Next Article in Journal
Telomere Dysfunction in Pediatric Patients with Differences/Disorders of Sexual Development
Previous Article in Journal
At the Intersection of Critical Care and Infectious Diseases: The Year in Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Insulin-like Growth Factor-1 Receptor in Tobacco Smoking-Associated Lung Cancer Development

by
Ayaz Shahid
1,*,
Shaira Gail Santos
2,†,
Carol Lin
1,† and
Ying Huang
1,*
1
Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766, USA
2
College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2024, 12(3), 563; https://doi.org/10.3390/biomedicines12030563
Submission received: 15 January 2024 / Revised: 23 February 2024 / Accepted: 29 February 2024 / Published: 2 March 2024
(This article belongs to the Section Cancer Biology and Oncology)

Abstract

:
Cancer remains a significant global health concern, with lung cancer consistently leading as one of the most common malignancies. Genetic aberrations involving receptor tyrosine kinases (RTKs) are known to be associated with cancer initiation and development, but RTK involvement in smoking-associated lung cancer cases is not well understood. The Insulin-like Growth Factor 1 Receptor (IGF-1R) is a receptor that plays a critical role in lung cancer development. Its signaling pathway affects the growth and survival of cancer cells, and high expression is linked to poor prognosis and resistance to treatment. Several reports have shown that by activating IGF-1R, tobacco smoke-related carcinogens promote lung cancer and chemotherapy resistance. However, the relationship between IGF-1R and cancer is complex and can vary depending on the type of cancer. Ongoing investigations are focused on developing therapeutic strategies to target IGF-1R and overcome chemotherapy resistance. Overall, this review explores the intricate connections between tobacco smoke-specific carcinogens and the IGF-1R pathway in lung carcinogenesis. This review further highlights the challenges in using IGF-1R inhibitors as targeted therapy for lung cancer due to structural similarities with insulin receptors. Overcoming these obstacles may require a comprehensive approach combining IGF-1R inhibition with other selective agents for successful cancer treatment.

1. Introduction

Cancer is among the leading causes of death worldwide and was responsible for nearly 10 million deaths in 2020 [1]. Among different types of cancer, lung cancer is the most common type of cancer globally [2], causing the most cancer-related deaths, with approximately 1.8 million deaths globally (18% of cancer-related deaths) [2,3]. In recent years, lung cancer has seen a rapid increase in both prevalence and mortality in industrially developed countries [4].
Receptor tyrosine kinases (RTKs) are a group of cell membrane receptors that play a vital role in multiple cellular processes, such as cell growth, differentiation, proliferation, and survival [5]. Many studies have shown that the dysregulation of RTK signaling can lead to various diseases, including cancer [6]. One of the members of the RTK family is the Insulin-like Growth Factor 1 Receptor (IGF-1R) [7], which plays a crucial role in mediating the biological effects of insulin-like growth factors (IGFs), specifically IGF-1 and partly IGF-2, which are produced in response to the release of growth hormone from the pituitary gland [7]. IGF-1R is also known for its oncogenic potential, which means that its abnormal activation or overexpression can contribute to the development and progression of various cancer types, including lung cancer [8,9,10,11]. Therefore, IGF-1R has been identified as a promising target for cancer treatment. The targeted inhibition of IGF-1R in cancer treatment yields more benefits than traditional non-selective treatments like chemotherapy and radiation therapy, which can harm normal cells [12]. However, IGF-1R inhibition can also lead to metabolic abnormalities, such as insulin dysregulation, through the action of IGF-1R/insulin receptor (IR) hybrids (as explained in Figure 1) [13,14]. Therefore, more investigations are needed in this area to determine the safest and most effective application of IGF/IGF-1R inhibitors as a therapy for lung cancer. Overall, the IGF/IGF-R signaling axis shows promise as a potential target for lung cancer therapies, but poses some challenges that cannot be overlooked.
Tobacco smoking is the leading cause of cancer-related deaths in both men and women in the United States [15]. Although extensive research has already been conducted, there are still unknown pathways that are activated by tobacco smoke-related carcinogens. Numerous studies are currently underway to address this research gap [16]. Targeting signaling pathways activated by tobacco smoke carcinogens is important in preventing lung cancer, as tobacco smoking causes one-third of all cancer-related deaths annually [17]. Tobacco smoke is comprised of thousands of chemicals, including at least 70 compounds known to cause cancer [18]. These cancer-causing agents can affect cell proliferation and promote uncontrolled cell growth by abnormally activating growth receptors [19,20]. Various studies have shown that tobacco smoke carcinogens can activate growth factor receptors, including IGF-1R, which play an important role in lung cancer development [21]. In this review article, we will discuss how tobacco smoke-specific carcinogens and IGF-1R signaling pathways are connected to lung cancer development, and how IGF-1R can serve as a potential target for lung cancer therapy.

2. What Is the IGF/IGF-R Signaling Axis?

The IGF/IGF-R signaling axis consists of two RTKs, IGF-1R and IGF-2R (Figure 1), along with their respective ligands, IGF-1/2, and binding proteins in the serum known as insulin-like growth factor binding proteins (IGFBPs) that act as transporter proteins [22,23]. The binding of IGFs to the IGF-Rs results in the subsequent downstream activation of several pathways that are crucial in cell proliferation and cell cycle progression [13] (Figure 2). Due to this mechanism, the IGF/IGF-R axis plays significant roles in many biological processes, such as normal cell growth and survival, and contributes to numerous pathological processes, including cancer development and progression [24].
The IGF/IGF-R axis is a highly conserved signaling pathway that has been around for millions of years, even before mammalian vertebrates existed [25,26]. While this pathway plays a crucial role in normal organismal growth and survival, it has also been shown to regulate animal carbohydrate and lipid metabolism [27,28,29,30,31,32]. The IGF-R is widespread in the body, and nearly every tissue can produce IGFs, making it an important research area due to its diverse biological effects and therapeutic potential [28,33,34]. It has also been demonstrated that the IGF/IGF-R axis is involved with numerous endocrine signaling pathways [28,35], which highlights the pleiotropic effects of IGF-R activation through the action of receptor hybrids composed of IGF-1R and IR monomers.
Similar to the IGF-R, IR is also a cell surface receptor that initiates downstream signaling cascades within the cell (Figure 1). However, both receptors respond to different ligands, as they differ slightly in two domains involved in ligand specificity: the first leucine-rich region, L1, and a cysteine-rich region, CR [34,36]. IR is mainly activated by insulin, whereas IGF-1R is activated by IGF-1 and IGF-2, leading to distinct signaling pathways upon activation of both receptors [37,38]. IR activation plays an important role in metabolic homeostasis by regulating glucose transport and glycogen synthesis [39,40]. Conversely, the activation of IGF-1R triggers PI3K and Ras-mediated pathways, primarily resulting in cell proliferation, growth, and survival [40,41] (Figure 2). The dysregulation of IGF-1R and IR signaling has been implicated in various diseases. For example, aberrant IR signaling is associated with type 1 and 2 diabetes [38]. As discussed earlier, abnormal IGF-1R activation has been associated with cancer initiation and development.
When exploring the IGF-1R signaling axis, it is imperative to consider the role of IR. Because of the structural homology between these two receptors, it has been found that IGF-1R monomers have the ability to dimerize with IR monomers, particularly the A isoform (IR-A), to create a functional hybrid receptor that can bind both IGFs and insulin [40] (Figure 1). IR-A is predominantly found in fetal and cancer cells and plays a crucial role in promoting cell cycle progression [34]. Ongoing research is being conducted to understand the roles of hybrid IGF-1R/IR receptors in various diseases and cellular processes, which is crucial for unraveling the complexities underlying cancer initiation and progression, as well as informing the development of targeted therapies.

3. Structure and Functions of IGF-1R and its Components

The IGF-1R is a crucial component of the IGF/IGF-R signaling system, composed of two extracellular α subunits (IGF-1Rα) and two transmembrane β subunits (IGF-1Rβ) [22] that are produced by a single gene located on chromosome 15q26.3 [42]. The IGF-1Rα and IGF-1Rβ subunits form a tetramer [33] that plays a vital role in cell survival and transformation due to its tyrosine kinase activity [23]. IGF-2R, also known as the mannose 6-phosphate receptor, does not possess tyrosine kinase or signal transduction activity. Instead, IGF-2R helps regulate IGF-2 levels in the body by capturing and transporting it to the lysosome for degradation [23,43,44] (Figure 1). IGF-1 and IGF-2 can bind to IGF-1R; however, IGF-1R has a 15- to 20-fold higher affinity for IGF-1 than IGF-2 [45]. The half-life and bioavailability of IGF-1 and IGF-2 in the circulation vary depending on the level of IGFBPs in the serum, which have different binding affinities and specificities [46,47]. A lack of IGF-1/2 ligands can hinder growth and lead to metabolic issues [48]. On the other hand, high levels of IGF-1/2 are linked to a higher risk of cancer and a lower lifespan [8,49].
When IGF-1 and IGF-2 bind to IGF-1R, they activate the receptor’s tyrosine kinase domain, phosphorylating receptor substrates. This activation leads to two main signaling pathways: (1) the PI3K pathway initiated by insulin receptor substrate (IRS), which mainly affects metabolic outcomes, and (2) the Ras/mitogen-activated protein kinase (MAPK) pathway initiated by Src homology and collagen (Shc) adaptor protein, which mainly affects cell growth and differentiation [13,50] (Figure 2). Thus, the activation of IGF-1R simultaneously promotes cell survival and proliferation while providing inhibitory protective mechanisms against apoptosis [51,52]. Studies have demonstrated that IGF-1R overexpression was associated with disease progression, poor prognosis, and treatment resistance in breast cancer, esophageal adenocarcinoma, colorectal cancer, and squamous cell carcinoma [53]. A meta-analysis has also shown that IGF-1R expression is an unfavorable factor for survival in non-small cell lung cancer (NSCLC) patients, and is associated with smoking status and tumor size [53].
The IGFBP family is evolutionarily ancient and highly conserved in vertebrates [54,55]. Six types of IGFBPs, designated IGFBP-1 through IGFBP-6, have been identified in the serum and serve to decrease the amount of free circulating IGFs through varying binding affinities and specificities [46]. The most critical binding protein, IGFBP-3, binds up to 80% of IGF-1 in the serum [23] with a higher affinity for IGF-1 than IGF-1R possesses [56]. Studies have found that high IGF-1 levels in the blood may increase the risk of lung cancer, while high levels of circulating IGFBP-3 may decrease risk [45,57,58]. Previous studies have demonstrated that p53 regulates the expression of IGFBP-3 and that increased transcription leads to higher expression levels [59,60]. However, when p53 is mutated, it cannot activate IGFBP-3, which thus fails to induce apoptosis [61,62,63]. While the expression of IGFBP-3 may not be completely absent, a decrease in its expression can cause an elevation in the local availability of IGF-1 in lung tissue, thereby increasing the risk of developing lung cancer [64,65]. These data suggest that IGFBP-3 could be a promising therapeutic option for cancer treatment.

4. IGF-1R Overexpression and Activation in Lung Cancer Initiation and Development

IGF-1R, an RTK activated by IGF-1 and IGF-2, contributes to lung cancer by initiating pathways involved in cell proliferation and survival [26]. Several studies have shown that IGF-1R activation leads to the opening of voltage-gated calcium channels, leading to an increase in intracellular Ca2+ levels [66,67]. The increased Ca2+ increases the phosphorylation of cAMP response element-binding protein (CREB) via the Erk5 pathway, resulting in the proliferation and transformation of lung epithelial cells [67]. This Ca2+ signaling mechanism leading to uncontrolled cell growth is crucial in promoting the formation and expansion of lung tumors [68,69]. Another study showed a significant increase in the expression of IGF-1 and IGF-1R in high-grade dysplastic bronchial tissues compared to normal tissues [70]. Additionally, the same study discovered that preneoplastic human bronchial epithelial (HBE) cells produce autocrine IGFs, which have the potential to elicit spontaneous lung tumor formation, especially after exposure to tobacco smoke carcinogens [70,71]. Some studies have also shown that IGFs are produced at high levels by the stromal tissue surrounding cancer cells, creating a favorable microenvironment for tumorigenesis and providing a prognostic factor for outcomes in lung cancer [10,72]. From these findings, it can be concluded that high IGF expression and signaling is a vital component of early lung carcinogenesis, transforming cells exposed to tobacco smoke carcinogens. The increased activity occurs through the activation of biochemical signaling pathways and the induction of DNA mutations, both caused by tobacco carcinogens [70].
In addition, IGF-1R has been well-studied for use in prognostic predictions in various malignancies, such as breast, prostate, lung, and colorectal cancers [73,74,75,76,77,78,79]. Two meta-analyses showed that a high expression of IGF-1R was associated with poor disease-free survival in non-small cell lung cancer (NSCLC) [53,80], a malignancy that is predominantly linked to tobacco smoking. However, one of these analyses found that poor overall survival in patients with NSCLC and small cell lung cancer (SCLC) could not be predicted based on IGF-1R expression [80]. Similarly, another study could not find a conclusive association between the prognosis of patients with SCLC and IGF-1R expression [81]. These findings suggest that IGF-1R may participate in developing NSCLC and could serve as a predictive prognostic factor for this specific cancer type.

5. Implication of IGF-1R Signaling Pathways in Tobacco Smoke-Associated Carcinogenesis

It is well-established that the overexpression of IGF-1R and IGFs, as well as the deregulation of IGF-1R signaling pathways, are closely associated with the development of lung cancer [70]. Given that tobacco smoking is the main risk factor for lung cancer, several studies have shown that tobacco smoke-specific carcinogens activate IGF-1R signaling pathways [82,83,84]. For example, a study reported that the NSCLC tumor tissues isolated from smokers showed significantly greater levels of phosphorylated IGF-1R (pIGF-1R) compared with tissues from non-smokers. They also found that nicotine-derived nitrosamine ketone, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a carcinogen specific to tobacco, stimulates the rapid Ca2+-mediated exocytosis of IGF-2, which can then bind to and activate IGF-1R [84]. Additionally, NNK was also shown to directly induce increased levels of pIGF-1R in NSCLC cells via β-adrenergic (β-AR) signaling. Treatment with β-AR antagonists resulted in the suppression of NNK-mediated cell transformation in human bronchial epithelial (HBE) cells and tumor formation in mouse models [83].
Furthermore, tobacco smoke works in conjunction with IGFs by accelerating its activity as an autocrine ligand for IGF-1R activation in lung epithelial cells [70,84]. Tobacco smoking also leads to disruptions in the renin-angiotensin system, which has been implicated in lung cancers and has been shown to mediate the activation of the IGF-1R signaling axis [82]. As described previously, the activation of IGF-1R can elicit spontaneous lung tumor formation through the aberrant activation of pathways involved in cell growth and survival. Thus, these findings suggest that tobacco smoking promotes lung tumorigenesis and the progression of malignancy through dysregulated IGF-1R activation. Below, we discuss potential downstream events after IGF-1R activation that contribute to transforming cells exposed to tobacco smoke carcinogens.

5.1. Overactivation of IGF-1R Increases Proliferation and Metastasis of Lung Cancer

It has been demonstrated that the expressions of IGF-1 and IGF-2 increase during lung carcinogenesis caused by tobacco smoking, and the expression and phosphorylation of IGF-1R are increased in high-grade dysplastic lesions compared to standard lung tissues [70]. Upon activation, IGF-1R is phosphorylated, resulting in a signaling cascade that stimulates downstream pathways involved in cell proliferation and cell cycle progression, including the P13K/Akt/mTOR and Ras/Raf/MAPK pathways [13]. The activation of IGF-1R has anti-apoptotic effects on cancer cells, which contributes to cancer development and progression [85,86]. For tumor cells to spread to distant organs, they must be able to detach from the extracellular matrix. IGF-1R signaling can prevent cancer cells from undergoing anoikis, a type of cell death that occurs when cells lose their attachment to the extracellular matrix and neighboring cells, possibly via inhibiting the activation of p53 and p21 [87]. The activation of IGF-1R also promotes anchorage-independent growth by recruiting RACK1-mediated STAT3, an important molecular mediator for cell growth in a non-adherent manner [82,88,89]. According to Yang et al., deficiency of IGF-1R in the lung tumor microenvironment resulted in decreased tumor initiation and progression, due to reduced proliferation, vascularization, fibrosis, and inflammation, as well as immunosuppression [90]. Therefore, it can be concluded that IGF-1R activation is crucial for the anchorage-independent growth of cancer cells, and it promotes tumor metastasis and the progression of malignancy [87].

5.2. Overactivation of IGF-1R Promotes Epithelial–Mesenchymal Transition (EMT) and Stemness of Cancer Cells

EMT is a critical event in the cellular evasion of death signals and an essential hallmark of cancer metastasis [91,92]. The significant changes during the EMT include the downregulation of epithelial markers, the rearrangement of the cytoskeleton, the loss of cell–cell adhesion and apical–basal polarity, and the acquisition of mesenchymal phenotype markers [93,94]. These cellular changes collectively drive cancer cells into a stem cell-like state, imparting the unique ability of self-renewal that significantly increases the difficulty of treating cancer [95].
Various studies have demonstrated that IGF-1R signaling induces EMT through multiple mechanisms in different cancers, including breast, prostate, and lung [96,97,98,99,100]. One proposed mechanism by which this occurs is through the IGF-1R activation of the PI3K/Akt/mTOR pathway, which leads to the upregulation of transcription factors like ZEB1 [95] and the stabilization of SLUG [101,102]. Both ZEB1 and SLUG function as negative regulators of E-cadherin expression, contributing to forming a mesenchymal phenotype [98]. As an alternative mechanism, it has also been demonstrated that IGF-1R may trigger EMT via interactions with STAT3, FAK, and NF-kB [101,103,104]. Given these findings, it appears that IGF-1R induces EMT and stem cell-like properties in cancer cells through numerous downstream targets, thereby increasing the chances of tumor metastasis to other sites in the body.

6. IGF-1R and Anti-Cancer Drug Resistance in Lung Cancer

Chemotherapy is commonly used to treat cancer, but resistance to chemotherapeutic agents often leads to treatment failure [105]. Some common resistance mechanisms include changes in the target proteins to decrease binding affinity, enhanced efflux pump to remove the drug from the cytoplasm, the absence of cellular mechanisms that result in apoptosis, and the use of alternative activating signaling pathways [105,106]. Accumulated evidence suggests that the activation of the IGF-1R signaling pathway plays an important role in chemotherapy resistance. Previous studies have shown that the overexpression of IGF-1R is associated with poor chemotherapy outcomes, while the inhibition of IGF-1R leads to improved chemotherapy responses in different types of cancers [9]. IGF-1R-mediated pathways promote cellular proliferation, inhibit apoptosis, alter drug targets, and increase the expression of transporter proteins to reduce intracellular drug concentration, all of which contribute to chemotherapeutic resistance [107].
The role of the IGF-1R signaling pathway is crucial in developing treatment resistance in lung cancer [108]. A study conducted on NSCLC cells that had developed resistance to cisplatin showed a decrease in the expression of IGFBP-3 and an increase in the activation of IGF-1R signaling, leading to the development of cisplatin resistance in NSCLC [109]. The resistance was reversed when human recombinant IGFBP-3 was used or when IGF-1R was inhibited using siRNA [109]. In another study on cisplatin resistance in NSCLC cells, the loss of IGFBP-3 expression resulted in reduced tumor cell sensitivity to cisplatin [110]. These findings indicate that components of the IGF-1R signaling axis, including IGFBP-3, play important roles in the cellular development of drug resistance to commonly used lung cancer therapies.
Furthermore, the IGF-1R signaling pathway is associated with resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) used for the treatment of NSCLC. EGFR is overexpressed in a majority of NSCLC cases; however, two available EGFR TKIs, erlotinib and gefitinib, have yielded poor responses in NSCLC patients with a history of smoking compared to patients who have never smoked [111]. It was also shown that the prolonged use of erlotinib or gefitinib for NSCLC resulted in acquired drug resistance mediated by the upregulation of IGF-1R [112,113]. According to Hurbin et al., the inhibition of IGF-1R with anti-IGF-1R antibodies re-sensitized resistant cells to gefitinib and induced apoptosis [114]. Acquired resistance to afatinib, another available EGFR TKI given to NSCLC patients with known EGFR mutations, is also attributed to the activation of the IGF-1R signaling pathway. The knockdown of IGF-1R sensitized resistant cells to afatinib and induced apoptosis in those cells [115]. In addition, NSCLC cells often develop resistance to osimertinib, a newer generation EGFR TKI used as a first- or second-line treatment for lung cancer. This resistance is mediated by IGF-1R activation through IGF-2 autocrine signaling [116]. These studies suggest that IGF-1R inhibition or a combination of EGFR/IGF-1R inhibition strategies could be potential therapeutic options for lung cancer patients who develop resistance to targeted EGFR inhibitors.
The inhibition of IGF-1R has also been revealed to improve therapeutic response to anti-cancer agents in cancers that do not involve EGFR mutations. In a study of a patient with anaplastic lymphoma receptor tyrosine kinase (ALK) fusion-positive lung cancer who had developed TKI resistance, combined ALK and IGF-1R inhibition improved therapeutic efficacy [117]. Another study also found that the overexpression of IGF-1R resulted in cells with acquired resistance to PI3K inhibitors [103]. The targeted knockdown of IGF-1R expression using specific siRNAs or IGF-1R TKIs reversed the resistance. These results suggest that long-term treatment with PI3K inhibitors may cause reversible drug resistance, and targeting IGF-1R provides a promising strategy to overcome the resistance [118]. Overall, the IGF-1R signaling pathway plays a crucial role in the cellular development of anti-cancer drug resistance, and the use of IGF-1R inhibitors in conjunction with other therapies offers a promising direction for lung cancer treatment.

7. The IGF/IGF-1R Signaling Axis Is a Potential Target for Cancer Therapy

The IGF-1R signaling pathway offers promising targets for cancer therapy. Using monoclonal antibodies, small molecule tyrosine kinase inhibitors, and IGF-1R mutations and silencing are four common anti-cancer strategies targeting the IGF signaling system. These strategies are used to treat various types of cancer, including lung cancer [11] (Figure 3). Since pre-clinical studies may contribute to the decision-making process for advancing these therapies to clinical trials, various pre-clinical studies have investigated IGF-1R-targeted therapies in lung cancer models, using monoclonal antibodies and tyrosine kinase inhibitors (Table 1). Monoclonal antibodies that target IGF-1R, such as cixutumumab, dalotuzumab, and figitumumab, are used for both in vitro and in vivo lung cancer treatments. Similarly, tyrosine kinase inhibitors like linsitinib, picropodophyllin, and BMS-754807 can also target IGF-1R for lung cancer therapy (Table 1). Clinical studies involving IGF-1R-targeted therapies in lung cancer patients have been conducted to evaluate the effectiveness of these therapies (Table 2). Presently, numerous clinical trials are underway to evaluate the effectiveness of targeting IGF-1R (Table 2). For example, a phase II clinical trial was conducted to investigate the effectiveness of figitumumab (CP-751,871), an IGF-1R monoclonal antibody, in combination with chemotherapy (carboplatin and paclitaxel) for treating advanced NSCLC in patients. Although pre-clinical studies demonstrated promising results, the results from clinical trials showed that the combination therapy did not significantly improve overall survival compared to chemotherapy alone. However, it did demonstrate potential benefits in specific patient subgroups, highlighting the complexity of patient selection for IGF-1R-targeted therapy [119].
Furthermore, another study found that cancer cells were sensitized to cisplatin upon IGF-1R inhibition, which can occur through IGF-1R inhibitors or siRNA-mediated IGF-1R suppression [151]. Combining IGF-1R inhibition with other selective inhibitors of PI3K or ATR, a kinase involved in the cellular response to DNA damage, displayed synergistic therapeutic effects [151,152]. Also, combining an IGF-1R monoclonal antibody and rapamycin led to complete tumor regression in pediatric sarcoma, surpassing the effects of single therapeutic agents that only induced modest growth delay [153]. Other pre-clinical studies utilizing IGF inhibitors have exhibited synergism when used with other chemotherapy agents or radiation therapy [23]. It has been found that some microRNAs (miRNAs) are likely to affect the expression of IGF-1R or its receptor signaling activity [154]. The findings of this study also indicate that PI3K and AKT, which are components downstream of the IR, play a crucial role in mediating signals for the IGF-1R pathway. Additionally, further investigations into miRNAs could provide valuable insights because of their indirect impact on the function of IGF-1R. Because insulin resistance involves the dysregulation of multiple growth factor signals, including IGF-1R, miRNAs affecting IR sensitivity are also likely to impact the IGF-1R pathway. These collective endeavors highlight the potential to use IGF-1R inhibition as a potent anti-cancer therapy, mainly when employed in conjunction with complementary treatment modalities.
As discussed previously, the IGF-1R signaling axis plays a critical role in many cellular processes. Consequently, IGF-1R-targeted therapies for any component of the IGF-1R pathway exhibit anti-cancer effects, such as inhibiting cell proliferation, inducing apoptosis, reducing angiogenesis, and reversing drug resistance (Table 3). All these mechanisms work together to synergistically enhance the therapeutic effectiveness of IGF-1R-targeted therapies in cancer treatment.
Unfortunately, despite these promising pre-clinical and early-phase clinical findings, numerous large-scale clinical trials involving NSCLC have yielded disappointing results, potentially due to a lack of patient selection markers [14,22,163]. One study on IGF-1R expression in NSCLC found that treatment with IGF-1R TKIs exhibited significant anti-tumor activity in NSCLC cells with wild-type EGFR and KRAS. Anti-tumor efficacy was not seen in NSCLC cells with mutations in those genes, indicating that EGFR and KRAS mutation status can be predictive markers of response to IGF-1R TKIs [164]. Other early-phase clinical trials have provided evidence that high levels of circulating IGF-1 in NSCLC cases can serve as a potential selection marker for response to IGF-1R inhibition [22,165]. These findings underscore the importance of refining patient selection criteria to harness the potential of IGF-1R inhibition in lung cancer treatment.

8. Challenges in Targeting the IGF/IGF-1R Signaling Axis

While the IGF/IGF-R signaling axis shows promise as a potential target for lung cancer therapies, previous studies have revealed that targeting this pathway presents challenges that cannot be overlooked. As discussed previously, the ability of IGF-1R monomers to dimerize with the IR-A isoform and bind both IGFs and insulin poses a considerable obstacle [14]. Given this, the targeted inhibition of IGF-1R can also lead to metabolic abnormalities, such as hyperglycemia and insulin dysregulation, through the action of IGF-1R/IR-A hybrids [13,14]. Thus, further investigations exploring the complex interplay between IGF-1R, and IR must be undertaken in order to understand the nuances associated with these signaling pathways. In addition, while many clinical trials have tested the efficacy of drugs targeting the IGF/IGF-1R signaling axis, little work has been done to elucidate specific patient biomarkers that can predict therapeutic response [163,166,167].
Consequently, previous clinical trials using monoclonal antibodies and tyrosine kinase inhibitors against IGF-1R have failed to demonstrate a significant benefit for cancer patients, likely due to a lack of patient selection [151,162,168]. Because many successful targeted anti-cancer therapies have relied heavily on the use of predictive biomarkers [166], more investigation into this area is crucial for determining the most effective application of IGF/IGF-1R inhibitors as a lung cancer therapy. These challenges emphasize that a simplistic approach will not suffice, and that drug-based inhibition of any component of the IGF-1R signaling system should be applied in conjunction with chemotherapy agents. Further exploration in this research field is required, primarily focusing on circumventing IGF-1R/IR-A hybrids and identifying specific patient biomarkers. However, despite these important considerations, the IGF/IGF-1R signaling axis still holds much promise as a potential candidate for future cancer therapeutic interventions, especially for lung cancer.

9. Conclusions

In this review, the significant role of the IGF/IGF-1R signaling axis in the development and progression of lung cancer, particularly in the context of tobacco smoke-induced carcinogenesis, is highlighted. Our findings emphasize the intricate mechanisms by which IGF-1R contributes to various aspects of lung cancer pathogenesis, including tumor proliferation, metastasis, EMT, stemness, and resistance to both chemotherapy and targeted therapies. Because of this, the IGF-1R signaling pathway serves as an attractive target for the development of lung cancer treatments. However, many challenges are associated with pursuing IGF-1R inhibitors, which have been highlighted by the disappointing results of recent clinical phase trials.
The setbacks observed in recent clinical trials with IGF-1R inhibitors highlight the need to refine patient selection criteria and identify predictive biomarkers to assess the efficacy of IGF-1R inhibition in clinical trials. Additionally, the development of successful cancer therapies requires a thorough understanding of the structural components and downstream effector pathways of the IGF/IGF-R axis, all of which offer potential avenues for novel therapeutic targets. Although many studies have explored IGF-1R inhibition as a treatment strategy, this system’s complexity and drug design challenges highlight the need for more nuanced and tailored approaches. Furthermore, the diverse functions of IGFBPs, which may operate independently of IGF signaling and are modulated by factors like p53, can provide new opportunities for therapeutic interventions.
The activation of IGF-1R by carcinogens in tobacco smoke can have significant downstream effects on lung cancer development, promoting cell growth, proliferation, and survival. The cumulative effect of these processes creates favorable circumstances for tumorigenesis within the lungs, as well as for metastasis due to tumor progression. When aberrantly activated, this pathway is also associated with the emergence of cancer stem cells. This can lead to tumor heterogeneity, therapy resistance, and a greater likelihood of disease recurrence. Gaining further clarity on the pivotal role of IGF-1R in lung cancer development is instrumental in identifying effective treatment strategies for this malignant disease.
In addition to the impact of IGF-1R on the development and progression of lung cancer, there is compelling evidence indicating that the IGF/IGF-1R axis plays a significant role in cancer treatment resistance. Dysregulation within this signaling pathway has been implicated in resistance to traditional chemotherapeutic agents and targeted therapies such as EGFR inhibitors. This underscores the need to address IGF-1R signaling within lung cancer treatment. Targeting the IGF/IGF-1R axis is a promising therapeutic approach, particularly when combined with other targeted agents or chemotherapy regimens. Pre-clinical studies have shown significant benefits can be derived from such a combinatorial strategy, suggesting that overcoming treatment resistance requires multifaceted and comprehensive interventions to counteract the tumor-promoting properties of IGF-1R.

10. Future Directions

Because of the structural homology between IGF-1R and IR, it is necessary to gain clarity on hybrid IGF-1R/IR receptors and their implications for disease progression and treatment response. Understanding the intricacies of these hybrid receptors is imperative for designing effective, targeted therapeutic interventions that minimize adverse side effects and metabolic disturbances. In addition, efforts should be directed towards identifying components of the IGF/IGF-1R axis that hold promise as predictive or prognostic biomarkers, thereby facilitating the development of personalized medicine tailored to individual patients. Setbacks observed in recent clinical trials exploring IGF-1R-targeted therapies indicate a need for more refined predictive biomarkers, such as EGFR or KRAS mutation status, to identify patients most likely to benefit from these therapies. Exploring the potential synergies between IGF-1R inhibition and established therapeutic modalities is also warranted, as these combinatorial approaches can overcome drug resistance and enhance treatment efficacy.
Furthermore, it is essential to delve into the IGF-independent functions of IGFBPs, which could reveal novel therapeutic targets with clinical implications beyond cancer treatment. Lastly, unraveling the complexities of IGR-1R signaling may reveal downstream targets beyond the receptor itself, which, if targeted, may present a powerful tool for circumventing resistance mechanisms and improving treatment strategies. Therefore, continued exploration of the intricate connections within the IGF/IGF-1R signaling axis is indispensable for realizing the full therapeutic potential of IGF-1R inhibition in the clinical setting, especially in cancer treatment.

Author Contributions

Conceptualization, A.S.; writing—original draft preparation, A.S., S.G.S. and C.L.; writing—review and editing, Y.H., A.S., S.G.S. and C.L.; supervision, Y.H.; funding acquisition, Y.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partly supported by the National Cancer Institute of the National Institutes of Health under award number R03CA256241 (Y.H.). This work was also supported by the Western University of Health Sciences Summer Student’s Fellowship Program (C.L.).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Wang, W.; Hao, Y.; Liu, Y.; Li, R.; Huang, D.B.; Pan, Y.Y. Nanomedicine in lung cancer: Current states of overcoming drug resistance and improving cancer immunotherapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2021, 13, e1654. [Google Scholar] [CrossRef]
  3. Rawal, S.; Patel, M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. Nanomicro Lett. 2021, 13, 142. [Google Scholar] [CrossRef]
  4. Walter, J.E.; Heuvelmans, M.A.; de Bock, G.H.; Yousaf-Khan, U.; Groen, H.J.M.; van der Aalst, C.M.; Nackaerts, K.; van Ooijen, P.M.A.; de Koning, H.J.; Vliegenthart, R.; et al. Relationship between the number of new nodules and lung cancer probability in incidence screening rounds of CT lung cancer screening: The NELSON study. Lung Cancer 2018, 125, 103–108. [Google Scholar] [CrossRef]
  5. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef]
  6. Lin, C.C.; Suen, K.M.; Lidster, J.; Ladbury, J.E. The emerging role of receptor tyrosine kinase phase separation in cancer. Trends. Cell Biol. 2023. [Google Scholar] [CrossRef]
  7. Girnita, L.; Worrall, C.; Takahashi, S.; Seregard, S.; Girnita, A. Something old, something new and something borrowed: Emerging paradigm of insulin-like growth factor type 1 receptor (IGF-1R) signaling regulation. Cell Mol. Life Sci. 2014, 71, 2403–2427. [Google Scholar] [CrossRef] [PubMed]
  8. Larsson, O.; Girnita, A.; Girnita, L. Role of Insulin-like Growth Factor 1 Receptor signalling in cancer. Br. J. Cancer 2005, 92, 2097–2101. [Google Scholar] [CrossRef] [PubMed]
  9. Yuan, J.; Yin, Z.; Tao, K.; Wang, G.; Gao, J. Function of Insulin-like Growth Factor 1 Receptor in cancer resistance to chemotherapy. Oncol. Lett. 2018, 15, 41–47. [Google Scholar] [CrossRef] [PubMed]
  10. Moschos, S.J.; Mantzoros, C.S. The role of the IGF system in cancer: From basic to clinical studies and clinical applications. Oncology 2002, 63, 317–332. [Google Scholar] [CrossRef] [PubMed]
  11. Arcaro, A. Targeting the insulin-like growth factor-1 receptor in human cancer. Front. Pharmacol. 2013, 4, 30. [Google Scholar] [CrossRef]
  12. Li, R.; Pourpak, A.; Morris, S.W. Inhibition of the insulin-like growth factor-1 receptor (IGF1R) tyrosine kinase as a novel cancer therapy approach. J. Med. Chem. 2009, 52, 4981–5004. [Google Scholar] [CrossRef] [PubMed]
  13. Ekyalongo, R.C.; Yee, D. Revisiting the IGF-1R as a breast cancer target. NPJ Precis Oncol. 2017, 1, 14. [Google Scholar] [CrossRef] [PubMed]
  14. Janssen, J.A.; Varewijck, A.J. IGF-IR Targeted Therapy: Past, Present and Future. Front. Endocrinol. 2014, 5, 224. [Google Scholar] [CrossRef]
  15. Warren, G.W.; Alberg, A.J.; Kraft, A.S.; Cummings, K.M. The 2014 Surgeon General’s report: "The health consequences of smoking--50 years of progress": A paradigm shift in cancer care. Cancer 2014, 120, 1914–1916. [Google Scholar] [CrossRef] [PubMed]
  16. Hecht, S.S. Progress and challenges in selected areas of tobacco carcinogenesis. Chem. Res. Toxicol. 2008, 21, 160–171. [Google Scholar] [CrossRef] [PubMed]
  17. Reducing Tobacco-Related Cancer Incidence and Mortality: Workshop Summary; National Academies Press: Washington, DC, USA, 2013.
  18. Available online: https://www.cancer.org/cancer/risk-prevention/tobacco/carcinogens-found-in-tobacco-products.html (accessed on 28 October 2020).
  19. Hecht, S.S. Tobacco smoke carcinogens and lung cancer. J. Natl. Cancer Inst. 1999, 91, 1194–1210. [Google Scholar] [CrossRef]
  20. Li, Y.; Hecht, S.S. Carcinogenic components of tobacco and tobacco smoke: A 2022 update. Food Chem. Toxicol. 2022, 165, 113179. [Google Scholar] [CrossRef] [PubMed]
  21. Witsch, E.; Sela, M.; Yarden, Y. Roles for growth factors in cancer progression. Physiology 2010, 25, 85–101. [Google Scholar] [CrossRef]
  22. Pillai, R.N.; Ramalingam, S.S. Inhibition of insulin-like growth factor receptor: End of a targeted therapy? Transl. Lung Cancer Res. 2013, 2, 14–22. [Google Scholar] [CrossRef]
  23. Velcheti, V.; Govindan, R. Insulin-like growth factor and lung cancer. J. Thorac. Oncol. 2006, 1, 607–610. [Google Scholar]
  24. Le Roith, D. Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors. N. Engl. J. Med. 1997, 336, 633–640. [Google Scholar] [CrossRef] [PubMed]
  25. Daughaday, W.H.; Kapadia, M.; Yanow, C.E.; Fabrick, K.; Mariz, I.K. Insulin-like growth factors I and II of nonmammalian sera. Gen. Comp. Endocrinol. 1985, 59, 316–325. [Google Scholar] [CrossRef] [PubMed]
  26. Rotwein, P. Insulinlike Growth Factor 1 Gene Variation in Vertebrates. Endocrinology 2018, 159, 2288–2305. [Google Scholar] [CrossRef] [PubMed]
  27. Sjogren, K.; Wallenius, K.; Liu, J.L.; Bohlooly, Y.M.; Pacini, G.; Svensson, L.; Tornell, J.; Isaksson, O.G.; Ahren, B.; Jansson, J.O.; et al. Liver-derived IGF-I is of importance for normal carbohydrate and lipid metabolism. Diabetes 2001, 50, 1539–1545. [Google Scholar] [CrossRef] [PubMed]
  28. Talia, C.; Connolly, L.; Fowler, P.A. The insulin-like growth factor system: A target for endocrine disruptors? Environ. Int. 2021, 147, 106311. [Google Scholar] [CrossRef]
  29. Itoh, K.; Balch, C.M.; Platsoucas, C.D. Monocyte-independent interleukin-2 production and proliferation of human T cells in response to murine hybridomas expressing the OKT3 monoclonal antibody: Interleukin-1 is not required for T-cell proliferation. Cell Immunol. 1988, 115, 36–56. [Google Scholar] [CrossRef]
  30. Wood, A.W.; Duan, C.; Bern, H.A. Insulin-like growth factor signaling in fish. Int. Rev. Cytol. 2005, 243, 215–285. [Google Scholar] [CrossRef]
  31. Rotwein, P. The insulin-like growth factor 2 gene and locus in nonmammalian vertebrates: Organizational simplicity with duplication but limited divergence in fish. J. Biol. Chem. 2018, 293, 15912–15932. [Google Scholar] [CrossRef]
  32. Randhawa, R.; Cohen, P. The role of the insulin-like growth factor system in prenatal growth. Mol. Genet. Metab. 2005, 86, 84–90. [Google Scholar] [CrossRef]
  33. Adams, T.E.; Epa, V.C.; Garrett, T.P.; Ward, C.W. Structure and function of the type 1 insulin-like growth factor receptor. Cell Mol. Life Sci. 2000, 57, 1050–1093. [Google Scholar] [CrossRef]
  34. Cao, J.; Yee, D. Disrupting Insulin and IGF Receptor Function in Cancer. Int. J. Mol. Sci. 2021, 22, 555. [Google Scholar] [CrossRef]
  35. Al-Samerria, S.; Radovick, S. The Role of Insulin-like Growth Factor-1 (IGF-1) in the Control of Neuroendocrine Regulation of Growth. Cells 2021, 10, 2664. [Google Scholar] [CrossRef]
  36. Lou, M.; Garrett, T.P.; McKern, N.M.; Hoyne, P.A.; Epa, V.C.; Bentley, J.D.; Lovrecz, G.O.; Cosgrove, L.J.; Frenkel, M.J.; Ward, C.W. The first three domains of the insulin receptor differ structurally from the Insulin-like Growth Factor 1 Receptor in the regions governing ligand specificity. Proc. Natl. Acad. Sci. USA 2006, 103, 12429–12434. [Google Scholar] [CrossRef]
  37. Hall, C.; Yu, H.; Choi, E. Insulin receptor endocytosis in the pathophysiology of insulin resistance. Exp. Mol. Med. 2020, 52, 911–920. [Google Scholar] [CrossRef] [PubMed]
  38. Boucher, J.; Kleinridders, A.; Kahn, C.R. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb. Perspect. Biol. 2014, 6. [Google Scholar] [CrossRef]
  39. Choi, E.; Bai, X.C. The Activation Mechanism of the Insulin Receptor: A Structural Perspective. Annu. Rev. Biochem. 2023, 92, 247–272. [Google Scholar] [CrossRef] [PubMed]
  40. Turvey, S.J.; McPhillie, M.J.; Kearney, M.T.; Muench, S.P.; Simmons, K.J.; Fishwick, C.W.G. Recent developments in the structural characterisation of the IR and IGF1R: Implications for the design of IR-IGF1R hybrid receptor modulators. RSC Med. Chem. 2022, 13, 360–374. [Google Scholar] [CrossRef]
  41. Iams, W.T.; Lovly, C.M. Molecular Pathways: Clinical Applications and Future Direction of Insulin-like Growth Factor-1 Receptor Pathway Blockade. Clin. Cancer Res. 2015, 21, 4270–4277. [Google Scholar] [CrossRef] [PubMed]
  42. Leffler, M.; Puusepp, S.; Zilina, O.; Zhu, Y.; Kuuse, K.; Bain, N.; Burgess, T.; Ounap, K.; Field, M. Two familial microduplications of 15q26.3 causing overgrowth and variable intellectual disability with normal copy number of IGF1R. Eur. J. Med. Genet. 2016, 59, 257–262. [Google Scholar] [CrossRef]
  43. Hebert, E. Mannose-6-phosphate/insulin-like growth factor II receptor expression and tumor development. Biosci. Rep. 2006, 26, 7–17. [Google Scholar] [CrossRef]
  44. Torrente, Y.; Bella, P.; Tripodi, L.; Villa, C.; Farini, A. Role of Insulin-Like Growth Factor Receptor 2 across Muscle Homeostasis: Implications for Treating Muscular Dystrophy. Cells 2020, 9, 441. [Google Scholar] [CrossRef]
  45. Dziadziuszko, R.; Camidge, D.R.; Hirsch, F.R. The insulin-like growth factor pathway in lung cancer. J. Thorac. Oncol. 2008, 3, 815–818. [Google Scholar] [CrossRef]
  46. Haywood, N.J.; Slater, T.A.; Matthews, C.J.; Wheatcroft, S.B. The insulin like growth factor and binding protein family: Novel therapeutic targets in obesity & diabetes. Mol. Metab. 2019, 19, 86–96. [Google Scholar] [CrossRef]
  47. Clemmons, D.R. Role of insulin-like growth factor binding proteins in controlling IGF actions. Mol. Cell Endocrinol. 1998, 140, 19–24. [Google Scholar] [CrossRef] [PubMed]
  48. Livingstone, C.; Borai, A. Insulin-like growth factor-II: Its role in metabolic and endocrine disease. Clin. Endocrinol. 2014, 80, 773–781. [Google Scholar] [CrossRef] [PubMed]
  49. Hankinson, S.E.; Willett, W.C.; Colditz, G.A.; Hunter, D.J.; Michaud, D.S.; Deroo, B.; Rosner, B.; Speizer, F.E.; Pollak, M. Circulating concentrations of insulin-like growth factor-I and risk of breast cancer. Lancet 1998, 351, 1393–1396. [Google Scholar] [CrossRef] [PubMed]
  50. Forbes, B.E.; Blyth, A.J.; Wit, J.M. Disorders of IGFs and IGF-1R signaling pathways. Mol. Cell Endocrinol. 2020, 518, 111035. [Google Scholar] [CrossRef] [PubMed]
  51. Baserga, R.; Resnicoff, M.; Dews, M. The IGF-I receptor and cancer. Endocrine 1997, 7, 99–102. [Google Scholar] [CrossRef] [PubMed]
  52. Baserga, R.; Peruzzi, F.; Reiss, K. The IGF-1 receptor in cancer biology. Int. J. Cancer 2003, 107, 873–877. [Google Scholar] [CrossRef] [PubMed]
  53. Zhao, S.; Qiu, Z.; He, J.; Li, L.; Li, W. Insulin-like growth factor receptor 1 (IGF1R) expression and survival in non-small cell lung cancer patients: A meta-analysis. Int. J. Clin. Exp. Pathol. 2014, 7, 6694–6704. [Google Scholar] [PubMed]
  54. Garcia de la Serrana, D.; Macqueen, D.J. Insulin-Like Growth Factor-Binding Proteins of Teleost Fishes. Front. Endocrinol. 2018, 9, 80. [Google Scholar] [CrossRef] [PubMed]
  55. Duan, C.; Xu, Q. Roles of insulin-like growth factor (IGF) binding proteins in regulating IGF actions. Gen. Comp. Endocrinol. 2005, 142, 44–52. [Google Scholar] [CrossRef] [PubMed]
  56. Cai, Q.; Dozmorov, M.; Oh, Y. IGFBP-3/IGFBP-3 Receptor System as an Anti-Tumor and Anti-Metastatic Signaling in Cancer. Cells 2020, 9, 1261. [Google Scholar] [CrossRef]
  57. Yu, H.; Spitz, M.R.; Mistry, J.; Gu, J.; Hong, W.K.; Wu, X. Plasma levels of insulin-like growth factor-I and lung cancer risk: A case-control analysis. J. Natl. Cancer Inst. 1999, 91, 151–156. [Google Scholar] [CrossRef]
  58. London, S.J.; Yuan, J.M.; Travlos, G.S.; Gao, Y.T.; Wilson, R.E.; Ross, R.K.; Yu, M.C. Insulin-like growth factor I, IGF-binding protein 3, and lung cancer risk in a prospective study of men in China. J. Natl. Cancer Inst. 2002, 94, 749–754. [Google Scholar] [CrossRef]
  59. Buckbinder, L.; Talbott, R.; Velasco-Miguel, S.; Takenaka, I.; Faha, B.; Seizinger, B.R.; Kley, N. Induction of the growth inhibitor IGF-binding protein 3 by p53. Nature 1995, 377, 646–649. [Google Scholar] [CrossRef]
  60. Hanafusa, T.; Shinji, T.; Shiraha, H.; Nouso, K.; Iwasaki, Y.; Yumoto, E.; Ono, T.; Koide, N. Functional promoter upstream p53 regulatory sequence of IGFBP3 that is silenced by tumor specific methylation. BMC Cancer 2005, 5, 9. [Google Scholar] [CrossRef] [PubMed]
  61. Grimberg, A.; Liu, B.; Bannerman, P.; El-Deiry, W.S.; Cohen, P. IGFBP-3 mediates p53-induced apoptosis during serum starvation. Int. J. Oncol. 2002, 21, 327–335. [Google Scholar] [CrossRef]
  62. Ludwig, R.L.; Bates, S.; Vousden, K.H. Differential activation of target cellular promoters by p53 mutants with impaired apoptotic function. Mol. Cell. Biol. 1996, 16, 4952–4960. [Google Scholar] [CrossRef] [PubMed]
  63. Grimberg, A. P53 and IGFBP-3: Apoptosis and cancer protection. Mol. Genet Metab. 2000, 70, 85–98. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, W.Y.; Kim, M.J.; Moon, H.; Yuan, P.; Kim, J.S.; Woo, J.K.; Zhang, G.; Suh, Y.A.; Feng, L.; Behrens, C.; et al. Differential impacts of insulin-like growth factor-binding protein-3 (IGFBP-3) in epithelial IGF-induced lung cancer development. Endocrinology 2011, 152, 2164–2173. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, C.; Cui, T.; Cai, R.; Wangpaichitr, M.; Mirsaeidi, M.; Schally, A.V.; Jackson, R.M. Growth Hormone-Releasing Hormone in Lung Physiology and Pulmonary Disease. Cells 2020, 9, 2331. [Google Scholar] [CrossRef] [PubMed]
  66. Gao, L.; Blair, L.A.; Salinas, G.D.; Needleman, L.A.; Marshall, J. Insulin-like growth factor-1 modulation of CaV1.3 calcium channels depends on Ca2+ release from IP3-sensitive stores and calcium/calmodulin kinase II phosphorylation of the alpha1 subunit EF hand. J. Neurosci. 2006, 26, 6259–6268. [Google Scholar] [CrossRef]
  67. Linnerth, N.M.; Baldwin, M.; Campbell, C.; Brown, M.; McGowan, H.; Moorehead, R.A. IGF-II induces CREB phosphorylation and cell survival in human lung cancer cells. Oncogene 2005, 24, 7310–7319. [Google Scholar] [CrossRef]
  68. Zha, J.; Lackner, M.R. Targeting the insulin-like growth factor receptor-1R pathway for cancer therapy. Clin. Cancer Res. 2010, 16, 2512–2517. [Google Scholar] [CrossRef]
  69. Bohula, E.A.; Playford, M.P.; Macaulay, V.M. Targeting the type 1 insulin-like growth factor receptor as anti-cancer treatment. Anticancer Drugs 2003, 14, 669–682. [Google Scholar] [CrossRef]
  70. Kim, W.Y.; Jin, Q.; Oh, S.H.; Kim, E.S.; Yang, Y.J.; Lee, D.H.; Feng, L.; Behrens, C.; Prudkin, L.; Miller, Y.E.; et al. Elevated epithelial insulin-like growth factor expression is a risk factor for lung cancer development. Cancer Res. 2009, 69, 7439–7448. [Google Scholar] [CrossRef]
  71. Moorehead, R.A.; Sanchez, O.H.; Baldwin, R.M.; Khokha, R. Transgenic overexpression of IGF-II induces spontaneous lung tumors: A model for human lung adenocarcinoma. Oncogene 2003, 22, 853–857. [Google Scholar] [CrossRef]
  72. Rajski, M.; Zanetti-Dallenbach, R.; Vogel, B.; Herrmann, R.; Rochlitz, C.; Buess, M. IGF-I induced genes in stromal fibroblasts predict the clinical outcome of breast and lung cancer patients. BMC Med. 2010, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, B.; Xiao, F.; Li, B.; Xie, B.; Zhou, J.; Zheng, J.; Zhang, W. The role of epithelial-mesenchymal transition and IGF-1R expression in prediction of gefitinib activity as the second-line treatment for advanced nonsmall-cell lung cancer. Cancer Investig. 2013, 31, 454–460. [Google Scholar] [CrossRef] [PubMed]
  74. Bielen, A.; Perryman, L.; Box, G.M.; Valenti, M.; de Haven Brandon, A.; Martins, V.; Jury, A.; Popov, S.; Gowan, S.; Jeay, S.; et al. Enhanced efficacy of IGF1R inhibition in pediatric glioblastoma by combinatorial targeting of PDGFRalpha/beta. Mol. Cancer Ther. 2011, 10, 1407–1418. [Google Scholar] [CrossRef] [PubMed]
  75. Bhatavdekar, J.M.; Patel, D.D.; Shah, N.G.; Karelia, N.H.; Vora, H.H.; Ghosh, N.; Suthar, T.P.; Balar, D.B. Prognostic value of insulin-like growth factor-1 receptors in patients with colon/rectal cancer: Correlation with plasma prolactin. Eur. J. Surg. Oncol. 1995, 21, 23–26. [Google Scholar] [CrossRef] [PubMed]
  76. Mountzios, G.; Kostopoulos, I.; Kotoula, V.; Sfakianaki, I.; Fountzilas, E.; Markou, K.; Karasmanis, I.; Leva, S.; Angouridakis, N.; Vlachtsis, K.; et al. Insulin-like Growth Factor 1 Receptor (IGF1R) expression and survival in operable squamous-cell laryngeal cancer. PLoS ONE 2013, 8, e54048. [Google Scholar] [CrossRef]
  77. Ozkan, E.E. Plasma and tissue insulin-like growth factor-I receptor (IGF-IR) as a prognostic marker for prostate cancer and anti-IGF-IR agents as novel therapeutic strategy for refractory cases: A review. Mol. Cell Endocrinol. 2011, 344, 1–24. [Google Scholar] [CrossRef] [PubMed]
  78. Obr, A.E.; Kumar, S.; Chang, Y.J.; Bulatowicz, J.J.; Barnes, B.J.; Birge, R.B.; Lazzarino, D.A.; Gallagher, E.; LeRoith, D.; Wood, T.L. Insulin-like growth factor receptor signaling in breast tumor epithelium protects cells from endoplasmic reticulum stress and regulates the tumor microenvironment. Breast. Cancer Res. 2018, 20, 138. [Google Scholar] [CrossRef]
  79. Yan, S.; Jiao, X.; Li, K.; Li, W.; Zou, H. The impact of IGF-1R expression on the outcomes of patients with breast cancer: A meta-analysis. OncoTargets Ther. 2015, 8, 279–287. [Google Scholar] [CrossRef]
  80. Xu, J.; Bie, F.; Wang, Y.; Chen, X.; Yan, T.; Du, J. Prognostic value of IGF-1R in lung cancer: A PRISMA-compliant meta-analysis. Medicine 2019, 98, e15467. [Google Scholar] [CrossRef]
  81. Badzio, A.; Wynes, M.W.; Dziadziuszko, R.; Merrick, D.T.; Pardo, M.; Rzyman, W.; Kowalczyk, A.; Singh, S.; Ranger-Moore, J.; Manriquez, G.; et al. Increased Insulin-like Growth Factor 1 Receptor protein expression and gene copy number in small cell lung cancer. J. Thorac. Oncol. 2010, 5, 1905–1911. [Google Scholar] [CrossRef]
  82. Boo, H.J.; Min, H.Y.; Hwang, S.J.; Lee, H.J.; Lee, J.W.; Oh, S.R.; Park, C.S.; Park, J.S.; Lee, Y.M.; Lee, H.Y. The tobacco-specific carcinogen NNK induces pulmonary tumorigenesis via nAChR/Src/STAT3-mediated activation of the renin-angiotensin system and IGF-1R signaling. Exp. Mol. Med. 2023, 55, 1131–1144. [Google Scholar] [CrossRef]
  83. Min, H.Y.; Boo, H.J.; Lee, H.J.; Jang, H.J.; Yun, H.J.; Hwang, S.J.; Smith, J.K.; Lee, H.J.; Lee, H.Y. Smoking-associated lung cancer prevention by blockade of the beta-adrenergic receptor-mediated insulin-like growth factor receptor activation. Oncotarget 2016, 7, 70936–70947. [Google Scholar] [CrossRef] [PubMed]
  84. Boo, H.J.; Min, H.Y.; Jang, H.J.; Yun, H.J.; Smith, J.K.; Jin, Q.; Lee, H.J.; Liu, D.; Kweon, H.S.; Behrens, C.; et al. The tobacco-specific carcinogen-operated calcium channel promotes lung tumorigenesis via IGF2 exocytosis in lung epithelial cells. Nat. Commun. 2016, 7, 12961. [Google Scholar] [CrossRef] [PubMed]
  85. Kooijman, R. Regulation of apoptosis by insulin-like growth factor (IGF)-I. Cytokine Growth Factor Rev. 2006, 17, 305–323. [Google Scholar] [CrossRef] [PubMed]
  86. Peruzzi, F.; Prisco, M.; Dews, M.; Salomoni, P.; Grassilli, E.; Romano, G.; Calabretta, B.; Baserga, R. Multiple signaling pathways of the Insulin-like Growth Factor 1 Receptor in protection from apoptosis. Mol. Cell Biol. 1999, 19, 7203–7215. [Google Scholar] [CrossRef] [PubMed]
  87. Ravid, D.; Maor, S.; Werner, H.; Liscovitch, M. Caveolin-1 inhibits cell detachment-induced p53 activation and anoikis by upregulation of insulin-like growth factor-I receptors and signaling. Oncogene 2005, 24, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
  88. Uttamsingh, S.; Zong, C.S.; Wang, L.H. Matrix-independent activation of phosphatidylinositol 3-kinase, Stat3, and cyclin A-associated Cdk2 Is essential for anchorage-independent growth of v-Ros-transformed chicken embryo fibroblasts. J. Biol. Chem. 2003, 278, 18798–18810. [Google Scholar] [CrossRef]
  89. Zhang, W.; Zong, C.S.; Hermanto, U.; Lopez-Bergami, P.; Ronai, Z.; Wang, L.H. RACK1 recruits STAT3 specifically to insulin and Insulin-like Growth Factor 1 Receptor for activation, which is important for regulating anchorage-independent growth. Mol. Cell Biol. 2006, 26, 413–424. [Google Scholar] [CrossRef]
  90. Alfaro-Arnedo, E.; Lopez, I.P.; Pineiro-Hermida, S.; Canalejo, M.; Gotera, C.; Sola, J.J.; Roncero, A.; Peces-Barba, G.; Ruiz-Martinez, C.; Pichel, J.G. IGF1R acts as a cancer-promoting factor in the tumor microenvironment facilitating lung metastasis implantation and progression. Oncogene 2022, 41, 3625–3639. [Google Scholar] [CrossRef]
  91. Hu, Y.Z.; Hu, Z.L.; Liao, T.Y.; Li, Y.; Pan, Y.L. LncRNA SND1-IT1 facilitates TGF-beta1-induced epithelial-to-mesenchymal transition via miR-124/COL4A1 axis in gastric cancer. Cell. Death Discov. 2022, 8, 73. [Google Scholar] [CrossRef]
  92. Cooper, J.; Giancotti, F.G. Integrin Signaling in Cancer: Mechanotransduction, Stemness, Epithelial Plasticity, and Therapeutic Resistance. Cancer Cell 2019, 35, 347–367. [Google Scholar] [CrossRef]
  93. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [PubMed]
  94. Roche, J. The Epithelial-to-Mesenchymal Transition in Cancer. Cancers 2018, 10, 52. [Google Scholar] [CrossRef]
  95. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef]
  96. Zhou, J.; Wang, J.; Zeng, Y.; Zhang, X.; Hu, Q.; Zheng, J.; Chen, B.; Xie, B.; Zhang, W.M. Implication of epithelial-mesenchymal transition in IGF1R-induced resistance to EGFR-TKIs in advanced non-small cell lung cancer. Oncotarget 2015, 6, 44332–44345. [Google Scholar] [CrossRef]
  97. Li, H.; Xu, L.; Zhao, L.; Ma, Y.; Zhu, Z.; Liu, Y.; Qu, X. Insulin-like growth factor-I induces epithelial to mesenchymal transition via GSK-3beta and ZEB2 in the BGC-823 gastric cancer cell line. Oncol. Lett. 2015, 9, 143–148. [Google Scholar] [CrossRef] [PubMed]
  98. Graham, T.R.; Zhau, H.E.; Odero-Marah, V.A.; Osunkoya, A.O.; Kimbro, K.S.; Tighiouart, M.; Liu, T.; Simons, J.W.; O’Regan, R.M. Insulin-like growth factor-I-dependent up-regulation of ZEB1 drives epithelial-to-mesenchymal transition in human prostate cancer cells. Cancer Res. 2008, 68, 2479–2488. [Google Scholar] [CrossRef]
  99. Li, H.; Batth, I.S.; Qu, X.; Xu, L.; Song, N.; Wang, R.; Liu, Y. IGF-IR signaling in epithelial to mesenchymal transition and targeting IGF-IR therapy: Overview and new insights. Mol. Cancer 2017, 16, 6. [Google Scholar] [CrossRef] [PubMed]
  100. Cevenini, A.; Orru, S.; Mancini, A.; Alfieri, A.; Buono, P.; Imperlini, E. Molecular Signatures of the Insulin-like Growth Factor 1-mediated Epithelial-Mesenchymal Transition in Breast, Lung and Gastric Cancers. Int. J. Mol. Sci. 2018, 19, 2411. [Google Scholar] [CrossRef]
  101. Hua, H.; Kong, Q.; Yin, J.; Zhang, J.; Jiang, Y. Insulin-like growth factor receptor signaling in tumorigenesis and drug resistance: A challenge for cancer therapy. J. Hematol. Oncol. 2020, 13, 64. [Google Scholar] [CrossRef]
  102. Sivakumar, R.; Koga, H.; Selvendiran, K.; Maeyama, M.; Ueno, T.; Sata, M. Autocrine loop for IGF-I receptor signaling in SLUG-mediated epithelial-mesenchymal transition. Int. J. Oncol. 2009, 34, 329–338. [Google Scholar]
  103. Kim, H.J.; Litzenburger, B.C.; Cui, X.; Delgado, D.A.; Grabiner, B.C.; Lin, X.; Lewis, M.T.; Gottardis, M.M.; Wong, T.W.; Attar, R.M.; et al. Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-kappaB and snail. Mol. Cell Biol. 2007, 27, 3165–3175. [Google Scholar] [CrossRef] [PubMed]
  104. Taliaferro-Smith, L.; Oberlick, E.; Liu, T.; McGlothen, T.; Alcaide, T.; Tobin, R.; Donnelly, S.; Commander, R.; Kline, E.; Nagaraju, G.P.; et al. FAK activation is required for IGF1R-mediated regulation of EMT, migration, and invasion in mesenchymal triple negative breast cancer cells. Oncotarget 2015, 6, 4757–4772. [Google Scholar] [CrossRef] [PubMed]
  105. Mansoori, B.; Mohammadi, A.; Davudian, S.; Shirjang, S.; Baradaran, B. The Different Mechanisms of Cancer Drug Resistance: A Brief Review. Adv. Pharm. Bull. 2017, 7, 339–348. [Google Scholar] [CrossRef] [PubMed]
  106. Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug Resistance in Cancer Chemotherapy. Int. J. Mol. Sci. 2020, 21, 3233. [Google Scholar] [CrossRef]
  107. Shi, W.J.; Gao, J.B. Molecular mechanisms of chemoresistance in gastric cancer. World J. Gastrointest Oncol. 2016, 8, 673–681. [Google Scholar] [CrossRef]
  108. Nwabo Kamdje, A.H.; Seke Etet, P.F.; Kipanyula, M.J.; Vecchio, L.; Tagne Simo, R.; Njamnshi, A.K.; Lukong, K.E.; Mimche, P.N. Insulin-like growth factor-1 signaling in the tumor microenvironment: Carcinogenesis, cancer drug resistance, and therapeutic potential. Front. Endocrinol. 2022, 13, 927390. [Google Scholar] [CrossRef]
  109. Sun, Y.; Zheng, S.; Torossian, A.; Speirs, C.K.; Schleicher, S.; Giacalone, N.J.; Carbone, D.P.; Zhao, Z.; Lu, B. Role of insulin-like growth factor-1 signaling pathway in cisplatin-resistant lung cancer cells. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, e563–e572. [Google Scholar] [CrossRef]
  110. Ibanez de Caceres, I.; Cortes-Sempere, M.; Moratilla, C.; Machado-Pinilla, R.; Rodriguez-Fanjul, V.; Manguan-Garcia, C.; Cejas, P.; Lopez-Rios, F.; Paz-Ares, L.; de CastroCarpeno, J.; et al. IGFBP-3 hypermethylation-derived deficiency mediates cisplatin resistance in non-small-cell lung cancer. Oncogene 2010, 29, 1681–1690. [Google Scholar] [CrossRef]
  111. Pao, W.; Miller, V.; Zakowski, M.; Doherty, J.; Politi, K.; Sarkaria, I.; Singh, B.; Heelan, R.; Rusch, V.; Fulton, L.; et al. EGF receptor gene mutations are common in lung cancers from "never smokers" and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc. Natl. Acad. Sci. USA 2004, 101, 13306–13311. [Google Scholar] [CrossRef]
  112. Murakami, A.; Takahashi, F.; Nurwidya, F.; Kobayashi, I.; Minakata, K.; Hashimoto, M.; Nara, T.; Kato, M.; Tajima, K.; Shimada, N.; et al. Hypoxia increases gefitinib-resistant lung cancer stem cells through the activation of Insulin-like Growth Factor 1 Receptor. PLoS ONE 2014, 9, e86459. [Google Scholar] [CrossRef]
  113. Hussmann, D.; Madsen, A.T.; Jakobsen, K.R.; Luo, Y.; Sorensen, B.S.; Nielsen, A.L. IGF1R depletion facilitates MET-amplification as mechanism of acquired resistance to erlotinib in HCC827 NSCLC cells. Oncotarget 2017, 8, 33300–33315. [Google Scholar] [CrossRef] [PubMed]
  114. Hurbin, A.; Wislez, M.; Busser, B.; Antoine, M.; Tenaud, C.; Rabbe, N.; Dufort, S.; de Fraipont, F.; Moro-Sibilot, D.; Cadranel, J.; et al. Insulin-like growth factor-1 receptor inhibition overcomes gefitinib resistance in mucinous lung adenocarcinoma. J. Pathol. 2011, 225, 83–95. [Google Scholar] [CrossRef] [PubMed]
  115. Lee, Y.; Wang, Y.; James, M.; Jeong, J.H.; You, M. Inhibition of IGF1R signaling abrogates resistance to afatinib (BIBW2992) in EGFR T790M mutant lung cancer cells. Mol. Carcinog. 2016, 55, 991–1001. [Google Scholar] [CrossRef] [PubMed]
  116. Manabe, T.; Yasuda, H.; Terai, H.; Kagiwada, H.; Hamamoto, J.; Ebisudani, T.; Kobayashi, K.; Masuzawa, K.; Ikemura, S.; Kawada, I.; et al. IGF2 Autocrine-Mediated IGF1R Activation Is a Clinically Relevant Mechanism of Osimertinib Resistance in Lung Cancer. Mol. Cancer Res. 2020, 18, 549–559. [Google Scholar] [CrossRef] [PubMed]
  117. Lovly, C.M.; McDonald, N.T.; Chen, H.; Ortiz-Cuaran, S.; Heukamp, L.C.; Yan, Y.; Florin, A.; Ozretic, L.; Lim, D.; Wang, L.; et al. Rationale for co-targeting IGF-1R and ALK in ALK fusion-positive lung cancer. Nat. Med. 2014, 20, 1027–1034. [Google Scholar] [CrossRef]
  118. Isoyama, S.; Dan, S.; Nishimura, Y.; Nakamura, N.; Kajiwara, G.; Seki, M.; Irimura, T.; Yamori, T. Establishment of phosphatidylinositol 3-kinase inhibitor-resistant cancer cell lines and therapeutic strategies for overcoming the resistance. Cancer Sci. 2012, 103, 1955–1960. [Google Scholar] [CrossRef]
  119. Langer, C.J.; Novello, S.; Park, K.; Krzakowski, M.; Karp, D.D.; Mok, T.; Benner, R.J.; Scranton, J.R.; Olszanski, A.J.; Jassem, J. Randomized, phase III trial of first-line figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin alone in patients with advanced non-small-cell lung cancer. J. Clin. Oncol. 2014, 32, 2059–2066. [Google Scholar] [CrossRef]
  120. Yeh, J.; Litz, J.; Hauck, P.; Ludwig, D.L.; Krystal, G.W. Selective inhibition of SCLC growth by the A12 anti-IGF-1R monoclonal antibody correlates with inhibition of Akt. Lung Cancer 2008, 60, 166–174. [Google Scholar] [CrossRef]
  121. Gong, Y.; Yao, E.; Shen, R.; Goel, A.; Arcila, M.; Teruya-Feldstein, J.; Zakowski, M.F.; Frankel, S.; Peifer, M.; Thomas, R.K.; et al. High expression levels of total IGF-1R and sensitivity of NSCLC cells in vitro to an anti-IGF-1R antibody (R1507). PLoS ONE 2009, 4, e7273. [Google Scholar] [CrossRef]
  122. Liang, S.; Galluzzo, P.; Sobol, A.; Skucha, S.; Rambo, B.; Bocchetta, M. Multimodality Approaches to Treat Hypoxic Non-Small Cell Lung Cancer (NSCLC) Microenvironment. Genes Cancer 2012, 3, 141–151. [Google Scholar] [CrossRef] [PubMed]
  123. Iwasa, T.; Okamoto, I.; Suzuki, M.; Hatashita, E.; Yamada, Y.; Fukuoka, M.; Ono, K.; Nakagawa, K. Inhibition of Insulin-like Growth Factor 1 Receptor by CP-751,871 radiosensitizes non-small cell lung cancer cells. Clin. Cancer Res. 2009, 15, 5117–5125. [Google Scholar] [CrossRef] [PubMed]
  124. Mulvihill, M.J.; Cooke, A.; Rosenfeld-Franklin, M.; Buck, E.; Foreman, K.; Landfair, D.; O’Connor, M.; Pirritt, C.; Sun, Y.; Yao, Y.; et al. Discovery of OSI-906: A selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Med. Chem. 2009, 1, 1153–1171. [Google Scholar] [CrossRef] [PubMed]
  125. McKinley, E.T.; Bugaj, J.E.; Zhao, P.; Guleryuz, S.; Mantis, C.; Gokhale, P.C.; Wild, R.; Manning, H.C. 18FDG-PET predicts pharmacodynamic response to OSI-906, a dual IGF-1R/IR inhibitor, in preclinical mouse models of lung cancer. Clin. Cancer Res. 2011, 17, 3332–3340. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, Q.; Pan, J.; Lubet, R.A.; Wang, Y.; You, M. Targeting the insulin-like growth factor-1 receptor by picropodophyllin for lung cancer chemoprevention. Mol. Carcinog. 2015, 54 (Suppl. 1), E129–E137. [Google Scholar] [CrossRef] [PubMed]
  127. de Miranda, L.B.L.; Lima, K.; Coelho-Silva, J.L.; Traina, F.; Kobayashi, S.S.; Machado-Neto, J.A. NT157 exerts antineoplastic activity by targeting JNK and AXL signaling in lung cancer cells. Sci. Rep. 2022, 12, 17092. [Google Scholar] [CrossRef] [PubMed]
  128. Franks, S.E.; Jones, R.A.; Briah, R.; Murray, P.; Moorehead, R.A. BMS-754807 is cytotoxic to non-small cell lung cancer cells and enhances the effects of platinum chemotherapeutics in the human lung cancer cell line A549. BMC Res. Notes 2016, 9, 134. [Google Scholar] [CrossRef]
  129. Zhang, C.; Zhao, X.; Wang, Z.; Gong, T.; Zhao, H.; Zhang, D.; Niu, Y.; Li, X.; Zhao, X.; Li, G.; et al. Dasatinib in combination with BMS-754807 induce synergistic cytotoxicity in lung cancer cells through inhibiting lung cancer cell growth, and inducing autophagy as well as cell cycle arrest at the G1 phase. Investig. New. Drugs 2023, 41, 438–452. [Google Scholar] [CrossRef]
  130. Friedbichler, K.; Hofmann, M.H.; Kroez, M.; Ostermann, E.; Lamche, H.R.; Koessl, C.; Borges, E.; Pollak, M.N.; Adolf, G.; Adam, P.J. Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycin. Mol. Cancer Ther. 2014, 13, 399–409. [Google Scholar] [CrossRef]
  131. Gao, J.; Chesebrough, J.W.; Cartlidge, S.A.; Ricketts, S.A.; Incognito, L.; Veldman-Jones, M.; Blakey, D.C.; Tabrizi, M.; Jallal, B.; Trail, P.A.; et al. Dual IGF-I/II-neutralizing antibody MEDI-573 potently inhibits IGF signaling and tumor growth. Cancer Res. 2011, 71, 1029–1040. [Google Scholar] [CrossRef]
  132. Lee, Y.J.; Imsumran, A.; Park, M.Y.; Kwon, S.Y.; Yoon, H.I.; Lee, J.H.; Yoo, C.G.; Kim, Y.W.; Han, S.K.; Shim, Y.S.; et al. Adenovirus expressing shRNA to IGF-1R enhances the chemosensitivity of lung cancer cell lines by blocking IGF-1 pathway. Lung Cancer 2007, 55, 279–286. [Google Scholar] [CrossRef]
  133. Akla, B.; Broussas, M.; Loukili, N.; Robert, A.; Beau-Larvor, C.; Malissard, M.; Boute, N.; Champion, T.; Haeuw, J.F.; Beck, A.; et al. Efficacy of the Antibody-Drug Conjugate W0101 in Preclinical Models of IGF-1 Receptor Overexpressing Solid Tumors. Mol. Cancer Ther. 2020, 19, 168–177. [Google Scholar] [CrossRef]
  134. Weickhardt, A.; Doebele, R.; Oton, A.; Lettieri, J.; Maxson, D.; Reynolds, M.; Brown, A.; Jackson, M.K.; Dy, G.; Adjei, A.; et al. A phase I/II study of erlotinib in combination with the anti-insulin-like growth factor-1 receptor monoclonal antibody IMC-A12 (cixutumumab) in patients with advanced non-small cell lung cancer. J. Thorac. Oncol. 2012, 7, 419–426. [Google Scholar] [CrossRef]
  135. Argiris, A.; Lee, J.W.; Stevenson, J.; Sulecki, M.G.; Hugec, V.; Choong, N.W.; Saltzman, J.N.; Song, W.; Hansen, R.M.; Evans, T.L.; et al. Phase II randomized trial of carboplatin, paclitaxel, bevacizumab with or without cixutumumab (IMC-A12) in patients with advanced non-squamous, non-small-cell lung cancer: A trial of the ECOG-ACRIN Cancer Research Group (E3508). Ann. Oncol. 2017, 28, 3037–3043. [Google Scholar] [CrossRef]
  136. Murakami, H.; Doi, T.; Yamamoto, N.; Watanabe, J.; Boku, N.; Fuse, N.; Yoshino, T.; Ohtsu, A.; Otani, S.; Shibayama, K.; et al. Phase 1 study of ganitumab (AMG 479), a fully human monoclonal antibody against the insulin-like growth factor receptor type I (IGF1R), in Japanese patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2012, 70, 407–414. [Google Scholar] [CrossRef] [PubMed]
  137. Soria, J.C.; Massard, C.; Lazar, V.; Ozoux, M.L.; Mery-Mignard, D.; Deslandes, A.; Tolcher, A.W. A dose finding, safety and pharmacokinetic study of AVE1642, an anti-insulin-like growth factor-1 receptor (IGF-1R/CD221) monoclonal antibody, administered as a single agent and in combination with docetaxel in patients with advanced solid tumours. Eur. J. Cancer 2013, 49, 1799–1807. [Google Scholar] [CrossRef] [PubMed]
  138. Ramalingam, S.S.; Spigel, D.R.; Chen, D.; Steins, M.B.; Engelman, J.A.; Schneider, C.P.; Novello, S.; Eberhardt, W.E.; Crino, L.; Habben, K.; et al. Randomized phase II study of erlotinib in combination with placebo or R1507, a monoclonal antibody to insulin-like growth factor-1 receptor, for advanced-stage non-small-cell lung cancer. J. Clin. Oncol. 2011, 29, 4574–4580. [Google Scholar] [CrossRef] [PubMed]
  139. Ellis, P.M.; Shepherd, F.A.; Laurie, S.A.; Goss, G.D.; Olivo, M.; Powers, J.; Seymour, L.; Bradbury, P.A. NCIC CTG IND.190 phase I trial of dalotuzumab (MK-0646) in combination with cisplatin and etoposide in extensive-stage small-cell lung cancer. J. Thorac. Oncol. 2014, 9, 410–413. [Google Scholar] [CrossRef] [PubMed]
  140. Moran, T.; Felip, E.; Keedy, V.; Borghaei, H.; Shepherd, F.A.; Insa, A.; Brown, H.; Fitzgerald, T.; Sathyanarayanan, S.; Reilly, J.F.; et al. Activity of dalotuzumab, a selective anti-IGF1R antibody, in combination with erlotinib in unselected patients with Non-small-cell lung cancer: A phase I/II randomized trial. Exp. Hematol. Oncol. 2014, 3, 26. [Google Scholar] [CrossRef] [PubMed]
  141. Huang, C.H.; Williamson, S.K.; Neupane, P.; Taylor, S.A.; Allen, A.; Smart, N.J.; Uypeckcuat, A.M.; Spencer, S.; Wick, J.; Smith, H.; et al. Impact Study: MK-0646 (Dalotuzumab), Insulin Growth Factor 1 Receptor Antibody Combined with Pemetrexed and Cisplatin in Stage IV Metastatic Non-squamous Lung Cancer. Front. Oncol. 2015, 5, 301. [Google Scholar] [CrossRef] [PubMed]
  142. Scagliotti, G.V.; Bondarenko, I.; Blackhall, F.; Barlesi, F.; Hsia, T.C.; Jassem, J.; Milanowski, J.; Popat, S.; Sanchez-Torres, J.M.; Novello, S.; et al. Randomized, phase III trial of figitumumab in combination with erlotinib versus erlotinib alone in patients with nonadenocarcinoma nonsmall-cell lung cancer. Ann. Oncol. 2015, 26, 497–504. [Google Scholar] [CrossRef] [PubMed]
  143. Di Maio, M.; Scagliotti, G.V. The lesson learned from figitumumab clinical program and the hope for better results in squamous lung cancer. Transl. Lung Cancer Res. 2015, 4, 15–17. [Google Scholar] [CrossRef] [PubMed]
  144. von Mehren, M.; Britten, C.D.; Pieslor, P.; Saville, W.; Vassos, A.; Harris, S.; Galluppi, G.R.; Darif, M.; Wainberg, Z.A.; Cohen, R.B.; et al. A phase 1, open-label, dose-escalation study of BIIB022 (anti-IGF-1R monoclonal antibody) in subjects with relapsed or refractory solid tumors. Investig. New Drugs 2014, 32, 518–525. [Google Scholar] [CrossRef] [PubMed]
  145. Chiappori, A.A.; Otterson, G.A.; Dowlati, A.; Traynor, A.M.; Horn, L.; Owonikoko, T.K.; Ross, H.J.; Hann, C.L.; Abu Hejleh, T.; Nieva, J.; et al. A Randomized Phase II Study of Linsitinib (OSI-906) Versus Topotecan in Patients With Relapsed Small-Cell Lung Cancer. Oncologist 2016, 21, 1163–1164. [Google Scholar] [CrossRef]
  146. Ciuleanu, T.E.; Ahmed, S.; Kim, J.H.; Mezger, J.; Park, K.; Thomas, M.; Chen, J.; Poondru, S.; VanTornout, J.M.; Whitcomb, D.; et al. Randomised Phase 2 study of maintenance linsitinib (OSI-906) in combination with erlotinib compared with placebo plus erlotinib after platinum-based chemotherapy in patients with advanced non-small cell lung cancer. Br. J. Cancer 2017, 117, 757–766. [Google Scholar] [CrossRef]
  147. Leighl, N.B.; Rizvi, N.A.; de Lima, L.G., Jr.; Arpornwirat, W.; Rudin, C.M.; Chiappori, A.A.; Ahn, M.J.; Chow, L.Q.; Bazhenova, L.; Dechaphunkul, A.; et al. Phase 2 Study of Erlotinib in Combination With Linsitinib (OSI-906) or Placebo in Chemotherapy-Naive Patients With Non-Small-Cell Lung Cancer and Activating Epidermal Growth Factor Receptor Mutations. Clin. Lung Cancer 2017, 18, 34–42.e32. [Google Scholar] [CrossRef]
  148. Ekman, S.; Harmenberg, J.; Frodin, J.E.; Bergstrom, S.; Wassberg, C.; Eksborg, S.; Larsson, O.; Axelson, M.; Jerling, M.; Abrahmsen, L.; et al. A novel oral insulin-like growth factor-1 receptor pathway modulator and its implications for patients with non-small cell lung carcinoma: A phase I clinical trial. Acta Oncol. 2016, 55, 140–148. [Google Scholar] [CrossRef]
  149. Park, K.; Tan, D.S.W.; Su, W.C.; Cho, B.C.; Kim, S.W.; Lee, K.H.; Wang, C.C.; Seto, T.; Huang, D.C.; Jung, H.H.; et al. Phase 1b Open-Label Trial of Afatinib Plus Xentuzumab (BI 836845) in Patients With EGFR Mutation-Positive NSCLC After Progression on EGFR Tyrosine Kinase Inhibitors. JTO Clin. Res. Rep. 2021, 2, 100206. [Google Scholar] [CrossRef]
  150. Haluska, P.; Menefee, M.; Plimack, E.R.; Rosenberg, J.; Northfelt, D.; LaVallee, T.; Shi, L.; Yu, X.Q.; Burke, P.; Huang, J.; et al. Phase I dose-escalation study of MEDI-573, a bispecific, antiligand monoclonal antibody against IGFI and IGFII, in patients with advanced solid tumors. Clin. Cancer Res. 2014, 20, 4747–4757. [Google Scholar] [CrossRef]
  151. O’Flanagan, C.H.; O’Shea, S.; Lyons, A.; Fogarty, F.M.; McCabe, N.; Kennedy, R.D.; O’Connor, R. IGF-1R inhibition sensitizes breast cancer cells to ATM-related kinase (ATR) inhibitor and cisplatin. Oncotarget 2016, 7, 56826–56841. [Google Scholar] [CrossRef]
  152. de Lint, K.; Poell, J.B.; Soueidan, H.; Jastrzebski, K.; Vidal Rodriguez, J.; Lieftink, C.; Wessels, L.F.; Beijersbergen, R.L. Sensitizing Triple-Negative Breast Cancer to PI3K Inhibition by Cotargeting IGF1R. Mol. Cancer Ther. 2016, 15, 1545–1556. [Google Scholar] [CrossRef] [PubMed]
  153. Kurmasheva, R.T.; Dudkin, L.; Billups, C.; Debelenko, L.V.; Morton, C.L.; Houghton, P.J. The insulin-like growth factor-1 receptor-targeting antibody, CP-751,871, suppresses tumor-derived VEGF and synergizes with rapamycin in models of childhood sarcoma. Cancer Res. 2009, 69, 7662–7671. [Google Scholar] [CrossRef]
  154. Chakraborty, C.; Doss, C.G.; Bandyopadhyay, S.; Agoramoorthy, G. Influence of miRNA in insulin signaling pathway and insulin resistance: Micro-molecules with a major role in type-2 diabetes. Wiley Interdiscip. Rev. RNA 2014, 5, 697–712. [Google Scholar] [CrossRef] [PubMed]
  155. Solomon, V.R.; Alizadeh, E.; Bernhard, W.; Makhlouf, A.; Hartimath, S.V.; Hill, W.; El-Sayed, A.; Barreto, K.; Geyer, C.R.; Fonge, H. Development and preclinical evaluation of cixutumumab drug conjugates in a model of insulin growth factor receptor I (IGF-1R) positive cancer. Sci. Rep. 2020, 10, 18549. [Google Scholar] [CrossRef]
  156. Burtrum, D.; Zhu, Z.; Lu, D.; Anderson, D.M.; Prewett, M.; Pereira, D.S.; Bassi, R.; Abdullah, R.; Hooper, A.T.; Koo, H.; et al. A fully human monoclonal antibody to the insulin-like growth factor I receptor blocks ligand-dependent signaling and inhibits human tumor growth in vivo. Cancer Res. 2003, 63, 8912–8921. [Google Scholar]
  157. Thomson, R.J.; Moshirfar, M.; Ronquillo, Y. Tyrosine Kinase Inhibitors. In StatPearls; StatPearls: St. Petersburg, FL, USA, 2024. [Google Scholar]
  158. Fuentes-Baile, M.; Ventero, M.P.; Encinar, J.A.; Garcia-Morales, P.; Poveda-Deltell, M.; Perez-Valenciano, E.; Barbera, V.M.; Gallego-Plazas, J.; Rodriguez-Lescure, A.; Martin-Nieto, J.; et al. Differential Effects of IGF-1R Small Molecule Tyrosine Kinase Inhibitors BMS-754807 and OSI-906 on Human Cancer Cell Lines. Cancers 2020, 12, 3717. [Google Scholar] [CrossRef] [PubMed]
  159. Girnita, A.; Girnita, L.; del Prete, F.; Bartolazzi, A.; Larsson, O.; Axelson, M. Cyclolignans as inhibitors of the insulin-like growth factor-1 receptor and malignant cell growth. Cancer Res. 2004, 64, 236–242. [Google Scholar] [CrossRef] [PubMed]
  160. Schmid, P.; Cortes, J.; Joaquim, A.; Janez, N.M.; Morales, S.; Diaz-Redondo, T.; Blau, S.; Neven, P.; Lemieux, J.; Garcia-Saenz, J.A.; et al. XENERA-1: A randomised double-blind Phase II trial of xentuzumab in combination with everolimus and exemestane versus everolimus and exemestane in patients with hormone receptor-positive/HER2-negative metastatic breast cancer and non-visceral disease. Breast Cancer Res. 2023, 25, 67. [Google Scholar] [CrossRef]
  161. de Bono, J.; Lin, C.C.; Chen, L.T.; Corral, J.; Michalarea, V.; Rihawi, K.; Ong, M.; Lee, J.H.; Hsu, C.H.; Yang, J.C.; et al. Two first-in-human studies of xentuzumab, a humanised insulin-like growth factor (IGF)-neutralising antibody, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1324–1332. [Google Scholar] [CrossRef] [PubMed]
  162. Chen, H.X.; Sharon, E. IGF-1R as an anti-cancer target--trials and tribulations. Chin. J. Cancer 2013, 32, 242–252. [Google Scholar] [CrossRef]
  163. Crudden, C.; Girnita, A.; Girnita, L. Targeting the IGF-1R: The Tale of the Tortoise and the Hare. Front. Endocrinol. 2015, 6, 64. [Google Scholar] [CrossRef]
  164. Kim, W.Y.; Prudkin, L.; Feng, L.; Kim, E.S.; Hennessy, B.; Lee, J.S.; Lee, J.J.; Glisson, B.; Lippman, S.M.; Wistuba, I.I.; et al. Epidermal growth factor receptor and K-Ras mutations and resistance of lung cancer to Insulin-like Growth Factor 1 Receptor tyrosine kinase inhibitors. Cancer 2012, 118, 3993–4003. [Google Scholar] [CrossRef] [PubMed]
  165. Habben, K.; Delmar, P.; Brownstein, C.M.; Koehler, W.; Kuenkele, K.; Splesis, O.; Ramalingam, S.S.; Engelman, J.A.; Chen, D. Investigation of predictive biomarkers for R1507, an anti-IGF1R antibody, in patients with advanced non-small cell lung cancer with progression after first-line chemotherapy. J. Clin. Oncol. 2011, 29. [Google Scholar] [CrossRef]
  166. Dolgin, E. IGF-1R drugs travel from cancer cradle to Graves’. Nat. Biotechnol. 2020, 38, 385–388. [Google Scholar] [CrossRef] [PubMed]
  167. Toretsky, J.A.; Gorlick, R. IGF-1R targeted treatment of sarcoma. Lancet Oncol. 2010, 11, 105–106. [Google Scholar] [CrossRef]
  168. Beckwith, H.; Yee, D. Minireview: Were the IGF Signaling Inhibitors All Bad? Mol. Endocrinol. 2015, 29, 1549–1557. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the functional connections between the IR and IGF-R signaling axis. The binding of insulin-like growth factors 1/2 (IGF-1/2) to their receptor insulin-like growth factor receptor 1 (IGF-1R) initiates a signaling cascade that ultimately leads to cell growth and division. In contrast, insulin binds with the insulin receptor (IR) to regulate glucose metabolism and maintain blood sugar levels. IR dimers and IGF-1R dimers can combine to form hybrid receptors capable of binding with insulin and IGF-1/2. These hybrid receptors, known as IR/IGF-1R hybrid receptors, play a crucial role in regulating various physiological processes such as cell growth, metabolism, and survival. IGF-2R does not activate any downstream signaling pathways.
Figure 1. Schematic representation of the functional connections between the IR and IGF-R signaling axis. The binding of insulin-like growth factors 1/2 (IGF-1/2) to their receptor insulin-like growth factor receptor 1 (IGF-1R) initiates a signaling cascade that ultimately leads to cell growth and division. In contrast, insulin binds with the insulin receptor (IR) to regulate glucose metabolism and maintain blood sugar levels. IR dimers and IGF-1R dimers can combine to form hybrid receptors capable of binding with insulin and IGF-1/2. These hybrid receptors, known as IR/IGF-1R hybrid receptors, play a crucial role in regulating various physiological processes such as cell growth, metabolism, and survival. IGF-2R does not activate any downstream signaling pathways.
Biomedicines 12 00563 g001
Figure 2. Schematic representation of the IGF/IGF-1R axis: (1) Insulin-like growth factor (IGF-1/2) binds to its receptor, insulin-like growth factor receptor 1 (IGF-1R), triggering a cascade of events that ultimately lead to the activation of the insulin receptor substrate (IRS). Once activated, IRS initiates the PI3K-AKT signaling pathway, which plays a crucial role in promoting cell survival. This is achieved through a reduction in apoptosis, a process of programmed cell death, and an increase in protein synthesis, which aids in cellular growth and maintenance. Overall, the activation of IRS and the subsequent PI3K-AKT signaling pathway are essential for maintaining healthy cellular functions. (2) The SHC protein acts as a signal transducer, triggering the activation of the Ras/MEK/ERK and JNK/MAPK pathways. These pathways, in turn, activate specific transcription factors that regulate the expression of target genes. By orchestrating this complex signaling cascade, the SHC protein plays a crucial role in coordinating cellular responses to diverse extracellular stimuli. This process is essential for maintaining cellular homeostasis and adapting to changing environmental conditions. (3) In addition to the activation of ligand-dependent signaling pathways, insulin-like growth factor-binding proteins (IGFBPs) 1–6 can also trigger ligand-independent signaling pathways. This means that IGFBPs can influence cellular processes even when not bound to IGFs. IGF-1R: Insulin-like growth factor receptor 1; PI3K: Phosphatidylinositol-3-kinase; AKT: Serine/threonine kinase, named protein kinase B (PKB); mTOR: Mammalian target of rapamycin; Bad: Bcl-2-associated death promoter; Bcl2: B-cell lymphoma 2; Shc: Adaptor protein; Ras: GTPase protein; JNK: c-Jun N-terminal kinase; MEK: Mitogen-activated protein kinase kinase; ERK: Extracellular regulated kinase; MAPK: Mitogen-activated protein kinase; ELK: ETS domain-containing protein.
Figure 2. Schematic representation of the IGF/IGF-1R axis: (1) Insulin-like growth factor (IGF-1/2) binds to its receptor, insulin-like growth factor receptor 1 (IGF-1R), triggering a cascade of events that ultimately lead to the activation of the insulin receptor substrate (IRS). Once activated, IRS initiates the PI3K-AKT signaling pathway, which plays a crucial role in promoting cell survival. This is achieved through a reduction in apoptosis, a process of programmed cell death, and an increase in protein synthesis, which aids in cellular growth and maintenance. Overall, the activation of IRS and the subsequent PI3K-AKT signaling pathway are essential for maintaining healthy cellular functions. (2) The SHC protein acts as a signal transducer, triggering the activation of the Ras/MEK/ERK and JNK/MAPK pathways. These pathways, in turn, activate specific transcription factors that regulate the expression of target genes. By orchestrating this complex signaling cascade, the SHC protein plays a crucial role in coordinating cellular responses to diverse extracellular stimuli. This process is essential for maintaining cellular homeostasis and adapting to changing environmental conditions. (3) In addition to the activation of ligand-dependent signaling pathways, insulin-like growth factor-binding proteins (IGFBPs) 1–6 can also trigger ligand-independent signaling pathways. This means that IGFBPs can influence cellular processes even when not bound to IGFs. IGF-1R: Insulin-like growth factor receptor 1; PI3K: Phosphatidylinositol-3-kinase; AKT: Serine/threonine kinase, named protein kinase B (PKB); mTOR: Mammalian target of rapamycin; Bad: Bcl-2-associated death promoter; Bcl2: B-cell lymphoma 2; Shc: Adaptor protein; Ras: GTPase protein; JNK: c-Jun N-terminal kinase; MEK: Mitogen-activated protein kinase kinase; ERK: Extracellular regulated kinase; MAPK: Mitogen-activated protein kinase; ELK: ETS domain-containing protein.
Biomedicines 12 00563 g002
Figure 3. Schematic representation of various therapeutic approaches to targeting IGF-1R receptors. The figure demonstrates how mutations in IGF-1R can affect its structure and downstream signaling pathways. (1) Mutations in IGF-1R can result in an incomplete receptor, causing changes in downstream signaling pathways. These modifications can have a considerable impact on cellular responses. (2) The use of small interfering RNA (siRNA) to silence the gene responsible for encoding the Insulin-like Growth Factor 1 Receptor (IGF-1R) leads to a significant reduction in the receptor levels. This reduction in IGF-1R levels effectively disrupts the signaling pathways associated with the receptor, thereby abrogating the downstream signaling. (3) The application of tyrosine kinase (TK) inhibitors suppresses the activation of IGF-1R, effectively blocking downstream signaling pathways. Inhibiting IGF-1R with TK inhibitors disrupts the signaling cascade, thereby preventing the transmission of crucial cellular signals and, ultimately, halting the cellular response driven by IGF-1R. (4) Applying anti-IGF-1R antibodies inhibits IGF-1R, effectively disrupting its downstream signaling pathways. Anti-IGF-1R antibodies act by binding to IGF-1R and preventing its activation, leading to the abrogation of downstream signaling pathways (RISC: RNA-induced silencing complex).
Figure 3. Schematic representation of various therapeutic approaches to targeting IGF-1R receptors. The figure demonstrates how mutations in IGF-1R can affect its structure and downstream signaling pathways. (1) Mutations in IGF-1R can result in an incomplete receptor, causing changes in downstream signaling pathways. These modifications can have a considerable impact on cellular responses. (2) The use of small interfering RNA (siRNA) to silence the gene responsible for encoding the Insulin-like Growth Factor 1 Receptor (IGF-1R) leads to a significant reduction in the receptor levels. This reduction in IGF-1R levels effectively disrupts the signaling pathways associated with the receptor, thereby abrogating the downstream signaling. (3) The application of tyrosine kinase (TK) inhibitors suppresses the activation of IGF-1R, effectively blocking downstream signaling pathways. Inhibiting IGF-1R with TK inhibitors disrupts the signaling cascade, thereby preventing the transmission of crucial cellular signals and, ultimately, halting the cellular response driven by IGF-1R. (4) Applying anti-IGF-1R antibodies inhibits IGF-1R, effectively disrupting its downstream signaling pathways. Anti-IGF-1R antibodies act by binding to IGF-1R and preventing its activation, leading to the abrogation of downstream signaling pathways (RISC: RNA-induced silencing complex).
Biomedicines 12 00563 g003
Table 1. Pre-clinical studies of IGF-1R-targeted therapies in lung cancer models.
Table 1. Pre-clinical studies of IGF-1R-targeted therapies in lung cancer models.
Drug TypesCompoundsLung Cancer ModelsOutcomesReferences
Monoclonal antibodyCixutumumab (IMC-A12)SCLC cell linesIncreased sensitivity to chemotherapies, decreased cell growth[120]
R1507NSCLC cell linesThe addition of R1507 to erlotinib inhibited cell growth and increased apoptosis[121]
Dalotuzumab (MK-0646)NSCLC xenograft tumors in miceIncreased median survival of treated mice compared with controls, increased sensitivity to erlotinib[122]
Figitumumab (CP-751, 871)NSCLC cell linesIncreased sensitivity to radiation therapy[123]
NSCLC xenograft tumors in miceAddition of CP-751, 871 to radiation therapy delayed tumor growth in vivo
Tyrosine kinase inhibitorLinsitinib (OSI-906)NSCLC cell linesInhibited IGF-1/IGF-2-mediated proliferation, increased apoptosis[124]
NSCLC xenograft tumors in miceInhibited tumor growth in cells expressing IGF-1R[125]
PicropodophyllinB(a)P-induced lung tumors in miceDecreased tumor multiplicity and load[126]
NSCLC cell linesDecreased cell viability and in vitro invasive capacity
NT157Lung cancer cell linesDecreased cell viability and oncogene expression, creating a tumor-suppressive signaling network[127]
BMS-754807NSCLC cell linesDecreased cell survival, increased apoptosis, enhanced cytotoxic effects of platinum chemotherapies[128]
Lung cancer cell linesThe addition of BMS-754807 to dasatinib inhibited cell growth, and induced autophagy and cell cycle arrest[129]
Ligand neutralizing monoclonal antibodyXentuzumab (BI 836845)NSCLC and SCLC cell linesDecreased cell proliferation, enhanced anti-tumor efficacy of rapamycin[130]
Dusigitumab (MEDI-573)Solid xenograft tumors in mice, including NSCLCInhibited IGF signaling pathways in tumors driven by autocrine IGF production[131]
shRNA-mediated Gene SilencingshIGF-1R (601, 801 and 3425)NSCLC cell linesIncreased sensitivity to chemotherapies, decreased cell colony formation[132]
Antibody–drug conjugatesW0101 (IGF-1R antibody-drug conjugate)Lung cancer cell linesPotent cytotoxic activity, inhibited tumor growth in cells expressing high levels of IGF-1R[133]
Abbreviations: B(a)P—benzo(a)pyrene; IGF—insulin-like growth factor; IGF-1R—insulin-like growth factor receptor 1; NSCLC—non-small-cell lung cancer; SCLC—small-cell lung cancer; shRNA—short hairpin RNA.
Table 2. Clinical trials of IGF-1R-targeted therapies in patients with lung cancer.
Table 2. Clinical trials of IGF-1R-targeted therapies in patients with lung cancer.
Drug TypesCompoundsPhaseLung Cancer TypesOutcomesReferences
Monoclonal antibodyCixutumumab (IMC-A12)IINSCLCThe addition of cixutumumab to other therapies increases toxicity without improving efficacy outcomes[134,135]
Ganitumab (AMG-479)IAdvanced solid tumorsSD > 6 weeks in 7 patients, including 2 with NSCLC[136]
AVE1642IAdvanced solid tumorsSD > 4 months in 11 patients, including 1 with NSCLC[137]
Teprotumumab (RV 001, R1507)IINSCLCThe addition of R1507 to erlotinib did not improve efficacy outcomes[138]
IINSCLCTerminated due to program termination (NCT00760929)[22,138]
Dalotuzumab (MK-0646)ISCLCThe addition of dalotuzumab to chemotherapies did not improve efficacy outcomes[139]
IINSCLCThe addition of dalotuzumab to erlotinib did not improve efficacy outcomes[140]
IINon-squamous lung cancerThe addition of dalotuzumab to chemotherapies did not improve efficacy outcomes[141]
Figitumumab (CP-751, 871)IIINSCLCThe addition of figitumumab to chemotherapies did not improve efficacy outcomes[119]
IIINSCLCDiscontinued due to futile HR, lack of improved efficacy outcomes, and serious adverse events[22,142,143]
BIIB022IAdvanced solid tumorsPreliminary evidence of biological activity in select patients; SD > 6 weeks in 20 patients[144]
Tyrosine kinase inhibitorLinsitinib (OSI-906)IISCLCLinsitinib did not improve efficacy outcomes[145]
NSCLCThe addition of linsitinib to erlotinib did not improve efficacy outcomes[146,147]
Picropodophyllin (AXL 1717)IAdvanced solid tumorsMedian PFS of 31 weeks and OS of 60 weeks in 15 patients with NSCLC[148]
Ligand neutralizing monoclonal antibodyXentuzumab (BI 836845)INSCLCThe addition of xentuzumab to afatinib did not substantially improve efficacy outcomes[149]
Dusigitumab (MEDI-573)IAdvanced solid tumorsPreliminary evidence (from 43 patients, including 1 with NSCLC) warrants further clinical evaluation[150]
Abbreviations: HR—hazard ratio; NSCLC—non-small cell lung cancer; OS—overall survival; PFS—progression-free survival; SCLC—small-cell lung cancer; SD—stable disease.
Table 3. Mechanisms of action for IGF-1R targeted therapies in anti-cancer treatment.
Table 3. Mechanisms of action for IGF-1R targeted therapies in anti-cancer treatment.
Molecule TypeAssociated Target(s) Examples Outcomes Intended Anti-Cancer Effects References
Monoclonal antibodyExtracellular ligand-binding α-subunit domain of IGF-1RCixutumumab (IMC-A12), R1507, Dalotuzumab (MK-0646), Figitumumab (CP-751, 871)Inhibits ligand binding to receptor and promotes receptor internalization and degradationAnti-proliferative, anti-growth, anti-metastatic, prevention of EMT transition, pro-apoptotic, increased sensitization to chemotherapy [121,155,156]
Tyrosine kinase inhibitorATP pocket of IGF-1R (ATP-competitive)Linsitinib (OSI-906),
BMS-754807, AG1024
Inhibits auto-phosphorylation of IGF-1R upon ligand binding, preventing recruitment of signaling proteins such as IRS and ShcAnti-proliferative, anti-growth, anti-metastatic,
pro-apoptotic, increased sensitization to chemotherapy
[127,157,158,159]
Allosteric site on IGF-1R (non-ATP competitive)XL228, Picropodophyllin, NT157Decreased tumor multiplicity and load [126]
Ligand-neutralizing monoclonal antibodyIGF-I/IIXentuzumab (BI 836845), Dusigitumab (MEDI-573)Prevents ligand binding to IGF-1RAnti-proliferative, anti-growth, pro-apoptotic, increased sensitization to chemotherapy[160,161,162]
shRNA-mediated gene silencingIGF-1R mRNA shIGF-1R(601, 801 and 3425)Prevents translation of IGF-1R mRNA, silencing gene expression Anti-tumorigenic, pro-apoptotic, increased sensitization to chemotherapy[132]
Antibody–drug conjugatesExtracellular domain of IGF-1RW0101 (IGF-1R antibody-drug conjugate)Promotes internalization of receptor and conjugated cytotoxic drug in cells expressing high levels of IGF-1R Anti-mitotic, anti-tumorigenic, cytotoxic [133]
Abbreviations: EMT—epithelial–mesenchymal transition; IGF—insulin-like growth factor; IGF-1R—insulin-like growth factor receptor 1; IRS—insulin receptor substrate; shRNA—short hairpin RNA; Shc—Src homology and collagen protein.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shahid, A.; Santos, S.G.; Lin, C.; Huang, Y. Role of Insulin-like Growth Factor-1 Receptor in Tobacco Smoking-Associated Lung Cancer Development. Biomedicines 2024, 12, 563. https://doi.org/10.3390/biomedicines12030563

AMA Style

Shahid A, Santos SG, Lin C, Huang Y. Role of Insulin-like Growth Factor-1 Receptor in Tobacco Smoking-Associated Lung Cancer Development. Biomedicines. 2024; 12(3):563. https://doi.org/10.3390/biomedicines12030563

Chicago/Turabian Style

Shahid, Ayaz, Shaira Gail Santos, Carol Lin, and Ying Huang. 2024. "Role of Insulin-like Growth Factor-1 Receptor in Tobacco Smoking-Associated Lung Cancer Development" Biomedicines 12, no. 3: 563. https://doi.org/10.3390/biomedicines12030563

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop