efficacy by addressing resistance mechanisms, particularly in aggressive malignancies. For instance, dual inhibition of IGF1R and (VEGF) pathways has been explored in , where preclinical models demonstrate that anti-VEGF agents like upregulate IGF1 signaling, and combined blockade suppresses tumor growth more effectively than monotherapy. Similarly, pairing IGF1R inhibitors with PD-1 checkpoint inhibitors shows promise in overcoming immunosuppressive microenvironments; an ongoing phase I trial (NCT06866548) as of November 2025 evaluates an anti-IGF1R with anti-PD-1 therapy in metastatic castration-resistant , aiming to boost T-cell infiltration and response rates. Completed trials like AEWS1221 (NCT02306161), which added ganitumab to in newly diagnosed metastatic , did not improve event-free survival but informed ongoing research into IGF pathway targeting in pediatric sarcomas as of 2025. Biomarker-driven approaches are critical for optimizing IGF1R therapies, focusing on expression, ligand levels, and pathway activation. (IHC) assessment of IGF1R expression in tumor biopsies serves as a primary predictive , with high membranous correlating to potential responsiveness in sarcomas and other IGF1R-dependent cancers. Circulating IGF-1 levels provide a non-invasive , as elevated IGF-1 is associated with increased IGF1R signaling and poorer in multiple tumor types, guiding enrollment. For pharmacodynamic monitoring, of IGF1R (p-IGF1R) in peripheral blood mononuclear cells or tumor tissue post-treatment confirms target engagement and inhibition, enabling dose adjustments in real-time during trials. Despite these advances, clinical challenges persist, limiting widespread adoption of IGF1R-targeted therapies. Dose-limiting toxicities, such as (observed in up to 28% of patients across anti-IGF1R trials), , and , necessitate careful management and often restrict dosing intensity. Compensatory upregulation of the (IR) or activation of alternative pathways, like PI3K/AKT, frequently emerges as a resistance mechanism, reducing long-term efficacy even in initially responsive tumors. Patient stratification remains a key barrier, as heterogeneous profiles and lack of robust predictive tools lead to inclusion of non-responders, inflating failure rates in unselected populations. Ongoing as of 2025 emphasizes integrating multi-omics to address these issues and improve therapeutic , with 2025 reviews noting promise in next-generation IGF1R inhibitors combined with .

References

  1. [1]
    IGF1R insulin like growth factor 1 receptor [ (human)] - NCBI
    Sep 27, 2025 · The insulin-like growth factor 1 receptor (IGF-1R) gene was found to be a potential downstream target of NUCKS1 in GC cells. In stressed corneal ...
  2. [2]
    The IGF1 Signaling Pathway: From Basic Concepts to Therapeutic ...
    Oct 4, 2023 · The present review article provides a concise overview of key ... Targeting insulin-like growth factor 1 receptor in sarcomas. Curr ...
  3. [3]
    IGF1R - Insulin-like growth factor 1 receptor - Homo sapiens (Human)
    Forms a hybrid receptor with INSR, the hybrid is a tetramer consisting of 1 alpha chain and 1 beta chain of INSR and 1 alpha chain and 1 beta chain of IGF1R.Missing: 903 464
  4. [4]
    The Structure of the Type 1 Insulin-Like Growth Factor Receptor - NCBI
    It consists of 4,989 nucleotides and codes for a 1,367 amino acid precursor (Fig. 1). The pre-proreceptor monomer includes a 30 residue signal peptide ...Missing: 903 | Show results with:903
  5. [5]
    IGF1R (insulin-like growth factor 1 receptor)
    Sep 1, 2008 · The mature heterotetramers have a β-α-α-β conformation. The α subunit is entirely extracellular and includes a cysteine-rich region and several ...Missing: composition | Show results with:composition
  6. [6]
    Recombinant Insulin-Like Growth Factor 1 Dimers: Receptor Binding ...
    Mar 4, 2023 · IGF-1R, as well as IR, can be described as (αβ)2 dimer of two αβ monomers. Each (αβ)2 dimer consists of α and β chains linked by several ...
  7. [7]
    Structural basis of the activation of type 1 insulin-like growth factor ...
    Oct 8, 2019 · We will use the amino acid numbering of human IGF1R to describe our ... α-CT (the C-terminus of the α-subunit). Full size image. The ...
  8. [8]
    Role of the activation loop tyrosines in regulation of the insulin-like ...
    Aug 18, 2006 · Ligand binding to the extracellular portion of IGF1R stimulates autophosphorylation at three sites (Tyr1131, Tyr1135, and Tyr1136) in the ...Missing: Y1135 Y1136 Y1141
  9. [9]
    Controlled Signaling—Insulin-Like Growth Factor Receptor ...
    Our recent study showed that IGF-1-promoted phosphorylation of the Tyr1250/1251 site in the IGF-1R C-terminal results in enhanced IGF-1R internalization and ...<|control11|><|separator|>
  10. [10]
    Insulin and its analogues and their affinities for the IGF1 receptor in
    On the other hand, the IGF1R binds the IGFs with a high affinity (10−10 M), but binds insulin with a 100-fold lower affinity (Blakesley et al. 1996).Binding of insulin analogues to... · What determines growth...
  11. [11]
  12. [12]
    Classification of Growth Factors and Their Receptors - NCBI - NIH
    The insulin receptor, IR, is the prototype for a family of RTKs whose distinctive structural feature is to function as a heterotetrameric aggregation of two α ...
  13. [13]
    Insulin Receptor Isoforms and Insulin Growth Factor-like Receptors
    Unlike most RTK receptors, the IR superfamily, including the IR and IGF-1R, exists as a covalent disulfide-linked dimer in the absence of a ligand.<|control11|><|separator|>
  14. [14]
  15. [15]
    Domain-dependent effects of insulin and IGF-1 receptors on ...
    Mar 27, 2017 · Overall, IR and IGF1R share more than 50% sequence homology and over 80% homology in the intracellular kinase domain. Both insulin and IGF-1 ...
  16. [16]
    Insulin-like growth factors: Ligands, binding proteins, and receptors
    The IGF system includes ligands (IGF-I, IGF-II, insulin), receptors (IGF-IR, M6P/IGF-IIR, and insulin receptor), and binding proteins (IGFBP-1-6).Missing: stoichiometry | Show results with:stoichiometry
  17. [17]
    The insulin-like growth factor system: A target for endocrine ...
    The insulin-like growth factor (IGF) system is a critical regulator of growth, especially during fetal development, while also playing a central role in ...
  18. [18]
    How ligand binds to the type 1 insulin-like growth factor receptor
    Feb 26, 2018 · Here, the authors present the crystal structures of the IGF-1R ectodomain both in its apo form and in complex with its ligand insulin-like ...Missing: homologs | Show results with:homologs
  19. [19]
    Small-molecule inhibition and activation-loop trans-phosphorylation ...
    Jun 19, 2008 · Ligand binding to IGF1R promotes autophosphorylation in trans (one kinase domain phosphorylating the other) of Tyr1135 and two other activation- ...Missing: Y1136 Y1141
  20. [20]
  21. [21]
    6PYH: Cryo-EM structure of full-length IGF1R-IGF1 ... - RCSB PDB
    Oct 23, 2019 · We report the cryo-EM structure of full-length IGF1R-IGF1 complex in the active state. This structure reveals that only one IGF1 molecule binds the Γ-shaped ...Missing: paper | Show results with:paper
  22. [22]
    How insulin-like growth factor I binds to a hybrid insulin receptor ...
    Aug 4, 2022 · The structure of an apo hybrid receptor is not yet available. Determination of the structure of apo IR and IGF-1R has thus far proved ...Missing: homology dIR
  23. [23]
    Specific insulin/IGF1 hybrid receptor activation assay ... - Nature
    Jan 21, 2015 · IR and IGF1R are highly homologous tetramers consisting of two alpha-beta subunits linked by disulfide bonds and containing tyrosine kinase ...Missing: heterotetrameric | Show results with:heterotetrameric
  24. [24]
    Insulin Receptor Isoforms and Insulin Receptor/Insulin-Like Growth ...
    Homology between IR and IGF-IR is high, ranging from 45–65% in the ligand binding domains to 60–85% in the tyrosine kinase and substrate recruitment domains (22 ...
  25. [25]
    emerging paradigm of insulin-like growth factor type 1 receptor (IGF ...
    The promising strategy of inhibiting IGF-1R kinase activity is complicated by IR cross-reactivity issues, as the kinase domain of the IGF-1R shares 85 % ...
  26. [26]
    IGF1R signaling cascade - Reactome
    The type 1 insulin-like growth factor receptor (IGF1R) binds and phosphorylates scaffold proteins, IRS1/2/4 and SHC1, which in turn bind effectors possessing ...Missing: STAT3 PLCγ PIP3 PDK1 mTOR S6K1 negative feedback
  27. [27]
    Insulin-like growth factor receptor signaling in tumorigenesis and ...
    Jun 3, 2020 · Mechanistically, activation of STAT3 contributes to the induction of EMT and cancer stemness by IGF-IR [30, 70].
  28. [28]
    The signaling landscape of insulin-like growth factor 1 - ScienceDirect
    IGFs activate several signaling pathways, including MAPK, PI3K/AKT, NO, and PKC, leading to endothelial cell migration, survival, nutrient uptake, and tube ...
  29. [29]
    Domain-dependent effects of insulin and IGF-1 receptors on ... - Nature
    Mar 27, 2017 · Both insulin and IGF-1 are able to bind to and activate each other's receptors, albeit with reduced affinity, and both IR and IGF1R also elicit ...
  30. [30]
    IGF-1 mediated phosphorylation of specific IRS-1 serines in Ames ...
    Phosphorylation of the IRS Ser residues inhibits Tyr phosphorylation thereby serving as a physiological negative-feedback control mechanism [11]. ... S6K1 ...
  31. [31]
    The IGF Pathway Regulates ERα through a S6K1-Dependent ...
    The initial objective was to determine whether IGF-induced negative feedback was linked to specific IRS isoforms. IRS-1 and IRS-2 are coexpressed in both ...
  32. [32]
    Mice carrying null mutations of the genes encoding insulin ... - PubMed
    Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r) ... Cell. 1993 Oct 8;75(1): ...Missing: knockout | Show results with:knockout
  33. [33]
    Mechanisms of IGF-1-Mediated Regulation of Skeletal Muscle ...
    IGF-1 has been shown to increase both proliferation and differentiation of cultured myoblasts [119]. When cells are in the proliferative stage, IGF-1 increased ...
  34. [34]
    Igf1 promotes longitudinal bone growth by insulin-like actions ...
    Insulin-like growth factor 1 (Igf1) is reputed to augment longitudinal bone growth by stimulating growth plate chondrocyte proliferation.
  35. [35]
    The hormonal action of IGF1 in postnatal mouse growth - PNAS
    Dec 9, 2008 · Our genetic study demonstrated that the endocrine IGF1 plays a very significant role in mouse growth, as its action contributes approximately30% of the adult ...Results · Growth Phenotypes · Endocrine Igf1 And Fertility
  36. [36]
    Tissue expression of IGF1R - Summary - The Human Protein Atlas
    Summary of IGF1R (CD221, IGFIR, IGFR, JTK13, MGC18216) expression in human tissue. Ubiquitous granular cytoplasmic expression, mostly in glandular cells.
  37. [37]
    IGF-I Receptor Mutations Resulting in Intrauterine and Postnatal ...
    Mutations in the IGF-IR gene that lead to abnormalities in the function or number of IGF-I receptors may also retard intrauterine and subsequent growth in ...Analysis Of Igf-I Receptor... · Patient 1 · Igf--Igfbp Axis
  38. [38]
    Insulin-Like Growth Factor 1 (IGF-1) Signaling in Glucose ...
    The direct effects of IGF-1 on glucose metabolism are mediated by the effect of IGF-1 on pancreatic insulin secretion or glucose uptake by muscle.
  39. [39]
    Insulin-like growth factor-1 deficiency and metabolic syndrome
    Jan 6, 2016 · Such results could indicate that hepatic derived IGF-1 plays a crucial role in skeletal muscle insulin signalling and glucose uptake. In murine ...
  40. [40]
    Differential Roles of Insulin and IGF-1 Receptors in Adipose Tissue ...
    Insulin and insulin-like growth factor 1 (IGF-1) play important roles in the development and differentiation of white and brown adipose tissue (WAT and BAT) (1, ...<|separator|>
  41. [41]
    Insulin-like growth factors 1 and 2 regulate expression of β-casein in ...
    It was found that both IGF-1 and IGF-2 activate the expression of milk protein β-casein in the presence of prolactin and hydrocortisone. It was found that β- ...
  42. [42]
    Decreased IGF Type 1 Receptor Signaling in Mammary Epithelium ...
    To study the functions of IGF-1R during pregnancy and lactation, we established transgenic mouse lines expressing a human dominant-negative kinase dead IGF-1R ( ...Missing: synergy | Show results with:synergy
  43. [43]
    IGF1R is a mediator of sex-specific metabolism in mice - Frontiers
    These results demonstrate that IGF1R is involved in metabolic homeostasis, with effects modulated by diet-induced obesity and aging in a sex dependent manner.Missing: milk maternal
  44. [44]
    Maternal Diabetes Impairs Insulin and IGF-1 Receptor Expression ...
    Mar 10, 2021 · An increased maternal blood glucose level during pregnancy is associated with an increased IGF-1R phosphorylation and IR-A expression in the placenta.
  45. [45]
    Mechanisms of Fetal Overgrowth in Gestational Diabetes - NIH
    Altered metabolism and hormone levels in GDM mothers contribute to excessive fetal growth by enhancing transplacental nutrient transfer through IGF-1 ...
  46. [46]
    Disorders of Growth Hormone in Childhood - Endotext - NCBI - NIH
    Mar 9, 2022 · The GH-IGF1 axis plays a major role in the childhood phase of growth with a significant role alongside sex steroids during puberty while in ...
  47. [47]
    40 YEARS OF IGF1: Insulin-like growth factors: actions on the ...
    ... height and weight above the 97th percentile and showed accelerated ... IGF1R in tissues correlate directly with body size (Holzenberger et al. 2001) ...
  48. [48]
    Genetic regulation of linear growth
    Mar 31, 2019 · ... polymorphisms in genes that affect height in the general population. ... 1) GH-IGF1 axis (IGF1R). The GH-IGF1 axis is one of the main ...<|separator|>
  49. [49]
    The role of insulin/IGF-like signaling in C. elegans longevity and aging
    Jul 7, 2010 · daf-2 encodes the only insulin/IGF-1 receptor expressed in worms, and age-1 is the catalytic subunit of the downstream phosphoinositide 3-kinase ...C. Elegans As A Premier... · Insulin/igf-1 Signaling · Endocrine Signaling And...
  50. [50]
    Studies of Caenorhabditis elegans DAF-2/insulin signaling reveal ...
    Here, we review recent studies that have uncovered new details about how insulin signaling affects lifespan in the nematode C. elegans.Missing: dIR | Show results with:dIR
  51. [51]
    Does Reduced IGF-1R Signaling in Igf1r +/− Mice Alter Aging? - PMC
    Nov 23, 2011 · Mutations in insulin/IGF-1 signaling pathway have been shown to lead to increased longevity in various invertebrate models.
  52. [52]
    Mutations Affecting Mammalian Aging: GH and GHR vs IGF-1 and ...
    This provides evidence that the association of GH deficiency and increased lifespan in Ames dwarf mice is causal (mechanistic). In contrast to the remarkable ...
  53. [53]
    Late-life targeting of the IGF-1 receptor improves healthspan and ...
    Jun 19, 2018 · Diminished growth factor signaling improves longevity in laboratory models, while a reduction in the somatotropic axis is favorably linked ...
  54. [54]
    IGF-I Receptor Mutations in Centenarians | PNAS
    Mar 4, 2008 · This article provides strong evidence that the IGF1R can be a genetic determinant of human longevity. The role of GH/IGF pathway in human ...
  55. [55]
    Human insulin/IGF-1 and familial longevity at middle age - Aging-US
    Second, offspring of sporadic female centenarians were shown to be smaller and display higher IGF-1 levels, indicative of IGF-1 insensitivity, while rare IGF-1R ...
  56. [56]
    Impaired IGF1R signaling in cells expressing longevity-associated ...
    The results indicate that MEFs expressing the human longevity-associated IGF1R mutations attenuated IGF1 signaling, as demonstrated by significant reduction in ...
  57. [57]
    Activation of the IGF1 pathway mediates changes in cellular ... - NIH
    In these cells, IGF1R mutations and IGF1 overexpression are found to correlate with skeletal and craniofacial defects (Cunningham et al., 2011; Guntur and Rosen ...
  58. [58]
    IGF1R Variants Associated with Isolated Single Suture ... - NIH
    These data suggest that IGF1R mutations may contribute to the risk and in some cases cause single suture craniosynostosis.
  59. [59]
    Various phenotypes of short stature with heterozygous IGF-1 ... - NIH
    Recombinant GH therapy might be effective in treating IGF1R-related short stature. ○ Higher IGF1 levels after rhGH treatment may indicate IGF1R gene mutations.Missing: overgrowth | Show results with:overgrowth
  60. [60]
    IGF1R curation results for Dosage Sensitivity - ClinGen Curated Genes
    Autosomal dominant mutations in IGF1R gene are reported to be associated with IGF1 insensitivity, in which the clinical presentation includes prenatal and ...
  61. [61]
    Beckwith-Wiedemann syndrome and the insulin-like growth factor-II ...
    Beckwith-Wiedemann syndrome and the insulin-like growth factor-II gene. Does the genotype explain the phenotype? · Full text · Selected References · ACTIONS.
  62. [62]
    Homozygous mutation of the IGF1 receptor gene in a patient with ...
    Dec 10, 2012 · The homozygous mutation of the IGF1R is associated with severe IUGR, dysmorphic features, and insulin resistance, while both parents were ...Missing: human | Show results with:human
  63. [63]
    Drugging IGF-1R in cancer: New insights and emerging opportunities
    At least 50% of breast cancer cases were found to express activated IGF-1R. These studies showed strong IGF-1R staining in the membrane and/or cytoplasm and ...
  64. [64]
    The impact of IGF-1R expression on the outcomes of patients ... - NIH
    It has been detected in 50%–93% of patients with breast cancer;5–8 however, the prognostic value of IGF-1R for breast cancer remains controversial.Missing: ERBB2 | Show results with:ERBB2
  65. [65]
    MicroRNA-Induced Regulation of the IGF-1 System in Various Types ...
    Sep 27, 2025 · The IGF-1 receptor (IGF-1R) is upregulated in colorectal, breast, prostate and lung cancers. It was also known to be a potential target in ...
  66. [66]
    Function of insulin-like growth factor 1 receptor in cancer resistance ...
    IGF1R-mediated resistance includes promoting cells to proliferate, inhibiting apoptosis, inducing increased expression of ABC transporter proteins on cell ...
  67. [67]
    Role of the IGF System in Cancer Growth and Metastasis
    Taken as a whole, however, these studies suggest that the IGFs can play a paracrine and/or autocrine role in promoting tumor growth in situ during tumor ...
  68. [68]
    Decoding the Role of Insulin-like Growth Factor 1 and Its Isoforms in ...
    Recent studies indicate that IGF signaling can promote EMT through IGF-1R-dependent and -independent mechanisms, involving interactions with the IR-A isoform ...<|separator|>
  69. [69]
    Potent Inhibition of Angiogenesis by the IGF-1 Receptor-Targeting ...
    Previously, we reported that a predominant action of an IGF-1R-targeted antibody was through inhibiting tumor-derived VEGF, and indirectly, angiogenesis.
  70. [70]
    Hypoxia‐induced ELF3 promotes tumor angiogenesis through IGF1 ...
    Jun 13, 2022 · ELF3 is a transcription factor that induces angiogenesis by transcriptionally increasing IGF1 expression. Hypoxia enhances the effect of ELF3 on angiogenesis.
  71. [71]
    The role of IGF-1 as a major growth factor for myeloma cell lines and ...
    Patients with IGF-1Rabsent MMC had a longer median event-free survival than patients with IGF-1Rpresent MMC (P = .006 and .005, Figures 6A and 6B). The same ...
  72. [72]
    20P Prognostic implications of IGF-1R expression in breast cancer ...
    Conclusions: In BC patients, high IGF1R tumor expression in cytoplasm and mem- brane was associated with increased risk of death, but not recurrence. In BC ...
  73. [73]
    Role of IGF1R in Breast Cancer Subtypes, Stemness, and ... - Frontiers
    In this study, these ERBB2-enriched tumors are the only subtype presenting a low patient prognosis in correlation with IGF1R expression (62). The risk of ...
  74. [74]
    Defective insulin secretion in pancreatic β cells lacking type 1 ... - JCI
    A potential role for IGF1R acting as a mediator of β cell proliferation through IRS-2 has been implied by observations indicating that the β cell failure ...
  75. [75]
    implications for the design of IR–IGF1R hybrid receptor modulators
    Feb 21, 2022 · The IR is activated through binding to insulin, whilst IGF1R is activated through binding to the hormones insulin-like growth factor (IGF) 1 and ...Igf1r Ligand Bound... · Ligand Binding · Igf1r Atp Competitive...
  76. [76]
    Uncovering the intricacies of IGF-1 in Alzheimer's disease - PubMed
    Dysregulation of IGF-1 signaling contributes to hyperphosphorylation of tau, NFT accumulation, increased β- and γ-secretase activity, elevated Aβ production, ...<|separator|>
  77. [77]
    Is insulin-like growth factor-1 involved in Parkinson's disease ...
    Feb 11, 2020 · From our experience, the mere partial IGF-1 deficiency in a murine model increases brain oxidative damage, inflammation, edema and apoptosis, ...
  78. [78]
  79. [79]
    Association between Insulin-Like Growth Factor-1 and Frailty among ...
    Conclusion: Lower serum IGF-1 levels were independently related with frailty in older adults. Keywords: IGF-1; disability; frailty. Publication types.
  80. [80]
    High-throughput sequencing analysis of nuclear-encoded ...
    Aug 10, 2022 · We hypothesize that humans with exceptional longevity harbor rare variants in nuclear-encoded mitochondrial genes (mitonuclear genes) that confer resistance.
  81. [81]
    [PDF] Population scale whole genome sequencing provides novel insights ...
    Jun 9, 2024 · Cell genomics 3, (2023). 647. 11. Gardner, E. J. et al. Damaging missense variants in IGF1R implicate a role for. 648. IGF-1 resistance in the etiology of type ...Missing: longevity | Show results with:longevity<|control11|><|separator|>
  82. [82]
    Superoxide anion radicals induce IGF-1 resistance through ...
    Inline graphic led to activation of the phosphatases PTP1B and PTEN, which via dephosphorylation of the IGF-1 receptor and phosphatidylinositol 3,4,5- ...
  83. [83]
    Protein-tyrosine Phosphatase 1B Antagonized Signaling by Insulin ...
    Mechanistically, the pro-survival IGF-1R signaling pathway was attenuated upon ectopic expression of PTP1B. This was due to dephosphorylation by PTP1B of IGF-1R ...Results · Ptp1b Directly... · Discussion
  84. [84]
    Role of Ubiquitination in IGF-1 Receptor Signaling and Degradation
    Apr 4, 2007 · Our data suggest important roles of ubiquitination in mediating IGF-1R signaling and degradation. Ubiquitination of IGF-1R requires receptor tyrosine kinase ...
  85. [85]
    emerging paradigm of insulin-like growth factor type 1 receptor (IGF ...
    Nov 26, 2013 · The α-subunit contains 710 amino acids (aa 1–710) and has in its structure two homologous domains (L1 and L2) separated by a cysteine-rich ...
  86. [86]
    Defective IGF-1 prohormone N-glycosylation and reduced IGF-1 ...
    Feb 24, 2022 · Each α and β subunit contains 11 and 5N-glycosylation sites ... glycosylation causes IGF-1R proreceptor instability and degradation [27].
  87. [87]
    Increased O-GlcNAcylation promotes IGF-1 receptor/PhosphatidyI ...
    Mar 16, 2022 · PTP1B is one of the main PTPases that control IGF1R phosphorylation. ... PTEN (phosphatase and tensin homolog) or PP2A (protein ...
  88. [88]
    SUMOylation mediates the nuclear translocation and signaling of ...
    Feb 9, 2010 · We report that IGF-1 promotes the modification of IGF-1R by small ubiquitin-like modifier protein-1 (SUMO-1) and its translocation to the nucleus.
  89. [89]
    Essential role of flotillin-1 palmitoylation in the intracellular ... - PubMed
    Jun 1, 2015 · Our data show that flotillin-1 palmitoylation is a new mechanism by which the intracellular localization and activation of IGF-1R are controlled.
  90. [90]
    The insulin receptor substrate (IRS) proteins - PubMed Central - NIH
    The IRS proteins play a central role in mediating the signals from the IR/IGF-1R that control tumor cell metabolism.
  91. [91]
    Lipid Rafts/Caveolae Are Essential for Insulin-like Growth Factor-1 ...
    In 3T3-L1 cells, IGF-1 receptor is located in lipid rafts/caveolae of the plasma membrane and can directly interact with caveolin-1, the major protein component ...
  92. [92]
    Functional antagonism of β-arrestin isoforms balance IGF-1R ...
    Jun 5, 2017 · β-arrestin recruitment and binding to the ligand-stimulated receptor is essential for desensitization, sequestration, recycling and ...
  93. [93]
    The insulin and IGF1 receptor kinase domains are functional dimers ...
    Mar 11, 2015 · Here we provide crystallographic, biophysical and biochemical evidence demonstrating that the phosphorylated kinase domains of IR and IGF1R form ...
  94. [94]
    The EGF receptor interacts with the type 1 IGF ... - ScienceDirect.com
    These results indicate novel evidence of crosstalk between two key cancer treatment targets, capable of modifying the stability of IGF1R protein.
  95. [95]
    IGF1R Result Summary - BioGRID
    Chemical Interactions 16; Network; PTM Sites 45. Showing 1 to 300 of 526 unique interactors.Missing: STRING | Show results with:STRING
  96. [96]
    The insulin-like growth factor-I receptor inhibitor figitumumab (CP ...
    Jul 13, 2010 · Figitumumab blocks the binding of IGF-1 to IGF-IR, inhibits the downstream signalling activated by both IGF-1 and IGF-2, and induces prolonged ...Materials And Methods · Pharmacokinetics · Pharmacodynamics
  97. [97]
    Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R ...
    Figitumumab (previously CP-751,871) is a highly specific, fully human IgG2 monoclonal antibody that inhibits IGF-1R autophosphorylation induced by IGF-I and IGF ...
  98. [98]
    Update of IGF-1 receptor inhibitor (ganitumab, dalotuzumab ...
    Apr 25, 2017 · ... Figitumumab (CP-751,871) (HR = 1.14, 95% CI = 1.00-1.29), indicating that figitumumab (CP-751,871) may be harmful on cancer treatment. Two ...
  99. [99]
    Phase I Dose-Escalation Study of Linsitinib (OSI-906) and Erlotinib ...
    Linsitinib (OSI-906) is a potent, orally bioavailable dual IGF1R and INSR tyrosine kinase inhibitor (TKI) with antiproliferative effects in a variety of tumor ...
  100. [100]
    Linsitinib (OSI-906) versus placebo for patients with ... - PubMed
    Linsitinib (OSI-906) versus placebo for patients with locally advanced or metastatic adrenocortical carcinoma: a double-blind, randomised, phase 3 study.Missing: mechanism | Show results with:mechanism
  101. [101]
    Antisense oligonucleotide targeting insulin-like growth factor-1 ...
    Apr 15, 2010 · In this study, we assess the potency and anti-cancer activity of a 2′-MOE-modified antisense oligonucleotide (ASO) targeting human IGF-IR, ...
  102. [102]
    Genistein induces apoptosis by the inactivation of the IGF-1R/p-Akt ...
    Furthermore, genistein induced the inactivation of IGF-1R and p-Akt and downregulated the Bcl-2/Bax protein ratio. These results suggest that genistein ...
  103. [103]
    Targeting the Insulin-Like Growth Factor (IGF) System Is Not as ...
    Most therapeutic strategies have been designed to specifically inhibit IGF-1R but have for the most part ignored the insulin receptor (IR).
  104. [104]
    Drugging IGF-1R in cancer: New insights and emerging opportunities
    Here we review the occurrence of IGF-1R aberrations and activation mechanisms in cancers, which justify the development of anti-IGF-1R therapies.
  105. [105]
    R1507, a Monoclonal Antibody to the Insulin-Like Growth Factor 1 ...
    Oct 24, 2011 · The overall complete response/partial response rate was 10% (95% CI, 4.9% to 16.5%). The median duration of response was 29 weeks (range, 12 to ...
  106. [106]
    Costs and Causes of Oncology Drug Attrition and Insulin-Like ...
    Jul 28, 2023 · It is possible that discovery and improved use of biomarkers for IGF-1R inhibitors could reduce these examples of clinical trial failure, ...Missing: NSCLC | Show results with:NSCLC
  107. [107]
    IGF-1R inhibitors in cancer: A review of available evidence and ...
    Clinical trials with IGF-1R inhibitors have mostly focused on breast cancer, gastrointestinal cancers, and NSCLC, with more than 100 clinical trials involving ...
  108. [108]
    Targeting the Insulin Growth Factor and the Vascular Endothelial ...
    Here, we report that the insulin growth factor-1 (IGF-1) is upregulated during treatment with bevacizumab of ovarian xenografts and show that dual anti-VEGF and ...In Vivo Anti-Vegf And Igf... · Results · Anti-Igf And -Vegf Therapy...
  109. [109]
    Safety and Efficacy Evaluation of Anti IGF-1R Monoclonal Antibody ...
    This trial is designed to evaluate the safety and efficacy of anti-IGF-1R mAb in combination with anti-PD-1 mAb in patients with mCRPC. Detailed Description.
  110. [110]
    [PDF] NCI Protocol #: 10003 Version Date: 04/06/2020 - Clinical Trials
    Jun 4, 2020 · antitumor effect by inhibiting IGF1R activation through blockade of ligand binding (IGF-1 and ... (51) = 0.30 nM), cynomolgus monkey (Kd = 0.15 nM) ...
  111. [111]
    A dose finding, safety and pharmacokinetic study of AVE1642, an ...
    Using immunohistochemistry assays (IHC), IGF-1R expression and occupancy by AVE1642 were evaluated. Two different antibodies were needed to perform this test.
  112. [112]
    IGF-1R targeting in cancer – does sub-cellular localization matter?
    Oct 20, 2023 · IGF-1R does not contain a nuclear localization signal (NLS), hence active mechanisms of nuclear import must contribute to nuclear localization.Missing: composition heterotetrameric
  113. [113]
    BI 885578, a Novel IGF1R/INSR Tyrosine Kinase Inhibitor with ...
    Abstract. Inhibition of the IGF1R, INSRA, and INSRB receptor tyrosine kinases represents an attractive approach of pharmacologic intervention in cancer,
  114. [114]
    The adverse events profile of anti-IGF-1R monoclonal antibodies in ...
    Our purpose was to conduct a systematic review of the adverse events (AEs) caused by anti-IGF-1R monoclonal antibodies in cancer therapy.
  115. [115]
    sticking points in understanding IGF1R function in cancer
    Discourse in the literature has acknowledged that a lack of patient stratification may have impacted success of IGF1R inhibitor trials. This argument alludes to ...<|control11|><|separator|>