
Insulin‑like growth factor in cancer: New perspectives (Review)
- Authors:
- Published online on: May 23, 2025 https://doi.org/10.3892/mmr.2025.13574
- Article Number: 209
-
Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
With the increasing standard of living, cancer has become a major contributor to the global burden of disease, which continues to grow worldwide, placing a serious economic burden on individuals, families and society (1). According to the Global Agency for Research on Cancer (GARC), 10 cancers accounted for two-thirds of new cases and mortalities worldwide in 2022. It is estimated that there will be >35 million new cancer cases in 2050, an increase of ~77% over the 2022 projections (2,3). In the face of the rapidly growing cancer burden, in addition to the urgent need for improvements in public preventive measures, the enhancement of cancer treatment strategies is equally crucial.
The IGF family consists mainly of two low-molecular-weight protein (IGF-1 and IGF-2), the corresponding receptors (IGF-1R and IGF-2R) and specific binding proteins (4). In 1963, Froesch et al (5) discovered the presence of a certain active substance in serum that could not be completely inhibited by insulin antibodies; this substance was subsequently purified by two scientists, Rinderknecht and Humbel (6), and named IGF-1 and IGF-2. IGF-1 is a small polypeptide consisting of 70 amino acids that is synthesized and secreted into the bloodstream mainly by the liver and some IGF-1 is acted upon by the kidneys or skeletal muscles in an autocrine or paracrine manner in their own tissues or periphery (7). The concentration of IGF1 in serum is regulated by insulin-like growth factor-binding proteins (IGFBPs). When IGFBPs are hydrolyzed by proteases, free IGF-1 binds to IGF-1R on the cell membrane to mediate the corresponding biological effects (8). In addition, IGF-1 levels are affected by a number of factors, such as age, nutritional status and the release of growth hormones (9). IGF-2 is composed of 67 amino acids and has growth-promoting activity similar to that of IGF-1, but its expression pattern is not controlled by growth hormone (10). IGF-2 is considered to play a critical role in fetal growth and development. Deficiency of IGF-1 inhibits the proliferation and protein synthesis of most cells in the body, predisposing the individual to various diseases, including metabolic bone disease, cardiovascular disease and neurodegenerative diseases (11,12). In addition to inhibiting brain development, reducing IGF-2 affects cell metabolism and stem cell self-renewal (13,14).
IGFs are involved in multiple stages of cancer development. IGF-1 serum levels are markedly higher in patients with advanced gastric cancer than in those with early-stage disease and are associated with a Helicobacter pylori positive status (15). Moreover, IGF1R activated by IGF-1 plays a role in epidermal growth factor receptor (EGFR)-mediated primary or secondary resistance to colorectal cancer by upregulating the PI3K/AKT signaling pathway (16). A prospective case-control study suggests that low serum IGF-2 levels are strongly associated with hepatocellular carcinoma risk (17). The IGFs system is involved in the regulation of cancer progression through a variety of signaling pathways, including the MAPK signaling pathway (18), the PI3K/AKT/mTOR signaling pathway (19) and the NF-κB signaling pathway (20). Therefore, strategies targeting the IGF system may be potential candidates for anticancer therapeutic options.
Relationship between IGFs and the malignant biological phenotypes of tumors
Tumor cells have a unique malignant biological phenotype, that manifests as a continuous proliferation signal, escape from growth inhibition, unlimited replication ability, continuous angiogenesis, resistance to cell death, invasion and metastasis, genomic instability and mutation, immune escape and other biological phenomena (21). The number of characteristics of cancer add to the complexity of cancer treatment. This section provides insights into the possible mechanisms by which IGFs influence the malignant biological behavior of cancers, providing new strategies for cancer therapy.
IGFs and tumor proliferation, invasion and metastasis
Continuous proliferation signals, invasion and metastasis are the main characteristics of the malignant phenotype of tumors (22). As an effective mitogen of various cell types, IGF-1 regulates various biological behaviors of tumor cells by binding to IGF-1R. Studies have shown that interferon-induced transmembrane protein (IFITM) is positively associated with gastric cancer progression, recurrence and mortality (23,24). Knocking down IFITM inhibits the proliferation, migration, invasion and epithelial mesenchymal transformation of gastric cancer cells. Further mechanistic studies suggest that IGF-1 induces IFITM2 expression through IGF-1R/STAT3 signal transduction, which ultimately affects tumor growth and metastasis (25). In melanoma, downregulation of IGF-1 reduces the dry character of melanoma initiating cells, including the expression of dry markers (SOX2, Oct-3/4, CD24 and CD133) and functional properties (melanosphere formation, aldehyde dehydrogenase activity and side population) and eventually inhibiting the proliferation and metastasis of tumor cells (26). Moreover, in breast cancer, IGF-1 mainly upregulates cysteine-rich 61 (Cyr61) by activating the PI3K/AKT pathway and an increase in Cyr61 promotes the growth and invasion of breast cancer cells (27). Notably, key transcription factors such as SOX2, which promote and maintain the dry characteristics of cancer cells, can upregulate the expression and autocrine activity of IGF-2, and IGF-2 then activates the IGF-1R/AKT signaling pathway to enhance the invasive and stemness characteristics of bladder cancer, forming a vicious cycle (28). IGFBP-2, an important member of the insulin-like growth factor binding protein family, is able to regulate a variety of cell signaling pathways to influence tumor progression. In oral cancer, IGFBP-2 promotes the upregulation of matrix metalloproteinase (MMP)2 and MMP9 through the activation of the PI3K/Akt/mTOR signaling pathway, which ultimately leads to the proliferation, migration and invasion of cancer cells (29). In addition, IGFBP-1 can be upregulated by H. pylori in a dose-dependent manner, promoting malignant biological processes such as the proliferation, migration and invasion of gastric cancer cells (30). Moreover, IGFBP-7 is also able to regulate the proliferation and migration of cancer cells through the JAK/PI3K signaling axis (31). In conclusion, these findings suggest that targeting IGFs is important for inhibiting cancer proliferation and aggressiveness.
IGFs and tumor angiogenesis
To meet the oxygen and nutrients requirements for continuous proliferation, tumors grow in a variety of ways to stimulate the formation of new blood vessels (32). Especially for solid tumors, new blood vessels are the key link between tumor invasion and metastasis, which also leads to difficulty in tumor treatment. Previous studies have shown that IGFs play an important role in endothelial cell physiology by promoting the expression of the vasodilators NO, VEGF and hypoxia-inducible factor (HIF) (33,34). IGF-1R is involved in most pathophysiological processes mediated by IGFs, including proangiogenic effects. However, IGF-2 also promotes angiogenesis through the insulin receptor (35). Studies have reported that the use of bisphosphonates can delay bone metastasis in patients with breast cancer and improve overall survival (36,37). Mechanistic exploration suggested that pamidronate and clodronate markedly inhibit IGF-1-induced HIF-1α protein accumulation and VEGF expression in breast cancer cells via the PI-3K/AKT/mTOR signaling pathway and ultimately eliminate IGF-1-induced tumor angiogenesis in vivo and in vitro (38). In hypoxic epithelial ovarian cancer, as a transcription factor of IGF-1, highly expressed ELF3-mediated secretion of IGF-1 and VEGF promoted endothelial cell proliferation, migration and tumor angiogenesis through activation of the tyrosine kinase pathway, whereas ELF3 silencing attenuated angiogenesis and tumorigenesis in a xenograft mouse model, demonstrating the pro-vascular effect of IGF-1 (39). Moreover, a variety of IGFBPs are also involved in angiogenesis (40,41). In glioblastoma, proteomic results suggest that IGFBP-1 is a key mediator of cancer cell secretion in response to the vascular production-promoting factor macrophage colony-stimulating factor (MCSF). When conditioned medium from cancer cells was added to human umbilical vein endothelial cells (HUVECs), the silencing of MCSF prevented blood vessel formation. Moreover, IGFBP-1 inhibition in cancer cells also blocked angiogenesis in HUVECs treated with conditioned medium (42). Furthermore, IGF-1R is also involved in the proangiogenic effect of IGFBP-2. IGFBP-2 causes the inactivation of protein tyrosine phosphatase β (RPTP-β) by binding to the RPTP-β receptor and subsequently inhibits the transcription of the tumor suppressor gene PTEN. Inhibition of PTEN mediates the activation of the IGF-I/PI3K/AKT signaling pathway, which in turn promotes vascular smooth muscle proliferation and tumor angiogenesis (43). In addition, other types of IGFBPs can promote or inhibit tumor angiogenesis (44,45). However, the pro-vascular effects of IGFBP seem to be independent of IGF, which provides a new direction for further exploration of the relationship between the IGF system and tumor angiogenesis.
IGFs and tumor autophagy
Autophagy plays a dual role in tumor growth. Early autophagy inhibits cancer progression, but with the continuous growth of tumors, autophagy provides nutrients and energy for tumor survival (46). The basal level autophagy flux is usually associated with tumor inhibition and it is often observed in breast cancer, prostate cancer gastric cancer, hepatocellular carcinoma and other types of cancer in which decreased expression of the-autophagy-associated protein Beclin 1 leads to increased proliferation of tumor cells (47–49). Moreover, deficiency of autophagy regulatory factors such as autophagy-related 4C cysteine peptidase (ATG4C) is more likely to cause cancer (50). However, a number of RAS mutated cancer cells maintain their own growth and metabolism through high levels of autophagy, including those of colorectal cancer and pancreatic cancer (51). IGF signal transduction can activate a number of intracellular kinases to activate and induce a series of reactions, related to apoptosis, autophagy and proliferation (52). In breast cancer cell lines (MCF-7), activation of IGF/PI3K signaling enhances mitochondrial homeostasis by increasing the number of new mitochondria and levels of oxidative phosphorylation and promotes the degradation of damaged mitochondria (mitochondrial autophagy) by increasing BNIP3, a protective mechanism that ultimately influences the cancer treatment response and evolution of the cancer phenotype (53). Moreover, the role of IGF-1 signaling in promoting autophagy has been demonstrated in breast cancer, prostate cancer and osteosarcoma (54). Furthermore, in colorectal cancer, IGF-2 is critical for cancer stem cell formation and IGF-2 preferentially interacts with insulin receptor isoform A rather than with IGF-1R to accelerate autophagy and metabolic remodeling in colorectal cancer (55). IGFBP-3, one of the major members of the insulin-like growth factor binding protein family, has growth inhibitory effects in vitro (56). However, high levels of IGFBP-3 in breast tumor tissues are associated with increased xenograft growth in mice and poor prognosis. Specifically, the binding of IGFBP3 to GRP78 increases autophagic site formation and autophagic system flux, thereby promoting breast cancer cell survival even under glucose starvation and hypoxic conditions (57). By contrast, IGFBP-3 has an oncogenic effect on ovarian cancer cells, reflecting the heterogeneity of tumor tissues and the diverse features of IGFBP-3 functions (58). The pro-autophagic effect of IGFBPs is also reflected in processes such as chemoresistance in hepatocellular carcinoma (59). These studies provide good prospects for in-depth exploration of the regulatory role of the insulin growth factor system in tumor autophagy.
IGFs and tumor metabolic remodeling
The proposed Warburg effect revealed the important role of metabolic reprogramming in cancer (60). To meet the increased demand for energy and substance synthesis, tumor cells change their flux by adjusting various metabolic pathways and targeting metabolic pathways has gradually become a focus of tumor therapy research (61). However, metabolic adaptability and heterogeneity caused by tumor heterogeneity and plasticity limit metabolic efficacy. IGFs, including IGF-1 and IGF-2, are involved in cellular metabolic signaling and influence glucose and cholesterol uptake and glycogen storage (62,63). Circulating levels of IGF-1 and certain IGFBPs are critical for the maintenance of glucose homeostasis (64). Studies have shown that chronic hyperglycemic diets increase the risk of colon cancer, in part by regulating the insulin/IGF-1 signaling axis by activating the downstream PI3K/AKT/mTOR, Ras/MAPK signaling pathways, glucose transporter proteins (GLUT1) and key enzymes of glycolysis (LDHA and HK2), thereby affecting glucose uptake and aerobic glycolysis in cancer cells (65). In breast cancer, the PPP1R1B truncated subtype (t-Darpp) is upregulated in trastuzumab resistant HER2+ breast cancer. t-Darpp activates IGF-1R/AKT signaling through heterodimerization with EGFR and HER2, promoting and stimulating glucose uptake, glycolysis and trastuzumab resistance in SK-BR-3 cells. Pharmacological inhibition and IGF-1R-targeted knockdown reverse the effects of t-Darpp on metabolic remodeling and drug resistance in tumor cells (66). IGFBP family proteins can bind to IGF-1 and IGF-2, thereby regulating the downstream conduction of IGF signals. IGFBP-1 plays an important role in the regulation of IGF-I signaling and influences a series of downstream biological events such as cell proliferation, survival, movement and metabolism (67). It is shown that IGFBP1 expression and secretion were significantly elevated in cancer cells, and secreted IGFBP-1 inhibits AKT1-mediated phosphorylation of ser27 of mitochondrial superoxide dismutase 2 (SOD2), thereby increasing the activity of SOD2 antioxidant enzymes. Increased SOD2 activity weakens the accumulation of mitochondrial reactive oxygen species (ROS) in spatially constrained cancer cells, thereby supporting the survival of tumor cells in blood vessels in lung tissue and accelerating tumor metastasis in mice (68). However, studies of the IGF system in cancer metabolism are still insufficient, not only the uptake and utilization of glucose, but also the mechanism of changes in lipid and amino acid metabolism still need to be explored.
IGFs and tumor immune escape
The tumor microenvironment (TME) is composed mainly of tumor cells, stromal cells, immune cells and the extracellular matrix, which assume the functions of material exchange, environmental stress and immune regulation (69). In the early stage of tumor colonization or growth, activated immune cells contribute to a tumor-suppressive inflammatory microenvironment that hinders tumor development, whereas long-term persistent antigenic stimulation, the activation of immunosuppressive cells and metabolic stress prompt the tumor to escape from the surveillance of the immune system and continue to grow, which is known as immune escape of the tumor (70,71). Modification changes in the tumor cells themselves and changes in the immune microenvironment lead to complexity of the immune escape process. Remodeling the positive immune microenvironment and stimulating or restoring the innate tumor suppression ability of the immune system are crucial for improving the malignant progression of tumors. IGF-1/IGF-1R was found to be critical in regulating the activity of a number of immune cells, including T cells. In a mouse model of hepatocellular carcinoma, regulatory T cells (Tregs) with high IGF-1R expression presented increased PI3K/AKT/mTOR signaling and were able to produce more ATP, lactate and ROS, which contribute to enhanced immunosuppressive effects (72). Tumor-associated macrophages (TAMs), important immunosuppressive cells in the microenvironment, are able to activate the Gli2/IGF-2/ERK1/2 signaling axis to promote TGF-β secretion and thus mediate the migration, invasion and EMT of hepatocellular carcinoma cells (Huh-7 cells) (73). Moreover, M2-type TAMs, through the secretion of IGF-1 and IGF-2, activate PI3K/AKT/mTOR signaling and enhance the malignant proliferation and stemness characteristics of cancer cells (74). In addition, adipose tissue in the microenvironment is also capable of secreting IGF-1 to support microenvironmental remodeling (75). IGF-1 serves as a key mediator that communicates between the microenvironment and cancer cells and targeting IGF-1 has become a critical part of the fight against tumor immunosuppression. Nevertheless, the relationships between IGFs and numerous microenvironmental components, such as NK cells, neutrophils and even the microbiota, remain to be elucidated and incorporating other targets or combination therapies to address the individual and tissue heterogeneity of tumors is crucial. Fig. 1 shows the role of IGF family members in tumor progression.
IGFs regulate signaling pathways associated with cancer progression
During the malignant progression of tumors, a variety of signaling pathways are activated to participate in the proliferation, migration, angiogenesis and metabolic remodeling of tumor cells, among which IGFs play important regulatory roles (76). This section summarizes the key signaling pathways involved in the regulation of IGFs in tumor progression.
IGFs regulate the PI3K/AKT signaling pathway
In an oncogenic context, the IGF family regulates a number of biological processes such as cancer cell proliferation, apoptosis, metabolism and protein synthesis, which are closely associated with the activation of PI3K/AKT signaling, which subsequently promotes the transcription of downstream pro-oncogenic target genes such as c-Myc and HIF-1α (77). In colorectal cancer, IGF-2 secreted by cancer-associated fibroblasts binds to IGF-1R on cancer cells to activate the PI3K/AKT/mTOR and Hippo-YAP1 signaling pathways to promote cancer cell proliferation, migration and invasion; after knockdown of IGF-1R or inhibition of IGF-1R with the IGF-1R inhibitor picropodophyllin, the tumor-promoting effects are reversed (78). In another study, IGF-1 was shown to regulate glucose metabolism in cancer cells with the involvement of kallikrein-related peptidase 10 (KLK10) and the knockdown of KLK10 markedly inhibited glucose metabolism and PI3K/Akt/mTOR signaling activation, a process that could be reversed by IGF-1, suggesting that IGF-1 and KLK10 serve as potential targets for regulating metabolic remodeling in colon cancer (79). As an important noncoding RNA, microRNA (miR)-186-3p is involved in the proliferation, migration and apoptosis of a number of cancer cells, especially cervical cancer cells. It can inhibit the activation of PI3K/AKT signaling through the inverse regulation of IGF-1 expression and ultimately suppress the tumorigenesis of cervical cancer cells (80). Moreover, IGF-1 mediates the activation of the PI3K/AKT/mTOR pathway in uterine smooth muscle tumors, gliomas and pancreatic cancer (81–83). Numerous components of the microenvironment are also involved in malignant tumor progression. M2 macrophages, as infiltration-rich immunosuppressive cells, are able to activate the PI3K/AKT/mTOR signaling axis by secreting IGF-1 and IGF-2, which promotes thyroid cancer cell invasion and the expression of stemness markers (Oct4, SOX2 and CD133), exacerbating the immunosuppressive effects of cancer (74). IGFBP-like protein 1 (IGFBP-L1), a key member of the IGFBP family, regulates its function by binding to IGF (84). In esophageal cancer, the methylation process of IGFBP-L1 was associated with tumor size and TNM stage. Relevant in vivo and in vitro experiments have confirmed that IGFBP-L1 methylation exerts a pro-oncogenic effect by promoting PI3K/AKT phosphorylation and IGFBP-L1 methylation has become a potential marker for the early detection of esophageal cancer, as well as a predictive marker for PI3K-targeted therapy in esophageal cancer (85).
IGFs regulate the NF-κB signaling pathway
NF-κB is a general term for a group of protein complexes, including mainly subunits such as RelA (p65), RelB, c-Rel, p50/p105 (NF-κB1) and p52/p100 (NF-κB2), which play important roles in cell proliferation, immune regulation and the stress response (86). High-level NF-κB activation is involved in tumorigenesis, angiogenesis, microenvironmental remodeling and chemoresistance (87). Activation of the NF-κB pathway under the influence of IGFs mediates the transcription of downstream signals and a range of tumorigenic activities. IGF-1, as a nutrient-responsive growth factor, activates NF-κB and the expression of downstream genes (Ccdn1, Vegf, Birc5 and Ptgs2) to promote the growth of pancreatic cancer in vitro and in vivo (88). Furthermore, the cross-talk between IGF-1 and ROS is involved in the occurrence and development of a variety of cancers, including liver, cervical and colorectal cancers. Further studies revealed that IGF-1 activates the inflammatory signals NF-κB and NLRP3 in cancer cells and that this activation depends on the accumulation of ROS and NOX2. The inhibition of the IGF-1 receptor substrates IRS-1 and NOX2 effectively prevents the development of cancer-related inflammation (89). IGFBP-3, a secreted glycoprotein, can regulate the mitogenic activity of IGF-1R. Recent studies have shown that high expression of IGFBP-3 can increase the radiosensitivity of cancer cells and induce their apoptosis by activating apoptosis-related proteins. Under irradiation, IGFBP-3 induces apoptosis and ROS production by activating NF-κB signaling and ROS further promote IGFBP-3 mediated signaling activity. The positive circuit of NF-κB activation and ROS production can accumulate more ROS in irradiated OSCC cells and this positive feedback regulation overcomes the pro-survival effect of NF-κB/IL-6 signaling (90). In another study, IGFBP-3 was able to enhance etoposide-induced cell growth inhibition by blocking the NF-κB signaling pathway in gastric cancer cells, confirming that IGFBP-3 has become a key target and marker for cancer therapy and further exploration of its in-depth regulatory mechanisms is worthwhile (91).
IGFs regulate the MAPK signaling pathway
MAPK is a key transmitter of signals from the cell surface to the nucleus and can be activated by factors such as cytokines, hormones, stressors and others to regulate cell growth, differentiation, inflammation and other physiological and pathological processes (92). In the context of cancer progression, MAPK signals are involved in various activities of cancer cells, including proliferation, apoptosis and immune escape. However, simply targeting MAPK-related signals has not been effective in treating cancer (93). The function of IGFs in the MAPK signaling pathway provides a new direction in the fight against cancer. IGF-1R is considered a potential cellular oncogene, especially in breast cancer, where high expression with IGF-1R is a driver of the malignant phenotype. Under continuous stimulation of IGF-1, IGF-1R/MAPK/PI3K signaling is activated, leading to resistance to estrogen tamoxifen and fluvestrant. Low doses of tamoxifen act as agonists in IGF-1-stimulated breast cancer cells and further increase IGF-1 expression. Key components involved in the IGF-1/IGF-1R signaling network have become potential targets for combined antiestrogen therapy (94). Ovarian cancer-associated antigen 66 (OVA66) was first shown to play a role in ovarian cancer by reducing IGF-1R expression and downstream phosphorylation of ERK1/2-Hsp27 signaling. In-depth mechanistic studies have shown that OVA66 can interact with MDM2 to coregulate the activation of the IGF-1R-ERK1/2 signaling pathway to promote tumorigenesis (95). Research has shown that type 2 diabetes (T2DM) is associated with an increased risk of colon cancer, along with increased insulin and IGF-1. Insulin and IGF-1 alone or in combination promoted the proliferation of MC38 colon cancer cells and reduced apoptosis. However, the use of ERK1/2 or JNK inhibitors inhibited the growth of colon cancer cells in vivo and in vitro, suggesting that the activation of ERK1/2 and JNK signaling by insulin and IGF-1 is at least partially responsible for the development of T2DM-associated colon cancer (96). These studies effectively confirmed the critical regulatory role of the IGF-1/IGF-1R/MAPK signaling axis in malignant tumor progression.
IGFs regulate the Wnt/β-catenin signaling pathway
The Wnt signaling pathway is mainly mediated by the activation of β-catenin and the sustained accumulation of β-catenin into the nucleus initiates the transcription of target genes by binding to T-cell factor (TCF)/lymphoid enhancer-binding factor transcription factors (97). The Wnt/β-catenin signaling pathway is closely related to stem cell differentiation and organ regeneration. Studies have shown that the activation of Wnt signaling in colorectal cancer is associated with the loss of function of the tumor regulator APC and the involvement of Wnt signaling has been reported in a variety of malignancies, including breast and stomach cancer (98,99). Nevertheless, targeting the Wnt pathway alone presents significant challenges for cancer therapy, including poor drug response and toxic side effects (100). As a receptor for IGF-1 and IGF-2, IGF1R-mediated phosphorylation of Akt and GSK3β promotes β-catenin stability and nuclear localization (101). Moreover, the nuclear localization of IGF-1R is mediated mainly by its C-terminal domain. Following its nuclear localization, IGF-1R promotes TCF-mediated β-catenin transcriptional activity, which has been confirmed in hepatocellular carcinoma cells (102). In colorectal cancer cell lines (HT-29 and SW620), knockdown of IGF-1R by small interfering RNA resulted in a blockade of the downstream PI3K/Akt and typical Wnt signaling pathways, which ultimately inhibited cancer cell proliferation and promoted apoptosis (103). To identify IGF-1-mediated miRNA regulatory networks that cause temozolomide (TMZ) to be insensitive to glioblastoma multiforme treatment, on the basis of comprehensive analysis of multiple databases and in vitro experiments, Chen et al (104) confirmed that IGF-1 upregulated miR-513a-5p signaling reduced the sensitivity of glioma cells to TMZ by inhibiting the NEDD4L-inactivated Wnt/β-catenin pathway. The mining of this signaling pathway provides a feasible target for improving the drug sensitivity of TMZ.
IGFs regulate the COX-2 signaling pathway
Cyclooxygenase-2 (COX-2) is a key regulatory molecule that catalyzes the synthesis of arachidonic acid to prostaglandin (PG), which is expressed in large quantities when cells are stimulated by inflammation, accelerating the occurrence of inflammatory storms (105). COX-2 is highly expressed in most tumors and promotes tumor proliferation, migration and angiogenesis (106). Moreover, the overexpression of COX-2 is often closely associated with chemotherapy resistance and immune escape processes in tumors, suggesting that COX-2 is an attractive therapeutic target in tumors (107). Nevertheless, the regulation of COX-2-related signaling pathways remains to be elucidated. The IGF-1/IGF-1R system has been shown to be an important promoter of tumor growth in various cancers, promoting tumor progression and metastasis by regulating multiple signaling pathways, such as those of PI3K/AKT and MAPK/ERK signaling (78,94). Notably, COX-2 expression is also affected by the IGF-1/IGF-1R signaling pathway (108). In a study of colon cancer cells, COX-2 expression and PGE2 synthesis were upregulated by the IGF-2/IGF-1R autocrine pathway and IGF-1R blockade decreased COX-2 activity and inhibited tumor cell proliferation and promoted apoptosis (103). Stoeltzing et al (109) reported that IGF-I selectively upregulates COX-2 through the MAPK/(ERK1/2) pathway in pancreatic cancer and that treatment with anti-IGF-IR antibodies can effectively inhibit IGF-IR and MAPK/ERK activation and reduce COX-2 expression in parental cells. Furthermore, in a BK5.IGF-1 mouse model of breast cancer, elevated levels of IGF-1 expression activated the COX-2/PGE2/EP3 signaling pathway, accompanied by increased VEGF expression and tumor angiogenesis (110). Celecoxib treatment resulted in a 45% reduction in mammary PGE2 levels and attenuated mast cell influx and angiogenesis, suggesting that COX-2-selective inhibitors may be useful in the prevention or treatment of breast cancer associated with elevated human IGF-1 levels (110). As a member of the IGFBP family, IGFBP-4 influences the inflammatory regulation of the tumor microenvironment. In lung cancer tissues, the expression of IGFBP-4 was markedly lower than that in normal tissues adjacent to the cancer, but the expression of COX-2 was greater in lung cancer tissues. In addition to inhibiting the expression of COX-2 in lung cancer cells, IGFBP-4 also inhibited the proliferation, migration and metastasis of cancer cells through the modulation of the PI3K/AKT, ERK and CREB pathways, which highlights the anticancer value of IGFBP-4 (111). These findings suggest that the regulation of the COX-2-related signaling axis by the IGF system could be a potential target for cancer therapy and deserves further exploration. Fig. 2 shows the key signaling pathways involved in the regulation of IGFs in tumor progression.
Application of IGFs in cancer therapy
The treatment of cancer is very complex and IGF-related signals affect multiple processes of cancer cell proliferation, invasion and metastasis. Moreover, IGFs also mediate tumor resistance to chemotherapy and confer resistance to immunotherapy (112). Therefore, IGFs are promising targets for cancer treatment. This section focusses on the application and related mechanisms of targeting IGFs in cancer therapy.
Targeted therapy
Targeting the IGF-1R signaling pathway is considered a potential breakthrough in anticancer therapy and a number of small molecule inhibitors and monoclonal antibodies targeting IGF-1R, such as BIIB-022, BMS-754807 and Teprotumumab, have been developed and evaluated in clinical trials (113–115). However, owing to their limited anticancer activity or drug toxicity, a number of clinical trials targeting IGF-1R have been abandoned and few drugs have actually entered clinical use. The unique advantages that natural compounds have in cancer progression make targeting IGF-1R possible. For example, the combination of curcumin and resveratrol inhibits NF-κB activity by targeting IGF-1R signaling, ultimately leading to apoptosis and cell cycle arrest in natural colon cancer cells, suggesting that IGF-1R may be an anticancer approach (116). Quercetin, which is abundant in fruits, vegetables, leaves and grains, can also inhibit skin cancer proliferation by targeting IGF-1R (117). Epigallocatechin gallate, a polyphenolic component of green tea, inhibits the progression of various cancers, including glioma, breast and liver cancers, by inhibiting IGF-1R via the phosphorylation of the tyrosine kinase IGF-1R (118–120). Targeting circulating IGF-1 and IGF-2 levels is another anticancer strategy. However, although serum IGF levels are closely associated with tumor progression, IGF also plays an important regulatory role in normal life activities, such as maintaining skeletal muscle growth and development, islet proliferation and cell metabolism. How to maximally inhibit tumor growth without interfering with normal physiological activities is a key issue in the development of IGF-1/2 inhibitors. For example, MEDI-573 inhibits IGF-1R signaling and tumor growth in vivo by neutralizing IGF-1 and IGF-2. MEDI-573 offers a potential targeted treatment strategy for cancer (121). Numerous studies have shown that IGFBPs can also affect the malignant biological behavior of tumors in an IGF-independent manner. Therefore, targeting IGFBPs has also become an IGFs-based cancer therapy (122,123). α1-Antitrypsin (AAT) is a member of the serine protease inhibitor superfamily and has anti-inflammatory and tissue protective effects. It can reduce colitis and chronic ileitis by inhibiting cytokine production and enhancing intestinal barrier function. In a mouse model of colon cancer, the level of IGFBP-3 decreased markedly under the action of serine protease and the use of AAT reversed this outcome, resulting anticancer effects (124). Several studies have shown that METTL3 plays important regulatory roles in prostate cancer proliferation, migration, invasion, apoptosis, drug resistance androgen-induced splicing and glycolipid metabolism maintenance (125,126). The inhibitor STM2457 can reduce the m6A level in cancer cells by inhibiting the IGFBP3/AKT signaling axis, thus exerting anticancer effects in vitro and in vivo (127). These studies confirm that targeting IGFs is an important strategy for cancer therapy.
Chemotherapy
For most cancers, chemotherapy is the mainstay of late-stage intervention, a treatment strategy that helps improve prognosis and overall survival. However, with the frequent occurrence of drug resistance, chemotherapeutic agents have become less effective in treating cancer (128,129). Therefore, researchers have begun to explore the potential mechanisms of tumor drug resistance with the aim of improving the efficacy of chemotherapy. Studies have confirmed that IGFs play important roles in drug resistance in tumors, including the IGF-1/IGF-2/IGF-1R signaling axis and the IGFBP family members that reduce the susceptibility of cancers such as lung and breast cancers to chemotherapy resistance (130,131). Therefore, targeting IGFs may help solve the problem of tumor drug resistance. Her2-positive breast cancer resistant to trastuzumab therapy severely affects prognosis. In trastuzumab-resistant Her2-positive breast cancer cells, IGFBP-3 expression was reduced, leading to the inhibition of Wnt signaling pathway release and increased Cullin7 expression mediated by TCF7L2. Cullin7 was subsequently involved in the degradation of IRS-1 in an mTOR/S6K-dependent manner to increase drug resistance. Intervention with IGFBP-3 or Cullin7 partially restored trastuzumab sensitivity in trastuzumab-resistant Her2-positive breast cancer cells, which is important for selecting the optimal therapeutic strategy for Her2-positive breast cancer (132). Tamoxifen, a selective estrogen receptor modulator and antagonist of ERα in breast tissue, is a commonly used adjuvant therapy for patients with ERα-positive breast cancer (133). However, tamoxifen resistance is becoming more common. Studies have shown that tamoxifen resistance is associated with IGFBP-1 accumulation and that the overexpression of IGFBP-1 promotes tamoxifen resistance in breast cancer cells by activating the ERK pathway, which can be reversed by knocking down IGFBP-1 (134). Moreover, in hepatocellular carcinoma (HCC), antiangiogenic tyrosine kinase inhibitors (TKIs) are effective therapeutic agents and the main therapeutic effect is to induce severe hypoxia in the tumor microenvironment (TME) through depletion of the vascular density of tumor. However, patients with HCC often develop resistance to TKIs and this resistance development is associated with increased IGFBP-1 expression, the aggregation mechanism manifested by TKI-induced hypoxia increasing IGFBP-1 expression through activation of HIF-1α and HIF-2α. Tumor-derived IGFBP-1 induces tumor angiogenesis by activating integrin α5β1/focal adhesion kinase/extracellular signal-regulated kinase signaling. The acquired resistance of tumor cells to TKIs is partially reversed by inhibiting IGFBP-1 and the combination of antiangiogenic TKIs and IGFBP-1 inhibitors may be a promising therapeutic strategy for HCC (135). IGFBP-2 is a secreted protein that prevents IGF-1/IGF-2 from binding to its receptor and it also participates in the regulation of the TME in a macrophage-dependent manner. IGFBP-2 is highly expressed in the blood of lung tumors and patients with lung cancer and high levels of IGFBP-2 are associated with poor survival and metastasis in patients with lung cancer. In vitro and in vivo experiments have shown that IGFBP-2 plays an important role in the acquisition of gefitinib resistance. Mechanistically, IGFBP-2 can activate STAT3 to increase the transcriptional activity of C-X-C motif ligand 1 (CXCL1), thereby increasing the intracellular expression level of CXCL1, which contributes to the survival of lung cancer cells in the gefitinib environment (136). The aforementioned results suggest the potential of IGFBP-2 as a biomarker of gefitinib resistance and a potential target for intervention. In addition, decreased survival of human lung cancer cells is associated with increased IGFBP-3 expression. IGFBP-3 plays an anticancer role by exerting cytotoxic effects on cell survival through a mechanism dependent on the interaction between the glycosaminoglycan hyaluronic acid (HA) and CD44. Loss of IGFBP-3 expression decreases the sensitivity of lung cancer cells to cisplatin. Casein kinase 2 (CK2) is an antiapoptotic kinase that maintains cell survival. Phosphorylation of IGFBP-3 by CK2 blocks the binding of IGFBP-3 to HA, activates HA-CD44 signaling and leads to reduced apoptosis, increased cell survival and cisplatin resistance. Blocking CK2 and IGFBP-3 phosphorylation may be an effective strategy to increase lung cancer susceptibility to cisplatin (137). Therefore, further exploration of the mechanism of IGFs in tumor drug resistance is important for improving tumor sensitivity to chemotherapy drugs.
Immunotherapy
Immunotherapy is another innovative cancer treatment strategy following surgery, chemoradiotherapy and targeted therapy, opening a new era of cancer treatment. This means that working on cancer cells alone will not achieve the goal of completely eliminating the tumor and new treatment strategies should consider the TME, that is, the surrounding immune cell components. Some inherent mechanisms of the tumor itself help tumor cells escape the surveillance and killing effects of the immune system, so tumor immune escape is also one of the bottlenecks to improving the current therapeutic effect on tumors (138). How to solve the immune escape and secondary drug resistance of tumor has become a difficult problem for the wide application of tumor immunotherapy. Ovarian cancer is the deadliest gynecological malignancy. Immune checkpoint inhibitors have shown good therapeutic efficacy in most malignancies, but have limited efficacy in patients with ovarian cancer. The main reason is that the large amount of extracellular matrix deposition in the ovarian cancer microenvironment leads to tumor vascular collapse, reduced vascular perfusion, poor drug delivery and blocked migration of cytotoxic T cells to the tumor area (139). As a widely used antihypertensive drug, losartan enhances vascular perfusion, thereby enhancing drug delivery and intratumoral invasion of immune effector cells and, on the other hand, enhances chemotherapy sensitivity by inhibiting IGF-1 signaling to reshape ovarian cancer and the microenvironment (140). Several studies have shown that IGF-2 in the tumor microenvironment is derived mainly from CAFs and that high levels of IGF-2 inhibit the infiltration and cytotoxicity of CD8+ T cells, exacerbating the immunosuppressive effect of tumors (78,141). Mechanistically, autocrine IGF-2 promotes self-activation by binding to the IGF-1 receptor (IGF-1R) on CAFs and activating PI3K/AKT signaling, followed by the secretion of various chemokines and cytokines (CCL5 and CXCL12) by CAFs to influence the infiltration of T cell. Furthermore, CAFs interact with T cells via the PD-1/PD-L1 and CD73/adenosine axes and inhibit their activation, proliferation and effector responses. Genetic inhibition or the targeted inhibitor of IGF-2, lincitinib, markedly enhances the response to immune checkpoint blockade, suggesting the potential of IGF-2 as a biomarker and therapeutic target in immunotherapy (141). In a mouse model of pancreatic cancer liver metastasis, the use of IGF-IR inhibitor IGF-Trap reshaped the local immunosuppressive microenvironment of liver tumors, reduced the recruitment of bone marrow-derived suppressor cells, reversed innate immune cell polarization and inhibited metastatic expansion. Moreover, when IGF-1R was combined with an anti-PD-1 antibody, the growth of experimental pancreatic ductal adenocarcinoma liver metastases was inhibited and the response of T cell was further enhanced. These results suggest that blocking IGFs has the dual effects of reshaping the immune microenvironment and enhancing immunotherapy (142). Current research confirms that single-agent immunotherapy is not advantageous in cancer treatment and that cotargeting IGFs offers a new therapeutic strategy to improve the efficacy of immunotherapy.
Radiotherapy
Radiotherapy plays a pivotal role in controlling and eradicating tumors as an adjuvant cancer treatment, either alone or in combination with other modalities (surgery, chemotherapy, immunotherapy and targeted therapy) (143). Despite the continuous progress in radiation technology, which allows for more precise radiotherapy of local tumor tissues while reducing the effect on normal tissues, problems such as radioresistance and tumor recurrence remain major challenges in the application of radiation therapy (144). Improving the sensitivity of tumor tissues to radiotherapy is the main direction of current research. Studies have shown that IGFs are closely related to radiation response and tumor radiosensitivity. Among them, IGF/IGF-1R signals can improve radiotherapy sensitivity by activating a series of signal transduction events involved in DNA damage repair. In colon cancer cells (HT-29 and SW480 cells), genetic knockout of IGF-1R or the use of the IGF-1R inhibitor NVP-ADW742 enhanced the killing effect of radiation on cancer cells, confirming that depletion of IGF-1R improves radiosensitivity to colon cancer therapy (145,146). Antrocin is a sesquiterpene lactone isolated from camphor that is used as a dietary supplement for cancer prevention and liver protection. In addition, antrocin has been shown to effectively antagonize a variety of cancers, including breast, lung, liver and colon cancers (147,148). In prostate cancer, the combination of antrocin and ionizing radiation (IR) synergistically inhibits the proliferation of radioresistant prostate cancer cells and induces apoptosis. Specifically, antrocin regulates the cell cycle and apoptosis through the inhibition of β-catenin mediated by IGF-1R, suggesting the potential value of antrocin as a potent therapeutic agent to overcome radioresistance (149). Due to the heterogeneity of tumor tissues, the challenges posed by radioresistance are increasingly significant. An in-depth exploration of the important role of IGFs in tumor radioresistance can provide potential therapeutic targets for improving tumor radiosensitivity. The application of targeted IGFs in cancer therapies and related mechanisms of action are summarized in Table I.
![]() | Table I.Summary of studies on the application of targeted IGFs in cancer therapies and related mechanisms of action. |
Summary and prospects
Cancer is a major social, economic and public safety issue in the 21st century and increasing morbidity and mortality rates have placed great burdens on the worldwide population. Despite advances in technology, the treatment and prevention of cancer are still in their infancy. Recently, as the function of IGFs has gradually been revealed, the important role that IGFs play in cancer progression has brought new hope for cancer treatment. The present review introduced the role of IGFs in cancer and their molecular mechanism, focusing on the application of IGFs in current cancer therapies, with the goal of providing a theoretical basis for comprehensive cancer diagnosis and treatment.
Inhibition of IGF-1R signaling is considered a promising strategy for inhibiting tumor growth and improving survival rates in a variety of cancers. However, several drugs, including teprotumumab and BIIB-022, which target IGF-1R, have not shown good therapeutic effects in clinical trials, possibly due to the lack of reliable IGF-1R inhibition biomarkers. Moreover, IGF-related molecular mechanisms not only play a role in tumor cells but also affect other nontumor cell components in the microenvironment, such as fibroblasts, macrophages and T cells, demonstrating that targeting IGFs alone cannot completely inhibit tumor growth and progression. Furthermore, owing to the heterogeneity and diversity of tumor cells themselves, the role of IGFs is not necessarily only as a tumor suppressor, so the development of related inhibitors must fully consider the impact on the tumor and its surrounding environment. With the advance of combination therapy, the combination of targeted IGFs and other technologies for the treatment of tumors has shown advantages, which not only increase the efficacy but also effectively prevent the occurrence of drug resistance. Future research should focus on exploring the use of combination therapy in cancer treatment. Moreover, with the advancement of multiomics technology, the importance of precision therapy and individualized treatment is increasingly emphasized and multidimensional and different levels of cancer-related mechanisms need to be explored to develop more reliable inhibitors, which, combined with a more rational target delivery mechanism, is the direction of cancer treatment in the future. In conclusion, targeting IGFs can provide more therapeutic options for cancer patients.
Acknowledgements
Not applicable.
Funding
The present study was supported by National Natural Science Foundation of China (grant no. 82260555) and The First Hospital of Lanzhou University Intra-Hospital Fund (grant no. ldyyyn2020-09).
Availability of data and materials
Not applicable.
Authors' contributions
DW prepared and wrote the original draft and was responsible for supervision and project administration. SD prepared and wrote the original draft and was responsible for visualization. WZ prepared and wrote the original draft and was responsible for supervision, project administration and funding acquisition. Data authentication is not applicable. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
IGF |
insulin-like growth factor |
IGFBP |
IGF-binding protein |
EGFR |
epidermal growth factor receptor |
IFITM |
interferon-induced transmembrane protein |
Cyr61 |
cysteine-rich 61 |
MMP |
matrix metalloproteinase |
MCSF |
macrophage colony-stimulating factor |
HUVEC |
human umbilical vein endothelial cells |
RPTP-β |
protein tyrosine phosphatase β |
ATG4C |
autophagy related 4C cysteine peptidase |
t-Darpp |
PPP1R1B truncated subtype |
SOD2 |
superoxide dismutase 2 |
ROS |
reactive oxygen species |
TAM |
tumor-associated macrophage |
ERα |
estrogen receptor α |
TME |
tumor microenvironment |
KLK10 |
kallikrein-related peptidase 10 |
IGFBP-L1 |
IGFBP-like protein 1 |
OVA66 |
ovarian cancer-associated antigen 66 |
T2DM |
type 2 diabetes |
TMZ |
temozolomide |
COX-2 |
cyclooxygenase-2 |
PG |
prostaglandin |
AAT |
α1-antitrypsin |
HCC |
hepatocellular carcinoma |
TKI |
tyrosine kinase inhibitor |
HA |
hyaluronic acid |
CK2 |
casein kinase 2 |
IR |
ionizing radiation |
References
Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024. View Article : Google Scholar : PubMed/NCBI | |
Jassim A, Rahrmann EP, Simons BD and Gilbertson RJ: Cancers make their own luck: Theories of cancer origins. Nat Rev Cancer. 23:710–724. 2023. View Article : Google Scholar : PubMed/NCBI | |
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI | |
Forbes BE, Blyth AJ and Wit JM: Disorders of IGFs and IGF-1R signaling pathways. Mol Cell Endocrinol. 518:1110352020. View Article : Google Scholar : PubMed/NCBI | |
Froesch ER, Buergi H, Ramseier EB, Bally P and Labhart A: antibody-suppressible and nonsuppressible insulin-like activities in human serum and their physiologic significance. an insulin assay with adipose tissue of increased precision and specificity. J Clin Invest. 42:1816–1834. 1963. View Article : Google Scholar : PubMed/NCBI | |
Rinderknecht E and Humbel RE: The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin. J Biol Chem. 253:2769–2776. 1978. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Zhu Q, Cao D, Peng Q, Zhang X, Li C, Zhang C, Zhou BO and Yue R: Bone marrow-derived IGF-1 orchestrates maintenance and regeneration of the adult skeleton. Proc Natl Acad Sci USA. 120:e22037791202023. View Article : Google Scholar : PubMed/NCBI | |
Guan X, Yan Q, Wang D, Du G and Zhou J: IGF-1 signaling regulates mitochondrial remodeling during myogenic differentiation. Nutrients. 14:12492022. View Article : Google Scholar : PubMed/NCBI | |
Matsushita M, Fujita K, Hatano K, De Velasco MA, Uemura H and Nonomura N: Connecting the dots between the gut-IGF-1-prostate axis: A role of IGF-1 in prostate carcinogenesis. Front Endocrinol (Lausanne). 13:8523822022. View Article : Google Scholar : PubMed/NCBI | |
LeRoith D, Holly JMP and Forbes BE: Insulin-like growth factors: Ligands, binding proteins and receptors. Mol Metab. 52:1012452021. View Article : Google Scholar : PubMed/NCBI | |
Dixit M, Poudel SB and Yakar S: Effects of GH/IGF axis on bone and cartilage. Mol Cell Endocrinol. 519:1110522021. View Article : Google Scholar : PubMed/NCBI | |
Vassilakos G, Lei H, Yang Y, Puglise J, Matheny M, Durzynska J, Ozery M, Bennett K, Spradlin R, Bonanno H, et al: Deletion of muscle IGF-I transiently impairs growth and progressively disrupts glucose homeostasis in male mice. FASEB J. 33:181–194. 2019. View Article : Google Scholar : PubMed/NCBI | |
Alberini CM and Chen DY: Memory enhancement: Consolidation, reconsolidation and insulin-like growth factor 2. Trends Neurosci. 35:274–283. 2012. View Article : Google Scholar : PubMed/NCBI | |
Alfares MN, Perks CM, Hamilton-Shield JP and Holly JMP: Insulin-like growth factor-II in adipocyte regulation: Depot-specific actions suggest a potential role limiting excess visceral adiposity. Am J Physiol Endocrinol Metab. 315:E1098–E1107. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ghafari F, Alizadeh AM, Agah S, Irani S and Mokhtare M: Insulin-like growth factor 1 serum levels in different stages of gastric cancer and their association with Helicobacter pylori status. Peptides. 158:1708922022. View Article : Google Scholar : PubMed/NCBI | |
Kasprzak A: Autophagy and the insulin-like growth factor (IGF) system in colonic cells: Implications for colorectal neoplasia. Int J Mol Sci. 24:36652023. View Article : Google Scholar : PubMed/NCBI | |
Adachi Y, Nojima M, Mori M, Himori R, Kubo T, Akutsu N, Lin Y, Kurozawa Y, Wakai K and Tamakoshi A; Japan Collaborative Cohort Study, : Insulin-like growth factor 2 and incidence of liver cancer in a nested case-control study. Cancer Epidemiol Biomarkers Prev. 30:2130–2135. 2021. View Article : Google Scholar : PubMed/NCBI | |
Stefani C, Miricescu D, Stanescu-Spinu II, Nica RI, Greabu M, Totan AR and Jinga M: Growth Factors, PI3K/AKT/mTOR and MAPK Signaling Pathways in Colorectal Cancer Pathogenesis: Where Are We Now? Int J Mol Sci. 22:102602021. View Article : Google Scholar : PubMed/NCBI | |
Feng L, Li B, Xi Y, Cai M and Tian Z: Aerobic exercise and resistance exercise alleviate skeletal muscle atrophy through IGF-1/IGF-1R-PI3K/Akt pathway in mice with myocardial infarction. Am J Physiol Cell Physiol. 322:C164–C176. 2022. View Article : Google Scholar : PubMed/NCBI | |
Fang WY, Tseng YT, Lee TY, Fu YC, Chang WH, Lo WW, Lin CL and Lo YC: Triptolide prevents LPS-induced skeletal muscle atrophy via inhibiting NF-κB/TNF-α and regulating protein synthesis/degradation pathway. Br J Pharmacol. 178:2998–3016. 2021. View Article : Google Scholar : PubMed/NCBI | |
de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 41:374–403. 2023. View Article : Google Scholar : PubMed/NCBI | |
Paredes F, Williams HC and San Martin A: Metabolic adaptation in hypoxia and cancer. Cancer Lett. 502:133–142. 2021. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Liu J, Tian Z, Zhang Z, Liu T, Chen C, Tang X and Zhu J: Highly expressed IFITM10 is associated with early diagnosis and T stage of gastric cancer. Transl Cancer Res. 10:382–392. 2021. View Article : Google Scholar : PubMed/NCBI | |
Seo GS, Lee JK, Yu JI, Yun KJ, Chae SC and Choi SC: Identification of the polymorphisms in IFITM3 gene and their association in a Korean population with ulcerative colitis. Exp Mol Med. 42:99–104. 2010. View Article : Google Scholar : PubMed/NCBI | |
Xu L, Zhou R, Yuan L, Wang S, Li X, Ma H, Zhou M, Pan C, Zhang J, Huang N, et al: IGF1/IGF1R/STAT3 signaling-inducible IFITM2 promotes gastric cancer growth and metastasis. Cancer Lett. 393:76–85. 2017. View Article : Google Scholar : PubMed/NCBI | |
Le Coz V, Zhu C, Devocelle A, Vazquez A, Boucheix C, Azzi S, Gallerne C, Eid P, Lecourt S and Giron-Michel J: IGF-1 contributes to the expansion of melanoma-initiating cells through an epithelial-mesenchymal transition process. Oncotarget. 7:82511–82527. 2016. View Article : Google Scholar : PubMed/NCBI | |
Sarkissyan S, Sarkissyan M, Wu Y, Cardenas J, Koeffler HP and Vadgama JV: IGF-1 regulates Cyr61 induced breast cancer cell proliferation and invasion. PLoS One. 9:e1035342014. View Article : Google Scholar : PubMed/NCBI | |
Chiu YF, Wu CC, Kuo MH, Miao CC, Zheng MY, Chen PY, Lin SC, Chang JL, Wang YH and Chou YT: Critical role of SOX2-IGF2 signaling in aggressiveness of bladder cancer. Sci Rep. 10:82612020. View Article : Google Scholar : PubMed/NCBI | |
Tsai YF, Chou HC, Liou MH, Liao EC, Cheng CT, Chang SJ and Chan HL: Role of IGFBP-2 in oral cancer metastasis. Biochim Biophys Acta Mol Basis Dis. 1867:1661432021. View Article : Google Scholar : PubMed/NCBI | |
Luo C, Sun F, Zhu H, Ni Y, Fang J, Liu Y, Shao S, Shen H and Hu J: Insulin-like growth factor binding protein-1 (IGFBP-1) upregulated by Helicobacter pylori and is associated with gastric cancer cells migration. Pathol Res Pract. 213:1029–1036. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mo W, Deng L, Cheng Y, Ge S and Wang J: IGFBP7 regulates cell proliferation and migration through JAK/STAT pathway in gastric cancer and is regulated by DNA and RNA methylation. J Cell Mol Med. 28:e700802024. View Article : Google Scholar : PubMed/NCBI | |
Liu ZL, Chen HH, Zheng LL, Sun LP and Shi L: Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 8:1982023. View Article : Google Scholar : PubMed/NCBI | |
Ren F, Wu K, Yang Y, Yang Y, Wang Y and Li J: Dandelion polysaccharide exerts anti-angiogenesis effect on hepatocellular carcinoma by regulating VEGF/HIF-1α expression. Front Pharmacol. 11:4602020. View Article : Google Scholar : PubMed/NCBI | |
Higashi Y, Pandey A, Goodwin B and Delafontaine P: Insulin-like growth factor-1 regulates glutathione peroxidase expression and activity in vascular endothelial cells: Implications for atheroprotective actions of insulin-like growth factor-1. Biochim Biophys Acta. 1832:391–399. 2013. View Article : Google Scholar : PubMed/NCBI | |
Bid HK, Zhan J, Phelps DA, Kurmasheva RT and Houghton PJ: Potent inhibition of angiogenesis by the IGF-1 receptor-targeting antibody SCH717454 is reversed by IGF-2. Mol Cancer Ther. 11:649–659. 2012. View Article : Google Scholar : PubMed/NCBI | |
Glass K, Fines C, Coulter P, Jena L, McCarthy HO and Buckley N: Development and characterization of a peptide-bisphosphonate nanoparticle for the treatment of breast cancer. Mol Pharm. 21:4970–4982. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ishikawa T: Differences between zoledronic acid and denosumab for breast cancer treatment. J Bone Miner Metab. 41:301–306. 2023. View Article : Google Scholar : PubMed/NCBI | |
Tang X, Zhang Q, Shi S, Yen Y, Li X, Zhang Y, Zhou K and Le AD: Bisphosphonates suppress insulin-like growth factor 1-induced angiogenesis via the HIF-1alpha/VEGF signaling pathways in human breast cancer cells. Int J Cancer. 126:90–103. 2010. View Article : Google Scholar : PubMed/NCBI | |
Seo SH, Hwang SY, Hwang S, Han S, Park H, Lee YS, Rho SB and Kwon Y: Hypoxia-induced ELF3 promotes tumor angiogenesis through IGF1/IGF1R. EMBO Rep. 23:e529772022. View Article : Google Scholar : PubMed/NCBI | |
Liu X, He H, Zhang F, Hu X, Bi F, Li K, Yu H, Zhao Y, Teng X, Li J, et al: m6A methylated EphA2 and VEGFA through IGF2BP2/3 regulation promotes vasculogenic mimicry in colorectal cancer via PI3K/AKT and ERK1/2 signaling. Cell Death Dis. 13:4832022. View Article : Google Scholar : PubMed/NCBI | |
Slater T, Haywood NJ, Matthews C, Cheema H and Wheatcroft SB: Insulin-like growth factor binding proteins and angiogenesis: From cancer to cardiovascular disease. Cytokine Growth Factor Rev. 46:28–35. 2019. View Article : Google Scholar : PubMed/NCBI | |
Nijaguna MB, Patil V, Urbach S, Shwetha SD, Sravani K, Hegde AS, Chandramouli BA, Arivazhagan A, Marin P, Santosh V and Somasundaram K: Glioblastoma-derived macrophage colony-stimulating factor (MCSF) induces microglial release of insulin-like growth factor-binding protein 1 (IGFBP1) to promote angiogenesis. J Biol Chem. 290:23401–23415. 2015. View Article : Google Scholar : PubMed/NCBI | |
Shen X, Xi G, Wai C and Clemmons DR: The coordinate cellular response to insulin-like growth factor-I (IGF-I) and insulin-like growth factor-binding protein-2 (IGFBP-2) is regulated through vimentin binding to receptor tyrosine phosphatase β (RPTPβ). J Biol Chem. 290:11578–11590. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ma YS, Shi BW, Guo JH, Liu JB, Yang XL, Xin R, Shi Y, Zhang DD, Lu GX, Jia CY, et al: microRNA-320b suppresses HNF4G and IGF2BP2 expression to inhibit angiogenesis and tumor growth of lung cancer. Carcinogenesis. 42:762–771. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wei LF, Weng XF, Huang XC, Peng YH, Guo HP and Xu YW: IGFBP2 in cancer: Pathological role and clinical significance (Review). Oncol Rep. 45:427–438. 2021. View Article : Google Scholar : PubMed/NCBI | |
Li X, He S and Ma B: Autophagy and autophagy-related proteins in cancer. Mol Cancer. 19:122020. View Article : Google Scholar : PubMed/NCBI | |
Li X, Yang KB, Chen W, Mai J, Wu XQ, Sun T, Wu RY, Jiao L, Li DD, Ji J, et al: CUL3 (cullin 3)-mediated ubiquitination and degradation of BECN1 (beclin 1) inhibit autophagy and promote tumor progression. Autophagy. 17:4323–4340. 2021. View Article : Google Scholar : PubMed/NCBI | |
Huangfu L, Wang X, Tian S, Chen J, Wang X, Fan B, Yao Q, Wang G, Chen C, Han J, et al: Piceatannol enhances Beclin-1 activity to suppress tumor progression and its combination therapy strategy with everolimus in gastric cancer. Sci China Life Sci. 66:298–312. 2023. View Article : Google Scholar : PubMed/NCBI | |
Filali-Mouncef Y, Hunter C, Roccio F, Zagkou S, Dupont N, Primard C, Proikas-Cezanne T and Reggiori F: The ménage à trois of autophagy, lipid droplets and liver disease. Autophagy. 18:50–72. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wen ZP, Zeng WJ, Chen YH, Li H, Wang JY, Cheng Q, Yu J, Zhou HH, Liu ZZ, Xiao J and Chen XP: Knockdown ATG4C inhibits gliomas progression and promotes temozolomide chemosensitivity by suppressing autophagic flux. J Exp Clin Cancer Res. 38:2982019. View Article : Google Scholar : PubMed/NCBI | |
Masliah-Planchon J, Garinet S and Pasmant E: RAS-MAPK pathway epigenetic activation in cancer: miRNAs in action. Oncotarget. 7:38892–38907. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chen PC, Kuo YC, Chuong CM and Huang YH: Niche modulation of IGF-1R signaling: its role in stem cell pluripotency, cancer reprogramming and therapeutic applications. Front Cell Dev Biol. 8:6259432021. View Article : Google Scholar : PubMed/NCBI | |
Lyons A, Coleman M, Riis S, Favre C, O'Flanagan CH, Zhdanov AV, Papkovsky DB, Hursting SD and O'Connor R: Insulin-like growth factor 1 signaling is essential for mitochondrial biogenesis and mitophagy in cancer cells. J Biol Chem. 292:16983–16998. 2017. View Article : Google Scholar : PubMed/NCBI | |
Riis S, Murray JB and O'Connor R: IGF-1 signalling regulates mitochondria dynamics and turnover through a conserved GSK-3β-Nrf2-BNIP3 pathway. Cells. 9:1472020. View Article : Google Scholar : PubMed/NCBI | |
Gao T, Liu X, He B, Pan Y and Wang S: IGF2 loss of imprinting enhances colorectal cancer stem cells pluripotency by promoting tumor autophagy. Aging (Albany NY). 12:21236–21252. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cai Q, Dozmorov M and Oh Y: IGFBP-3/IGFBP-3 receptor system as an anti-tumor and anti-metastatic signaling in cancer. Cells. 9:12612020. View Article : Google Scholar : PubMed/NCBI | |
Grkovic S, O'Reilly VC, Han S, Hong M, Baxter RC and Firth SM: IGFBP-3 binds GRP78, stimulates autophagy and promotes the survival of breast cancer cells exposed to adverse microenvironments. Oncogene. 32:2412–2420. 2013. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Shao C, Liu J, Sun H, Yao B, Ma C, Xu H and Zhu W: ULK2 suppresses ovarian cancer cell migration and invasion by elevating IGFBP3. PeerJ. 12:e176282024. View Article : Google Scholar : PubMed/NCBI | |
Lin JC, Liu TP, Chen YB and Yang PM: PF-429242 exhibits anticancer activity in hepatocellular carcinoma cells via FOXO1-dependent autophagic cell death and IGFBP1-dependent anti-survival signaling. Am J Cancer Res. 13:4125–4144. 2023.PubMed/NCBI | |
Xia P, Zhang H, Lu H, Xu K, Jiang X, Jiang Y, Gongye X, Chen Z, Liu J, Chen X, et al: METTL5 stabilizes c-Myc by facilitating USP5 translation to reprogram glucose metabolism and promote hepatocellular carcinoma progression. Cancer Commun (Lond). 43:338–364. 2023. View Article : Google Scholar : PubMed/NCBI | |
Jing Z, Liu Q, He X, Jia Z, Xu Z, Yang B and Liu P: NCAPD3 enhances Warburg effect through c-myc and E2F1 and promotes the occurrence and progression of colorectal cancer. J Exp Clin Cancer Res. 41:1982022. View Article : Google Scholar : PubMed/NCBI | |
Okuyama T, Kyohara M, Terauchi Y and Shirakawa J: The roles of the IGF axis in the regulation of the metabolism: interaction and difference between insulin receptor signaling and IGF-I receptor signaling. Int J Mol Sci. 22:68172021. View Article : Google Scholar : PubMed/NCBI | |
Ravera S, Puddu A, Bertola N, Verzola D, Russo E, Maggi D and Panfoli I: IGF-1 signaling modulates oxidative metabolism and stress resistance in ARPE-19 cells through PKM2 function. Int J Mol Sci. 25:136402024. View Article : Google Scholar : PubMed/NCBI | |
Stanley TL, Fourman LT, Zheng I, McClure CM, Feldpausch MN, Torriani M, Corey KE, Chung RT, Lee H, Kleiner DE, et al: Relationship of IGF-1 and IGF-binding proteins to disease severity and glycemia in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 106:e520–e533. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kasprzak A: Insulin-Like growth factor 1 (IGF-1) signaling in glucose metabolism in colorectal cancer. Int J Mol Sci. 22:64342021. View Article : Google Scholar : PubMed/NCBI | |
Lenz G, Hamilton A, Geng S, Hong T, Kalkum M, Momand J, Kane SE and Huss JM: t-Darpp Activates IGF-1R signaling to regulate glucose metabolism in trastuzumab-resistant breast cancer cells. Clin Cancer Res. 24:1216–1226. 2018. View Article : Google Scholar : PubMed/NCBI | |
Lung Cancer Cohort Consortium (LC3), . The blood proteome of imminent lung cancer diagnosis. Nat Commun. 14:30422023. View Article : Google Scholar : PubMed/NCBI | |
Cai G, Qi Y, Wei P, Gao H, Xu C, Zhao Y, Qu X, Yao F and Yang W: IGFBP1 sustains cell survival during spatially-confined migration and promotes tumor metastasis. Adv Sci (Weinh). 10:e22065402023. View Article : Google Scholar : PubMed/NCBI | |
Onkar SS, Carleton NM, Lucas PC, Bruno TC, Lee AV, Vignali DAA and Oesterreich S: The great immune escape: Understanding the divergent immune response in breast cancer subtypes. Cancer Discov. 13:23–40. 2023. View Article : Google Scholar : PubMed/NCBI | |
Leuzzi G, Vasciaveo A, Taglialatela A, Chen X, Firestone TM, Hickman AR, Mao W, Thakar T, Vaitsiankova A, Huang JW, et al: SMARCAL1 is a dual regulator of innate immune signaling and PD-L1 expression that promotes tumor immune evasion. Cell. 187:861–881.e32. 2024. View Article : Google Scholar : PubMed/NCBI | |
De Martino M, Rathmell JC, Galluzzi L and Vanpouille-Box C: Cancer cell metabolism and antitumour immunity. Nat Rev Immunol. 24:654–669. 2024. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Huang L, Zhu J, Wu Y, Shi J and Dai K: Differential expression of insulin-like growth factor type 1 receptor identifies heterogeneous intrahepatic regulatory T subsets in mouse hepatocellular carcinoma. Clin Exp Immunol. 208:47–59. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu M, Zhong YB, Shao J, Zhang C and Shi C: Tumor-associated macrophages promote human hepatoma Huh-7 cell migration and invasion through the Gli2/IGF-II/ERK1/2 axis by secreting TGF-β1. Cancer Biol Ther. 21:1041–1050. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lv J, Liu C, Chen FK, Feng ZP, Jia L, Liu PJ, Yang ZX, Hou F and Deng ZY: M2-like tumour-associated macrophage-secreted IGF promotes thyroid cancer stemness and metastasis by activating the PI3K/AKT/mTOR pathway. Mol Med Rep. 24:6042021. View Article : Google Scholar : PubMed/NCBI | |
Uehara H, Kobayashi T, Matsumoto M, Watanabe S, Yoneda A and Bando Y: Adipose tissue:Critical contributor to the development of prostate cancer. J Med Invest. 65:9–17. 2018. View Article : Google Scholar : PubMed/NCBI | |
Manzella L, Massimino M, Stella S, Tirrò E, Pennisi MS, Martorana F, Motta G, Vitale SR, Puma A, Romano C, et al: Activation of the IGF axis in thyroid cancer: Implications for tumorigenesis and treatment. Int J Mol Sci. 20:32582019. View Article : Google Scholar : PubMed/NCBI | |
Xie X, Zhu Y, Cheng H, Li H, Zhang Y, Wang R, Li W and Wu F: BPA exposure enhances the metastatic aggression of ovarian cancer through the ERα/AKT/mTOR/HIF-1α signaling axis. Food Chem Toxicol. 176:1137922023. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Chen B, Li H, Wang Y, Liu X, Wong KY, Chan WN, Chan AK, Cheung AH, Leung KT, et al: Cancer-associated fibroblasts potentiate colorectal cancer progression by crosstalk of the IGF2-IGF1R and Hippo-YAP1 signaling pathways. J Pathol. 259:205–219. 2023. View Article : Google Scholar : PubMed/NCBI | |
Wei H, Dong C and Shen Z: Kallikrein-related peptidase (KLK10) cessation blunts colorectal cancer cell growth and glucose metabolism by regulating the PI3K/Akt/mTOR pathway. Neoplasma. 67:889–897. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lu X, Song X, Hao X, Liu X, Zhang X, Yuan N, Ma H and Zhang Z: miR-186-3p attenuates the tumorigenesis of cervical cancer via targeting insulin-like growth factor 1 to suppress PI3K-Akt signaling pathway. Bioengineered. 12:7079–7092. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Sun Y, Cong S and Zhang F: Insulin-like growth factor-1 promotes human uterine leiomyoma cell proliferation via PI3K/AKT/mTOR pathway. Cells Tissues Organs. 212:194–202. 2023. View Article : Google Scholar : PubMed/NCBI | |
Gao C, He XF, Xu QR, Xu YJ and Shen J: Sevoflurane downregulates insulin-like growth factor-1 to inhibit cell proliferation, invasion and trigger apoptosis in glioma through the PI3K/AKT signaling pathway. Anticancer Drugs. 30:e07442019. View Article : Google Scholar : PubMed/NCBI | |
Rieder S, Michalski CW, Friess H and Kleeff J: Insulin-like growth factor signaling as a therapeutic target in pancreatic cancer. Anticancer Agents Med Chem. 11:427–433. 2011. View Article : Google Scholar : PubMed/NCBI | |
Guo C, Cho KS, Li Y, Tchedre K, Antolik C, Ma J, Chew J, Utheim TP, Huang XA, Yu H, et al: IGFBPL1 regulates axon growth through IGF-1-mediated signaling cascades. Sci Rep. 8:20542018. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Zhang M, He T, Yang W, Wang L, Zhang L and Guo M: Epigenetic silencing of IGFBPL1 promotes esophageal cancer growth by activating PI3K-AKT signaling. Clin Epigenetics. 12:222020. View Article : Google Scholar : PubMed/NCBI | |
Guo Q, Jin Y, Chen X, Shen X, Lin M, Zeng C, Zhou T and Zhang J: NF-κB in biology and targeted therapy: New insights and translational implications. Signal Transduct Target Ther. 9:532024. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Dou X, Zheng Z, Ye C, Lu TX, Liang HL, Wang L, Weichselbaum RR and He C: YTHDF2/m6 A/NF-κB axis controls anti-tumor immunity by regulating intratumoral Tregs. EMBO J. 42:e1131262023. View Article : Google Scholar : PubMed/NCBI | |
Harvey AE, Lashinger LM, Hays D, Harrison LM, Lewis K, Fischer SM and Hursting SD: Calorie restriction decreases murine and human pancreatic tumor cell growth, nuclear factor-κB activation and inflammation-related gene expression in an insulin-like growth factor-1-dependent manner. PLoS One. 9:e941512014. View Article : Google Scholar : PubMed/NCBI | |
Wang C, An Y, Wang Y, Shen K, Wang X, Luan W, Ma F, Ni L, Liu M and Yu L: Insulin-like growth factor-I activates NFκB and NLRP3 inflammatory signalling via ROS in cancer cells. Mol Cell Probes. 52:1015832020. View Article : Google Scholar : PubMed/NCBI | |
Wang SH, Chen YL, Hsiao JR, Tsai FY, Jiang SS, Lee AY, Tsai HJ and Chen YW: Insulin-like growth factor binding protein 3 promotes radiosensitivity of oral squamous cell carcinoma cells via positive feedback on NF-κB/IL-6/ROS signaling. J Exp Clin Cancer Res. 40:952021. View Article : Google Scholar : PubMed/NCBI | |
Kim MS and Lee DY: Insulin-like growth factor binding protein-3 enhances etoposide-induced cell growth inhibition by suppressing the NF-κB activity in gastric cancer cells. Mol Cell Biochem. 403:107–113. 2015. View Article : Google Scholar : PubMed/NCBI | |
Cao Y, Chen J, Ren G, Zhang Y, Tan X and Yang L: Punicalagin prevents inflammation in LPS-Induced RAW264.7 macrophages by inhibiting FoxO3a/autophagy signaling pathway. Nutrients. 11:27942019. View Article : Google Scholar : PubMed/NCBI | |
Peluso I, Yarla NS, Ambra R, Pastore G and Perry G: MAPK signalling pathway in cancers: Olive products as cancer preventive and therapeutic agents. Semin Cancer Biol. 56:185–195. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Moerkens M, Ramaiahgari S, de Bont H, Price L, Meerman J and van de Water B: Elevated insulin-like growth factor 1 receptor signaling induces antiestrogen resistance through the MAPK/ERK and PI3K/Akt signaling routes. Breast Cancer Res. 13:R522011. View Article : Google Scholar : PubMed/NCBI | |
Rao W, Li H, Song F, Zhang R, Yin Q, Wang Y, Xi Y and Ge H: OVA66 increases cell growth, invasion and survival via regulation of IGF-1R-MAPK signaling in human cancer cells. Carcinogenesis. 35:1573–1581. 2014. View Article : Google Scholar : PubMed/NCBI | |
Teng JA, Wu SG, Chen JX, Li Q, Peng F, Zhu Z, Qin J and He ZY: The activation of ERK1/2 and JNK MAPK signaling by insulin/IGF-1 Is responsible for the development of colon cancer with type 2 diabetes Mellitus. PLoS One. 11:e01498222016. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Xiao Q, Xiao J, Niu C, Li Y, Zhang X, Zhou Z, Shu G and Yin G: Wnt/β-catenin signalling: Function, biological mechanisms and therapeutic opportunities. Signal Transduct Target Ther. 7:32022. View Article : Google Scholar : PubMed/NCBI | |
Yu F, Yu C, Li F, Zuo Y, Wang Y, Yao L, Wu C, Wang C and Ye L: Wnt/β-catenin signaling in cancers and targeted therapies. Signal Transduct Target Ther. 6:3072021. View Article : Google Scholar : PubMed/NCBI | |
Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y and Sun R: Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci. 81:792024. View Article : Google Scholar : PubMed/NCBI | |
Chatterjee A, Paul S, Bisht B, Bhattacharya S, Sivasubramaniam S and Paul MK: Advances in targeting the WNT/β-catenin signaling pathway in cancer. Drug Discov Today. 27:82–101. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hsieh CH, Cheng LH, Hsu HH, Ho TJ, Tu CC, Lin YM, Chen MC, Tsai FJ, Hsieh YL and Huang CY: Apicidin-resistant HA22T hepatocellular carcinoma cells strongly activated the Wnt/β-catenin signaling pathway and MMP-2 expression via the IGF-IR/PI3K/Akt signaling pathway enhancing cell metastatic effect. Biosci Biotechnol Biochem. 77:2397–2404. 2013. View Article : Google Scholar : PubMed/NCBI | |
Jamwal G, Singh G, Dar MS, Singh P, Bano N, Syed SH, Sandhu P, Akhter Y, Monga SP and Dar MJ: Identification of a unique loss-of-function mutation in IGF1R and a crosstalk between IGF1R and Wnt/β-catenin signaling pathways. Biochim Biophys Acta Mol Cell Res. 1865:920–931. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang QY, Wang L, Song ZY and Qu XJ: Knockdown of type I insulin-like growth factor receptor inhibits human colorectal cancer cell growth and downstream PI3K/Akt, WNT/β-catenin signal pathways. Biomed Pharmacother. 73:12–18. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chen KC, Chen PH, Ho KH, Shih CM, Chou CM, Cheng CH and Lee CC: IGF-1-enhanced miR-513a-5p signaling desensitizes glioma cells to temozolomide by targeting the NEDD4L-inhibited Wnt/β-catenin pathway. PLoS One. 14:e02259132019. View Article : Google Scholar : PubMed/NCBI | |
Hashemi Goradel N, Najafi M, Salehi E, Farhood B and Mortezaee K: Cyclooxygenase-2 in cancer: A review. J Cell Physiol. 234:5683–5699. 2019. View Article : Google Scholar : PubMed/NCBI | |
Peng Y, Wang Y, Tang N, Sun D, Lan Y, Yu Z, Zhao X, Feng L, Zhang B, Jin L, et al: Andrographolide inhibits breast cancer through suppressing COX-2 expression and angiogenesis via inactivation of p300 signaling and VEGF pathwa. J Exp Clin Cancer Res. 37:2482018. View Article : Google Scholar : PubMed/NCBI | |
Huang R, Yu J, Zhang B, Li X, Liu H and Wang Y: Emerging COX-2 inhibitors-based nanotherapeutics for cancer diagnosis and treatment. Biomaterials. 315:1229542025. View Article : Google Scholar : PubMed/NCBI | |
Song KH, Kang JH, Woo JK, Nam JS, Min HY, Lee HY, Kim SY and Oh SH: The novel IGF-IR/Akt-dependent anticancer activities of glucosamine. BMC Cancer. 14:312014. View Article : Google Scholar : PubMed/NCBI | |
Stoeltzing O, Liu W, Fan F, Wagner C, Stengel K, Somcio RJ, Reinmuth N, Parikh AA, Hicklin DJ and Ellis LM: Regulation of cyclooxygenase-2 (COX-2) expression in human pancreatic carcinoma cells by the insulin-like growth factor-I receptor (IGF-IR) system. Cancer Lett. 258:291–300. 2007. View Article : Google Scholar : PubMed/NCBI | |
Tian J, Lambertz I, Berton TR, Rundhaug JE, Kiguchi K, Shirley SH, Digiovanni J, Conti CJ, Fischer SM and Fuchs-Young R: Transgenic insulin-like growth factor-1 stimulates activation of COX-2 signaling in mammary glands. Mol Carcinog. 51:973–983. 2012. View Article : Google Scholar : PubMed/NCBI | |
Li W, Sun D, Lv Z, Wei Y, Zheng L, Zeng T and Zhao J: Insulin-like growth factor binding protein-4 inhibits cell growth, migration and invasion and downregulates COX-2 expression in A549 lung cancer cells. Cell Biol Int. 41:384–391. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mengzhe G, Shanshan X, Di Z and Shihua W: Editorial: The role of the IGF axis in tumorigenesis and cancer treatment: From genes to metabolites. Front Endocrinol (Lausanne). 13:11239622023. View Article : Google Scholar : PubMed/NCBI | |
Wang P, Mak VC and Cheung LW: Drugging IGF-1R in cancer: New insights and emerging opportunities. Genes Dis. 10:199–211. 2022. View Article : Google Scholar : PubMed/NCBI | |
Fuentes-Baile M, Ventero MP, Encinar JA, García-Morales P, Poveda-Deltell M, Pérez-Valenciano E, Barberá VM, Gallego-Plazas J, Rodríguez-Lescure Á, Martín-Nieto J and Saceda M: Differential effects of IGF-1R small molecule tyrosine kinase inhibitors BMS-754807 and OSI-906 on human cancer cell lines. Cancers (Basel). 12:37172020. View Article : Google Scholar : PubMed/NCBI | |
Stan MN and Krieger CC: The adverse effects profile of teprotumumab. J Clin Endocrinol Metab. 108:e654–e662. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Wen G, Sun L, Yuan W, Wang R, Zeng Q, Zhang G and Yu B: Cryptotanshinone inhibits cellular proliferation of human lung cancer cells through downregulation of IGF-1R/PI3K/Akt signaling pathway. Oncol Rep. 40:2926–2934. 2018.PubMed/NCBI | |
Jung M, Bu SY, Tak KH, Park JE and Kim E: Anticarcinogenic effect of quercetin by inhibition of insulin-like growth factor (IGF)-1 signaling in mouse skin cancer. Nutr Res Pract. 7:439–445. 2013. View Article : Google Scholar : PubMed/NCBI | |
Le CT, Leenders WPJ, Molenaar RJ and van Noorden CJF: Effects of the green tea polyphenol epigallocatechin-3-gallate on glioma: A critical evaluation of the literature. Nutr Cancer. 70:317–333. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu F, Ye S, Zhao L and Niu Q: The role of IGF/IGF-1R signaling in the regulation of cancer stem cells. Clin Transl Oncol. 26:2924–2934. 2024. View Article : Google Scholar : PubMed/NCBI | |
Ding F and Yang S: Epigallocatechin-3-gallate inhibits proliferation and triggers apoptosis in colon cancer via the hedgehog/phosphoinositide 3-kinase pathways. Can J Physiol Pharmacol. 99:910–920. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gao J, Chesebrough JW, Cartlidge SA, Ricketts SA, Incognito L, Veldman-Jones M, Blakey DC, Tabrizi M, Jallal B, Trail PA, et al: Dual IGF-I/II-neutralizing antibody MEDI-573 potently inhibits IGF signaling and tumor growth. Cancer Res. 71:1029–1040. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kim JH, Choi DS, Lee OH, Oh SH, Lippman SM and Lee HY: Antiangiogenic antitumor activities of IGFBP-3 are mediated by IGF-independent suppression of Erk1/2 activation and Egr-1-mediated transcriptional events. Blood. 118:2622–2631. 2011. View Article : Google Scholar : PubMed/NCBI | |
Gao S, Sun Y, Zhang X, Hu L, Liu Y, Chua CY, Phillips LM, Ren H, Fleming JB, Wang H, et al: IGFBP2 Activates the NF-κB pathway to drive epithelial-mesenchymal transition and invasive character in pancreatic ductal adenocarcinoma. Cancer Res. 76:6543–6554. 2016. View Article : Google Scholar : PubMed/NCBI | |
Cai Q, Kim M, Harada A, Idowu MO, Sundaresan G, Zweit J and Oh Y: Alpha-1 Antitrypsin inhibits tumorigenesis and progression of colitis-associated colon cancer through suppression of inflammatory neutrophil-activated serine proteases and IGFBP-3 proteolysis. Int J Mol Sci. 23:137372022. View Article : Google Scholar : PubMed/NCBI | |
Wei X, Huo Y, Pi J, Gao Y, Rao S, He M, Wei Q, Song P, Chen Y, Lu D, et al: METTL3 preferentially enhances non-m6A translation of epigenetic factors and promotes tumourigenesis. Nat Cell Biol. 24:1278–1290. 2022. View Article : Google Scholar : PubMed/NCBI | |
He P, Liu X, Yu G, Wang Y, Wang S, Liu J and An Y: METTL3 facilitates prostate cancer progression via inducing HOXC6 m6A modification and stabilizing its expression through IGF2BP2-dependent mechanisms. Mol Cell Biochem. 479:1707–1720. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Wang M, Wang H, Yang J, Li X, Zhang R, Ding X, Hou H, Zhou J and Wu M: METTL3 inhibitor suppresses the progression of prostate cancer via IGFBP3/AKT pathway and synergizes with PARP inhibitor. Biomed Pharmacother. 179:1173662024. View Article : Google Scholar : PubMed/NCBI | |
Pomeroy AE, Schmidt EV, Sorger PK and Palmer AC: Drug independence and the curability of cancer by combination chemotherapy. Trends Cancer. 8:915–929. 2022. View Article : Google Scholar : PubMed/NCBI | |
Dias MP, Moser SC, Ganesan S and Jonkers J: Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nat Rev Clin Oncol. 18:773–791. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ravi P, Wang V, Fichorova RN, McGregor B, Wei XX, Basaria S and Sweeney CJ: IGF-1 axis changes with ADT and docetaxel in metastatic prostate cancer. Endocr Relat Cancer. 30:e2302412023. View Article : Google Scholar : PubMed/NCBI | |
Codony-Servat J, Cuatrecasas M, Asensio E, Montironi C, Martínez-Cardús A, Marín-Aguilera M, Horndler C, Martínez-Balibrea E, Rubini M, Jares P, et al: Nuclear IGF-1R predicts chemotherapy and targeted therapy resistance in metastatic colorectal cancer. Br J Cancer. 117:1777–1786. 2017. View Article : Google Scholar : PubMed/NCBI | |
Qiu N, He YF, Zhang SM, Zhan YT, Han GD, Jiang M, He WX, Zhou J, Liang HL, Ao X, et al: Cullin7 enhances resistance to trastuzumab therapy in Her2 positive breast cancer via degrading IRS-1 and downregulating IGFBP-3 to activate the PI3K/AKT pathway. Cancer Lett. 464:25–36. 2019. View Article : Google Scholar : PubMed/NCBI | |
Xu Z, Wang X, Sun W, Xu F, Kou H, Hu W, Zhang Y, Jiang Q, Tang J and Xu Y: RelB-activated GPX4 inhibits ferroptosis and confers tamoxifen resistance in breast cancer. Redox Biol. 68:1029522023. View Article : Google Scholar : PubMed/NCBI | |
Zheng Y, Sowers JY and Houston KD: IGFBP-1 expression promotes tamoxifen resistance in breast cancer cells via Erk pathway activation. Front Endocrinol (Lausanne). 11:2332020. View Article : Google Scholar : PubMed/NCBI | |
Suzuki H, Iwamoto H, Seki T, Nakamura T, Masuda A, Sakaue T, Tanaka T, Imamura Y, Niizeki T, Nakano M, et al: Tumor-derived insulin-like growth factor-binding protein-1 contributes to resistance of hepatocellular carcinoma to tyrosine kinase inhibitors. Cancer Commun (Lond). 43:415–434. 2023. View Article : Google Scholar : PubMed/NCBI | |
Lu H, Ai J, Zheng Y, Zhou W, Zhang L, Zhu J, Zhang H and Wang S: IGFBP2/ITGA5 promotes gefitinib resistance via activating STAT3/CXCL1 axis in non-small cell lung cancer. Cell Death Dis. 15:4472024. View Article : Google Scholar : PubMed/NCBI | |
Coleman KL, Chiaramonti M, Haddad B, Ranzenberger R, Henning H, Al Khashali H, Ray R, Darweesh B, Guthrie J, Heyl D and Evans HG: Phosphorylation of IGFBP-3 by casein kinase 2 blocks its interaction with hyaluronan, enabling HA-CD44 signaling leading to increased NSCLC cell survival and cisplatin resistance. Cells. 12:4052023. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Lu Q, Zhou H, Liu J, Nadorp B, Lasry A, Sun Z, Lai B, Rona G, Zhang J, et al: A membrane-associated MHC-I inhibitory axis for cancer immune evasion. Cell. 186:3903–3920.e21. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yu L, Ding Y, Wan T, Deng T, Huang H and Liu J: Significance of CD47 and Its association with tumor immune microenvironment heterogeneity in ovarian cancer. Front Immunol. 12:7681152021. View Article : Google Scholar : PubMed/NCBI | |
Sun Y, Yin Z, Li S, Wu L, Zhang Y, Zhao Y, Gomes Dos Santos IL, Subudhi S, Lei P, Muzikansky A, et al: Losartan rewires the tumor-immune microenvironment and suppresses IGF-1 to overcome resistance to chemo-immunotherapy in ovarian cancer. Br J Cancer. 131:1683–1693. 2024. View Article : Google Scholar : PubMed/NCBI | |
Song D, Wu Y, Li J, Liu J, Yi Z, Wang X, Sun J, Li L, Wu Q, Chen Y, et al: Insulin-like growth factor 2 drives fibroblast-mediated tumor immunoevasion and confers resistance to immunotherapy. J Clin Invest. 134:e1833662024. View Article : Google Scholar : PubMed/NCBI | |
Hashimoto M, Konda JD, Perrino S, Celia Fernandez M, Lowy AM and Brodt P: Targeting the IGF-Axis potentiates immunotherapy for pancreatic ductal adenocarcinoma liver metastases by altering the immunosuppressive microenvironment. Mol Cancer Ther. 20:2469–2482. 2021. View Article : Google Scholar : PubMed/NCBI | |
Pointer KB, Pitroda SP and Weichselbaum RR: Radiotherapy and immunotherapy: Open questions and future strategies. Trends Cancer. 8:9–20. 2022. View Article : Google Scholar : PubMed/NCBI | |
Hessels AC, Langendijk JA, Gawryszuk A, Heersters MAAM, van der Salm NLM, Tissing WJE, van der Weide HL and Maduro JH: Review-Late toxicity of abdominal and pelvic radiotherapy for childhood cancer. Radiother Oncol. 170:27–36. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zong R, Chen X, Feng J and Xu S: IGF-1R depletion sensitizes colon cancer cell lines to radiotherapy. Cancer Biomark. 32:199–206. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen L, Zhu Z, Gao W, Jiang Q, Yu J and Fu C: Systemic analysis of different colorectal cancer cell lines and TCGA datasets identified IGF-1R/EGFR-PPAR-CASPASE axis as important indicator for radiotherapy sensitivity. Gene. 627:484–490. 2017. View Article : Google Scholar : PubMed/NCBI | |
Chen JH, T H Wu A, T W Tzeng D, Huang CC, Tzeng YM and Chao TY: Antrocin, a bioactive component from Antrodia cinnamomea, suppresses breast carcinogenesis and stemness via downregulation of β-catenin/Notch1/Akt signaling. Phytomedicine. 52:70–78. 2019. View Article : Google Scholar : PubMed/NCBI | |
Su YH, Wu JS, Dai YZ, Chen YT, Lin YX, Tzeng YM and Liao JW: Anti-oxidant, anti-mutagenic activity and safety evaluation of antrocin. Toxics. 11:5472023. View Article : Google Scholar : PubMed/NCBI | |
Chen YA, Tzeng DTW, Huang YP, Lin CJ, Lo UG, Wu CL, Lin H, Hsieh JT, Tang CH and Lai CH: Antrocin sensitizes prostate cancer cells to radiotherapy through inhibiting PI3K/AKT and MAPK signaling pathways. Cancers (Basel). 11:342018. View Article : Google Scholar : PubMed/NCBI |