IMR Press / FBL / Volume 28 / Issue 8 / DOI: 10.31083/j.fbl2808163
Open Access Review
The Relationship between IGF Pathway and Acquired Resistance to Tyrosine Kinase Inhibitors in Cancer Therapy
Show Less
1 Cancer Research Center, Capital Medical University Affiliated Beijing Chest Hospital, 101149 Beijing, China
*Correspondence: jinjingtan@ccmu.edu.cn (Jinjing Tan)
Front. Biosci. (Landmark Ed) 2023, 28(8), 163; https://doi.org/10.31083/j.fbl2808163
Submitted: 14 December 2022 | Revised: 10 March 2023 | Accepted: 24 March 2023 | Published: 11 August 2023
(This article belongs to the Special Issue Signaling Pathways in Cancers)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The tyrosine kinase signaling pathway is an important pathway for cell signal transduction, and is involved in regulating cell proliferation, cell cycle, apoptosis and other essential biological functions. Gene mutations involved in the tyrosine kinase signaling pathway often lead to the development of cancers. Epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor-2 (HER2) are well known receptor tyrosine kinases (RTKs), which belong to the ERBB family and have high mutation frequency in cancers. Tyrosine kinase inhibitors (TKI) targeting EGFR and HER2 have been widely used in the clinical treatment of lung and breast cancers. However, after a period of treatment, patients will inevitably develop resistance to TKI. The insulin-like growth factor (IGF) receptor family, like the ERBB receptor family, belongs to the receptor tyrosine kinase superfamily, which also conducts an important cell signal transduction function. There is an overlap between IGF signaling and EGFR signaling in biological functions and downstream signals. In this review, we summarize the current state of knowledge of how IGF signaling interacts with EGFR signaling can influence cell resistance to EGFR/HER2-TKI. We also summarize the current drugs designed for targeting IGF signaling pathways and their research progress, including clinical trials and preclinical studies. Altogether, we aimed to discuss the future therapeutic strategies and application prospects of IGF signaling pathway targeted therapy.

Keywords
insulin-like growth factor
tyrosine kinase inhibitors
drug resistance
cancer
EGFR
HER2
1. Introduction

Cancer is one of the major challenges to human health. Female breast cancer has the highest incidence (accounting for 11.7% of de novo tumors), followed by lung cancer (11.4%), which is the leading cause of cancer death (accounting for 18.0% of total cancer deaths) [1]. Thanks to the progress of life science research, we now have had a deep understanding of the mechanism of cancer, such as intracellular signal transduction, cell cycle regulation and apoptosis induction. It has been found that tyrosine kinases play a significant role in tumorigenesis and development [2]. Mutations in some genes can lead to sustained activation of tyrosine kinases, which results in uncontrolled cell growth. These genes, which can cause cells to become immortalized or cancerous, are called tumor driven genes [3]. Tyrosine kinase has become an important therapeutic target in the treatment of cancer. The increased use of tyrosine kinase inhibitors (TKIs), which are often small molecule inhibitors targeting tyrosine kinases, are a prelude to targeted therapy for cancer treatment, and have the potential to effectively prolong the survival of cancer patients [4]. A retrospective study analyzed the median survival (95% CI) of patients with lung adenocarcinoma, and found that the median survival of patients with tumor driven genes mutation who received targeted drug therapy was 3.49 months (3.02–4.33), which was significantly higher than that of patients with tumor driven genes mutation but receiving conventional treatment (surgery, radiotherapy or chemotherapy) (2.38 months (1.81–2.93)) and patients without tumor driven genes mutation and receiving conventional treatment (2.08 (1.84–2.46)) [5]. Another study suggested that 60%–80% of patients with chronic myeloid leukemia treated with imatinib achieved clinical benefits lasting more than a decade [6]. Although the use of targeted kinase inhibitors has brought significant benefits to many cancer patients, it is disappointing to note that these drugs are not curative and most only delay tumor progression, because acquired resistance ultimately develops in most patients with advanced metastatic disease [7]. Solving the problem of drug resistance and enhancing drug sensitivity are the key to improving the therapeutic effect and the prognosis of cancer patients.

In this review, we introduce two representative TKIs, epidermal growth factor receptor (EGFR)-TKIs and human epidermal growth factor receptor-2 (HER2)-TKIs, for the treatment of lung and breast cancer. We describe the clinical application of EGFR-TKI and HER2-TKI in these cancers, the current results of these therapies, and the status of the development of drug resistance for these TKIs. The mechanism of TKI resistance is complicated, and several studies have found that the insulin-like growth factor (IGF) signaling pathway plays a crucial role in TKI resistance [8, 9, 10]. In this review, we describe the relationship between the IGF pathway and acquired resistance to TKIs.

2. Application of TKIs in Tumor Therapy

Protein kinase/phosphatase-regulated signaling pathways are involved in the occurrence and development of almost all types of cancer and have become one of the most important drug targets in the 21st century [4]. The tyrosine residues of the receptor protein are phosphorylated and interact with downstream signaling molecules to activate the Ras/Raf/MAPK or PI3K/Akt signaling pathways and regulate the survival, proliferation and metastasis of tumor cells. Receptor tyrosine kinases (RTKs), which have become the key targets for cancer treatment, are composed of multiple receptor families, including the ERBB receptor family (such as EGFR, HER2), the IGF receptor family (such as IGF-1R, InsR), the VEGF receptor family, the FGF receptor family, the c-MET receptor, and the ALK receptor. Since the end of 2022, 78 small molecule protein kinases-targeted inhibitors have been approved by the Food and Drug Administration (FDA), of which 18 inhibitors target non-receptor tyrosine kinases and 38 inhibitors target receptor tyrosine kinases (Table 1, Ref. [11, 12]). The main targets of TKIs include EGFR/HER, VEGFR, ALK, MET, JAK, FGFR, ABL, KIT [12], among which the most widely and successfully used are EGFR and HER2. EGFR and HER2 are members of the transmembrane RTKs ERBB family.

Table 1.FDA-approved small molecule inhibitors or monoclonal antibodies (mAb) targeting protein kinase4.
Drug Primary targets1 The type of target/drugs2 Year approved Therapeutic indications3
Abemaciclib CDK4/6 CMGC 2017 Combination therapy with an aromatase inhibitor or with fulvestrant or as a monotherapy for breast cancers
Abrocitinib JAK TK 2022 Atopic dermatitis
Acalabrutinib BTK TK 2017 Mantle cell lymphomas, CLL, SLL
Afatinib EGFR/HER TK 2013 NSCLC
Alectinib ALK TK 2015 ALK-positive metastatic NSCLC that progressed on or is intolerant to crizotinib
Alpelisib PI3K LK 2019 Combination therapy with fulvestrant for advanced or metastatic breast cancer with hormone receptor-positive, HER2-negative (HR+/HER2-) and PIK3CA-mutation, in postmenopausal women or men
Amivantamab-vmjw EGFR/MET ADCs 2021 NSCLC with EGFR exon 20 insertion mutation
Asciminib ABL TK 2021 Ph+ CML in chronic phase (CP), previously treated with 2 or more tyrosine kinase inhibitors (TKIs)
Avapritinib KIT TK 2020 GIST with PDGFRα exon 18 mutation
Axitinib VEGFR TK 2012 RCC
Baricitinib JAK TK 2017 Extensive alopecia
Binimetinib MEK1/2 STE 2018 Combination therapy with encorafenib for BRAF V600E/K melanomas
Bosutinib ABL TK 2012 Newly diagnosed chronic phase Ph+ CML
Brigatinib ALK TK 2017 ALK-positive metastatic NSCLC that have progressed or are intolerant to crizotinib
Cabozantinib RET TK 2012 Medullary thyroid cancers, RCC, HCC
Capmatinib MET TK 2020 NSCLC with MET exon 14 skipping
Ceritinib ALK TK 2014 ALK-positive metastatic NSCLC that progressed on or is intolerant to crizotinib
Cetuximab EGFR/HER mAb 2004 EGFR-expressing metastatic colorectal carcinoma intolerant to irinotecan-based chemotherapy
Cobimetinib MEK1/2 STE 2015 BRAF V600E/K melanomas in combination with vemurafenib
Copanlisib PI3K LK 2017 Adult patients with recurrent marginal zone lymphoma who have received at least two-line treatment
Crizotinib ALK TK 2011 Locally advanced or metastatic NSCLC with ALK-positive
Dabrafenib BRAF TKL 2013 BRAF V600E/K melanomas, BRAF V600E NSCLC, BRAF V600E anaplastic thyroid cancers
Dacomitinib EGFR/HER TK 2018 EGFR-mutant NSCLC
Dasatinib ABL TK 2006 CML
Deucravacitinib TYK2 TK 2022 Adult patients with moderate to severe plaque psoriasis suitable for systemic therapy or phototherapy
Duvelisib PI3K LK 2018 SLL, CLL, FL
Encorafenib BRAF TKL 2018 Combination therapy with binimetinib for BRAF V600E/K melanomas
Entrectinib TRK TK 2019 Solid tumors with NTRK fusion proteins, ROS1-positive NSCLC
Erdafitinib FGFR TK 2019 Urothelial bladder cancers
Erlotinib EGFR/HER TK 2004 NSCLC, pancreatic cancers
Everolimus mTOR AK 2009 HER2-negative breast cancers, pancreatic neuroendocrine tumors, RCC, angiomyolipomas, subependymal giant cell astrocytomas
Fam-trastuzumab deruxtecan-nxki HER2 ADCs 2022 Unresectable or metastatic non-small cell lung cancer with HER2 mutation
Fedratinib JAK TK 2019 Myelofibrosis
Fostamatinib SYK TK 2018 Immunothrombocytopenia
Futibatinib FGFR TK 2022 Locally advanced or metastatic cholangiocarcinoma with FGFR2 gene rearrangement/fusion
Gefitinib EGFR/HER TK 2003 NSCLC
Gilteritinib FLT3 TK 2018 AML
Ibrutinib BTK TK 2013 CLL, mantle cell lymphomas, marginal zone lymphomas, graft vs. host disease, Waldenstrom macroglobulinemia
Idelalisib PI3K LK 2014 SLL, CLL, FL
Imatinib ABL TK 2001 Ph+ CML or ALL, aggressive systemic mastocytosis, chronic eosinophilic leukemias, dermatofibrosarcoma protuberans, hypereosinophilic syndrome, GIST, myelodysplastic/ myeloproliferative disease
Infigratinib FGFR TK 2021 Cholangiocarcinomas with FGFR2 fusion proteins
Lapatinib EGFR/HER TK 2007 In combination with letrozole for post-menopausal women with HR-positive metastatic breast cancer that overexpresses the HER2 receptor
Larotrectinib TRK TK 2018 Solid tumors with NTRK fusion proteins
Lenvatinib VEGFR TK 2015 Differentiated thyroid cancers
Lorlatinib ALK TK 2018 ALK-positive metastatic NSCLC that has progressed on crizotinib and at least one other ALK inhibitor or alectinib/ceritinib as the first ALK inhibitor therapy
Margetuximab HER2 mAb 2020 Metastatic HER2-positive breast cancer
Midostaurin FLT3 TK 2017 AML, mastocytosis, mast cell leukemias
Mobocertinib EGFR/HER TK 2021 NSCLC for EGFR-positive exon 21 insertions
Necitumumab EGFR/HER mAb 2015 Combined with gemcitabine and cisplatin for the first-line treatment of metastatic squamous NSCLC
Neratinib EGFR/HER TK 2017 HER2-positive breast cancers
Netarsudil ROCK1/2 AGC 2017 Glaucoma
Nilotinib ABL TK 2007 Ph+ CML
Nintedanib VEGFR TK 2014 Idiopathic pulmonary fibrosis
Olaratumab PDGFR-α mAb 2016 Combined with doxorubicin for adult soft tissue sarcomas
Osimertinib EGFR/HER TK 2015 Metastatic EGFR T790M mutation-positive NSCLC that progressed on or after EGFR TKI therapy
Pacritinib JAK TK 2022 Myelofibrosis
Palbociclib CDK4/6 CMGC 2015 In combination with letrozole for postmenopausal women with ER-positive, HER2-negative advanced breast cancer for metastatic disease
Panitumumab EGFR/HER mAb 2006 EGFR-expressing metastatic colorectal carcinoma
Pazopanib VEGFR TK 2009 RCC, soft tissue sarcomas
Pemigatinib FGFR TK 2020 Advanced cholangiocarcinoma with a FGFR2 fusion or rearrangement
Pertuzumab HER2 mAb 2012 In combination with trastuzumab and docetaxel for neoadjuvant treatment of HER2-positive locally advanced inflammatory or early-stage breast cancer
Pexidartinib CSF1R TK 2019 Tenosynovial giant cell tumors
Ponatinib ABL TK 2012 Ph+ CML or ALL
Pralsetinib RET TK 2020 RET-fusion NSCLC, medullary thyroid cancer, thyroid cancer
Ramucirumab VEGFR mAb 2014 Gastric or gastroesophageal junction adenocarcinoma, metastatic non-small cell lung cancer, colorectal cancer
Regorafenib BRAF TKL 2012 Colorectal cancers
Ribociclib CDK4/6 CMGC 2017 Combination therapy with an aromatase inhibitor for breast cancers
Ripretinib KIT TK 2020 Fourth-line treatment for GIST
Ruxolitinib JAK TK 2011 Myelofibrosis, polycythemia vera, atopic dermatitis
Selpercatinib RET TK 2020 Metastatic RET fusion-positive NSCLC, thyroid cancers, RET mutant medullary thyroid cancers
Selumetinib MEK1/2 STE 2020 Neurofibromatosis type I
Sirolimus mTOR AK 1999 Kidney transplants, lymphangioleiomyomatosis
Sorafenib BRAF TKL 2005 HCC, RCC, thyroid cancer (differentiated)
Sunitinib VEGFR TK 2006 GIST, pancreatic neuroendocrine tumors, RCC
Temsirolimus mTOR AK 2007 RCC
Tepotinib MET TK 2021 NSCLC with MET mutations
Tirbanibulin SRC TK 2020 Actinic keratosis
Tivozanib VEGFR TK 2021 Third-line treatment of RCC
Tofacitinib JAK TK 2012 Rheumatoid arthritis, psoriatic arthritis, ulcerative colitis
Trametinib MEK1/2 STE 2013 In combination with dabrafenib for unresectable or metastatic melanoma with BRAF V600E or V600K mutations
Trastuzumab HER2 mAb 1998 HER2-positive breast cancer
Trastuzumab–deruxtecan HER2 ADCs 2019 HER2 low expression metastatic breast cancer
Trastuzumab–emtansine HER2 ADCs 2013 HER2 positive breast cancer
Trilaciclib CDK4/6 CMGC 2021 Chemotherapy-induced myelosuppression
Tucatinib EGFR/HER TK 2020 Combination second-line treatment for HER2-positive breast cancers
Umbralisib PI3K/CK1 LK/CK 2021 Marginal zone lymphoma, FL
Upadacitinib JAK TK 2019 Second-line treatment for rheumatoid arthritis
Vandetanib VEGFR TK 2011 Medullary thyroid cancers
Vemurafenib BRAF TKL 2011 BRAF V600E melanomas
Zanubrutinib BTK TK 2019 Mantle cell lymphomas

1 Although many of these drugs are multikinase inhibitors, only the primary therapeutic targets are given here. ALK, anaplastic lymphoma kinase; BTK, Bruton tyrosine kinase; CDK4/6, cyclin-​dependent kinase 4/6; CK1, casein kinases; CSF1R, colony-​stimulating factor 1 receptor; FGFR, fibroblast growth factor receptors; FLT3, Fms-like TyrosineKinase-3; JAK, Janus kinase; MET, mesenchymal-epithelial transition factor; mTOR, mammalian target of rapamycin; PDGFR-α, platelet-​derived growth factor receptor-α; PI3K, phosphatidylinositol 4,5-​bisphosphate 3-​kinase; ROCK1/2, Rho-​associated coiled-​coil-​containing protein kinases 1/2;SYK, spleen tyrosine kinase; TRK, tropomyosin receptor kinase; TYK2, tyrosine kinase 2; VEGFR, vascular endothelial growth factor receptor.

2 ADCs, antibody-drug conjugates; AGC, protein kinase A/G/C; AK, atypical kinases; CMGC, cyclin-dependent kinases, MAP kinases, glycogen synthase kinases, CDC-like kinases; LK, lipid kinases; mAb, monoclonal antibody; STE, homologues of sterile 7; TK, tyrosine kinases; TKL, tyrosine kinase like.

3 CLL, chronic lymphocytic leukemias; CML, chronic myelogenous leukemias; SLL, small lymphocytic lymphomas; PH, Philadelphia chromosome; RCC, renal cell carcinomas; ALL, acute lymphoblastic leukemias; GIST, gastrointestinal stromal tumors; FL, follicular lymphoma; NSCLC, non-small cell lung cancer; HCC, hepatocellular carcinoma; AML, acute myelogenous leukemias.

4 Adapted from Ref. [11, 12], and some information is obtained from https://www.fda.gov/.

The ERBB family, also known as the EGF receptor family or Type I receptor family, is a transmembrane RTKs family, whose members include ERBB1 (EGFR/HER-1), ERBB2 (Neu/HER-2), ERBB3 (HER-3) and ERBB4 (HER-4) [13]. ERBB receptors are activated after homodimerization or heterodimerization, which sequentially activates intracellular signaling cascades. There are two main downstream pathways, the RAS-RAF-MEK-MAPK pathway and the PI3K-AKT-mTOR pathway [14]. The former controls gene transcription, cell cycle progression and cell proliferation, while the latter activates anti-apoptotic and pro-survival signaling cascades [15]. About 20% of non-small cell lung cancer (NSCLC) patients have EGFR activating mutations. The main subtype includes exon 19 deletion (Ex19del) and exon 21 L858R mutation [16]. HER2 mutations are found in approximately 25–30% of tumors in invasive breast cancer [17]. Mutations in the EGFR and HER2 genes lead to persistent activation of the ERBB signaling pathway, resulting in uncontrolled cell growth, immortalization of cells and transformation into cancer cells. Using targeted drugs to specifically inhibit abnormally expressed genes can effectively inhibit the process of tumor growth, metastasis and invasion, and have the potential to treat several malignancies. To accomplish this, scientists have designed a variety of small molecule inhibitors targeting EGFR and HER2. In this review, we will introduce the clinical application of EGFR-TKI and HER2-TKI in the treatment of lung and breast cancers.

2.1 The Clinical Application of EGFR-TKI in Lung Cancer

Lung cancer is the leading cause of cancer-related deaths worldwide, with more than half (57%) of lung cancer patients having metastases at the time of diagnosis, with only a 5% 5-year survival rate [18]. NSCLC is the most dominant subtype of lung cancer, accounting for 85% of all lung cancer cases [19]. EGFR is one of the most common driven gene in NSCLC. The proportion of EGFR activation mutation in NSCLC patients is about 20% [16]. Females (69.7%), non-smokers (66.6%), and patients with adenocarcinoma (80.9%), have a higher frequency of EGFR mutations (p < 0.001 for all comparisons) [20]. EGFR-TKIs, which target EGFR activating mutations, are recommended by current treatment guidelines for the first-line treatment of advanced NSCLC patients with activating mutations of EGFR [21, 22]. Representative drugs of the first generation of EGFR-TKIs include gefitinib, erlotinib, icotinib. These are reversible inhibitors that compete with adenosine triphosphate (ATP) to bind to the tyrosine kinase domain of EGFR and reversibly inhibit the kinase activity of EGFR [23]. Gefitinib was approved by the FDA in 2015 as a first-line treatment for advanced NSCLC patients with EGFR activating mutations [24]. The second generation of EGFR-TKIs, including afatinib and dacomitinib, were originally designed to overcome resistance of the first generation of EGFR-TKIs and are irreversible inhibitors of EGFR. Afatinib is also a dual specific inhibitor against EGFR/HER2, which can covalently combine with EGFR and HER2. Dacomitinib is an irreversible pan-HER inhibitor. Compared with the first-generation EGFR-TKIs, the broad-spectrum activity of the second-generation drugs enhances their ability to inhibit EGFR-dependent tumor growth. However, the second generation EGFR-TKIs have a higher incidence of adverse events compared with gefitinib, such as enhancive skin and gastrointestinal toxicity and decreased tolerance to maximum therapeutic doses, limiting their use in patients resistant to first-generation drugs [25]. After 9~13 months of treatment with the first- or second-generation EGFR-TKIs, most patients inevitably develop acquired resistance, among which 50%–60% have the EGFR exon 20 T790M mutation [26, 27, 28]. Therefore, the T790M mutation is known as the “gatekeeper” mutation, which increases the steric hindrance of the EGFR tyrosine kinase domain. Sequentially, it increases the affinity of ATP to the mutant EGFR receptor and increases the competition between ATP and reversible EGFR-TKIs, ultimately decreasing the effectiveness of the first- and second-generation EGFR-TKIs [26]. The representative drug of the third generation of EGFR-TKIs is osimertinib, which can effectively overcome EGFR-TKIs resistance caused by the T790M mutation and achieves increased clinical activity and tolerance. Compared with chemotherapy, osimertinib improved progression-free survival (PFS) and overall survival (OS) for patients with disease progression during treatment with first-generation EGFR-TKIs and concurrently with T790M mutation [28]. Osimertinib has dominated the treatment of EGFR-positive NSCLC worldwide, and is currently the first-line treatment for T790M-positive NSCLC patients and the first choice of patients who have become resistance to first or second-generation EGFR-TKIs [28, 29]. But there are always new EGFR mutations that make the tumor resistant to current generation of EGFR-TKIs, such as the recently discovered EGFR C797S mutation [30]. In response to this problem, the fourth generation EGFR-TKIs are currently being developed, such as EAI045, BLU-945, TQB3804, grigatinib, but their clinical efficacy is still uncertain. Additional clinical trials may be needed in the future to verify the efficacy of the novel EGFR-TKIs in patients with advanced lung cancer.

From the history of multigenerational EGFR-TKIs development, it can be learnt that simply optimizing TKIs to improve the efficacy of targeted drugs may not be the best way to manage drug resistance, as the newly occurred mutations may be endless. Alternative approaches to managing patient resistance should be considered, such as combining immunotherapy or chemotherapy, using downstream signaling pathway inhibitors or targeting non-target-dependent resistance mechanisms.

2.2 The Clinical Application of HER2-TKI in Breast Cancer

According to the latest statistics, the morbidity of breast cancer is now the highest of all cancers and exceeds that of lung cancer [1]. Breast cancer can be divided into separate tumor subtypes according to the expression of prognostic factors such as estrogen receptor (ER), progesterone receptor (PR), and HER2, which are closely relevant to treatment strategies [31]. ER-positive tumor patients mainly receive endocrine therapy; HER2-positive tumors can be treated with antibodies or small molecule inhibitors targeting HER2; while patients with triple-negative tumors generally receive chemotherapy [32]. HER2 (ERBB2/Neu) is one of the most typical oncogenes involved in the occurrence of breast cancer. HER2-positive breast cancer, characterized by HER2 protein overexpression or gene amplification, has been found in approximately 25% to 30% of invasive breast cancers, and is especially associated with a poor prognosis [17]. Unlike other members of the ERBB family, HER2 lacks natural ligands in vivo, and HER2 exerts its biological effects primarily by forming dimers with itself or other growth factor receptors [14]. The main dimerizing chaperones include EGFR/HER1, HER3, IGF-1R, IGF-2R, and c-MET [33]. Similar to EGFR, HER2 has ATP-dependent tyrosine kinase domains and extracellular domains, enabling it to be targeted with small molecule inhibitors or monoclonal antibodies (mAb).

There are several HER2-targeting drugs now used in clinical practice. Trastuzumab (Herceptin), a humanized mAb targeting the extracellular subdomain IV of HER2 [34], which induces antibody-dependent cell-mediated cytotoxicity (ADCC), is widely used as a targeted therapy in patients with HER2 overexpression. Lapatinib, a small molecule inhibitor that targets both EGFR and HER2, has been approved for HER2-positive breast cancer patients treated with trastuzumab with disease progression [35]. Pertuzumab is a second-generation recombinant humanized mAb, which can bind to the extracellular dimerization domain II of HER2, thereby preventing HER2 from forming heterodimer with HER1, HER3, HER4, and IGF-1R, consequently inhibiting cell proliferation [36]. In addition, trastuzumab emtansine (T-DM1) is an antibody-drug conjugates (ADCs) that combines the anti-HER2 effect of trastuzumab with the cytotoxicity of the anti-microtubule agent DM1 in order to selectively introduce potent cytotoxic agents into HER2-overexpressing cells by binding to HER2 [37].

3. The IGF Signaling Pathway is Involved in Acquired Resistance to TKIs

The application of TKIs has brought great hope to a large number of patients suffering from lung, breast, and colorectal cancers, leukemia and other tumors; however, most patients will inevitably develop drug resistance after the use of targeted drugs. NSCLC patients treated with EGFR-TKIs develop drug resistance in an average of 6 to 13 months [38]. The majority of patients who achieve an initial response to trastuzumab-based regimens develop resistance within 1 year [39]. A comprehensive understanding of the resistance mechanisms of TKIs will contribute to the development of a new generation of targeted drugs and follow-up therapy, which has the potential to effectively prolong patient survival. The resistance mechanisms of TKIs are complicated and varied, and are divided into target-dependent resistance mechanisms and non-target-dependent resistance mechanisms. The former is usually induced by a secondary mutation in the ATP-binding pocket of the receptor kinase, which alters its molecular conformation and declines its combination with TKI [40]. Similar to the development of the third generation EGFR-TKI, this type of drug resistance can be effectively improved by changing the chemical conformation of pharmaceutical molecules. Non-target-dependent drug resistance mechanisms are more complex and diverse, including compensation of other signaling pathways, sustained activation of downstream pathways, and phenotypic transformation [40, 41]. For example, in spite of the capacity of EGFR-TKIs that can still inhibit the phosphorylation of mutant EGFR, drug-resistant cells maintain activation of survival and proliferation signals through other pathways.

Several studies have shown that the activation of the IGF signaling pathway can impact the sensitivity of cells to TKIs. Choi et al. [42] found that inhibition of IGF-1R enhanced the growth inhibition and pro-apoptotic effects of gefitinib on H1650 cells. Increased phosphorylation of IGF-1R and AKT was found in hepatocellular carcinoma cells after gefitinib treatment [43]. The expression of IGF-1 was also significantly increased in erlotinib-resistant glioblastoma cell lines, but not in sensitive cell lines [44]. Seropositivity of IGF-1 was found to be an independent adverse prognostic factor in NSCLC patients treated with gefitinib [45]. The specificity of predicting gefitinib resistance with high levels of total IGF-1R was 76%, and the positive predictive value was 81%. The specificity of predicting gefitinib resistance with high levels of phosphorylated IGF-1R was 100%, the sensitivity was 41%, the positive predictive value was 100%, and the negative predictive value was 23%, implying that the expression of IGF-1R can be used as a biomarker to predict the emerging resistance of NSCLC to gefitinib [46]. These findings suggest that the use of TKIs may promote the activation of the IGF pathway, and the activation of the IGF pathway is involved in the resistance of cells to TKIs.

The IGF pathway is an intricate network, mainly consisting of the following three components: three ligands, insulin, IGF-1, IGF-2; three cell surface receptors, IGF-1R, IGF-2R, insulin receptor (InsR); and six high-affinity insulin-like growth factor binding proteins (IGFBPs) [47]. IGFs are synthesized in the liver prevailingly, and the level of IGFs is regulated by IGFBPs principally [8, 48]. There are 6 types of IGFBPs with high affinity in vivo, which bind to 98% of circulating IGF-1 and act as carrier proteins to regulate the transport of IGF-1 and prolong its relatively short half-life [49, 50]. InsR, IGF-1R and IGR-2R are all transmembrane dimer receptors. IGF-1R and InsR belong to RTKs family. Since they are of the same domain, IGF-1R and InsR usually exist on the cell surface in the form of a homodimer or heterodimer [51]. Similarly, the extracellular domain structure of EGFR, which belongs to RTKs as well, is similar to that of IGF-1R [52], suggesting the possibility of IGF-1R/EGFR heterodimer formation [53]. The prime activating ligands of IGF-1R are IGF-1 and IGF-2. After combining with the ligands, IGF-1R activates its tyrosine kinase activity, phosphorylates some insulin receptor substrate (IRS) proteins, and then triggers multiple downstream signaling pathways, including the RAS-RAF-MEK-MAPK pathway, the PI3K-AK-mTOR pathway, TAK/STAT and Src pathways, which are closely associated with the proliferation, invasion, metastasis, epithelial mesenchymal transition (EMT) and drug resistance of tumor cells [8, 54]. There are two subtypes of InsR, InsR-A and InsR-B, which differ in their ligand binding and signal transduction properties. InsR-A is associated with the mitotic pathway and binds to IGF-2 and insulin, mainly expressed in cancer and fetal cells; while InsR-B only combines with insulin at physiological concentrations, and mainly regulates glucose homeostasis, leading to major metabolic effects, that are prevailingly expressed in metabolic tissues [55, 56]. Unlike IGF-1R and InsR, due to the loss of intracellular domain, IGF-2R lacks kinase activity, which removes IGF-2 mainly through receptor-mediated endocytosis and lysosomal degradation, preventing it from activating InsR and IGF-1R, thereby disrupting IGF-2 signal transduction [57, 58] (Fig. 1).

Fig. 1.

IGF, insulin and their receptors. IGF is secreted by liver cells, tumor cells or other cells, while insulin is secreted by islet-β cells. IGF-1R can heterodimerize with InsR-A or InsR-B to form hybrid receptors, which is activated by binding to IGF-1 or IGF-2. IGF-2R can be used to isolate IGF-2 (red line), considering its lack of kinase activity. IGF-2 is also able to bind and activate InsR-A. IGFBPs usually serve as a serum bank for IGFs. This illustration was created by Figdraw.

IGFBPs, a type of circulating protein with sophisticated functions, are considered to be a carrier protein regulating the activity of IGFs and can be used as a serum bank for IGFs [9]. In addition to binding IGF through common IGF binding domains, several IGFBPs can also combine with specific membrane receptors and attach to the cell surface or extracellular matrix, ultimately performing a variety of intricate functions through IGF-dependent or IGF-independent mechanisms, including cell proliferation, movement, and tissue remodeling [49, 59, 60]. IGFBP2 is the second most abundant IGFBP in the circulation. Several studies have shown that IGFBP2 is overexpressed in various tumors [60, 61, 62, 63, 64]. However, another study demonstrated that the expression of IGFBP2 and IGFBP4 in PC-9DR2 cell lines with dual resistance to MET-TKI and gefitinib is deceased [65], indicating that the role of IGFBP2 is complicated. IGFBP3 is the staple IGF carrier protein in serum [66], with both stimulating and inhibiting effects on the biological activity of IGF, but little is known about these biological switches. IGFBP3 has long been considered a strong negative regulator of IGF-1R activity [67]. Studies have shown that deficiency of IGFBP3 expression results in resistance to osimertinib in NSCLC [68]. In contrast, other studies have demonstrated that enhancive expression of IGFBP3 results in resistance to afatinib in lung adenocarcinoma [66]. The increase of IGFBP3 and IGFBP5 was also found to contribute to the resistance of neuroblastomas to TKI [69]. Therefore, the effect of IGFBP3 on TKIs resistance is complicated. The function of IGFBP7, similar to that of IGFBP2 or IGFBP3, is complex. Studies have found that IGFBP7 can compete with IGFs to combine with IGF-1R, thereby inhibiting the activation of IGF-1R [70]. Another study found that the content of IGFBP7 is reduced in half of metastatic breast cancers, and the abnormal activation of IGFBP7 can inhibit the activation of the MAPK pathway [35], implying that IGFBP7 may negatively regulate the IGF pathway. On the contrary, overexpression of IGFBP7 has been shown to prevent tumor cell apoptosis, resulting in EGFR-TKIs resistance in lung cancer cells [71]. These findings demonstrate that IGFBPs have dual effects on tumor development, drug resistance of TKIs, and activation of the IGF pathway. The relevant biological switch is still unclear, and future studies need to further explore the mechanism. A variety of IGFBPs with similar structure and function have been shown in separate studies to be involved in regulating cell sensitivity to EGFR-TKI along. It is not clear whether their relationship is independent, synergistic, or antagonistic in this context. Therefore, we need more systematic and comprehensive research.

4. The Mechanism of the IGF Pathway Involved in Acquired Resistance to TKIs

The IGF pathway is involved in various processes of tumor growth, such as tumor proliferation, invasion, metastasis, and EMT [8]. Nuclear IGF-IR can also regulate gene expression [9], which is related to the involvement of the IGF pathway in resistance to chemotherapy, radiotherapy and targeted therapy. In this section, we discuss the mechanism of the IGF pathway to acquired resistance to TKIs from three aspects: (1) The IGF pathway is involved in the activation of the EGFR compensatory signaling pathway. Although EGFR-TKIs can still inhibit the phosphorylation of mutant EGFR, drug-resistant cells maintain activation of downstream signaling pathways through other pathways. (2) The IGF pathway promotes the activation of EMT and integrin pathways, which in turn promotes tumor growth, metastasis and drug resistance, leading to tumor progression. (3) Activation of the IGF pathway can inhibit apoptosis or cell cycle arrest caused by targeted drugs, leading to sustained growth of tumor cells and resistance to TKIs.

4.1 Activation of Compensatory Signaling Pathway

When the dominant pathway is blocked, cancer cells are able to activate alternative signaling pathways to maintain cell survival and proliferation [72]. Key downstream signaling pathways shared by many receptor and non-receptor tyrosine kinases such as ERK or AKT, can be activated by a variety of RTKs. As a result, when the EGFR pathway is blocked by TKIs, other RTKs, including the IGF signaling pathway, are alternatively upregulated, which directly activate downstream ERK or AKT signaling pathways to compensate for the lack of EGFR signal transduction function. The resistance to TKIs mediated by this process is independent of EGFR [73]. For example, a study has shown that the IGF pathway can activate AKT and phosphorylate PRAS40 (an AKT substrate, 40 kD and rich in proline) to stop its inhibition of mTOR, and then enhance mTOR signaling [27], resulting in continued cell proliferation and resistance to EGFR-TKIs [74]. Another study also found that the increased expression of IGFBP3 can enhance the expression of MET and phosphorylation of ERK by activating the IGF-1R pathway, which causes afatinib resistance in lung adenocarcinoma [66].

In addition, some tyrosine kinases can also be activated by a portion of overexpressed RTKs. It has been shown that overexpression of IGF-1R, EGFR, HER2 and HER3 can activate Src, a non-receptor tyrosine kinase, which has been verified to be a key mediator of trastuzumab resistance in HER2-positive breast cancer [75, 76]. In conclusion, IGF-1R can induce trastuzumab resistance in HER2+ breast cancer by enhancing Src kinase activity [17].

In addition to its IGF-dependent mechanisms, IGFBPs can perform diversified functions through non-IGF-dependent mechanisms [49, 59]. IGFBP3 has been shown to activate the TGF-β-Smad2/3 signaling pathway independently of the IGF pathway [77]. The Met promoter region contains a putative Smad2/3 binding site [78], suggesting that overexpression of IGFBP3 may enhance the expression of Met through the TGFb-Smad2/3 signaling pathway, which may result in part, to the acquired resistance of Met-TKI and EGFR-TKI [65].

IGF-1R can interact with the receptors of the ERBB family, and EGFR and IGF-1R can form heterodimers due to their similar extracellular domain structure [52]. Studies have shown that gefitinib [79] and erlotinib [80] can induce a direct interaction between IGF-1R and EGFR, which promotes the formation of the IGF-1R/EGFR heterodimer, thereby activating the IGF-1R pathway, causing an increase in the expression of survivin, and ultimately, resulting in acquired resistance to EGFR-TKIs. Closer binding of heterodimers results in stronger recovery of downstream signaling pathways, which negatively affects the efficacy of inhibitors [53]. The basal level of IGF-1R expression is important for initiating the formation of this heterodimer [79]. A study also found that IGF-2 is overexpressed in Herceptin-resistant breast cancer cells [81]. As a result, a large number of IGF-2 binds to IGF-1R to facilitate IGF-1R phosphorylation and activation, resulting in changes in its configuration, which promotes the interaction between activated IGF-1R and HER2 to form a heterodimer on the surface of the cell and accelerate its phosphorylation, activate its tyrosine kinase activity and downstream signaling pathways, and ultimately affect the sensitivity of cells to trastuzumab. Similarly, the heterodimerization also leads to gefitinib resistance in colon cancer cells [82]. Other than heterodimer, HER2/HER3/IGF-1R can also form heterotrimer [83], resulting in enhanced activity of downstream PI3K/Akt, Src kinase and other signaling pathways.

4.2 Promote the Activation of EMT and Integrin Pathways

It has been demonstrated that EMT is closely related to tumor metastasis and drug resistance [84]. The IGF system can regulate the EMT process [85]. Several studies have verified that activation of the IGF pathway may facilitate drug resistance by promoting EMT. For example, a study found that in PC-9/GR (gefitinib resistance) cells and H460/ER (erlotinib resistance) cells, IGF-1R can upregulate the expression of Snail by activating the ERK/AKT pathway (Snail is the main regulator of EMT [85]), which promotes the transport of β-catenin from the cell membrane to the nucleus, thereby resulting in direct inhibition of the expression of E-cadherin (a cell junction-related protein) and enhanced EMT in NSCLC cells [86]. Another study has shown that carcinoma-associated fibroblasts (CAFs) can overexpress IGF-1 and hepatocyte growth factor (HGF) through paracrine mechanisms, and activate the corresponding receptors IGF-1R and C-MET respectively, which increase the expression and phosphorylation of annexin A2 (ANXA2), and ultimately improve EMT and EGFR-TKIs resistance in vitro and in vivo [87]. This mechanism was also found in cholangiocarcinoma and gastric cancers, and mediated EGFR-TKIs resistance in these tumors [88, 89, 90].

IGFBP2 has been found to be involved in integrin pathways [91, 92]. Integrins mediate cell adhesion and transmit mechanical and chemical signals to the interior of the cell to drive multiple stem cell functions, including tumor initiation, epithelial plasticity, metastatic reactivation, and resistance to therapies targeting oncogene and immune checkpoint. In tumor tissues, various mechanisms can deregulate integrin signaling, allowing tumor cells to proliferate unchecked, invade across tissue boundaries, and survive in heterogeneous microenvironments [93]. IGFBP2 contains Gly-Arg-Asp (RGD) and heparin-binding sequences, which directly bind integrins and extracellular matrix to trigger biological behaviors independent of IGFs, that promote tumor progression through activation of integrin pathways [94, 95]. Focal adhesion kinase (FAK) is an important member of integrin-mediated signal transduction [96]. A study has shown that overexpression of IGFBP2 promotes FAK phosphorylation, leading to dasatinib resistance in NSCLC cells [60].

4.3 Regulation of the Cell Cycle and Apoptosis

Evidence accumulated in recent years indicates that, in addition to typical cell-surface localization pattern and ligand-activated mechanism of action, IGF-1R is present in the cell nucleus of both normal and cancer cells [97, 98, 99]. Nuclear translocation of IGF-1R is attained via proteosomal, lysosomal and endocytic pathways, and importin-β is a key factor in nuclear mobilization [100, 101]. Nuclear IGF-1R exhibits a number of biological activities similar to those of typical transcription factors, and regulates cellular metabolism, tumor growth, metastasis, and resistance to anti-cancer therapies [101]. Guerard et al. [102] demonstrated that gefitinib induces IGF-1R internalization and the intracellular redistribution of amphiregulin in mucinous lung adenocarcinoma. This promotes the formation of amphiregulin/IGF-1R/importin-β1 complexes, allowing the nuclear transport of IGF-1R. Nuclear IGF-1R subsequently leads to accumulation of p21Waf1 (a cyclin-dependent kinase inhibitor) and G1 phase arrest, which in turn inhibits gefitinib-induced apoptosis and contributes to gefitinib resistance [102].

In addition to the nuclear IGF-1R pathway, IGF-1R on the cell surface can also activate the AKT-dependent survival pathway through the classic ligand-activated mechanism of action, and participate in the resistance of tumor cells to EGFR-TKIs [102]. It is known that p27 is a cell cycle regulator and it can induce cell cycle arrest by impairing the ability of cyclin E to promote G1-S phase transition, which is mainly regulated by the amount of nuclear p27. It has been shown that TKIs can induce cell cycle arrest by promoting the expression of nuclear p27, thus preventing the continuous proliferation of tumor cells [103]. The IGF pathway can activate AKT, which phosphorylates p27 at T157, resulting in cytoplasmic isolation of p27 and a significant decrease in the number of nuclear p27, ultimately contributing to continued cell proliferation and EGFR-TKIs resistance [104]. Furthermore, PRAS40, a substrate of AKT, can be phosphorylated as a result of the activation of AKT by the activated IGF pathway, and that phosphorylated PRAS40 can interact with forkhead box O3 (FOXO3a) to decrease the transcription of apoptotic genes [28], ultimately leading to continued cell proliferation and EGFR-TKIs resistance [74].

It has also been demonstrated that the overexpressed IGFBP7 can inhibit the activity of pro-apoptotic protein caspase and the expression of B-cell lymphoma-2-like 11 (BIM), thus restraining the apoptosis of tumor cells, and finally, leading to the resistance of EGFR-TKIs in lung cancer [71].

The above reported drug resistance mechanisms are related to the role of the IGF pathway in promoting tumorigenesis and development, promoting EMT, and regulating gene expression with atypical functions of nuclear IGF-1R, which belong to the non-target-dependent resistance mechanism of TKIs. Whether the IGF pathway can regulate the expression of target genes through other mechanisms, and then lead to cell resistance to TKIs through a target-dependent mechanism is still unknown. In addition, the mechanism by which the expression levels of various molecules in the IGF pathway are altered in tumor tissues is another topic worthy of discussion. Since the IGF pathway molecules can regulate the resistance of cells to TKIs in different mechanisms, future studies can focus on whether the factors or diseases that affect the expression levels of IGFBPs and IGF-1/2 in vivo will affect the therapeutic effect of TKIs, so as to help better guide medication and predict efficacy.

5. Therapeutics Targeting the IGF Pathway

The activation of the IGF signaling pathway is related to the acquired resistance to TKIs in cells. As a result, the combination of IGF pathway inhibitors and TKIs has emerged as an effective strategy to prevent drug resistance. A number of studies have demonstrated that blocking the IGF pathway can reverse the resistance of TKIs. Jung-Min Choi et al. [105] demonstrated that in EI24-mediated PC9 resistance to gefitinib, the efficacy of EGFR-TKI could be improved by co-inhibiting the IGF-1R pathway by using the IGF-1R small molecule inhibitor PQ-401 or AG-1024. They also found that AG-1024 enhanced the growth inhibition and pro-apoptotic effect of gefitinib in drug-resistant H1650 cells [42]. In addition, Juan Zhou et al. [86] found that the IC50 of gefitinib and erlotinib in PC-9 derived gefitinib resistant cells and H460 derived erlotinib resistant cells were significantly reduced after silencing IGF-1R expression by siRNA, suggesting that IGF-1R plays an important role in restoring the sensitivity of cells to gefitinib or erlotinib. Browne et al. [106] observed that in trastuzumab-resistant or sensitive HER2+ breast cancer cells (BT474, BT474/Tr, SKBR3, SKBR3/Tr), the efficacy of trastuzumab was improved by using siRNA or IGF-1R TKI NVP-AEW541 to inhibit the expression of IGF-1R. These studies suggest that blocking the IGF pathway may improve the efficacy of TKIs in drug-resistant cells. However, in vivo evidence for this conclusion is lacking. Relevant animal experiments can be designed for future research, including using IGF inhibitors.

These studies have demonstrated that the IGF signaling pathway affects drug resistance of tumor cells. There are three major strategies to design drugs that block the IGF signaling pathway for clinical anti-tumor treatment: receptor blocking, kinase inhibition, and ligand isolation (Table 2, Ref. [107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146]) [147]. Among them, mAb is the most commonly used strategy for receptor blocking in clinical studies. The ideal IGF-1R antagonist mAb, with strong specificity in epitope recognition, does not bind to InsR, which avoids side effects such as insulin resistance and hyperglycemia caused by targeting InsR-B. Nevertheless, IGF-1R mAb does not block the activation of InsR-A [148]. InsR-A is capable of binding to IGF-2 to activate downstream signaling pathways, which is a potential cause of drug resistance to IGF-1R mAb. Currently, commonly used IGF-1R mAbs are: cixutumumab (IMC-A12) and ganitumab (AMG-479). Inhibition of tyrosine kinases is another therapeutic strategy targeting the IGF pathway. Since the major sequences of kinase domains of the IGF system receptors are similar, and the sequences of their ATP-binding pocket are almost absolutely conformed [149], kinase inhibitors can theoretically inhibit virtually all IGF receptors. This suggests the possibility of synthesizing drugs that target both the IGF pathway and the EGFR pathway. Such drugs include linsitinib (OSI-906) and BMS-754807. OSI-906 is a dual IGF-1R/InsR inhibitor. Since kinase inhibitors may impair the action of InsR-B, they may cause adverse effects on normal metabolism. The third strategy is ligand isolation using anti-ligand mAb or recombinant IGFBPs, such as dusigitumab (MEDI-573), and xentuzumab (BI 836845). Compared with the other two anti-IGF pathway strategies, ligand isolation does not result in severe metabolic disorders in preclinical animal models [150].

Table 2.Strategies of targeting IGF pathway.
Drug type Compound Preclinical studies
Monoclonal antibodies Cixutumumab (IMC-A12) [107, 108, 109]
Ganitumab (AMG-479) [110, 111, 112, 113]
Figitumumab (CP-751871) [114, 115, 116, 117]
Dalotuzumab (MK-0646) [118]
Teprotumumab (R1507) [119, 120]
Small molecule inhibitors Linsitinib (OSI-906) [121, 122, 123, 124]
BMS-754807 [122, 125, 126, 127]
Picropodophyllin (AXL1717) [128, 129, 130, 131]
NT157 [132, 133]
AG-1024 [134]
NVP-AEW541 [135, 136, 137]
NVP-ADW742 [138, 139, 140]
GSK1904529A [141, 142, 143]
Ligand neutralization Dusigitumab (MEDI-573) [144]
Xentuzumab (BI 836845) [145, 146]

Many clinical trials have begun to evaluate the efficacy of drugs targeting the IGF pathway (Table 3, Ref. [151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173]). Several clinical trials using combined therapy with cetuximab in the treatment of lung or breast cancer have shown no significant clinical benefit. Notably, combined treatment significantly increases the incidence of adverse hyperglycemic reactions (NCT01232452, NCT00870870, NCT00955305, NCT00684983). Another clinical trial demonstrated that the survival rate of patients with recurrent small cell lung cancer treated with linsitinib (OSI-906) alone was significantly lower than that treated with the chemotherapeutic drug topotecan (NCT01533181) [174]. Furthermore, other clinical studies on OSI-906 have not yet resulted in satisfactory outcomes [151, 152, 175, 176]. A clinical trial on the combination of xentuzumab, a dual anti-IGF-1/2 neutralizing antibody, in breast cancer patients showed that the combination of xentuzumab on the basis of everolimus or exemestane did not improve the PFS of the overall population, resulting in an early cessation of the trial (NCT03659136) [153]. Therefore, IGF-1R mAb treatment is largely ineffective in unselected cancer patients. A few clinical trials have suggested that IGF-1R targeted therapy may be beneficial only in specific cancer patients [154, 177]. Several preclinical studies suggested potential predictive biomarkers, such as BRCA1, β-catenin/TCF activity, BACH1, TMPRSS2-ERG fusion gene, IRS2 copy number gain, InsR-A/IGF-IR overexpression, mutation of BRAF and KRAS [178, 179, 180, 181, 182]. There should be clinical trials to evaluate these biomarkers.

Table 3.Clinical trial of drugs targeting IGF pathway in cancer patients.
Drugs Combination Cancer type Phase Participants Results1 Ref. or trial ID
Dalotuzumab Irinotecan, cetuximab Chemorefractory, KRAS exon 2 mutant colorectal cancer II/III 69 ORR, 5.6%. Median PFS, OS were not statistically significantly different with dalotuzumab alone or placebo. [155]
Dalotuzumab Erlotinib Refractory advanced non-small-cell lung cancer I/II I 20 II, ORR, 2.7%. No significant difference in PFS, OS, compared to erlotinib. [156]
II 75
Dalotuzumab Pemetrexed, cisplatin Untreated non-squamous lung cancer stage IV II 26 PR, 25%. SD, 33.33%. PD, 33.33%. [157]
NCT00799240
Dusigitumab None Advanced solid tumors I 43 CR, 0. PR, 0. SD, 33.33%. [158]
Figitumumab Erlotinib Stage IIIB/IV or recurrent disease with nonadenocarcinoma histology II 583 OS, 5.7 months. Median PFS, 6.2 months. No improvement, compared to erlotinib. [159]
NCT00673049
Figitumumab Paclitaxel, carboplatin Stage IIIB/IV or recurrent NSCLC disease with nonadenocarcinoma histology III 681 OS, 8.6 months. Median PFS, 4.7 months. ORR, 33%. No improvement, compared to paclitaxel, plus carboplatin. [160]
Figitumumab Docetaxel or prednisone Progressing castration-resistant prostate cancer II 204 Median PFS was 4.9 months, with HR = 1.44 (95% CI, 1.06–1.96), compared to docetaxel/prednisone alone. [161]
Ganitumab None Metastatic progressive carcinoid or pancreatic neuroendocrine tumors II 60 Median PFS, 6.3 months (95% CI, 4.2–12.6). OS at 12 months was 66% (95% CI, 52–77%). [162]
Ganitumab Gemcitabine Previously untreated metastatic pancreatic adenocarcinoma II 125 OS at 6 months was 57% (95% CI, 41–70%). [163]
Teprotumumab Erlotinib Progressing advanced-stage non-small-cell lung cancer (NSCLC) II 172 12-week PFS rates were 39%, 37%, and 44%, and median OS was 8.1, 8.1, and 12.1 months for the three groups (erlotinib plus placebo, or R1507 weekly, or R1507 every 3 weeks), respectively. [154]
Teprotumumab None Recurrent or refractory soft tissue sarcomas II 163 ORR, 2.5% (95% CI, 0.7–6.2%). Median PFS, 5.7 weeks. Median OS, 11 months. [164]
Teprotumumab None Refractory or recurrent Ewing sarcomas II 115 CR or PR, 10% (95% CI, 4.9–16.5%). Median OS, 7.6 months (95% CI, 6–9.7 months). [165]
Xentuzumab Afatinib Previously treated EGFR mutation-positive NSCLC I 32 ORR, 0. Median SD, 2.3 months (95% CI, 0.8–10.9 months). [166]
NCT02191891
Xentuzumab Everolimus, exemestane Hormone receptor-positive/HER2-negative locally advanced and/or metastatic breast cancer Ib/II II 140 Median PFS, 7.3 months, with HR = 0.21 (95% CI, 0.05–0.98, p = 0.0293), compared to everolimus plus exemestane. [153]
I, NCT02123823
II, NCT03659136
Linsitinib None Locally advanced or metastatic adrenocortical carcinoma III 139 Median OS, 323 days (95% CI, 256–507 days). No significant difference, compared to placebo. [151]
NCT00924989
Linsitinib None Wild type gastrointestinal stromal tumors II 20 ORR, 0. CBR at 9 months was 40%. 9 months PFS, 52%. 9 months OS, 80%. [152]
Picropodophyllin None Advanced solid tumors Ia/b Ia 20 For 15 NSCLC patients, the median PFS was 31 weeks and the OS was 60 weeks. [172]
Ib 39 NCT01062620
Picropodophyllin None Previously treated, locally advanced or metastatic NSCLC II 99 12-weeks PFS, 29%, ORR, 0. Estimated median OS, 38.7 weeks. No improvement, compared to docetaxel. [167]
Cixutumumab Capecitabine, lapatinib HER2-positive metastatic breast cancer previously treated with trastuzumab II 68 Median PFS, 5.0 months. Median OS, 21.9 months. No improvement, compared to capecitabine plus lapatinib. [168]
NCT00684983
Cixutumumab Bevacizumab Stage IV or recurrent non-squamous, NSCLC II 175 Median PFS, 7.0 months. ORR, 58.7%. Median OS, 16.1 months. The addition of cixutumumab increased toxicity without improving efficacy. [169]
NCT00955305
Cixutumumab Doxorubicin Advanced soft tissue sarcoma I 30 Estimated PFS, 5.3 months (95% CI, 3.0–6.3 months). [173]
NCT00720174
Cixutumumab Androgen deprivation (AD) New metastatic hormone-sensitive prostate cancer II 210 NO statistical difference between AD and AD plus Cixutumumab in OS or PFS. [171]
Cixutumumab Cetuximab Recurrent/metastatic head and neck squamous cell carcinoma II 91 Median PFS, 2.0 months. CBR, 15.3%. No improvement, compared to cetuximab alone. [170]

1 ORR, objective response rate; CBR, clinical benefit rate; PFS, progression-free survival; OS, overall survival; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; HR, hazard ratio; CI, confidence interval; Trial ID registered number at https://ClinicalTrials.gov

Though the anticancer efficacy of the drugs targeting the IGF pathway is promising in preclinical models, the results of clinical trials have been disappointing. Up to now, there has not been any anti-IGF-1R mAb approved as a cancer drug treatment by the FDA. The following three issues need to be addressed when considering targeting the IGF pathway as a therapeutic strategy. (1) The poor response of selective IGF-1R targeting drugs may be primarily due to the compensation of the activated InsR pathway or the reduction of cellular IGF-1R [51]. As a consequence, inhibition of InsR appears to be imperative, and future research will focus on developing InsR inhibitors that target cancer cells. (2) There is a lack of predictive biomarkers to select patients who will benefit from targeted therapy. Therefore, a large number of marker studies are needed to confirm the appropriate screening method for selecting patients suitable for IGF pathway-targeted therapy. (3) Drugs can cause adverse effects on normal metabolism by impairing the physiological action of InsR, leading to side effects such as metabolic disorders and hyperglycemia. Therefore, when targeting IGF-1R or InsR to treat cancers, alterations in InsR-B function should be avoided. When these issues are resolved, targeting InsR-A may become a promising strategy to exert antitumor effects without causing severe metabolic side effects.

6. Conclusions and Perspectives

TKIs can interfere with the signal transduction of tumor cells, and inhibit the proliferation of tumor cells and neovascularization, with little effect on normal cells [2], making it a promising clinical therapy for cancer patients. A growing number of TKI are being approved by the FDA to treat tumors. The most widely and successful used TKIs are EGFR-TKIs and HER2-TKIs, which are mainly used in the targeted therapy of lung and breast cancers. Although the application of TKIs has brought great hope to a large number of patients, most will inevitably develop drug resistance. The resistance mechanisms of TKIs are complicated, which can be divided into target-dependent resistance mechanisms and non-target-dependent resistance mechanisms. It has been shown that the activation of the IGF signaling pathway can impact the sensitivity of cells to TKIs. The IGF pathway contributes to acquired resistance to TKIs primarily by promoting the activation of compensatory signaling pathways, promoting the activation of EMT and integrin pathways, and regulating the cell cycle and apoptosis. A variety of drugs targeting the IGF pathway have been designed for cancer therapy. Although the anticancer efficacy of the drugs targeting the IGF pathway is promising in preclinical models, the results of clinical trials have been disappointing, limiting the current clinical application of IGF pathway inhibitors.

In order to address drug resistance to EGFR-TKIs caused by the IGF signaling pathway, simultaneously targeting the IGF pathway and EGFR may be an effective strategy. One option is to combine drugs to augment the response time of targeted agents, such as the combination of IGF pathway inhibitors and TKIs for cancer treatment. In addition, single-molecule drugs that inhibit multiple correlative kinases can be developed, such as small molecule inhibitors that target both the IGF and EGFR pathways, to improve efficacy and reduce toxicity. This will require more in-depth basic and clinical research studies.

Author Contributions

YP wrote the original draft, draw the figures, and acquired, initially analyzed, and visualized data from the reviewed literature. JT edited the review, conceptualized and interpreted the data from the reviewed literature. Both authors have read and agreed to the published version of the manuscript. Both authors contributed to editorial changes in the manuscript. Both authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

The author thanks current members of the laboratory for their valuable contributions.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021; 71: 209–249.
[2]
Qing-yuan J, Lei-fu Y, Li-ming H. Small molecule tyrosine kinase inhibitors and their application in cancer treatment: research advances. Journal of Pharmaceutical Research International. 2020; 47: 1047–1056.
[3]
Martínez-Jiménez F, Muiños F, Sentís I, Deu-Pons J, Reyes-Salazar I, Arnedo-Pac C, et al. A compendium of mutational cancer driver genes. Nature Reviews. Cancer. 2020; 20: 555–572.
[4]
Roskoski R, Jr. A historical overview of protein kinases and their targeted small molecule inhibitors. Pharmacological Research. 2015; 100: 1–23.
[5]
Kris MG, Johnson BE, Berry LD, Kwiatkowski DJ, Iafrate AJ, Wistuba II, et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA. 2014; 311: 1998–2006.
[6]
Khorashad JS, Deininger MWN. Selection of therapy: rational decisions based on molecular events. Hematology/oncology Clinics of North America. 2011; 25: 1009–1023, vi.
[7]
Cohen P, Cross D, Jänne PA. Kinase drug discovery 20 years after imatinib: progress and future directions. Nature Reviews. Drug Discovery. 2021; 20: 551–569.
[8]
Liu L, Li X. A Review of IGF1 Signaling and IGF1-related Long Noncoding RNAs in Chemoresistance of Cancer. Current Cancer Drug Targets. 2020; 20: 325–334.
[9]
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. Journal of Hematology & Oncology. 2020; 13: 64.
[10]
Rosenzweig SA. Acquired Resistance to Drugs Targeting Tyrosine Kinases. Advances in Cancer Research. 2018; 138: 71–98.
[11]
Roskoski R, Jr. Properties of FDA-approved small molecule protein kinase inhibitors: A 2022 update. Pharmacological Research. 2022; 175: 106037.
[12]
Attwood MM, Fabbro D, Sokolov AV, Knapp S, Schiöth HB. Trends in kinase drug discovery: targets, indications and inhibitor design. Nature Reviews. Drug Discovery. 2021; 20: 839–861.
[13]
Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Critical Reviews in Oncology/hematology. 1995; 19: 183–232.
[14]
Wang Z. ErbB Receptors and Cancer. Methods in Molecular Biology (Clifton, N.J.). 2017; 1652: 3–35.
[15]
Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. The New England Journal of Medicine. 2008; 358: 1160–1174.
[16]
Roskoski R, Jr. Small molecule inhibitors targeting the EGFR/ErbB family of protein-tyrosine kinases in human cancers. Pharmacological Research. 2019; 139: 395–411.
[17]
Lyu H, Yang XH, Edgerton SM, Thor AD, Wu X, He Z, et al. The erbB3- and IGF-1 receptor-initiated signaling pathways exhibit distinct effects on lapatinib sensitivity against trastuzumab-resistant breast cancer cells. Oncotarget. 2016; 7: 2921–2935.
[18]
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA: A Cancer Journal for Clinicians. 2020; 70: 7–30.
[19]
Testa U, Castelli G, Pelosi E. Lung Cancers: Molecular Characterization, Clonal Heterogeneity and Evolution, and Cancer Stem Cells. Cancers. 2018; 10: 248.
[20]
Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. The New England Journal of Medicine. 2009; 361: 958–967.
[21]
Tan DSW, Yom SS, Tsao MS, Pass HI, Kelly K, Peled N, et al. The International Association for the Study of Lung Cancer Consensus Statement on Optimizing Management of EGFR Mutation-Positive Non-Small Cell Lung Cancer: Status in 2016. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer. 2016; 11: 946–963.
[22]
Chen P, Liu Y, Wen Y, Zhou C. Non-small cell lung cancer in China. Cancer Communications (London, England). 2022; 42: 937–970.
[23]
Huang L, Fu L. Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharmaceutica Sinica. B. 2015; 5: 390–401.
[24]
Kazandjian D, Blumenthal GM, Yuan W, He K, Keegan P, Pazdur R. FDA Approval of Gefitinib for the Treatment of Patients with Metastatic EGFR Mutation-Positive Non-Small Cell Lung Cancer. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2016; 22: 1307–1312.
[25]
He J, Huang Z, Han L, Gong Y, Xie C. Mechanisms and management of 3rd generation EGFR TKI resistance in advanced non small cell lung cancer (Review). International Journal of Oncology. 2021; 59: 90.
[26]
Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2018; 29: i10–i19.
[27]
Passaro A, Guerini-Rocco E, Pochesci A, Vacirca D, Spitaleri G, Catania CM, et al. Targeting EGFR T790M mutation in NSCLC: From biology to evaluation and treatment. Pharmacological Research. 2017; 117: 406–415.
[28]
Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. The New England Journal of Medicine. 2017; 376: 629–640.
[29]
Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. The New England Journal of Medicine. 2018; 378: 113–125.
[30]
Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nature Medicine. 2015; 21: 560–562.
[31]
Luque-Bolivar A, Pérez-Mora E, Villegas VE, Rondón-Lagos M. Resistance and Overcoming Resistance in Breast Cancer. Breast Cancer (Dove Medical Press). 2020; 12: 211–229.
[32]
Waks AG, Winer EP. Breast Cancer Treatment: A Review. JAMA. 2019; 321: 288–300.
[33]
Vernieri C, Milano M, Brambilla M, Mennitto A, Maggi C, Cona MS, et al. Resistance mechanisms to anti-HER2 therapies in HER2-positive breast cancer: Current knowledge, new research directions and therapeutic perspectives. Critical Reviews in Oncology/Hematology. 2019; 139: 53–66.
[34]
Junttila TT, Akita RW, Parsons K, Fields C, Lewis Phillips GD, Friedman LS, et al. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 2009; 15: 429–440.
[35]
Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. The New England Journal of Medicine. 2006; 355: 2733–2743.
[36]
Swain SM, Baselga J, Kim SB, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. The New England Journal of Medicine. 2015; 372: 724–734.
[37]
Esparís-Ogando A, Montero JC, Arribas J, Ocaña A, Pandiella A. Targeting the EGF/HER Ligand-Receptor System in Cancer. Current Pharmaceutical Design. 2016; 22: 5887–5898.
[38]
Zhao Y, Wang H, He C. Drug resistance of targeted therapy for advanced non-small cell lung cancer harbored EGFR mutation: from mechanism analysis to clinical strategy. Journal of Cancer Research and Clinical Oncology. 2021; 147: 3653–3664.
[39]
Nahta R, Yu D, Hung MC, Hortobagyi GN, Esteva FJ. Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer. Nature Clinical Practice. Oncology. 2006; 3: 269–280.
[40]
Xu J, Wang J, Zhang S. Mechanisms of resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors and therapeutic strategies in non-small cell lung cancer. Oncotarget. 2017; 8: 90557–90578.
[41]
Cooper AJ, Sequist LV, Lin JJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nature Reviews. Clinical Oncology. 2022; 19: 499–514.
[42]
Choi YJ, Rho JK, Jeon BS, Choi SJ, Park SC, Lee SS, et al. Combined inhibition of IGFR enhances the effects of gefitinib in H1650: a lung cancer cell line with EGFR mutation and primary resistance to EGFR-TK inhibitors. Cancer Chemotherapy and Pharmacology. 2010; 66: 381–388.
[43]
Bodzin AS, Wei Z, Hurtt R, Gu T, Doria C. Gefitinib resistance in HCC mahlavu cells: upregulation of CD133 expression, activation of IGF-1R signaling pathway, and enhancement of IGF-1R nuclear translocation. Journal of Cellular Physiology. 2012; 227: 2947–2952.
[44]
Löw S, Vougioukas VI, Hielscher T, Schmidt U, Unterberg A, Halatsch ME. Pathogenetic pathways leading to glioblastoma multiforme: association between gene expressions and resistance to erlotinib. Anticancer Research. 2008; 28: 3729–3732.
[45]
Masago K, Fujita S, Togashi Y, Kim YH, Hatachi Y, Fukuhara A, et al. Clinical significance of epidermal growth factor receptor mutations and insulin-like growth factor 1 and its binding protein 3 in advanced non-squamous non-small cell lung cancer. Oncology Reports. 2011; 26: 795–803.
[46]
Peled N, Wynes MW, Ikeda N, Ohira T, Yoshida K, Qian J, et al. Insulin-like growth factor-1 receptor (IGF-1R) as a biomarker for resistance to the tyrosine kinase inhibitor gefitinib in non-small cell lung cancer. Cellular Oncology (Dordrecht). 2013; 36: 277–288.
[47]
Chang HP, Yang SF, Wang SL, Su PH. Associations among IGF-1, IGF2, IGF-1R, IGF-2R, IGFBP-3, insulin genetic polymorphisms and central precocious puberty in girls. BMC Endocrine Disorders. 2018; 18: 66.
[48]
Bruchim I, Sarfstein R, Werner H. The IGF Hormonal Network in Endometrial Cancer: Functions, Regulation, and Targeting Approaches. Frontiers in Endocrinology. 2014; 5: 76.
[49]
Jones JI, Clemmons DR. Insulin-like growth factors and their binding proteins: biological actions. Endocrine Reviews. 1995; 16: 3–34.
[50]
Jehle PM, Schulten K, Schulz W, Jehle DR, Stracke S, Manfras B, et al. Serum levels of insulin-like growth factor (IGF)-I and IGF binding protein (IGFBP)-1 to -6 and their relationship to bone metabolism in osteoporosis patients. European Journal of Internal Medicine. 2003; 14: 32–38.
[51]
Cao J, Yee D. Disrupting Insulin and IGF Receptor Function in Cancer. International Journal of Molecular Sciences. 2021; 22: 555.
[52]
Garrett TP, McKern NM, Lou M, Frenkel MJ, Bentley JD, Lovrecz GO, et al. Crystal structure of the first three domains of the type-1 insulin-like growth factor receptor. Nature. 1998; 394: 395–399.
[53]
Wang DD, Ma L, Wong MP, Lee VHF, Yan H. Contribution of EGFR and ErbB-3 Heterodimerization to the EGFR Mutation-Induced Gefitinib- and Erlotinib-Resistance in Non-Small-Cell Lung Carcinoma Treatments. PLoS ONE. 2015; 10: e0128360.
[54]
Beckwith H, Yee D. Insulin-like growth factors, insulin, and growth hormone signaling in breast cancer: implications for targeted therapy. Endocrine Practice: Official Journal of the American College of Endocrinology and the American Association of Clinical Endocrinologists. 2014; 20: 1214–1221.
[55]
Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, et al. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocrine Reviews. 2017; 38: 379–431.
[56]
Kido Y, Nakae J, Accili D. Clinical review 125: The insulin receptor and its cellular targets. The Journal of Clinical Endocrinology and Metabolism. 2001; 86: 972–979.
[57]
Maris C, D’Haene N, Trépant AL, Le Mercier M, Sauvage S, Allard J, et al. IGF-IR: a new prognostic biomarker for human glioblastoma. British Journal of Cancer. 2015; 113: 729–737.
[58]
Kornfeld S. Structure and function of the mannose 6-phosphate/insulinlike growth factor II receptors. Annual Review of Biochemistry. 1992; 61: 307–330.
[59]
Kelley KM, Oh Y, Gargosky SE, Gucev Z, Matsumoto T, Hwa V, et al. Insulin-like growth factor-binding proteins (IGFBPs) and their regulatory dynamics. The International Journal of Biochemistry & Cell Biology. 1996; 28: 619–637.
[60]
Lu H, Wang L, Gao W, Meng J, Dai B, Wu S, et al. IGFBP2/FAK pathway is causally associated with dasatinib resistance in non-small cell lung cancer cells. Molecular Cancer Therapeutics. 2013; 12: 2864–2873.
[61]
Probst-Hensch NM, Steiner JHB, Schraml P, Varga Z, Zürrer-Härdi U, Storz M, et al. IGFBP2 and IGFBP3 protein expressions in human breast cancer: association with hormonal factors and obesity. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2010; 16: 1025–1032.
[62]
Mishra L, Bass B, Ooi BS, Sidawy A, Korman L. Role of insulin-like growth factor-I (IGF-I) receptor, IGF-I, and IGF binding protein-2 in human colorectal cancers. Growth Hormone & IGF Research: Official Journal of the Growth Hormone Research Society and the International IGF Research Society. 1998; 8: 473–479.
[63]
Zhang L, Huang W, Chen J, Zhou X, Lu Z, Zhou H. Expression of IGFBP2 in gastric carcinoma and relationship with clinicopathologic parameters and cell proliferation. Digestive Diseases and Sciences. 2007; 52: 248–253.
[64]
He Y, Zhou Z, Hofstetter WL, Zhou Y, Hu W, Guo C, et al. Aberrant expression of proteins involved in signal transduction and DNA repair pathways in lung cancer and their association with clinical parameters. PLoS ONE. 2012; 7: e31087.
[65]
Yamaoka T, Ohmori T, Ohba M, Arata S, Kishino Y, Murata Y, et al. Acquired Resistance Mechanisms to Combination Met-TKI/EGFR-TKI Exposure in Met-Amplified EGFR-TKI-Resistant Lung Adenocarcinoma Harboring an Activating EGFR Mutation. Molecular Cancer Therapeutics. 2016; 15: 3040–3054.
[66]
Yamaoka T, Ohmori T, Ohba M, Arata S, Murata Y, Kusumoto S, et al. Distinct Afatinib Resistance Mechanisms Identified in Lung Adenocarcinoma Harboring an EGFR Mutation. Molecular Cancer Research: MCR. 2017; 15: 915–928.
[67]
Lee HY, Chun KH, Liu B, Wiehle SA, Cristiano RJ, Hong WK, et al. Insulin-like growth factor binding protein-3 inhibits the growth of non-small cell lung cancer. Cancer Research. 2002; 62: 3530–3537.
[68]
Hayakawa D, Takahashi F, Mitsuishi Y, Tajima K, Hidayat M, Winardi W, et al. Activation of insulin-like growth factor-1 receptor confers acquired resistance to osimertinib in non-small cell lung cancer with EGFR T790M mutation. Thoracic Cancer. 2020; 11: 140–149.
[69]
MacFarland SP, Naraparaju K, Iyer R, Guan P, Kolla V, Hu Y, et al. Mechanisms of Entrectinib Resistance in a Neuroblastoma Xenograft Model. Molecular Cancer Therapeutics. 2020; 19: 920–926.
[70]
Evdokimova V, Tognon CE, Benatar T, Yang W, Krutikov K, Pollak M, et al. IGFBP7 binds to the IGF-1 receptor and blocks its activation by insulin-like growth factors. Science Signaling. 2012; 5: ra92.
[71]
Wu SG, Chang TH, Tsai MF, Liu YN, Hsu CL, Chang YL, et al. IGFBP7 Drives Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibition in Lung Cancer. Cancers. 2019; 11: 36.
[72]
Jones HE, Gee JMW, Hutcheson IR, Knowlden JM, Barrow D, Nicholson RI. Growth factor receptor interplay and resistance in cancer. Endocrine-related Cancer. 2006; 13 Suppl 1: S45–S51.
[73]
Zhou J, Ji Q, Li Q. Resistance to anti-EGFR therapies in metastatic colorectal cancer: underlying mechanisms and reversal strategies. Journal of Experimental & Clinical Cancer Research: CR. 2021; 40: 328.
[74]
Dougherty MI, Lehman CE, Spencer A, Mendez RE, David AP, Taniguchi LE, et al. PRAS40 Phosphorylation Correlates with Insulin-Like Growth Factor-1 Receptor-Induced Resistance to Epidermal Growth Factor Receptor Inhibition in Head and Neck Cancer Cells. Molecular Cancer Research: MCR. 2020; 18: 1392–1401.
[75]
Muthuswamy SK. Trastuzumab resistance: all roads lead to SRC. Nature Medicine. 2011; 17: 416–418.
[76]
Zhang S, Huang WC, Li P, Guo H, Poh SB, Brady SW, et al. Combating trastuzumab resistance by targeting SRC, a common node downstream of multiple resistance pathways. Nature Medicine. 2011; 17: 461–469.
[77]
Fanayan S, Firth SM, Baxter RC. Signaling through the Smad pathway by insulin-like growth factor-binding protein-3 in breast cancer cells. Relationship to transforming growth factor-beta 1 signaling. The Journal of Biological Chemistry. 2002; 277: 7255–7261.
[78]
Kajihara I, Jinnin M, Makino T, Masuguchi S, Sakai K, Fukushima S, et al. Overexpression of hepatocyte growth factor receptor in scleroderma dermal fibroblasts is caused by autocrine transforming growth factor β signaling. Bioscience Trends. 2012; 6: 136–142.
[79]
Morgillo F, Kim WY, Kim ES, Ciardiello F, Hong WK, Lee HY. Implication of the insulin-like growth factor-IR pathway in the resistance of non-small cell lung cancer cells to treatment with gefitinib. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2007; 13: 2795–2803.
[80]
Morgillo F, Woo JK, Kim ES, Hong WK, Lee HY. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Research. 2006; 66: 10100–10111.
[81]
Zhang M, Li Z, Liu X. MiR-98-5p/IGF2 Axis Influence Herceptin Sensitivity through IGF1R/HER2 Heterodimer Formation and AKT/mTOR Signal Pathway in HER2 Positive Breast Cancer. Asian Pacific Journal of Cancer Prevention: APJCP. 2021; 22: 3693–3703.
[82]
Yang L, Li J, Ran L, Pan F, Zhao X, Ding Z, et al. Phosphorylated insulin-like growth factor 1 receptor is implicated in resistance to the cytostatic effect of gefitinib in colorectal cancer cells. Journal of Gastrointestinal Surgery: Official Journal of the Society for Surgery of the Alimentary Tract. 2011; 15: 942–957.
[83]
Huang X, Gao L, Wang S, McManaman JL, Thor AD, Yang X, et al. Heterotrimerization of the growth factor receptors erbB2, erbB3, and insulin-like growth factor-i receptor in breast cancer cells resistant to herceptin. Cancer Research. 2010; 70: 1204–1214.
[84]
Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nature Reviews. Molecular Cell Biology. 2019; 20: 69–84.
[85]
Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nature Reviews. Molecular Cell Biology. 2014; 15: 178–196.
[86]
Zhou J, Wang J, Zeng Y, Zhang X, Hu Q, Zheng J, et al. Implication of epithelial-mesenchymal transition in IGF1R-induced resistance to EGFR-TKIs in advanced non-small cell lung cancer. Oncotarget. 2015; 6: 44332–44345.
[87]
Yi Y, Zeng S, Wang Z, Wu M, Ma Y, Ye X, et al. Cancer-associated fibroblasts promote epithelial-mesenchymal transition and EGFR-TKI resistance of non-small cell lung cancers via HGF/IGF-1/ANXA2 signaling. Biochimica et Biophysica Acta. Molecular Basis of Disease. 2018; 1864: 793–803.
[88]
Vaquero J, Lobe C, Clapéron A, Mergey M, Desbois-Mouthon C, Praz F, et al. Insulin/insulin-like growth factor-1 receptors mediate acquired resistance to anti-EGFR therapy in human cholangiocarcinoma cells by regulating an epithelial to mesenchymal transition/cancer stem cell axis. European Journal of Cancer. 2016; 61: S131.
[89]
Vaquero J, Lobe C, Tahraoui S, Clapéron A, Mergey M, Merabtene F, et al. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2018; 24: 4282–4296.
[90]
Zhang Z, Wang J, Ji D, Wang C, Liu R, Wu Z, et al. Functional genetic approach identifies MET, HER3, IGF1R, INSR pathways as determinants of lapatinib unresponsiveness in HER2-positive gastric cancer. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2014; 20: 4559–4573.
[91]
Schütt BS, Langkamp M, Rauschnabel U, Ranke MB, Elmlinger MW. Integrin-mediated action of insulin-like growth factor binding protein-2 in tumor cells. Journal of Molecular Endocrinology. 2004; 32: 859–868.
[92]
Wang GK, Hu L, Fuller GN, Zhang W. An interaction between insulin-like growth factor-binding protein 2 (IGFBP2) and integrin alpha5 is essential for IGFBP2-induced cell mobility. The Journal of Biological Chemistry. 2006; 281: 14085–14091.
[93]
Cooper J, Giancotti FG. Integrin Signaling in Cancer: Mechanotransduction, Stemness, Epithelial Plasticity, and Therapeutic Resistance. Cancer Cell. 2019; 35: 347–367.
[94]
Kawai M, Breggia AC, DeMambro VE, Shen X, Canalis E, Bouxsein ML, et al. The heparin-binding domain of IGFBP-2 has insulin-like growth factor binding-independent biologic activity in the growing skeleton. The Journal of Biological Chemistry. 2011; 286: 14670–14680.
[95]
Russo VC, Schütt BS, Andaloro E, Ymer SI, Hoeflich A, Ranke MB, et al. Insulin-like growth factor binding protein-2 binding to extracellular matrix plays a critical role in neuroblastoma cell proliferation, migration, and invasion. Endocrinology. 2005; 146: 4445–4455.
[96]
Sieg DJ, Hauck CR, Ilic D, Klingbeil CK, Schaefer E, Damsky CH, et al. FAK integrates growth-factor and integrin signals to promote cell migration. Nature Cell Biology. 2000; 2: 249–256.
[97]
Chen CW, Oberley TD, Roy D. Inhibition of stilbene estrogen-induced cell proliferation of renal epithelial cells through the modulation of insulin-like growth factor-I receptor expression. Cancer Letters. 1996; 105: 51–59.
[98]
Sun H, Tu X, Prisco M, Wu A, Casiburi I, Baserga R. Insulin-like growth factor I receptor signaling and nuclear translocation of insulin receptor substrates 1 and 2. Molecular Endocrinology (Baltimore, Md.). 2003; 17: 472–486.
[99]
Sarfstein R, Belfiore A, Werner H. Identification of Insulin-Like Growth Factor-I Receptor (IGF-IR) Gene Promoter-Binding Proteins in Estrogen Receptor (ER)-Positive and ER-Depleted Breast Cancer Cells. Cancers. 2010; 2: 233–261.
[100]
Vecchione A, Marchese A, Henry P, Rotin D, Morrione A. The Grb10/Nedd4 complex regulates ligand-induced ubiquitination and stability of the insulin-like growth factor I receptor. Molecular and Cellular Biology. 2003; 23: 3363–3372.
[101]
Werner H, Sarfstein R, Laron Z. The Role of Nuclear Insulin and IGF1 Receptors in Metabolism and Cancer. Biomolecules. 2021; 11: 531.
[102]
Guerard M, Robin T, Perron P, Hatat AS, David-Boudet L, Vanwonterghem L, et al. Nuclear translocation of IGF1R by intracellular amphiregulin contributes to the resistance of lung tumour cells to EGFR-TKI. Cancer Letters. 2018; 420: 146–155.
[103]
Di Gennaro E, Barbarino M, Bruzzese F, De Lorenzo S, Caraglia M, Abbruzzese A, et al. Critical role of both p27KIP1 and p21CIP1/WAF1 in the antiproliferative effect of ZD1839 (’Iressa’), an epidermal growth factor receptor tyrosine kinase inhibitor, in head and neck squamous carcinoma cells. Journal of Cellular Physiology. 2003; 195: 139–150.
[104]
Jameson MJ, Taniguchi LE, VanKoevering KK, Stuart MM, Francom CR, Mendez RE, et al. Activation of the insulin-like growth factor-1 receptor alters p27 regulation by the epidermal growth factor receptor in oral squamous carcinoma cells. Journal of Oral Pathology & Medicine: Official Publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology. 2013; 42: 332–338.
[105]
Choi JM, Jang JY, Choi YR, Kim HR, Cho BC, Lee HW. Reduced expression of EI24 confers resistance to gefitinib through IGF-1R signaling in PC9 NSCLC cells. Lung Cancer (Amsterdam, Netherlands). 2015; 90: 175–181.
[106]
Browne BC, Crown J, Venkatesan N, Duffy MJ, Clynes M, Slamon D, et al. Inhibition of IGF1R activity enhances response to trastuzumab in HER-2-positive breast cancer cells. Annals of Oncology. 2011; 22: 68–73.
[107]
Zamykal M, Martens T, Matschke J, Günther HS, Kathagen A, Schulte A, et al. Inhibition of intracerebral glioblastoma growth by targeting the insulin-like growth factor 1 receptor involves different context-dependent mechanisms. Neuro-oncology. 2015; 17: 1076–1085.
[108]
Riesterer O, Yang Q, Raju U, Torres M, Molkentine D, Patel N, et al. Combination of anti-IGF-1R antibody A12 and ionizing radiation in upper respiratory tract cancers. International Journal of Radiation Oncology, Biology, Physics. 2011; 79: 1179–1187.
[109]
Attias-Geva Z, Bentov I, Ludwig DL, Fishman A, Bruchim I, Werner H. Insulin-like growth factor-I receptor (IGF-IR) targeting with monoclonal antibody cixutumumab (IMC-A12) inhibits IGF-I action in endometrial cancer cells. European Journal of Cancer. 2011; 47: 1717–1726.
[110]
Mendivil A, Zhou C, Cantrell LA, Gehrig PA, Malloy KM, Blok LJ, et al. AMG 479, a novel IGF-1-R antibody, inhibits endometrial cancer cell proliferation through disruption of the PI3K/Akt and MAPK pathways. Reproductive Sciences. 2011; 18: 832–841.
[111]
Beltran PJ, Mitchell P, Chung YA, Cajulis E, Lu J, Belmontes B, et al. AMG 479, a fully human anti-insulin-like growth factor receptor type I monoclonal antibody, inhibits the growth and survival of pancreatic carcinoma cells. Molecular Cancer Therapeutics. 2009; 8: 1095–1105.
[112]
Beltran PJ, Chung YA, Moody G, Mitchell P, Cajulis E, Vonderfecht S, et al. Efficacy of ganitumab (AMG 479), alone and in combination with rapamycin, in Ewing’s and osteogenic sarcoma models. The Journal of Pharmacology and Experimental Therapeutics. 2011; 337: 644–654.
[113]
Beltran PJ, Calzone FJ, Mitchell P, Chung YA, Cajulis E, Moody G, et al. Ganitumab (AMG 479) inhibits IGF-II-dependent ovarian cancer growth and potentiates platinum-based chemotherapy. Clinical Cancer Research. 2014; 20: 2947–2958.
[114]
Pavlicek A, Lira ME, Lee NV, Ching KA, Ye J, Cao J, et al. Molecular predictors of sensitivity to the insulin-like growth factor 1 receptor inhibitor Figitumumab (CP-751,871). Molecular Cancer Therapeutics. 2013; 12: 2929–2939.
[115]
Juergens H, Daw NC, Geoerger B, Ferrari S, Villarroel M, Aerts I, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. Journal of Clinical Oncology. 2011; 29: 4534–4540.
[116]
Ii M, Li H, Adachi Y, Yamamoto H, Ohashi H, Taniguchi H, et al. The efficacy of IGF-I receptor monoclonal antibody against human gastrointestinal carcinomas is independent of k-ras mutation status. Clinical Cancer Research. 2011; 17: 5048–5059.
[117]
Chakraborty AK, Zerillo C, DiGiovanna MP. In vitro and in vivo studies of the combination of IGF1R inhibitor figitumumab (CP-751,871) with HER2 inhibitors trastuzumab and neratinib. Breast Cancer Research and Treatment. 2015; 152: 533–544.
[118]
Becker MA, Hou X, Tienchaianada P, Haines BB, Harrington SC, Weroha SJ, et al. Ridaforolimus (MK-8669) synergizes with Dalotuzumab (MK-0646) in hormone-sensitive breast cancer. BMC Cancer. 2016; 16: 814.
[119]
Kolb EA, Kamara D, Zhang W, Lin J, Hingorani P, Baker L, et al. R1507, a fully human monoclonal antibody targeting IGF-1R, is effective alone and in combination with rapamycin in inhibiting growth of osteosarcoma xenografts. Pediatric Blood & Cancer. 2010; 55: 67–75.
[120]
Huang HJ, Angelo LS, Rodon J, Sun M, Kuenkele KP, Parsons HA, et al. R1507, an anti-insulin-like growth factor-1 receptor (IGF-1R) antibody, and EWS/FLI-1 siRNA in Ewing’s sarcoma: convergence at the IGF/IGFR/Akt axis. PLoS ONE. 2011; 6: e26060.
[121]
Kuhn DJ, Berkova Z, Jones RJ, Woessner R, Bjorklund CC, Ma W, et al. Targeting the insulin-like growth factor-1 receptor to overcome bortezomib resistance in preclinical models of multiple myeloma. Blood. 2012; 120: 3260–3270.
[122]
Fuentes-Baile M, Ventero MP, Encinar JA, García-Morales P, Poveda-Deltell M, Pérez-Valenciano E, 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.
[123]
Liu L, Liang C, Zhuo C, Jiang H, Ye H, Ruan T, et al. OSI-906 restores the sensitivity of ovarian clear cell carcinoma to cisplatin by targeting the IGF1R/AKT pathway. Medical Oncology. 2022; 39: 26.
[124]
Wu Q, Tian AL, Kroemer G, Kepp O. Autophagy induction by IGF1R inhibition with picropodophyllin and linsitinib. Autophagy. 2021; 17: 2046–2047.
[125]
Hou X, Huang F, Macedo LF, Harrington SC, Reeves KA, Greer A, et al. Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hormonal agents in treatment of estrogen-dependent breast cancer. Cancer Research. 2011; 71: 7597–7607.
[126]
Halvorson KG, Barton KL, Schroeder K, Misuraca KL, Hoeman C, Chung A, et al. A high-throughput in vitro drug screen in a genetically engineered mouse model of diffuse intrinsic pontine glioma identifies BMS-754807 as a promising therapeutic agent. PLoS ONE. 2015; 10: e0118926.
[127]
Franks SE, Jones RA, Briah R, Murray P, Moorehead RA. 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 Research Notes. 2016; 9: 134.
[128]
Sun R, Tanino R, Tong X, Haque EF, Amano Y, Isobe T, et al. Picropodophyllin inhibits the growth of pemetrexed-resistant malignant pleural mesothelioma via microtubule inhibition and IGF-1R-, caspase-independent pathways. Translational Lung Cancer Research. 2022; 11: 543–559.
[129]
Liao F, Chen Y, Wu Q, Wen J, Chen X, Wang W, et al. Selective elimination of CML stem/progenitor cells by picropodophyllin in vitro and in vivo is associated with p53 activation. Biochemical and Biophysical Research Communications. 2021; 579: 1–7.
[130]
Tarnowski M, Tkacz M, Zgutka K, Bujak J, Kopytko P, Pawlik A. Picropodophyllin (PPP) is a potent rhabdomyosarcoma growth inhibitor both in vitro and in vivo. BMC Cancer. 2017; 17: 532.
[131]
Girnita A, All-Ericsson C, Economou MA, Aström K, Axelson M, Seregard S, et al. The insulin-like growth factor-I receptor inhibitor picropodophyllin causes tumor regression and attenuates mechanisms involved in invasion of uveal melanoma cells. Clinical Cancer Research. 2006; 12: 1383–1391.
[132]
Fenerich BA, Fernandes JC, Rodrigues Alves APN, Coelho-Silva JL, Scopim-Ribeiro R, Scheucher PS, et al. NT157 has antineoplastic effects and inhibits IRS1/2 and STAT3/5 in JAK2V617F-positive myeloproliferative neoplasm cells. Signal Transduction and Targeted Therapy. 2020; 5: 5.
[133]
Flashner-Abramson E, Klein S, Mullin G, Shoshan E, Song R, Shir A, et al. Targeting melanoma with NT157 by blocking Stat3 and IGF1R signaling. Oncogene. 2016; 35: 2675–2680.
[134]
Wen B, Deutsch E, Marangoni E, Frascona V, Maggiorella L, Abdulkarim B, et al. Tyrphostin AG 1024 modulates radiosensitivity in human breast cancer cells. British Journal of Cancer. 2001; 85: 2017–2021.
[135]
Tanno B, Mancini C, Vitali R, Mancuso M, McDowell HP, Dominici C, et al. Down-regulation of insulin-like growth factor I receptor activity by NVP-AEW541 has an antitumor effect on neuroblastoma cells in vitro and in vivo. Clinical Cancer Research. 2006; 12: 6772–6780.
[136]
Scotlandi K, Manara MC, Nicoletti G, Lollini PL, Lukas S, Benini S, et al. Antitumor activity of the insulin-like growth factor-I receptor kinase inhibitor NVP-AEW541 in musculoskeletal tumors. Cancer Research. 2005; 65: 3868–3876.
[137]
Manara MC, Landuzzi L, Nanni P, Nicoletti G, Zambelli D, Lollini PL, et al. Preclinical in vivo study of new insulin-like growth factor-I receptor–specific inhibitor in Ewing’s sarcoma. Clinical Cancer Research. 2007; 13: 1322–1330.
[138]
Zhou H, Rao J, Lin J, Yin B, Sheng H, Lin F, et al. The insulin-like growth factor-I receptor kinase inhibitor NVP-ADW742 sensitizes medulloblastoma to the effects of chemotherapy. Oncology Reports. 2011; 25: 1565–1571.
[139]
Warshamana-Greene GS, Litz J, Buchdunger E, García-Echeverría C, Hofmann F, Krystal GW. The insulin-like growth factor-I receptor kinase inhibitor, NVP-ADW742, sensitizes small cell lung cancer cell lines to the effects of chemotherapy. Clinical Cancer Research. 2005; 11: 1563–1571.
[140]
He Y, Zhang J, Zheng J, Du W, Xiao H, Liu W, et al. The insulin-like growth factor-1 receptor kinase inhibitor, NVP-ADW742, suppresses survival and resistance to chemotherapy in acute myeloid leukemia cells. Oncology Research. 2010; 19: 35–43.
[141]
Zhou Q, Zhang J, Cui Q, Li X, Gao G, Wang Y, et al. GSK1904529A, an insulin-like growth factor-1 receptor inhibitor, inhibits glioma tumor growth, induces apoptosis and inhibits migration. Molecular Medicine Reports. 2015; 12: 3381–3385.
[142]
Sabbatini P, Rowand JL, Groy A, Korenchuk S, Liu Q, Atkins C, et al. Antitumor activity of GSK1904529A, a small-molecule inhibitor of the insulin-like growth factor-I receptor tyrosine kinase. Clinical Cancer Research. 2009; 15: 3058–3067.
[143]
Fei HD, Yuan Q, Mao L, Chen FL, Cui ZH, Tao S, et al. Assessment of GSK1904529A as a promising anti-osteosarcoma agent. Oncotarget. 2017; 8: 49646–49654.
[144]
Zhong H, Fazenbaker C, Breen S, Chen C, Huang J, Morehouse C, et al. MEDI-573, alone or in combination with mammalian target of rapamycin inhibitors, targets the insulin-like growth factor pathway in sarcomas. Molecular Cancer Therapeutics. 2014; 13: 2662–2673.
[145]
Weyer-Czernilofsky U, Hofmann MH, Friedbichler K, Baumgartinger R, Adam PJ, Solca F, et al. Antitumor Activity of the IGF-1/IGF-2-Neutralizing Antibody Xentuzumab (BI 836845) in Combination with Enzalutamide in Prostate Cancer Models. Molecular Cancer Therapeutics. 2020; 19: 1059–1069.
[146]
Friedbichler K, Hofmann MH, Kroez M, Ostermann E, Lamche HR, Koessl C, et al. Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycin. Molecular Cancer Therapeutics. 2014; 13: 399–409.
[147]
Weroha SJ, Haluska P. IGF-1 receptor inhibitors in clinical trials–early lessons. Journal of Mammary Gland Biology and Neoplasia. 2008; 13: 471–483.
[148]
Pandini G, Wurch T, Akla B, Corvaia N, Belfiore A, Goetsch L. Functional responses and in vivo anti-tumour activity of h7C10: a humanised monoclonal antibody with neutralising activity against the insulin-like growth factor-1 (IGF-1) receptor and insulin/IGF-1 hybrid receptors. European Journal of Cancer. 2007; 43: 1318–1327.
[149]
Munshi S, Hall DL, Kornienko M, Darke PL, Kuo LC. Structure of apo, unactivated insulin-like growth factor-1 receptor kinase at 1.5 A resolution. Acta Crystallographica. Section D, Biological Crystallography. 2003; 59: 1725–1730.
[150]
Ekyalongo RC, Yee D. Revisiting the IGF-1R as a breast cancer target. NPJ Precision Oncology. 2017; 1: 14.
[151]
Fassnacht M, Berruti A, Baudin E, Demeure MJ, Gilbert J, Haak H, et al. Linsitinib (OSI-906) versus placebo for patients with locally advanced or metastatic adrenocortical carcinoma: a double-blind, randomised, phase 3 study. The Lancet Oncology. 2015; 16: 426–435.
[152]
von Mehren M, George S, Heinrich MC, Schuetze SM, Yap JT, Yu JQ, et al. Linsitinib (OSI-906) for the Treatment of Adult and Pediatric Wild-Type Gastrointestinal Stromal Tumors, a SARC Phase II Study. Clinical Cancer Research. 2020; 26: 1837–1845.
[153]
Schmid P, Sablin MP, Bergh J, Im SA, Lu YS, Martínez N, et al. A phase Ib/II study of xentuzumab, an IGF-neutralising antibody, combined with exemestane and everolimus in hormone receptor-positive, HER2-negative locally advanced/metastatic breast cancer. Breast Cancer Research. 2021; 23: 8.
[154]
Ramalingam SS, Spigel DR, Chen D, Steins MB, Engelman JA, Schneider CP, 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. Journal of Clinical Oncology. 2011; 29: 4574–4580.
[155]
Sclafani F, Kim TY, Cunningham D, Kim TW, Tabernero J, Schmoll HJ, et al. Dalotuzumab in chemorefractory KRAS exon 2 mutant colorectal cancer: Results from a randomised phase II/III trial. International Journal of Cancer. 2017; 140: 431–439.
[156]
Moran T, Felip E, Keedy V, Borghaei H, Shepherd FA, Insa A, 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. Experimental Hematology & Oncology. 2014; 3: 26.
[157]
Huang CH, Williamson SK, Neupane P, Taylor SA, Allen A, Smart NJ, 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. Frontiers in Oncology. 2016; 5: 301.
[158]
Haluska P, Menefee M, Plimack ER, Rosenberg J, Northfelt D, LaVallee T, 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. Clinical Cancer Research. 2014; 20: 4747–4757.
[159]
Scagliotti GV, Bondarenko I, Blackhall F, Barlesi F, Hsia TC, Jassem J, et al. Randomized, phase III trial of figitumumab in combination with erlotinib versus erlotinib alone in patients with nonadenocarcinoma nonsmall-cell lung cancer. Annals of Oncology. 2015; 26: 497–504.
[160]
Langer CJ, Novello S, Park K, Krzakowski M, Karp DD, Mok T, et al. 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. Journal of Clinical Oncology. 2014; 32: 2059–2066.
[161]
de Bono JS, Piulats JM, Pandha HS, Petrylak DP, Saad F, Aparicio LMA, et al. Phase II randomized study of figitumumab plus docetaxel and docetaxel alone with crossover for metastatic castration-resistant prostate cancer. Clinical Cancer Research. 2014; 20: 1925–1934.
[162]
Strosberg JR, Chan JA, Ryan DP, Meyerhardt JA, Fuchs CS, Abrams T, et al. A multi-institutional, phase II open-label study of ganitumab (AMG 479) in advanced carcinoid and pancreatic neuroendocrine tumors. Endocrine-Related Cancer. 2013; 20: 383–390.
[163]
Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ, Jr, Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Annals of Oncology. 2012; 23: 2834–2842.
[164]
Pappo AS, Vassal G, Crowley JJ, Bolejack V, Hogendoorn PCW, Chugh R, et al. A phase 2 trial of R1507, a monoclonal antibody to the insulin-like growth factor-1 receptor (IGF-1R), in patients with recurrent or refractory rhabdomyosarcoma, osteosarcoma, synovial sarcoma, and other soft tissue sarcomas: results of a Sarcoma Alliance for Research Through Collaboration study. Cancer. 2014; 120: 2448–2456.
[165]
Pappo AS, Patel SR, Crowley J, Reinke DK, Kuenkele KP, Chawla SP, et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: results of a phase II Sarcoma Alliance for Research through Collaboration study. Journal of Clinical Oncology. 2011; 29: 4541–4547.
[166]
Park K, Tan DSW, Su WC, Cho BC, Kim SW, Lee KH, 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 Clinical and Research Reports. 2021; 2: 100206.
[167]
Bergqvist M, Holgersson G, Bondarenko I, Grechanaya E, Maximovich A, Andor G, et al. Phase II randomized study of the IGF-1R pathway modulator AXL1717 compared to docetaxel in patients with previously treated, locally advanced or metastatic non-small cell lung cancer. Acta Oncologica. 2017; 56: 441–447.
[168]
Haddad TC, He J, O’Sullivan CC, Chen B, Northfelt D, Dueck AC, et al. Randomized Phase II Trial of Capecitabine and Lapatinib with or without IMC-A12 (Cituxumumab) in Patients with HER2-Positive Advanced Breast Cancer Previously Treated with Trastuzumab and Chemotherapy: NCCTG N0733 (Alliance). Breast Cancer Research and Treatment. 2021; 188: 477–487.
[169]
Argiris A, Lee JW, Stevenson J, Sulecki MG, Hugec V, Choong NW, 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). Annals of Oncology. 2017; 28: 3037–3043.
[170]
Ferrarotto R, William WN, Jr, Tseng JE, Marur S, Shin DM, Murphy B, et al. Randomized phase II trial of cixutumumab alone or with cetuximab for refractory recurrent/metastatic head and neck squamous cell carcinoma. Oral Oncology. 2018; 82: 83–90.
[171]
Wong RL, Duong MT, Tangen CM, Agarwal N, Cheng HH, Vogelzang NJ, et al. Survival outcomes and risk group validation from SWOG S0925: a randomized phase II study of cixutumumab in new metastatic hormone-sensitive prostate cancer. Prostate Cancer and Prostatic Diseases. 2020; 23: 486–493.
[172]
Ekman S, Harmenberg J, Frödin JE, Bergström S, Wassberg C, Eksborg S, 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 Oncologica. 2016; 55: 140–148.
[173]
Chugh R, Griffith KA, Davis EJ, Thomas DG, Zavala JD, Metko G, et al. Doxorubicin plus the IGF-1R antibody cixutumumab in soft tissue sarcoma: a phase I study using the TITE-CRM model. Annals of Oncology. 2015; 26: 1459–1464.
[174]
Chiappori AA, Otterson GA, Dowlati A, Traynor AM, Horn L, Owonikoko TK, et al. A Randomized Phase II Study of Linsitinib (OSI-906) Versus Topotecan in Patients With Relapsed Small-Cell Lung Cancer. The Oncologist. 2016; 21: 1163–1164.
[175]
Bendell JC, Jones SF, Hart L, Spigel DR, Lane CM, Earwood C, et al. A phase Ib study of linsitinib (OSI-906), a dual inhibitor of IGF-1R and IR tyrosine kinase, in combination with everolimus as treatment for patients with refractory metastatic colorectal cancer. Investigational New Drugs. 2015; 33: 187–193.
[176]
Barata P, Cooney M, Tyler A, Wright J, Dreicer R, Garcia JA. A phase 2 study of OSI-906 (linsitinib, an insulin-like growth factor receptor-1 inhibitor) in patients with asymptomatic or mildly symptomatic (non-opioid requiring) metastatic castrate resistant prostate cancer (CRPC). Investigational New Drugs. 2018; 36: 451–457.
[177]
Sclafani F, Kim TY, Cunningham D, Kim TW, Tabernero J, Schmoll HJ, et al. A Randomized Phase II/III Study of Dalotuzumab in Combination With Cetuximab and Irinotecan in Chemorefractory, KRAS Wild-Type, Metastatic Colorectal Cancer. Journal of the National Cancer Institute. 2015; 107: djv258.
[178]
Huang F, Chang H, Greer A, Hillerman S, Reeves KA, Hurlburt W, et al. IRS2 copy number gain, KRAS and BRAF mutation status as predictive biomarkers for response to the IGF-1R/IR inhibitor BMS-754807 in colorectal cancer cell lines. Molecular Cancer Therapeutics. 2015; 14: 620–630.
[179]
Mancarella C, Casanova-Salas I, Calatrava A, Ventura S, Garofalo C, Rubio-Briones J, et al. ERG deregulation induces IGF-1R expression in prostate cancer cells and affects sensitivity to anti-IGF-1R agents. Oncotarget. 2015; 6: 16611–16622.
[180]
Xie M, Sun M, Ji X, Li D, Chen X, Zhang B, et al. Overexpression of BACH1 mediated by IGF2 facilitates hepatocellular carcinoma growth and metastasis via IGF1R and PTK2. Theranostics. 2022; 12: 1097–1116.
[181]
Lee H, Kim N, Yoo YJ, Kim H, Jeong E, Choi S, et al. β-catenin/TCF activity regulates IGF-1R tyrosine kinase inhibitor sensitivity in colon cancer. Oncogene. 2018; 37: 5466–5475.
[182]
Cohen-Sinai T, Cohen Z, Werner H, Berger R. Identification of BRCA1 As a Potential Biomarker for Insulin-Like Growth Factor-1 Receptor Targeted Therapy in Breast Cancer. Frontiers in Endocrinology. 2017; 8: 148.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top