Open Access

Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review)

  • Authors:
    • Miguel Morales-Rodríguez
    • Lucero Paniagua-García
    • Jayanthi Narayanan
    • Horacio Zamudio-Meza
    • Ramón Víctor Moreno-Torres
    • Carlo César Cortés-González
    • José Antonio Juanico-Lorán
    • Beatriz Martínez-Pérez
    • Jorge Fernández-Retana
  • View Affiliations

  • Published online on: February 10, 2023     https://doi.org/10.3892/ol.2023.13714
  • Article Number: 128
  • Copyright: © Morales-Rodríguez et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cervical cancer is among the most frequently occurring neoplasms worldwide, and it particularly affects individuals in developing countries. Factors such as the low quality of screening tests, the high incidence of locally advanced cancer stages and the intrinsic resistance of certain tumors are the main causes of failure in the treatment of this neoplasm. Due to advances in the understanding of carcinogenic mechanisms and bioengineering research, advanced biological nanomaterials have been manufactured. The insulin‑like growth factor (IGF) system comprises multiple growth factor receptors, including IGF receptor 1. These receptors are activated by binding to their respective growth factor ligands, IGF‑1 and IGF‑2, and insulin, and play an important role in the development, maintenance, progression, survival and treatment resistance of cervical cancer. In the present review, the role of the IGF system in cervical cancer and three nanotechnological applications that use elements of this system are described, namely Trap decoys, magnetic iron oxide nanoparticles and protein nanotubes. Their use in the treatment of resistant cervical cancer tumors is also discussed.

Introduction

Cervical cancer has the fourth highest incidence and mortality among gynecological neoplasms worldwide. In 2018, the annual estimates for cervical cancer were ~570,000 new cases and 311,000 mortalities (1). However, this neoplasm affects each country according to its degree of economic development, social factors and lifestyle, and is an imminent and serious crisis for developing countries. In Mexico from 2011 to 2015, the mortality rate for cervical cancer was 6.45 per 100,000 women (2), indicating that it a highly prevalent health issue. Furthermore, the use of screening programs based on cytology, known as pap smear testing, has not been successful in developing countries due to the poor quality of the tests resulting in high rates of false negatives (3,4). Additionally, patients frequently first present at health centers with advanced lesions and are diagnosed with locally advanced stages IB2 to IVA according to the FIGO classification (5). The suggested treatment comprises cisplatin (CDDP)-based chemotherapy concomitant with radiation therapy plus brachytherapy, which represents the standard of care in patients with locally advanced disease (69). The average prognosis for 5-year survival is 56% (5,10).

Although patients with a poor response to standard treatment are treated with secondary systemic therapies (7,8), there is no standard treatment for patients with progressive or metastatic cervical cancer due to its heterogeneous manifestations (11). Notably, chemotherapeutic treatments for cervical cancer have shown limited success due to the lack of specificity associated with systemic administration. In addition, higher doses are required to achieve a therapeutic effect, which increases the adverse cytotoxic effects that exacerbate those of the first treatment and may reduce the physical integrity of the patient; therefore, survival is limited. The resistance of cancer cells to physical and chemical methods, low efficiency of drug delivery and highly heterogeneous tumor microenvironments represent significant impediments in clinical oncology. Furthermore, even when drug administration is optimized, the efficiency of chemotherapy has several challenges, one of which is the typical hypovascularization of cervical cancer tissues (12), which reduces the efficiency of systemic drug distribution. The cellular origin of cervical cancer also contributes to the development and diversity of the tumor microenvironment, which creates different obstacles to drug transport, even in tumors of the same size and stage. Additionally, it has been shown that the density of the tumor cells and formation of intercellular junctions serve key roles in the pharmacokinetics of chemotherapeutic agents in solid tumors (13).

Nano-oncology is a subdivision of nanomedicine in which nanotechnology is used in the treatment of cancer (14,15). Specific delivery strategies for anticancer agents have been developed, generally in the nanoscale range, using materials such as organic nanoparticles made from lipids, polymers, liposomes, polymeric micelles, dendrimers and engineered peptides and nucleic acids, and inorganic nanoparticles such as carbon, metal and metal oxide nanoparticles (16,17). Nanomaterials have distinctive physical, chemical and optical properties and may be modified with biological molecules to direct them toward specific targets. In this regard, membrane receptors and their ligands have great relevance as biomarkers and therapeutic targets in the treatment of different neoplasms. The insulin-like growth factor (IGF) system has been reported in epithelial and glandular tumors, including prostate cancer, breast cancer and colon cancer, and is an excellent target for nano-oncology (1820).

The present review provides a brief overview of the IGF system, its relevance in cervical cancer and the development of new nanotechnology-based therapies targeting IGF complex molecules for the treatment of cervical cancer.

IGF axis

The IGF system is a complex network comprising growth factors IGF-1 and −2, cell surface receptors IGF-1R and −2R, the IGF binding protein (IGFBP) family of high-affinity specific binding proteins (IGFBP-1 to −6) and IGFBP proteases, as well as molecules that interact with IGFBP to regulate and disseminate the actions of IGF in tissues (21). IGFs are peptide hormones from a family that also includes insulin. While the main role of IGF-2 is as a regulator of embryonic and fetal development, IGF-1 is maintained throughout life as a broad-spectrum growth factor (22). These factors bind with a specific receptor on the cell surface and stimulate different signaling pathways.

The sequence of IGF-1R has 60% homology with that of the insulin receptor. It is generated as a polypeptide precursor that is post-translationally modified by glycosylation, proteolytic cleavage and dimerization to form a heterotetramer comprising two α subunits and two β subunits bonded together via α-α and β-β disulfide linkages. The α subunits are located outside the cell and contain the ligand binding site, while the β subunits have extracellular and transmembrane domains and an intracellular portion that contains the tyrosine kinase catalytic domain (Fig. 1) (23). The binding of an IGF ligand to its receptor activates the tyrosine kinase domain, which induces a conformational change allowing autophosphorylation at the Tyr950 site. This phosphorylation site is a docking point for substrates such as the insulin receptor substrate 1–4 proteins, where activation of the PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signaling pathways occurs. The former regulates cell survival and protein synthesis, while the latter regulates gene expression, cell proliferation and differentiation (Fig. 2A) (24). IGF-1R has a tyrosine-kinase domain, whereas IGF-2R does not. Due to this fact and its high affinity for the IGF-2 ligand, it is proposed that the function of IGF-2R is to limit the interaction of IGF-2 with IGF-1R, thereby acting as a tumor suppressor (25,26). The covalent attachment of a small ubiquitin-like modifier (SUMO) family protein to three lysine residues in the b-subunit of IGF-1R via SUMOylation induces its translocation to the nucleus in a ligand-independent manner after (27). In the nucleus, IGF-1R and T cell factor/lymphoid enhancer factor act as transcriptional coactivators to increase the promoter activity and expression of downstream target genes, including cyclin D1 and Axin2 (Fig. 2B), which promote cell cycle progression (19,27).

The actions of IGFs are regulated by interaction with soluble IGFBPs and IGFBP proteases. The IGFBP family comprises six IGFBPs that bind to IGF with high affinity and specificity, and IGFBP-related proteins that are structurally similar to IGFBPs but have lower IGF-binding affinity (22). The six IGFBP proteins are structurally similar to each other but not to cell surface receptors. Each of these binding proteins is the product of different genes and has different functional properties; however, they are all mostly present as high-molecular-weight complexes with IGF-1 and IGF-2 in the circulation and extracellular space, for example, as ~150-kDa complexes with IGFBP-3 and the acid-labile subunit (28,29). These complexes inhibit extravascular transit and help to retain the IGF-1 ligand in the circulation. The IGFBP proteases are critical for modulating the availability of IGF-1 at the cellular level and regulating its half-life via the degradation of IGF-1-containing complexes (Fig. 3A). The dynamic balance of IGF-1, IGFBP and IGFBP proteases constitutes the IGF-1 axis that ultimately determines the extent of the cellular effects dependent on this hormone (3032). Following dissociation of the ternary complex, the IGFBP/IGF binary complexes are cleared from the circulation via the endothelium, from where they are delivered to tissues and interact with cell surface receptors (Fig. 3B). Since the binding affinity of IGFs for IGFBPs is higher than that for their receptors, IGFBPs in tissues inhibit the interaction of IGF with its receptors and thereby regulate the action of IGF, promoting a microenvironment that functions as a reservoir for the slow release of ligands. This prolongs the half-life of IGFs in the circulation and prevents them from crossing the capillary barrier (28,33). IGFBP-3 is the most abundant binding protein in human serum; it is present in several glycosylated forms weighing between 40 to 44 kDa and has been shown to regulate the apoptosis induced by p53 (34).

IGF axis in cervical cancer

Human papillomavirus (HPV) infection is the primary etiological factor of cervical carcinogenesis (35). The HPV E6 and E7 viral proteins serve well-established oncogenic functions: E6 binds to p53 in a trimeric complex with E6-associated protein, a ubiquitin-ligase, which induces the degradation of p53 in proteasomes (36), while E7 binds to hypophosphorylated retinoblastoma-associated protein (pRB), which is rapidly degraded by proteasomes and constitutively releases the transcription factor E2F (37,38). The HPV-induced loss of function of these two tumor suppressors is a fundamental cause of cervical cancer carcinogenesis. However, additional elements are involved in the inactivation of p53 and pRB (39).

Studies have demonstrated the relationships between viral proteins and members of the IGF system during the neoplastic process. In a study conducted by Kuramoto et al (39), it was shown that the expression of IGF-1R is gradually upregulated in cervical intraepithelial neoplasia (CIN) 3 and invasive cancer lesions while its expression is moderate in CIN 1 and 2. The study also suggested that the viral oncoprotein E6 represses p53 and causes transcriptional dysregulation by activating the upregulation of the expression of this receptor. Furthermore, it confirmed that the phosphorylation of IGF-1R increases as the disease progresses. The phosphorylation of IGF-1R activates the MAPK (Ras/Raf/MEK/ERK) and PI3K survival signaling pathways, which contribute to cell survival and drug resistance and thereby serve an important role in progression of the neoplasia (39). It has also been observed in other human neoplasms, including clear cell kidney cancer, colorectal carcinoma and pediatric glioma, that the nuclear translocation of IGF-1R is associated with advanced disease and poor prognosis (19,40). In the study of Codony-Servat et al (19), it was observed that the treatment of patients with metastatic colorectal cancer using IGF-1R blocking antibodies induced an increase in nuclear translocation, suggesting that receptor nuclear sequestration may contribute to resistance. In another study, in which the upregulation of IGF-1R was shown to be associated with resistance to radiotherapy in patients with HPV-16-positive cervical cancer, IGF-1R was proposed as a predictive biomarker of the response to radiation (41). Similarly, in a recent study IGF-2R was proposed as a poor prognostic biomarker for patients with cervical cancer since it may be involved in the recurrence of the disease. In that study, Takeda et al (42) describe an oncogenic mechanism of IGF-2R, in which it participates in the regulation of lysosomal transport via Golgi bodies, together with cathepsins B and L loaded with mannose-6-phosphate, resulting in increased lysosomal homeostasis and decreased apoptosis. Thus, IGF-2R appears to have a dual oncogenic role in cervical cancer.

The upregulation of the IGF receptors in tumors resistant to radiation therapy indicates that they are potential targets for alternative therapies. Furthermore, the expression of ligands of the IGF system has been reported in different events that contribute to the pathogenesis and progression of various neoplasms (43,44). In non-small cell lung cancer, a study reported that the expression of IGF-1 and IGF-1R was upregulated and associated with progression and poor prognosis, and suggested that the autocrine/paracrine activity of IGF-1 may play an important role in the development of lung cancer (45). In cervical cancer, a review of the IGF axis indicated that the presence of IGF-1 may contribute to each stage of tumor progression, from malignant transformation, tumor growth, local invasion, distal metastasis and resistance to treatment (46). Elevated levels of IGF-1 and IGF-2 promote signaling via the stimulation of IGF-1R in cervical cancer from the CIN phase (47,48), with a dose-dependent effect on the growth and invasiveness of tumor cells, mainly mediated by IGF-1. Furthermore, an unexpected role of IGF-1 as a stimulator of the invasion and proliferation of cervical cells through interaction with IGF-1R with the cooperation of integrin αvβ3 has been reported (49). It is important to note that relatively low IGF-2 mRNA levels have been reported in primary tumor samples and cervical tumor cell models (29,50,51). Therefore, it appears that the production of IGF-2 by cervical epithelial cells is insufficient to transduce a strong mitogenic signal. Nevertheless, Steller et al (50) proposed that the autocrine function of IGF-2 in cervical cancer cells involves the mitogenic signaling of epidermal growth factor (EGF).

Studies on IGF-binding proteins in cervical neoplasia have mainly reported on IGFBP-2 and −3. The role of IGFBP-2 in tumorigenesis is complex and multifaceted, as it can both promote and suppress tumors. The prolonged expression of HPV16 E6 and E7 suppresses IGFBP-2 expression; IGFBP-2 generally inhibits the actions of IGF and thereby inhibits mitogenesis, differentiation, survival and other cellular processes, which may be due to the ability of IGFBP-2 to compete with IGF-1R or −2R for the binding of IGF-1 or −2 ligands (47,52). However, IGFBP-2 has also been demonstrated to interact with integrins to exert oncogenic effects that promote cell proliferation and invasion and suppress apoptosis. Specifically, studies have shown that IGFBP-2 is associated with metastasis and uses integrin-dependent mechanisms to reduce cell adhesion and promote invasion, suggesting that IGFBP-2 has IGF-independent oncogenic effects (5254). By contrast, IGFBP-3 is known to protect against cancer via the p53-mediated activation of apoptosis. However, IGFBP-3 upregulation is a late event after E6/E7 expression in infected cells, after which E6 inhibits p53 activity and consequently blocks apoptosis (55). Additionally, E7 impedes the ability of IGFBP-3 to induce apoptosis. This appears to be mediated via the binding of E7 to the nuclear localization sequence of IGFBP-3 in the nucleus, which reduces the half-life of nuclear IGFBP3 and subsequently induces the polyubiquitination and proteolysis of IGFBP-3 in cervical cancer cells (28,56). However, the functions of IGFBP-3 in the nucleus are not clearly understood, although it may regulate transcription and modify cellular functions through intranuclear pathways (57). Notably, a study of 226 patients found that a high nuclear concentration of IGFBP-3 was a powerful predictor of recurrence in prostate cancer (57,58).

IGF axis members as therapeutic targets in cervical cancer

As explained above, the components of the IGF system are activated in an aberrant way during carcinogenesis and, importantly, the expression of certain components confers resistance to the treatments used for this neoplasia, making them a key target for new therapeutic strategies. Several approaches have been used to target components of the IGF system, in particular IGF-1R, due to its involvement in cancer cell growth. These include interference RNAs, antisense oligonucleotides and RNAs, triple helix-forming oligonucleotides, specific kinase inhibitors, single chain antibodies and humanized anti-IGF-1R monoclonal antibodies. Tyrosine kinase inhibitors and monoclonal antibodies are among the most useful; they include ganitumab (AMG-479), dalotuzumab (MK-0646), cixutumumab (IMC-A12), teprotumumab (R1507) and figitumumab (CP-751,871), which are fully human recombinant monoclonal antibodies commonly used to target IGF-1R. They prevent IGF-1 from binding to IGF-1R and inhibit downstream signaling via the PI3K/Akt pathway (18,5963). The PI3K/Akt pathway is known to promote cell growth and survival in response to extracellular signals. However, a study investigating advances in the treatment of solid tumors with these IGF-1R inhibitory antibodies, alone or in combination with other therapies, revealed they had non-significant effects on overall survival and progression-free survival, and furthermore, adverse effects were observed for dalotuzumab in the breast, colorectal and prostate cancer subgroups (63). Although monoclonal antibodies are highly selective, their development as therapeutic agents is challenging due to their poor tumor penetration and high production costs (28).

Extracellular domain of IGF-1R used as a trap nanoparticle

The action of cell surface receptors can be effectively blocked via the use of soluble decoys that specifically bind to a ligand with high affinity, thereby limiting the bioavailability of the ligand and the signaling it would otherwise mediate at the membrane receptor (64,65). Furthermore, other studies have demonstrated that the efficiency of these decoys is significantly improved by the addition of the Fc domain of human IgG1 to form a more stable chimeric protein known as a ‘Trap’. Specific Traps have been used to treat various diseases, including rheumatoid arthritis (66), cryopyrin-associated periodic syndromes (67), wet macular degeneration and metastatic colorectal cancer (65). In addition, an EGFR-Fc fusion decoy comprising the truncated extracellular domains of EGFR/ErbB-1 and ErbB-4 was shown to have high affinity for EGF-like growth factor and inhibit the proliferation, invasion and metastasis of breast cancer cells (64,68).

The identification of elements of the IGF system as therapeutic targets in different tumors has stimulated the development of decoys based on the IGF receptor system. A study conducted by Samani et al (69) initially designed a truncated protein of IGF-1R comprising the first 933 amino acids of the native receptor and encompassing its extracellular domain. This protein was expressed in H-59 highly metastatic murine lung carcinoma cells and detected as a secreted heterotetramer (βm-α-α-βm) that exogenously neutralized the IGF-1 ligand and inhibited the proliferation, invasion and resistance to apoptosis of the cells via the regulation of IGF-1R signaling. Similarly, the expression of this protein markedly reduced the metastatic potential of the H-59 cells following their intrasplenic/portal inoculation in mice, reducing the formation of liver metastases by 90% and significantly extending the disease-free survival time. In a second study, a gutless adenovirus expressing soluble IGF-1R (sIGFIR) was intravenously injected into mice, which led to the production of measurable plasma levels of sIGFIR for up to 21 days and significantly inhibited liver metastasis (70). Subsequently, to optimize this soluble decoy for translation to the clinic, its pharmacokinetic properties and therapeutic potential were improved via fusion with the Fc portion of human IgG1 to form sIGFIR/hFc-IgG1. The addition of the Fc fragment did not alter the binding kinetics of the recombinant protein. Furthermore, this IGF-Trap decoy had high binding affinity for hIGF-1, moderately lower affinity for mouse IGF-2 and IGF-1, and a three-log lower affinity for insulin (20). IGF-Trap displayed similar effects to sIGFIR, with the ability to inhibit IGF-1, IGF-2 and IGF-1R-regulated cell signaling and functions in various types of carcinoma cells in vitro, including breast, lung and colon carcinoma cells. However, the pharmacokinetic profile of IGF-TRAP was more favorable than that of sIGFIR in vivo, as demonstrated by half-lives of 47.5 and 21.9 h, respectively, which confirmed that the two Fc domains improved the stability of the protein in vivo (20,64).

A frequent limitation of fusion proteins is that they may form high-molecular-weight complexes via the formation of disulfide bonds between Fc fragments. This is an issue for the IGF-Trap decoy, a tetramer that comprises two subunits each fused to an IgG1 Fc domain, in which the proximity of adjacent Fc domains facilitates the formation of disulfide bonds and large molecular complexes. For this reason, the IGF-Trap decoy was redesigned by the replacement of cysteine with serine in the hinge region of the Fc fragment of human IgG1, and the introduction of a longer, more flexible linker between the IGF-1R ectodomain and the Fc domain (Fig. 4). This modification decreased the formation of high molecular weight complexes by this Trap and increased its stability, thereby improving its pharmacodynamic properties (64,71). Using the kinase receptor activation (KIRA) assay, it was shown that the serum bioavailability of IGF-1 is closely associated with the pharmacokinetic/pharmacodynamic profile of the IGF-Trap. In this assay, the bioavailability of the ligand was measured via quantification of the phosphorylated IGF-1 receptor. Unlike traditional endpoint bioassays that measure the downstream effects of IGF-1R activation, the KIRA assay directly measures receptor activation, thereby eliminating the confounding effects of other factors that may also activate downstream signaling pathways. In addition, since the bioavailability and bioactivity of IGF-1 are affected by IGF-BP and naturally occurring proteases in the circulation, the KIRA assay provides a more accurate measure of bioactive ligands (72). The aforementioned studies indicate that IGF-Trap has high specificity for IGF-1 and IGF-2 and low affinity for insulin, and therefore should minimally influence the physiological functions of insulin. In addition, the penetration and diffusion of IGF-Trap into solid tumors may exert beneficial effects via the neutralization of locally produced IGFs. Furthermore, reducing the bioavailability of IGFs using IGF-Trap may affect various components of the tumor microenvironment and thereby provide an enhanced growth inhibiting effect. These data also suggest that IGF-Trap could provide a surrogate marker for response assessment and a potential tool for the classification of patients with resistant cervical tumors.

Nanoparticles targeting IGF-1R with theragnostic advantages

Magnetic nanoparticles (MNPs) have shown promising results in the personalized therapy and clinical management of patients with resistant tumors. Due to the unique physicochemical properties of MNPs, they may be used for multiple applications simultaneously, particularly for theragnostic purposes, such as imaging combined with the administration of therapeutic drugs. Magnetic iron oxide nanoparticles (IONPs) are biocompatible and biodegradable with low toxicity. Therefore, various types of IONPs have been used clinically and have been shown to be safe. Furthermore, IONPs have unique paramagnetic properties that provide T2- and T2*-weighted images with a strong contrast, and a T1 effect at very low concentrations (73,74).

Biodegradable IONPs have been generated and directed against different target receptors, including the urokinase plasminogen activator (uPA) receptor (uPAR). In one study, amphiphilic polymer-coated IONPs were conjugated to the amino-terminal fragment of uPA, the natural high-affinity ligand for uPAR (75). In addition, the polymer coating was modified to allow the encapsulation of hydrophobic chemotherapeutic drugs to form nanoparticulate drug delivery vehicles that are also sensitive to magnetic resonance imaging (MRI). The fluorescent hydrophobic drug doxorubicin (Dox) was efficiently encapsulated into the IONPs to form compact Dox-loaded nanoparticles that were stable at pH 7.4 but released Dox at an acidic pH of 4.0-5.0 within 2 h. These Dox-encapsulating IONPs were observed to retain their T2 MRI contrast effect following their internalization in tumor cells (75). Notably, this IONP system can be conjugated with different ligands and thus be directed to different target receptors to perform theranostic functions. IGF-1R appears to be an ideal target receptor due to its upregulation in tumor cells resistant to treatments. In another study, Zhou et al (76) aimed to exploit the theranostic capacities of IONPs directed at this receptor by loading IONPs with Dox and conjugating them with recombinant human IGF-1 for targeting purposes (Fig. 5). The efficacy of these theranostic IONPs, referred to as IBF-1-IONP-Dox, was evaluated using human patient-derived xenograft (PDX) models in which pancreatic cancer tissue was implanted into severe combined immunodeficient mice. The repeated systemic administration of the IGF-1-targeted theragnostic IONPs was monitored by optical imaging and near infrared magnetic resonance, and the results revealed that IGF-1-IONP-Dox induced a significantly greater reduction in PDX growth than was achieved using free Dox or undirected IONP-Dox in both subcutaneous and orthotopic locations. In summary, theragnostic nanoparticles that can easily be modified using a variety of targeting molecules and therapeutic agents, such as antibodies, peptides, small molecules and aptamers, via several conjugation strategies have been directed to specific targets including IGF-1R.

These IONPs constitute a novel model for the imaging and targeted administration of drugs for the treatment of tumors (77). Human pancreatic PDX models, which are highly similar to tumors in patients in terms of their intratumoral heterogeneity, histological features and tumor microenvironments, were used to assess the effect of IONPs. The strategy of using IGF-1 for the targeted therapy of pancreatic cancer is promising. Although this system has not been tested in cervical tumors, it appears to be a promising innovation for the management of resistant tumors.

Protein nanotubes

The self-assembly of peptides to form nanostructured materials is a research area in which the non-covalent interactions within or between peptide building blocks have been investigated for their contribution to the self-assembly process (78). Based on the role of IGFBPs in the initiation, development, progression and survival of cancer and their function as natural antagonists of IGFs, IGFBP mimetics have been created as potential alternative therapies for cancer treatment using IGFBP-2 as a template. It was observed that by fragmenting the IGFBP-2 protein at the single tryptophan residue within the conserved CWCV motif, the carboxyl terminal fragment was stable and able to inhibit the binding of IGF-1 to IGF 1R (79). Therefore, this fragment was subjected to further investigation.

The native sequence of the hIGFBP-2249-289 fragment includes two cysteine residues in its primary sequence, and cysteine-rich regions have been observed to increase the specificity of the ligand (79). Previously, in a study by Binkert et al (80), the amino acid sequences of the mature forms of human IGFBP-1, IGFBP-2 and the rat BRL-BP proteins were aligned, and they observed that the three IGFBPs share a cysteine-rich region homologous at its amino terminus, plus an RGD motif embedded in a conserved pentapeptide. However, there are differences between the three proteins of this family. IGFBP-2 has the highest number of cysteines at its carboxyl end and carries an Arg-Gly-Asp (RGD) motif embedded in a conserved pentapeptide, which implies a structural or functional relevance (80). Therefore, following the addition of an extra cysteine at residue 281, an hIGFBP-2249-289 (R281C) polypeptide with an odd number of cysteines was obtained (79,81,82), which spontaneously self-assembled to form soluble nanotubular structures via the formation of intermolecular disulfide bonds. The formation and disassembly of the nanotubes can be controlled by the choice of appropriate redox conditions. Furthermore, the polypeptide fragment contains an RGD motif in its sequence (81,82), and an RGD array is present on the surface of the nanotubes, which serves as a site for the active targeting of cancer cells via integrin binding. RGD is an adhesive peptide widely studied in the field of biomaterials. It has been established that RGD is very effective in promoting the attachment of numerous types of cells to various materials. It constitutes the main binding domain of integrins present in the extracellular matrix, including fibronectin, vitronectin, fibrinogen, osteopontin and bone sialoprotein (83,84).

An interesting application of this protein nanotube system was reported in the study by Asampille et al (81). The interior of the nanotubes was loaded with Dox as a representative hydrophobic cytotoxic drug (Fig. 6A) or with the dye fluorescein isothiocyanate as a representative imaging agent (Fig. 6B). In order to determine the ability of the multi-RGD moieties to specifically deliver the nanotubes to cancer cells, integrins were overexpressed on HeLa and MDA-MB-231 cell lines in vitro. Confocal microscopy showed that the nanotubes remained attached to the membrane of these cells, while flow cytometry revealed an increase in apoptosis caused by the action of Dox at the cell periphery (Fig. 7b-2). These results demonstrate the theragnostic potential of these nanotubes (28,81) in resistant tumors, including cervical cancer.

Conclusions

Cervical cancer is a public health issue that particularly affects developing countries. The lack of efficiency in screening methods, the prevalence of locally advanced stages and intrinsic resistance to common treatments are the main reasons for the failure to control this neoplasm. The conventional treatment recommended by The International Federation of Gynecology and Obstetrics, which comprises 50-Gy radiotherapy concomitant with CDDP-based chemotherapy and brachytherapy, is applied indiscriminately to the majority of patients (8,9). A prediction system has been proposed that indicates the response to treatment and/or the risk of metastasis via the molecular analysis of transcriptional gene signatures (7,85,86), which are molecular tools that enable oncologists to select the optimum therapeutic strategy for each patient. However, the poor prognosis of cervical cancer to conventional treatment necessitates the development of novel therapeutic alternatives that are more efficient in eliminating resistant tumors. Nanotechnology has been used to prepare dual or theragnostic systems that can be manufactured using various materials, including nanogels, polymeric micelles, liposomes and targeting agents such as cell-penetrating peptides (81,8792), functionalized with drugs and combined with bioactive cellular molecules that increase the specificity and effectiveness of diagnosis and treatment. Furthermore, advances in the manufacture of advanced biological materials such as protein nanomaterials have highlighted their potential in bioengineering and biomedical applications (93) (Fig. 7). The present review emphasizes the participation of three elements of the IGF system (Fig. 7A), which actively participate in tumor survival and resistance mechanisms, summarizing their use as bioactive molecules and/or therapeutic targets of nanocarriers (Table I). It may be concluded that they represent a breakthrough in nano-oncology and have potential in the treatment of resistant cervical tumors.

Table I.

Nanotherapy therapeutics and diagnostics based on elements of the IGF-axis.

Table I.

Nanotherapy therapeutics and diagnostics based on elements of the IGF-axis.

AuthorsNanoparticleIGF-axis targetTheragnostic capacityBioactive moleculesActivityAction mechanism(Refs.)
Chen et al, 2020; Samani et al, 2004; Vaniotis et al, 2018;Trap decoysIGF-1 and IGF-2 ligandsTherapeuticChimeric proteinSystemicBlocks the binding of IGF-1 and −2 with cell receptors(64,69,71)
Yang et al, 2008; Zhou et al, 2016Magnetic iron oxide therapeuticIGF-1RDiagnostic and nanoparticlesMagnetic nanoparticles conjugated with IGF-1Targeted membrane receptorsNanoparticles loaded with drug or fluorescent molecules, targeting IGF-1R in the cell membrane(75,76)
Kibbey et al, 2006; Binkert et al, 1989; Asampille et al, 2018; Swain et al, 2010;Protein nanotubesTumor cell membrane proteins, i.e., integrinsDiagnostic and therapeuticIGFBP-2 carboxyl end (repeated RGD motif) conjugated to leader molecule (ligands)Targeted membrane receptorsNanotubes loaded with drug or fluorescent molecules, targeting membrane proteins(7982)

[i] IGF, insulin-like growth factor; IGF-1R, IGF 1 receptor; IGFBP, IGF binding protein; RGD, Arg-Gly-Asp.

Acknowledgements

The authors acknowledge Professor Alejandro Ariel García-Arriaga (Division of Health, Biological and Environmental Sciences, Open and Distance University of Mexico, Mexico City, México) and Professor Fernando Ferrara-Suárez (Nanotechnology and Biotechnology Engineering Division, Polytechnic University of the Valley of Mexico. Tultitlán, Mexico State, México) for language editing of the manuscript. Parts of the figures were drawn by using pictures from Servier Medical Art. Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/).

Funding

This work was supported by the Research Support of the Polytechnic University of the Valley of Mexico.

Availability of data and materials

Not applicable.

Authors' contributions

JFR and MMR conceived the study, wrote the introduction and conclusions sections, and reviewed and edited the manuscript. LPG, HZM and CCCG wrote the IGF axis, IGF axis in cervical cancer and IGF axis members as therapeutic targets in cervical cancer sections. JN and BMP wrote the extracellular domain of IGF-1R used as a trap nanoparticle and nanoparticles targeting IGF-1R with theragnostic advantages sections. JFR and RVMT wrote the protein nanotubes section. MMR and JAJL participated in the acquisition of data on the IGF axis, extracellular domain of IGF-1R used as a trap nanoparticle, nanoparticles targeting IGF-1R with theragnostic advantages, and protein nanotubes and worked on the development of all figures. Data authentication is not applicable. All authors have 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.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Aldaco-Sarvide F, Pérez-Pérez P, Cervantes-Sánchez G, Torrecillas-Torres L, Erazo-Valle-Solís AA, Cabrera-Galeana P, Motola-Kuba D, Anaya P, Rivera-Rivera S and Cárdenas-Cárdenas E: Mortalidad por cáncer en México: Actualización 2015. Gac Mex Oncol. 17:28–34. 2018.

3 

Granados-García V, Flores YN, Pérez R, Rudolph SE, Lazcano-Ponce E and Salmerón J: Cost of the cervical cancer screening program at the Mexican social security institute. Salud Publica Mex. 56:502–510. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Murillo R, Almonte M, Pereira A, Ferrer E, Gamboa OA, Jerónimo J and Lazcano-Ponce E: Cervical cancer screening programs in Latin America and the Caribbean. Vaccine. 26 (Suppl 11):L37–L48. 2008. View Article : Google Scholar : PubMed/NCBI

5 

McCormack M, Kadalayil L, Hackshaw A, Hall-Craggs MA, Symonds RP, Warwick V, Simonds H, Fernando I, Hammond M, James L, et al: A phase II study of weekly neoadjuvant chemotherapy followed by radical chemoradiation for locally advanced cervical cancer. Br J Cancer. 108:2464–2469. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Gadducci A and Cosio S: Neoadjuvant chemotherapy in locally advanced cervical cancer: Review of the literature and perspectives of clinical research. Anticancer Res. 40:4819–4828. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Fernandez-Retana J, Lasa-Gonsebatt F, Lopez-Urrutia E, Coronel-Martínez J, Cantu De Leon D, Jacobo-Herrera N, Peralta-Zaragoza O, Perez-Montiel D, Reynoso-Noveron N, Vazquez-Romo R and Perez-Plasencia C: Transcript profiling distinguishes complete treatment responders with locally advanced cervical cancer. Transl Oncol. 8:77–84. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Monk BJ, Enomoto T, Kast WM, McCormack M, Tan DSP, Wu X and González-Martín A: Integration of immunotherapy into treatment of cervical cancer: Recent data and ongoing trials. Cancer Treatment Reviews. 106:1023852022. View Article : Google Scholar : PubMed/NCBI

9 

Bhatla N, Aoki D, Sharma DN and Sankaranarayanan R: Cancer of the cervix uteri: 2021 Update. Int J Gynecol Obstet. 155 (Suppl 1):S28–S44. 2021. View Article : Google Scholar

10 

Naga Ch P, Gurram L, Chopra S and Mahantshetty U: The management of locally advanced cervical cancer. Curr Opin Oncol. 30:323–329. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Li H, Wu X and Cheng X: Advances in diagnosis and treatment of metastatic cervical cancer. J Gynecol Oncol. 27:e432016. View Article : Google Scholar : PubMed/NCBI

12 

Höckel S, Schlenger K, Vaupel P and Höckel M: Association between host tissue vascularity and the prognostically relevant tumor vascularity in human cervical cancer. Int J Oncol. 19:827–832. 2001.PubMed/NCBI

13 

Sims LB, Curry KC, Parupalli S, Horner G, Frieboes HB and Steinbach-Rankins JM: Efficacy of surface-modified PLGA nanoparticles as a function of cervical cancer Type. Pharm Res. 36:662019. View Article : Google Scholar : PubMed/NCBI

14 

Sau S, Alsaab HO, Bhise K, Alzhrani R, Nabil G and Iyer AK: Multifunctional nanoparticles for cancer immunotherapy: A groundbreaking approach for reprogramming malfunctioned tumor environment. J Control Release. 274:24–34. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Chaturvedi VK, Singh A, Singh VK and Singh MP: Cancer nanotechnology: A new revolution for cancer diagnosis and therapy. Curr Drug Metab. 20:416–429. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Buzea C, Pacheco II and Robbie K: Nanomaterials and nanoparticles: Sources and toxicity. Biointerphases. 2:MR17–MR71. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Zhu L, Zhou Z, Mao H and Yang L: Magnetic nanoparticles for precision oncology: Theranostic magnetic iron oxide nanoparticles for image-guided and targeted cancer therapy. Nanomedicine (Lond). 12:73–87. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Hewish M, Chau I and Cunningham D: Insulin-like growth factor 1 receptor targeted therapeutics: Novel compounds and novel treatment strategies for cancer medicine. Recent Pat Anticancer Drug Discov. 4:54–72. 2009. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Wang N, Rayes RF, Elahi SM, Lu Y, Hancock MA, Massie B, Rowe GE, Aomari H, Hossain S, Durocher Y, et al: The IGF-Trap: Novel inhibitor of carcinoma growth and metastasis. Mol Cancer Ther. 14:982–993. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Lelbach A, Muzes G and Feher J: The insulin-like growth factor system: IGFs, IGF-binding proteins and IGFBP-proteases. Acta Physiol Hung. 92:97–107. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Schaffer A, Koushik A, Trottier H, Duarte-Franco E, Mansour N, Arseneau J, Provencher D, Gilbert L, Gotlieb W, Ferenczy A, et al: Insulin-like growth factor-I and risk of high-grade cervical intraepithelial neoplasia. Cancer Epidemiol Biomarkers Prev. 16:716–722. 2007. View Article : Google Scholar : PubMed/NCBI

23 

De Meyts P and Whittaker J: Structural biology of insulin and IGF1 receptors: Implications for drug design. Nat Rev Drug Discov. 1:769–783. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Hakuno F and Takahashi SI: IGF1 receptor signaling pathways. J Mol Endocrinol. 61:T69–T86. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Liefers-Visser JAL, Meijering RAM, Reyners AKL, van der Zee AGJ and de Jong S: IGF system targeted therapy: Therapeutic opportunities for ovarian cancer. Cancer Treat Rev. 60:90–99. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Huang Z, Wen Y, Shandilya R, Marks JR, Berchuck A and Murphy SK: High throughput detection of M6P/IGF2R intronic hypermethylation and LOH in ovarian cancer. Nucleic Acids Res. 34:555–563. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Sehat B, Tofigh A, Lin Y, Trocmé E, Liljedahl U, Lagergren J and Larsson O: SUMOylation mediates the nuclear translocation and signaling of the IGF-1 receptor. Sci Signal. 3:ra102010. View Article : Google Scholar : PubMed/NCBI

28 

Brahmkhatri VP, Prasanna C and Atreya HS: Insulin-like growth factor system in cancer: Novel targeted therapies. Biomed Res Int. 2015:5380192015. View Article : Google Scholar : PubMed/NCBI

29 

Mathur SP, Mathur RS and Young RC: Cervical epidermal growth factor-receptor (EGF-R) and serum insulin-like growth factor II (IGF-II) levels are potential markers for cervical cancer. Am J Reprod Immunol. 44:222–230. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Bayes-Genis A, Conover CA and Schwartz RS: The insulin-like growth factor axis: A review of atherosclerosis and restenosis. Circ Res. 86:125–130. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Blat C, Villaudy J and Binoux M: In vivo proteolysis of serum insulin-like growth factor (IGF) binding protein-3 results in increased availability of IGF to target cells. J Clin Invest. 93:2286–2290. 1994. View Article : Google Scholar : PubMed/NCBI

32 

Rajah R, Katz L, Nunn S, Solberg P, Beers T and Cohen P: Insulin-like growth factor binding protein (IGFBP) proteases: Functional regulators of cell growth. Prog Growth Factor Res. 6:273–284. 1995. View Article : Google Scholar : PubMed/NCBI

33 

Butt AJ and Williams AC: IGFBP-3 and apoptosis-a licence to kill? Apoptosis. 6:199–205. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Grimberg A, Liu B, Bannerman P, El-Deiry WS and Cohen P: IGFBP-3 mediates p53-induced apoptosis during serum starvation. Int J Oncol. 21:327–335. 2002.PubMed/NCBI

35 

zur Hausen H: Papillomaviruses and cancer: From basic studies to clinical application. Nat Rev Cancer. 2:342–350. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Scheffner M, Werness BA, Huibregtse JM, Levine AJ and Howley PM: The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell. 63:1129–1136. 1990. View Article : Google Scholar : PubMed/NCBI

37 

Boyer SN, Wazer DE and Band V: E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res. 56:4620–4624. 1996.PubMed/NCBI

38 

Jones DL, Thompson DA and Münger K: Destabilization of the RB tumor suppressor protein and stabilization of p53 contribute to HPV type 16 E7-induced apoptosis. Virology. 239:97–107. 1997. View Article : Google Scholar : PubMed/NCBI

39 

Kuramoto H, Hongo A, Liu YX, Ojima Y, Nakamura K, Seki N, Kodama J and Hiramatsu Y: Immunohistochemical evaluation of insulin-like growth factor I receptor status in cervical cancer specimens. Acta Med Okayama. 62:251–259. 2008.PubMed/NCBI

40 

Aleksic T, Chitnis MM, Perestenko OV, Gao S, Thomas PH, Turner GD, Protheroe AS, Howarth M and Macaulay VM: Type 1 insulin-like growth factor receptor translocates to the nucleus of human tumor cells. Cancer Res. 70:6412–6419. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Moreno-Acosta P, Vallard A, Carrillo S, Gamboa O, Romero-Rojas A, Molano M, Acosta J, Mayorga D, Rancoule C, Garcia MA, et al: Biomarkers of resistance to radiation therapy: A prospective study in cervical carcinoma. Radiat Oncol. 12:1202017. View Article : Google Scholar : PubMed/NCBI

42 

Takeda T, Komatsu M, Chiwaki F, Komatsuzaki R, Nakamura K, Tsuji K, Kobayashi Y, Tominaga E, Ono M, Banno K, et al: Upregulation of IGF2R evades lysosomal dysfunction-induced apoptosis of cervical cancer cells via transport of cathepsins. Cell Death Dis. 10:8762019. View Article : Google Scholar : PubMed/NCBI

43 

Scagliotti GV and Novello S: The role of the insulin-like growth factor signaling pathway in non-small cell lung cancer and other solid tumors. Cancer Treat Rev. 38:292–302. 2012. View Article : Google Scholar : PubMed/NCBI

44 

You L, Liu C, Tang H, Liao Y and Fu S: Advances in targeting insulin-like growth factor signaling pathway in cancer treatment. Curr Pharm Des. 20:2899–2911. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Fu S, Tang H, Liao Y, Xu Q, Liu C, Deng Y, Wang J, Wang J and Fu X: Expression and clinical significance of insulin-like growth factor 1 in lung cancer tissues and perioperative circulation from patients with non-small-cell lung cancer. Curr Oncol. 23:12–19. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Durzyńska J: IGF axis and other factors in HPV-related and HPV-unrelated carcinogenesis (review). Oncol Rep. 32:2295–2306. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Pickard A, Durzynska J, McCance DJ and Barton ER: The IGF axis in HPV associated cancers. Mutat Res Rev Mutat Res. 772:67–77. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Wu X, Tortolero-Luna G, Zhao H, Phatak D, Spitz MR and Follen M: Serum levels of insulin-like growth factor I and risk of squamous intraepithelial lesions of the cervix. Clin Cancer Res. 9:3356–3361. 2003.PubMed/NCBI

49 

Shen MR, Hsu YM, Hsu KF, Chen YF, Tang MJ and Chou CY: Insulin-like growth factor 1 is a potent stimulator of cervical cancer cell invasiveness and proliferation that is modulated by alphavbeta3 integrin signaling. Carcinogenesis. 27:962–971. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Steller MA, Delgado CH, Bartels CJ, Woodworth CD and Zou Z: Overexpression of the insulin-like growth factor-1 receptor and autocrine stimulation in human cervical cancer cells. Cancer Res. 56:1761–1765. 1996.PubMed/NCBI

51 

van der Veeken J, Oliveira S, Schiffelers RM, Storm G, van Bergen En Henegouwen PM and Roovers RC: Crosstalk between epidermal growth factor receptor- and insulin-like growth factor-1 receptor signaling: Implications for cancer therapy. Curr Cancer Drug Targets. 9:748–760. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Pickard A, McDade SS, McFarland M, McCluggage WG, Wheeler CM and McCance DJ: HPV16 down-regulates the insulin-like growth factor binding protein 2 to promote epithelial invasion in organotypic cultures. PLoS Pathog. 11:e10049882015. View Article : Google Scholar : PubMed/NCBI

53 

Kaur G, Balasubramaniam SD and Lee YJ: IGFBP-2 in cervical cancer development. Exp Mol Pathol. 113:1043622020. View Article : Google Scholar : PubMed/NCBI

54 

Schütt BS, Langkamp M, Rauschnabel U, Ranke MB and Elmlinger MW: Integrin-mediated action of insulin-like growth factor binding protein-2 in tumor cells. J Mol Endocrinol. 32:859–868. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Berger AJ, Baege A, Guillemette T, Deeds J, Meyer R, Disbrow G and Schlegel R and Schlegel R: Insulin-like growth factor-binding protein 3 expression increases during immortalization of cervical keratinocytes by human papillomavirus type 16 E6 and E7 proteins. Am J Pathol. 161:603–610. 2002. View Article : Google Scholar : PubMed/NCBI

56 

Mannhardt B, Weinzimer SA, Wagner M, Fiedler M, Cohen P, Jansen-Dürr P and Zwerschke W: Human papillomavirus type 16 E7 oncoprotein binds and inactivates growth-inhibitory insulin-like growth factor binding protein 3. Mol Cell Biol. 20:6483–6495. 2000. View Article : Google Scholar : PubMed/NCBI

57 

Baxter RC: Nuclear actions of insulin-like growth factor binding protein-3. Gene. 569:7–13. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Seligson DB, Yu H, Tze S, Said J, Pantuck AJ, Cohen P and Lee KW: IGFBP-3 nuclear localization predicts human prostate cancer recurrence. Horm Cancer. 4:12–23. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Arcaro A: Targeting the insulin-like growth factor-1 receptor in human cancer. Front Pharmacol. 4:302013. View Article : Google Scholar : PubMed/NCBI

60 

King ER and Wong KK: Insulin-like growth factor: Current concepts and new developments in cancer therapy. Recent Pat Anticancer Drug Discov. 7:14–30. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Navarro M and Baserga R: Limited redundancy of survival signals from the type 1 insulin-like growth factor receptor. Endocrinology. 142:1073–1081. 2001. View Article : Google Scholar : PubMed/NCBI

62 

Park S, Chapuis N, Tamburini J, Bardet V, Cornillet-Lefebvre P, Willems L, Green A, Mayeux P, Lacombe C and Bouscary D: Role of the PI3K/AKT and mTOR signaling pathways in acute myeloid leukemia. Haematologica. 95:819–828. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Qu X, Wu Z, Dong W, Zhang T, Wang L, Pang Z, Ma W and Du J: Update of IGF-1 receptor inhibitor (ganitumab, dalotuzumab, cixutumumab, teprotumumab and figitumumab) effects on cancer therapy. Oncotarget. 8:29501–29518. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Chen YM, Qi S, Perrino S, Hashimoto M and Brodt P: Targeting the IGF-axis for cancer therapy: Development and validation of an IGF-Trap as a potential drug. Cells. 9:10982020. View Article : Google Scholar : PubMed/NCBI

65 

Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, Boland P, Leidich R, Hylton D, Burova E, et al: VEGF-Trap: A VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA. 99:11393–11398. 2002. View Article : Google Scholar : PubMed/NCBI

66 

Messori A, Santarlasci B and Vaiani M: New drugs for rheumatoid arthritis. N Engl J Med. 351:937–938. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Hoffman HM, Throne ML, Amar NJ, Sebai M, Kivitz AJ, Kavanaugh A, Weinstein SP, Belomestnov P, Yancopoulos GD, Stahl N and Mellis SJ: Efficacy and safety of rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated periodic syndromes: Results from two sequential placebo-controlled studies. Arthritis Rheum. 58:2443–2452. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Lindzen M, Carvalho S, Starr A, Ben-Chetrit N, Pradeep CR, Köstler WJ, Rabinkov A, Lavi S, Bacus SS and Yarden Y: A recombinant decoy comprising EGFR and ErbB-4 inhibits tumor growth and metastasis. Oncogene. 31:3505–3515. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Samani AA, Chevet E, Fallavollita L, Galipeau J and Brodt P: Loss of tumorigenicity and metastatic potential in carcinoma cells expressing the extracellular domain of the type 1 insulin-like growth factor receptor. Cancer Res. 64:3380–3385. 2004. View Article : Google Scholar : PubMed/NCBI

70 

Wang N, Lu Y, Pinard M, Pilotte A, Gilbert R, Massie B and Brodt P: Sustained production of a soluble IGF-I receptor by gutless adenovirus-transduced host cells protects from tumor growth in the liver. Cancer Gene Ther. 20:229–236. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Vaniotis G, Moffett S, Sulea T, Wang N, Elahi SM, Lessard E, Baardsnes J, Perrino S, Durocher Y, Frystyk J, et al: Enhanced anti-metastatic bioactivity of an IGF-TRAP re-engineered to improve physicochemical properties. Sci Rep. 8:173612018. View Article : Google Scholar : PubMed/NCBI

72 

Sadick MD, Intintoli A, Quarmby V, McCoy A, Canova-Davis E and Ling V: Kinase receptor activation (KIRA): A rapid and accurate alternative to end-point bioassays. J Pharm Biomed Anal. 19:883–891. 1999. View Article : Google Scholar : PubMed/NCBI

73 

Bulte JWM and Kraitchman DL: Iron oxide MR contrast agents for molecular and cellular imaging. NMR Biomed. 17:484–499. 2004. View Article : Google Scholar : PubMed/NCBI

74 

Miller-Kleinhenz JM, Bozeman EN and Yang L: Targeted nanoparticles for image-guided treatment of triple-negative breast cancer: Clinical significance and technological advances. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 7:797–816. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Yang L, Cao Z, Sajja HK, Mao H, Wang L, Geng H, Xu H, Jiang T, Wood WC, Nie S and Wang YA: Development of receptor targeted magnetic iron oxide nanoparticles for efficient drug delivery and tumor imaging. J Biomed Nanotechnol. 4:439–449. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Zhou H, Qian W, Uckun FM, Zhou Z, Wang L, Wang A, Mao H and Yang L: IGF-1 receptor targeted nanoparticles for image-guided therapy of stroma-rich and drug resistant human cancer. Proc SPIE Int Soc Opt Eng. Apr 17–2016.(Epub ahead of print).

77 

Yu MK, Park J and Jon S: Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics. 2:3–44. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Gao X and Matsui H: Peptide-based nanotubes and their applications in bionanotechnology. Adv Mater. 17:2037–2050. 2005. View Article : Google Scholar : PubMed/NCBI

79 

Kibbey MM, Jameson MJ, Eaton EM and Rosenzweig SA: Insulin-like growth factor binding protein-2: Contributions of the C-terminal domain to insulin-like growth factor-1 binding. Mol Pharmacol. 69:833–845. 2006. View Article : Google Scholar : PubMed/NCBI

80 

Binkert C, Landwehr J, Mary JL, Schwander J and Heinrich G: Cloning, sequence analysis and expression of a cDNA encoding a novel insulin-like growth factor binding protein (IGFBP-2). EMBO J. 8:2497–2502. 1989. View Article : Google Scholar : PubMed/NCBI

81 

Asampille G, Verma BK, Swain M, Shettar A, Rosenzweig SA, Kondaiah P and Atreya HS: An ultra-stable redox-controlled self-assembling polypeptide nanotube for targeted imaging and therapy in cancer. J Nanobiotechnology. 16:1012018. View Article : Google Scholar : PubMed/NCBI

82 

Swain M, Thirupathi R, Krishnarjuna B, Eaton EM, Kibbey MM, Rosenzweig SA and Atreya HS: Spontaneous and reversible self-assembly of a polypeptide fragment of insulin-like growth factor binding protein-2 into fluorescent nanotubular structures. Chem Commun (Camb). 46:216–218. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Arnaout MA, Mahalingam B and Xiong JP: Integrin structure, allostery, and bidirectional signaling. Annu Rev Cell Dev Biol. 21:381–410. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Bellis SL: Advantages of RGD peptides for directing cell association with biomaterials. Biomaterials. 32:4205–4210. 2011. View Article : Google Scholar : PubMed/NCBI

85 

Pedroza-Torres A, López-Urrutia E, García-Castillo V, Jacobo-Herrera N, Herrera LA, Peralta-Zaragoza O, López-Camarillo C, De Leon DC, Fernández-Retana J, Cerna-Cortés JF and Pérez-Plasencia C: MicroRNAs in cervical cancer: Evidences for a miRNA profile deregulated by HPV and its impact on radio-resistance. Molecules. 19:6263–6281. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Fernandez-Retana J, Zamudio-Meza H, Rodriguez-Morales M, Pedroza-Torres A, Isla-Ortiz D, Herrera L, Jacobo-Herrera N, Peralta-Zaragoza O, López-Camarillo C, Morales-Gonzalez F, et al: Gene signature based on degradome-related genes can predict distal metastasis in cervical cancer patients. Tumour Biol. Jun 22–2017.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

87 

Cuggino JC, Molina M, Wedepohl S, Igarzabal CIA, Calderón M and Gugliotta LM: Responsive nanogels for application as smart carriers in endocytic pH-triggered drug delivery systems. Eur Polym J. 78:14–24. 2016. View Article : Google Scholar

88 

Patel SG, Sayers EJ, He L, Narayan R, Williams TL, Mills EM, Allemann RK, Luk LYP, Jones AT and Tsai YH: Cell-penetrating peptide sequence and modification dependent uptake and subcellular distribution of green florescent protein in different cell lines. Sci Rep. 9:62982019. View Article : Google Scholar : PubMed/NCBI

89 

Poshteh Shirani M, Rezaei B, Khayamian T, Dinari M, Karami K, Mehri-Lighvan Z, Hosseini Shamili F, Ramazani M and Alibolandi M: Folate receptor-targeted multimodal fluorescence mesosilica nanoparticles for imaging, delivery palladium complex and in vitro G-quadruplex DNA interaction. J Biomol Struct Dyn. 36:4156–4169. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Tomitaka A, Arami H, Huang Z, Raymond A, Rodriguez E, Cai Y, Febo M, Takemura Y and Nair M: Hybrid magneto-plasmonic liposomes for multimodal image-guided and brain-targeted HIV treatment. Nanoscale. 10:184–194. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Trujillo-Nolasco M, Cruz-Nova P, Ferro-Flores G, Gibbens-Bandala B, Morales-Avila E, Aranda-Lara L, Vargas M and Ocampo-García B: Development of 177Lu-DN(C19)-CXCR4 ligand nanosystem for combinatorial therapy in pancreatic cancer. J Biomed Nanotechnol. 17:263–278. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Wei T, Chen C, Liu J, Liu C, Posocco P, Liu X, Cheng Q, Huo S, Liang Z, Fermeglia M, et al: Anticancer drug nanomicelles formed by self-assembling amphiphilic dendrimer to combat cancer drug resistance. Proc Natl Acad Sci USA. 112:2978–2983. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Sun H, Li Y, Yu S and Liu J: Hierarchical self-assembly of proteins through rationally designed supramolecular interfaces. Front Bioeng Biotechnol. 8:2952020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 25 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Morales-Rodríguez M, Paniagua-García L, Narayanan J, Zamudio-Meza H, Moreno-Torres RV, Cortés-González CC, Juanico-Lorán JA, Martínez-Pérez B and Fernández-Retana J: Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review). Oncol Lett 25: 128, 2023
APA
Morales-Rodríguez, M., Paniagua-García, L., Narayanan, J., Zamudio-Meza, H., Moreno-Torres, R.V., Cortés-González, C.C. ... Fernández-Retana, J. (2023). Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review). Oncology Letters, 25, 128. https://doi.org/10.3892/ol.2023.13714
MLA
Morales-Rodríguez, M., Paniagua-García, L., Narayanan, J., Zamudio-Meza, H., Moreno-Torres, R. V., Cortés-González, C. C., Juanico-Lorán, J. A., Martínez-Pérez, B., Fernández-Retana, J."Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review)". Oncology Letters 25.3 (2023): 128.
Chicago
Morales-Rodríguez, M., Paniagua-García, L., Narayanan, J., Zamudio-Meza, H., Moreno-Torres, R. V., Cortés-González, C. C., Juanico-Lorán, J. A., Martínez-Pérez, B., Fernández-Retana, J."Insulin‑like growth factor axis: A potential nanotherapy target for resistant cervical cancer tumors (Review)". Oncology Letters 25, no. 3 (2023): 128. https://doi.org/10.3892/ol.2023.13714