Combination therapy approaches to target insulin-like growth factor receptor signaling in breast cancer

in Endocrine-Related Cancer
Authors:
Aleksandra M Ochnik Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia

Search for other papers by Aleksandra M Ochnik in
Current site
Google Scholar
PubMed
Close
and
Robert C Baxter Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, New South Wales, Australia

Search for other papers by Robert C Baxter in
Current site
Google Scholar
PubMed
Close

Free access

Sign up for journal news

Insulin-like growth factor receptor (IGF1R) signaling as a therapeutic target has been widely studied and clinically tested. Despite the vast amount of literature supporting the biological role of IGF1R in breast cancer, effective clinical translation in targeting its activity as a cancer therapy has not been successful. The intrinsic complexity of cancer cell signaling mediated by many tyrosine kinase growth factor receptors that work together to modulate each other and intracellular downstream mediators in the cell highlights that studying IGF1R expression and activity as a prognostic factor and therapeutic target in isolation is certainly associated with problems. This review discusses the current literature and clinical trials associated with IGF-1 signaling and attempts to look at new ways of designing novel IGF1R-directed breast cancer therapy approaches to target its activity 
and/or intracellular downstream signaling pathways in IGF1R-expressing breast cancers.

Abstract

Insulin-like growth factor receptor (IGF1R) signaling as a therapeutic target has been widely studied and clinically tested. Despite the vast amount of literature supporting the biological role of IGF1R in breast cancer, effective clinical translation in targeting its activity as a cancer therapy has not been successful. The intrinsic complexity of cancer cell signaling mediated by many tyrosine kinase growth factor receptors that work together to modulate each other and intracellular downstream mediators in the cell highlights that studying IGF1R expression and activity as a prognostic factor and therapeutic target in isolation is certainly associated with problems. This review discusses the current literature and clinical trials associated with IGF-1 signaling and attempts to look at new ways of designing novel IGF1R-directed breast cancer therapy approaches to target its activity 
and/or intracellular downstream signaling pathways in IGF1R-expressing breast cancers.

Introduction

A key strategy in precision cancer therapy is the targeting of cell-surface tyrosine kinase receptors that may drive tumor cell survival, proliferation and migration. In some cases, this has been remarkably successful, for example, in the inhibition of the human epidermal growth factor (EGF) receptor-2 (HER2) in women with HER2-positive breast cancer and the blockade of EGF receptor kinase activity in non-small-cell lung cancer. In contrast, the insulin-like growth factor (IGF) type 1 receptor (IGF1R) has, to date, proved to be a much less successful target. The IGF1R has two ligands, IGF-1 and IGF-2, and they are polypeptides of approximately 7.5 kDa that are structurally and functionally related to insulin. Some tumors also produce IGF-2 in larger precursor forms with variable receptor reactivity (Greenall et al. 2013). In addition to their high-affinity interaction with IGF1R, the IGFs bind with much lower affinity (about 1%) to the structurally related insulin receptor (InsR) in its main metabolically active isoform (isoform B), whereas IGF-2 and insulin itself are potent activators of the more mitogenic InsR isoform A (Morcavallo et al. 2014). Activation of IGF1R and/or InsR-A has been documented in many cancer types, making these receptors attractive therapeutic targets (Pollak 2012).

IGF-1 and IGF-2 circulate in adults at about 15–30 nM and 50–100 nM, respectively, concentrations high enough to saturate both their anabolic and mitogenic receptors. Their bioactivity is controlled by six IGF-binding proteins (IGFBPs), the most abundant of which is IGFBP-3, the main carrier of IGF-1 and IGF-2 in the circulation. IGFBPs are also found in the pericellular space, where they regulate IGF access to their cell-surface receptors (Baxter 2014). Cancer therapies that modulate the concentration or activity of any component of this complex system of receptors, binding proteins and ligands are likely to perturb the equilibrium across this system, sometimes in unanticipated ways. This article aims to provide a contemporary overview of the use of IGF1R-directed therapies in breast cancer clinical practice, with particular emphasis on the combination treatments directed toward IGF1R and other cellular targets.

Breast cancer and therapy resistance

Breast cancer remains the most common form of cancer in women, and GLOBOCAN statistics report ~1.7 million cases and 522,000 deaths worldwide in 2012 (Tao et al. 2014). There are predictions that breast cancer rates will reach 3.2 million cases by 2050 (Tao et al. 2014). An ongoing clinical problem is the large number of women who do not either respond or develop resistance to current breast cancer therapies. Circumvention of this issue is critical to the management of breast cancer, which urgently requires the development of more sophisticated combinatorial breast cancer therapies that effectively inhibit key cancer-driving signaling pathways (Zanardi et al. 2015).

Breast cancer is a highly heterogeneous disease and is treated clinically based on its molecular profile. The different molecular breast cancer subtypes include luminal A/B basal-like and HER2 breast cancers. Within these subtypes, both steroid receptors, including the estrogen receptor (ER) and progesterone receptor (PR), and tyrosine kinase growth factor receptors such as the IGF1R, EGFR and HER2 can be positive or negative in expression and integral to the cells’ behavior (Perou et al. 2000, 2011). For ER-positive luminal A/B breast cancers, therapies to reduce ERα activity at the receptor level using selective ER antagonists (fulvestrant) and aromatase inhibitors (exemestane, letrozole and anastrozole) to reduce estrogen levels are used. Conversely, triple-negative breast cancers (TNBCs – defined as ER, PR and HER2), that overlap strongly with the basal-like molecular subtype (Lehmann et al. 2011), are currently reliant on cytotoxic chemotherapy (e.g. doxorubicin, 5-fluorouracil and docetaxel), which is not reflective of the intrinsic molecular profiles of these cancers (Pal et al. 2011). For HER2-amplified breast cancers, a number of HER2-directed therapies, including the monoclonal antibodies (mAbs) trastuzumab and pertuzumab, which inhibit HER2/HER3 dimerization and activity, the dual tyrosine kinase inhibitor – lapatinib – which inhibits HER2 and EGFR activity and T-DMI (an antibody drug-conjugate of the mAb, trastuzumab and the chemotherapy drug DMI (emtansine)) (Verma et al. 2012, Mendes et al. 2015), have been developed and are clinically used. Currently, there are no targeted therapies for TNBC, which are widely used although many drugs designed to inhibit the emerging pathways are in clinical development (Kalimutho et al. 2015).

Of most concern is that the median overall survival in women diagnosed with metastatic breast cancer for estrogen-targeted therapies is 4 years, for HER2-targeted therapies is 3 years and for cytotoxic chemotherapy and radiotherapy is 12 months (Andre et al. 2004, Dawood et al. 2010, Pal et al. 2011). Despite the impressive clinical improvements associated with current breast cancer-targeted therapies such as the development of the HER2 inhibitor trastuzumab, innovative treatment strategies to manage resistant disease are still required.

IGF1R-targeted therapies: mechanism 
of action

Receptor tyrosine kinase inhibitors (TKIs), antireceptor monoclonal antibodies (mAbs) and mAbs against the IGFs have been developed to inhibit IGF1R activity and its downstream signaling (Yang & Yee 2012, King et al. 2014). IGF1R tyrosine kinase inhibitors (TKIs) are small molecules that either bind to the receptor competitively with ATP, e.g. PQIP, the forerunner of OSI-906 (Linsitinib) (Ji et al. 2007, Mulvihill et al. 2009), the structurally-related compound NVP-AEW541 (Brockhoff et al. 2012), BMS-754807 (Carboni et al. 2009) and BI 885578 (Sanderson 2015) or inhibit in a non-ATP-competitive manner, e.g. picropodophyllin or PPP (Girnita et al. 2004). Owing to substantial structural similarity between the IGF1R and the InsR (Lawrence et al. 2007), these inhibitors generally show some cross-reactivity with the InsR. Relative potencies, measured as the inhibition of receptor autophosphorylation in living cells, range from about 5% (Garcia-Echeverria et al. 2004, Ji et al. 2007) to 50–100% (Sanderson et al. 2015). Interestingly, inhibitors with a high degree of selectivity in intact cells may still show almost full cross-reactivity between IGF1R and InsR in biochemical kinase assays in vitro (Garcia-Echeverria et al. 2004, Ji et al. 2007), suggesting that receptor conformation in live cells contributes to the specificity. IGF1R TKIs generally do not downregulate the receptor, an exception being PPP (Vasilcanu et al. 2008). 
mAbs against IGF1R typically inhibit ligand-dependent receptor signaling by binding to the receptor competitively with its ligands IGF-1 and IGF-2 (Cohen et al. 2005, Wang et al. 2010a). An important component of their mode of action is also receptor downregulation (Burtrum et al. 2003, Cohen et al. 2005, Wang et al. 2010a). As a consequence of receptor blockade, circulating IGF-1 is markedly elevated (Yin et al. 2013), although unable to elicit a receptor-mediated response. Because they interact with extracellular receptor sequences rather than the intracellular ATP-binding domain, IGF1R mAbs can be selected to avoid the cross-reactivity with InsR seen for the TKIs (Goetsch et al. 2005), while still recognizing IGF1R–InsR hybrids (Cohen et al. 2005). However, as IGF-2 is a potent activator of the mitogenic isoform A InsR (Belfiore et al. 2009), the concomitant downregulation of InsR together with IGF1R may provide a therapeutic advantage (Sachdev et al. 2006). Humanized or fully human IGF1R mAbs include dalotuzumab (MK-0646), figitumumab 
(CP-751,871), cixutumumab (IMC-A12), robatumumab (SCH 717454), ganitumab (AMG179), AVE1642 and R1507, which inhibit the dimerization of the receptor. At the time of publication, the majority of trials involving these drugs are either completed/terminated or inactive.

A different approach to inhibit IGF1R signaling is by targeting the ligands that activate the receptor – IGF-1 and IGF-2. IGF-1/2 mAbs such as MEDI-573 (Haluska et al. 2014) and BI 836845 (Friedbichler et al. 2014) act by neutralizing the IGF-1 and IGF-2 ligands, thus potentially blocking the activation of both IGF1R and InsR-A. Such agents greatly increase circulating IGF concentrations, but the majority appear to be complexed to antibody and therefore biologically inert (Mireuta et al. 2014). In addition to direct approaches to inhibit IGF1R activation, whether by receptor or ligand blockade, numerous targeted therapies that inhibit IGF1R-driven downstream effectors involved in the regulation of protein translation, such as mTOR inhibitors (i.e. rapamycin, everolimus and temsirolimus), have been developed and are also of clinical interest as discussed in subsequent sections.

Indirectly targeting IGF1R signaling can also be achieved by using somatostatin or its analogues such as pasireotide (SOM230), which is approved as a therapy in the United States and Europe for conditions including Cushing’s disease and acromegaly, and it acts to reduce growth hormone production and consequently IGF-1 production and signaling (Kleinberg et al. 2011, Lewis et al. 2014). In another indirect approach, metformin, a standard treatment for type 2 diabetes, is shown to inhibit crosstalk between InsR/IGF1R and EGFR and as a result has indirect effects on IGF-1 signaling (Rozengurt et al. 2010, Liu et al. 2011).

Predictive biomarkers of IGF1R 
therapy response

Although high tumor IGF1R expression is reported to be a poor prognostic indicator in a variety of cancers including gastric (Matsubara et al. 2008), colorectal (Takahari et al. 2009), renal cell carcinoma (Sichani et al. 2010), and in some breast cancer subtypes (Taunk et al. 2010, Hartog et al. 2011), few proposed markers to predict responsiveness to IGF1R-targeting therapies have progressed beyond preliminary reports. Cell-line studies in sarcoma and neuroblastoma show that sensitivity toward TKI BMS-536924 was associated with high IGF-1, IGF-2 and IGF1R expression, with drug resistance associated with the expression of IGFBP-3 and IGFBP-6 (Huang et al. 2009). Other cell studies in breast and lung cancer have similarly pointed to a possible role of high IGF1R expression in predicting both TKI and mAb responsiveness (Gong et al. 2009, Litzenburger et al. 2011). The glycosylation state of IGF1R has also been found to correlate with CP-751,871 sensitivity in gastric and hepatocellular carcinoma cell lines, possibly related to the formation of IGF1R/InsR hybrid receptors (Kim et al. 2012). In a comprehensive screen of CP-751,871 sensitivity across 93 cancer cell lines, gene expression levels of IGF-axis components and MYB were found to have the strongest predictive value (Pavlicek et al. 2013). A more complex classifier for 
OSI-906 responsiveness was developed by integrating gene expression corresponding to drug sensitivity, IGF1R expression by in situ hybridization and K-RAS status, leading to a test claimed to have 100% predictive accuracy (Pitts et al. 2010).

However, despite promising leads from cell biology, measurement of IGF1R and other IGF-axis components has had relatively little success as a predictive marker in clinical studies. In a phase 1 study of patients with prostate cancer treated with CP-751,871 plus docetaxel, IGF1R detectable on circulating tumor cells appeared to correlate with treatment response (measured as PSA decline), although no statistical analysis was presented (de Bono et al. 2007). Contrasting with this finding and numerous cell biology studies, IGF1R expression, detected by immunohistochemistry, was unable to predict clinical outcome in patients with bone or soft-tissue sarcomas treated with cixutumumab (IMC-A12) in combination with the mTOR inhibitor temsirolimus (Schwartz et al. 2013). In patients with metastatic pancreatic cancer treated with gemcitabine, increased overall survival in response to ganitumab (AMG179) was predicted by circulating IGF and IGFBP-3 levels – notably high IGF-2 (McCaffery et al. 2013). The conclusion to be drawn from these and other clinical studies is that there is currently no clinically well-validated biomarker to predict treatment response for IGF1R pathway blockade, by treatment with either a IGF1R mAb or IGF1R-TKI, that could be applied to breast cancer trials.

IGF1R-directed combination therapies: specific molecular subtypes of breast cancer

Estrogen receptor-positive breast cancer

IGF1R and ERα crosstalk pathways in breast cancer have been widely studied for therapeutic intervention in ER-positive breast cancers (Fagan & Yee 2008). The IGF 
signaling pathway is intrinsically involved in the regulation of the ERα after phosphorylation of the protein. Given that ER-positive breast cancers are 
the most common subtype, comprising approximately 70% of all diagnosed breast cancers (Jeselsohn et al. 2015), and oncogenic activity in this molecular subtype is contributed to by IGF signaling (Becker et al. 2011), many clinical trials have been undertaken to test IGF1R-directed therapies in ER-positive breast cancer. The lack of clinical success of these trials, which did not lead to any improvements in clinical outcomes, highlights that targeting IGF1R in isolation is not sufficient to overcome tumor growth (Yee 2012). There is now re-ignited hope that IGF1R therapies combined with other FDA-approved drug treatments and emerging drug therapies will be more effective and lead to better clinical outcomes by preventing re-activation of IGF1R and/or activation of compensatory signaling pathways that can overcome the drug effects (Beckwith & Yee 2014).

As previously outlined, identification of better predictive biomarkers that reflect IGF1R therapy response is required to determine which individuals will or will not benefit from IGF1R-targeted therapies. Moreover, the complexity of IGF1R signaling dictated by the ability of the cell to either re-activate the IGF1R-signaling pathway and/or drive oncogenesis via the activation of alternative growth factor signaling pathways such as the InsR, EGFR, HER2 or vascular endothelial growth factor receptor (VEGFR), further heightens the difficulty in designing effective IGF1R combination targeted therapies.

IGF1R and chemotherapy

The role of IGF1R in promoting resistance to chemotherapy is well established (Bohula et al. 2003). Cell-line studies using the ER-positive MCF7 and Bcap-37 breast cancer cell lines show that the cell-permeable plant-derived flavonoid wogonin, that is known to possess anti-carcinogenic and anti-inflammatory properties, increases the sensitivity to the chemotherapy agent doxorubicin via the suppression of IGF1R/AKT signaling pathway (Fu et al. 2015). Furthermore, in vitro studies in MCF7 cells reported that the effectiveness of using the highly potent and selective IGF1R tyrosine kinase inhibitor PQIP, an analogue of OSI-906, in combination with chemotherapy (i.e. gemcitabine) depends on the sequence of drug administration (Khatri et al. 2012). Enhancement of the response to the IGF1R mAbs AVE1642 or BMS-754807 by gemcitabine has been demonstrated using established BxPC-3 human pancreatic tumor xenografts (Maloney et al. 2003, Awasthi et al. 2012), suggesting that these types of IGF1R and chemotherapy combination therapies may also have clinical benefit in other cancers including breast.

The phase 3 NEOZOTAC clinical trial completed in 2013 reported that reduced IGF1R expression during and after neo-adjuvant chemotherapy treatment is associated with improved pathological outcomes in HER2-negative breast cancer (Tables 1 and 2: Ref #16; Nbib1099436) (de Groot et al. 2016). Similarly, reduced IGF1R expression was predictive of a complete pathologic response in ER-positive breast cancers treated with neoadjuvant chemotherapy (Bhargava et al. 2011), highlighting that IGF1R expression modulates the chemotherapy response. Significantly elevated IGF1R protein and mRNA expression have additionally been shown in naturally occurring cisplatin-resistant SKOV3 and HEY ovarian cancer cells compared with those in the more sensitive A2780 and OAW42 ovarian cancer cell lines. Most importantly, chemoresistance was reversed in the early-stage cancer by the addition of the IGF1R inhibitor – small molecule inhibitor PPP. This proves as the evidence that co-treatment with chemotherapy and IGF1R inhibitors has clinical potential (Singh et al. 2014b).

A phase 1 clinical trial investigated the pharmacokinetic (PK) profiles and safety of the chemotherapy agent gemcitabine in conjunction with AMG479 in advanced solid tumors (Table 1: Ref #20, Nbib974896), which has been deemed to be tolerable and safe leading to improved survival outcomes in a placebo-controlled phase 2 trial for metastatic pancreatic cancer (Kindler et al. 2012). A combination therapy comprising cisplatin and etoposide (CE) in conjunction with the IGF1R inhibitor cixutumumab has been evaluated in a phase 2 trial for small-cell lung carcinoma (Table 1: Ref #26, Nbib887159), but was reported not to be associated with any clinical improvements in progression-free survival (PFS) compared with CE therapy alone (Belani et al. 2013). Nonetheless, given that IGF1R inhibition has been shown to positively influence the chemotherapy response, a therapy approach of this nature may still have benefit for breast cancer and is worth further investigation possibly with other FDA-approved therapies.

Table 1

IGF1R-directed therapy clinical trials/studies for breast cancer and solid tumors. Phase 1, 2 and 3 clinical trials/studies using insulin-like growth factor receptor (IGF1R)-directed therapies either as single-agent therapies or combinatorial therapies for breast cancer, and solid tumors were obtained from the clinical trials U.S. National Institutes of Health website (https://clinicaltrials.gov/) and listed in reverse chronological order from the estimated completion dates.

Ref # Clinical trials identifier Study title Phase Estimated completion Breast cancer grade/molecular subtype Published drug doses and results
#1 Nbib1042379 I-SPY 2 TRIAL: neoadjuvant and personalized adaptive novel agents to treat breast cancer (I-SPY 2) (Barker et al. 2009) 2 Sept-2017 Histologically confirmed invasive cancer of the breast, any tumor 
ER/PR status, any HER-2/neu status NA
#2 Nbib1446159 Study of MEDI-573 plus standard endocrine therapy for women with hormone-sensitive metastatic breast cancer 1.2 Sept-2017 ER or PR positive, or both; HER2 negative NA
#3 Nbib2045368 Study of IGF-methotrexate conjugate in the treatment of advanced tumors expressing IGF-1R 1 Jan-2017 IGF1R positive NA
#4 Nbib1776008 Akt Inhibitor MK-2206 and anastrozole with or without goserelin acetate in treating patients with stage II–III breast cancer 2 Dec-2016 Clinical stage 2 or 3 PIK3CA mutant ER-positive and HER2-ve invasive breast cancer NA
#5 Nbib1111825 Temsirolimus plus neratinib for patients with metastatic HER2-amplified or triple negative breast cancer 1.2 Dec-2016 Metastatic HER2-amplified or triple negative NA
#6 Nbib1650506 Study of erlotinib and metformin in triple negative breast cancer 1 Sept-2016 Triple-negative breast cancer, prior diagnosis of ER- or PR-positive breast cancer (HER2 negative) NA
#7 Nbib1708161 A phase Ib/II study of the combination of BYL719 plus AMG 479 in adult patients with selected solid tumors 1.2 Jun-2016 PIK3CA mutated or amplified; hormone receptor-positive breast carcinoma NA
#8 Nbib699491 Cixutumumab and temsirolimus in treating patients with locally recurrent or metastatic breast cancer 1.2 Jun-2016 Metastatic or locally recurrent disease (locally recurrent disease should be stage IV) NA
#9 Nbib1733004 A phase 1 study of MM-141 in patients with advanced solid tumors 1 Dec-2015 Advanced solid tumors NA
#10 Nbib1372618 Breast cancer chemoprevention by SOM230, 
an IGF-I action inhibitor 2 Oct-2015 ER positive, DCIS NA
#11 Nbib1295632 Safety and tolerability of different dose combinations of ridaforolimus with MK-2206 or MK-0752 for participants with advanced cancer (MK-8669-049) (Gupta et al. 2015, Piha-Paul et al. 2015) 1 Aug-2015 Histologically-confirmed breast cancer: a low RAS-gene signature and a high Ki67 index label if ER positive Drug doses: MTD: ridaforolimus = 10 mg/day + 
MK-2206 = 90 mg/week.
Results: 2/16 (12.5%) = PR; 2/14 (14.3%) = CR; no clinical response detected in prostate cancer patients. SD for two patients, 416 and 285 days (Gupta et al. 2015).
Results: 2/15 (13%) responded; one PR and one CR. SD for one patient ≥6 months (Piha-Paul et al. 2015).
#12 Nbib728949 A study for safety and effectiveness of IMC-A12 by itself or combined with antiestrogens to treat breast cancer (Gradishar et al. 2015) 2 Feb-2015 ER and/or PR positive Drug doses: Arm 1 (n = 62): CIX = 10 mg/kg + anti-estrogen; Arm B (n = 31): CIX = 10 mg/kg. CIX well tolerated.
Results: DCR: Arm A = 40.3% (95% CI: 28.1, 536); Arm B = 51.6% (95% CI: 33.1, 69.8). Low IR, IR-A and IR-B mRNA expression associated with significant increase in PFS and OS.
#13 Nbib1225172 Study of BMS-754807 combined with letrozole or BMS-754807 alone in patients with hormone receptor-positive breast cancer and resistance to non-steroidal aromatase inhibitors 2 Nov-2014 Postmenopausal women with HR-positive and HER-2-negative locally advanced or metastatic breast cancer NA
#14 Nbib1245205 Akt inhibitor MK2206 in combination with lapatinib ditosylate in patients with advanced or metastatic solid tumors or breast cancer 1 Aug-2014 Advanced and unresectable or metastatic HER2-positive breast cancer NA
#15 Nbib684983 Capecitabine and lapatinib with or without cixutumumab in treating patients with previously treated HER2-positive stage IIIB, stage IIIC, or stage IV breast cancer 2 Jan-2014 HER2-positive stage IIIB-IV breast cancer NA
#16 Nbib1099436 Neo-adjuvant chemotherapy (TAC) with or without zoledronic acid in treating 
HER2-negative breast cancer patients (NEO-ZOTAC) (de Groot et al. 2016) 3 Sept-2013 HER2-negative large resectable or locally advanced breast cancer Drug doses: TAC chemotherapy = 75mg/m2 docetaxel; 50 mg/m2 doxorubicin and 500 mg/m2 cyclophosphamide ± 4 mg zoledronic acid.
Results: 47.2% significant reduction in IGF1R expression after TAC; better pathological response to TAC in patients with reduced IGF1R (P = 0.006; during therapy) and (P = 0.020; after therapy) and patients with 3129G >T polymorphism (P = 0.032), multivariate analysis.
#17 Nbib739453 A phase 1 dose-escalation study of OSI-906 and erlotinib (Tarceva) (Macaulay et al. 2016) 1 Dec-2011 Advanced solid tumors Drug doses: linsitinib schedules (S): S1 = 50–600 mg/QD; S2 = 50–400 mg/QD; S3 = 100/150 mg/BID in conjunction with 100–150 mg/QD erlotinib. Linsitinib/erlotinib combination was well tolerated.
Results: 5/75 (7%) = PR and 38/75 (51%) = DCR.
#18 Nbib963547 A study of MK2206 in combination with trastuzumab and lapatinib for the treatment of HER2+ solid tumors (2206-015) 
(Hudis et al. 2013) 1 Dec-2011 Locally advanced or metastatic HER2-positive solid tumor Drug doses: trastuzumab = 8 mg/kg day 1 and 6 mg/kg every 3 weeks in conjunction with MK-2206 = 45 or 60 mg QOD (two cohorts) or 135 and 200 mg dose (two cohorts). Trastuzumab and MK-2206 in general was well tolerated.
Results: 1/31 = PR; 1/31 = CR and 5/31 = SD (up to 4 months).
#19 Nbib1372644 Breast cancer chemoprevention by SOM230, an IGF-I action inhibitor: a proof of principle trial (Singh et al. 2014a) 1.2 Jun-2011 Atypical ductal hyperplasia, lobular carcinoma in situ, and/or atypical lobular hyperplasia Drug doses: pasireotide = 600 µg/BID.
Results: AH lesions = reduced proliferation (3.6 ± 2.6% to 1.3 ± 1.2%) and increased apoptosis (0.3 ± 0.2% to 1.3 ± 0.6%). DCIS response was similar. IGF1R, ERK1/2 and AKT phosphorylation were significantly reduced.
#20 Nbib974896 Study of AMG 479 with biologics or chemotherapy for subjects with advanced solid tumors 1 Apr-2011 Advanced solid tumors NA
#21 Nbib1205685 Endocrine therapy + OSI-906 with or without erlotinib for hormone-sensitive metastatic breast cancer 2 Jul-2011 Clinical stage IV invasive mammary carcinoma, ER- and/or PR-positive NA; terminated
#22 Nbib372996 Study of CP-751,871 in combination with exemestane in postmenopausal women with hormone receptor positive advanced breast cancer 2 May-2011 HR-positive advanced breast cancer NA, terminated
#23 Nbib730379 A combination study with ridaforolimus (MK8669) and dalotuzumab (MK0646) in patients with advanced cancer (8669-004) (Di Cosimo et al. 2015) 1 Nov-2010 Advanced cancer Drug doses: ridaforolimus = 10–40 mg/day and dalotuzumab = 10 mg/kg/week or 7.5 mg/kg every other week.
Results: PR = 6/87 (7%), 40/87 (46%) = SD and 27/87 (31%) = PD. 10/23 (43%) breast cancer patients showed anti-tumor activity of which 6/11 (55%) were ER+/Ki67 high expressing.
#24 Nbib774878 Study of AVE1642 (IGF-1R/CD221) in combination with fulvestrant (Faslodex) in postmenopausal patients with advanced hormone-dependent breast cancer 2 Nov-2010 Advanced hormone-dependent breast cancer, HER2-positive excluded NA, Terminated
Other solid cancers
#25 Nbib955305 Paclitaxel, carboplatin, and bevacizumab with or without cixutumumab in treating patients with stage IV or recurrent non-small cell lung cancer 2 Dec-2015 NA NA
#26 Nbib887159 Cisplatin and etoposide with or without vismodegib or cixutumumab in treating patients with extensive-stage small cell lung cancer (Belani et al. 2013) 2 Sept-2014 NA Drug doses: CE alone ((75 mg/m2 D1 and E 100 mg/m2 D1-3) Arm A) combined with either V 
((150 mg/day) Arm B) or CIX 
((6 mg/kg weekly IV) Arm C).
Results: no significant improvement in PFS was detected for CE combined with either V or CIX, compared with CE alone.
#27 Nbib906373 A study of IMC-A12 in combination with sorafenib in patients with advanced cancer of the liver 2 May-2014 NA NA
#28 Nbib617708 S0727 gemcitabine hydrochloride and erlotinib hydrochloride with or without monoclonal antibody therapy in treating patients with metastatic pancreatic cancer that cannot be removed by surgery 1/2 Feb-2014 NA NA

DCIS, ductal carcinoma in situ; ER, estrogen receptor; HER, human epidermal receptor; HR, hormone receptor; PIK3CA, phosphatidylinositol 3-kinase; PR, progesterone receptor; Ref, reference; TNBC, triple negative breast cancer;

AH, atypical hyperplasia; BID, twice daily; CIX, cixutumumab; CR, complete response; DCR, disease control rate; MTD, maximum tolerated dose; 
NA, not available; NR, not relevant; OS, overall survival; PD, progressive disease; PFS, progression free survival; PR, partial response; QD, once daily; QOD, every other day; QW, once weekly; 
SD, stable disease; TAC, (T – Docetaxel (also called Taxotere), A – Doxorubicin (originally called Adriamycin) and C – Cyclophosphamide).

IGF1R and anti-estrogens

Early studies using MCF7 and T47D breast cancer cell lines and mouse xenograft studies using MCF7 cells provide evidence that IGF1R inhibitors including the TKI, NVP-AEW541 and the mAb CP-751,871 in combination with ER-targeted therapies such as letrozole or tamoxifen lead to more effective anti-tumor responses compared with the single agents (Cohen et al. 2005, Lisztwan et al. 2008). Similarly, in vitro and in vivo studies using BMS-754807 in combination with either of the hormonal agents: 4-hydroxytamoxifen, fulvestrant or letrozole have also demonstrated a strong synergistic effect on tumor inhibitory responses in ER-positive breast cancer (Table 2) (Hou et al. 2011). As IGF-1 can regulate ER phosphorylation and activity in breast cancer (Becker et al. 2011, Ochnik et al. 2016) and the indirect IGF-1-inhibitor (somatostatin analogue) SOM230 has been shown to reduce proliferation and increase apoptosis via an IGF1R-mediated effect in pre-malignant breast lesions (i.e. atypical hyperplasia) in a phase 1 proof-of-principle trial (Table 1: Ref #19, Nbib1372644) (Singh et al. 2014a), an additional treatment phase 1 study was completed to determine whether SOM230 can also be used as chemoprevention for early-stage ER-positive ductal carcinoma in situ (Table 1: Ref #10, Nbib1372618).

Table 2

Current and future directions of IGF1R-targeted breast cancer therapies. The table summarizes the preclinical studies and clinical testing of the various IGF1R-targeted – direct and/or indirect (i.e. IGF1R-downstream mediators) – as detailed in this review based on ER-positive, ER-negative, HER2-positive, BRCA-mutated and PI3K-amplified/mutated molecular subtypes.

Breast cancer molecular subtype IGF1R-targeted (direct or indirect) combination treatments
ER+ IGF1R and chemotherapy
Wogonin plus chemotherapy (Fu et al. 2015)
IGF1Ri plus chemotherapy (IGF1R polymorphisms) (Ref #16 – Nbib10994436) (de Groot et al. 2016)
IGF1R and anti-estrogens
BMS-754807 plus letrozole (Ref #13 – Nbib1225172) or 4-hydroxytamoxifen or fulvestrant (Hou et al. 2011)
MEDI-573 plus aromatase inhibitor (Ref #2 – Nbib1446159)
MEK inhibitor and anti-estrogens (Periyasamy-Thandavan et al. 2012)
IGF1R and other
aIGF1Ri and SphK1i plus anti-estrogens (luminal A) (Fig. 2) (Pitson et al. 2003, Granata et al. 2007).
ER− IGF1R and chemotherapy
BMS-754807 plus chemotherapy (Litzenburger et al. 2011)
IGF1R and EGFR
aIGF1R/EGFR bispecific antibodies (Dong et al. 2011, Croasdale et al. 2012)
aPI3K-inhibitor (i.e. SF1126) plus EGFR inhibitor (gefitinib/erlotinib) (basal) (Deng et al. 2015)
Temsirolimus plus gefitinib/erlotinib (basal) (Madden et al. 2014)
Metformin and erlotinib (Ref #6 – Nbib1650506)
AG1024 plus gefitinib (mesenchymal) (Camirand et al. 2005)
OSI-906 plus erlotinib (Ref #17 Nbib739453) (Macaulay et al. 2016)
Cixutumumab, gemcitabine and erlotinib (Ref #28 – Nbib617708)
aSelumetinib and gefitinib (Maiello et al. 2015)
IGF1R and VEGF
Chemotherapy plus cixutumumab ± bevacizumab (Ref #25 – Nbib955305)
R1507 or AMG479 plus bevacizumab (Ref #20 – Nbib974896) (Mahadevan et al. 2014)
IGF2 and other
aMAB292 plus NVP-AEW541 (Mancini et al. 2014)
aERβ antagonist, PHTPP plus bevacizumab and/or erlotinib (Mancini et al. 2014)
IGF1R and FAK
aFAK/ROCK inhibitors plus chemotherapy (mesenchymal) (Kurio et al. 2011, Taliaferro-Smith et al. 2015) IGF1R and other
AMG479 plus metformin (Ref #1 – Nbib1042379)
aIGF1Ri, SphK1i plus chemotherapy (basal) (Fig. 2) (Wang et al. 2010, Fu et al. 2015, de Groot et al. 2016)
HER2+ IGF1R, HER2 and chemotherapy
aMM-141 plus chemotherapy (Fitzgerald et al. 2014)
aTrastuzumab or lapatinib plus PI3K inhibitors (Chakrabarty et al. 2012)
aFigitumumab, trastuzumab and neratinib (Chakraborty et al. 2015)
Capecitabine and lapatinib ± cixutumumab (Ref #15 – Nbib684983)
AKT inhibitor, MM-2206, trastuzumab ± paclitaxel (Ref #1 – Nbib1042379) (Chien et al. 2016)
MK-2206 plus lapatinib (Ref #14 – Nbib1245205) or trastuzumab (Ref #18 – Nbib963547) (Hudis et al. 2013)
Temsirolimus and neratinib (Ref #5 – Nbib1111825)
IGF1R, Src and HER2
Sorafenib, IMC-A12 plus trastuzumab/lapatinib (Ref #27 – Nbib906373) (Browne et al. 2012)
BRCA mutated PARP, chemotherapy and IGF1R
aOlaparib/veliparib, chemotherapy plus BMS-536924 (Amin et al. 2015)
Veliparib plus chemotherapy (Nbib2595905)
PI3K amplified and/or mutated IGF1R and PI3K
aAMG479 plus BYL719 (Ref #7 – Nbib1708161)
PI3K and EGFR
aSF1126/PI-103 plus EGFR inhibitor (Yi et al. 2013, Deng et al. 2015)
AKT and other
MK-2206 plus goserelin acetate (pre-menopausal) or anastrozole (post-menopausal) (Ref #4 – Nbib1776008)
Not specified IGF1R and other
Ridaforolimus, MK-2206 and MK-0752 (Ref #11 – Nbib1295632)
Ridaforolimus and MK0646 (Ref #1 – Nbib730379) (Di Cosimo et al. 2015)
aCixutumumab, gemcitabine and erltonib (Mahadevan et al. 2014)
aCixutumumab, paclitaxel and bevacizumab (Mahadevan et al. 2014)
aCixutumumab, carboplatin and etoposide (Mahadevan et al. 2014)
aCixutumumab, mFOLFOX6 and bevacizumab (Mahadevan et al. 2014)
aCixutumumab, capecitabine and trastuzumab (Mahadevan et al. 2014)
aCixutumumab and sorafenib (Mahadevan et al. 2014)

IGF1R-combination therapies that have preclinical potential and could be priority therapies for future clinical testing.

A phase 2 clinical trial completed in 2015, testing IMC-A12 alone or in combination with anti-estrogen therapies in women who had progressed on anti-estrogen therapy, deemed the combination to be safe and tolerable, but the dual therapy did not show any significant clinical outcomes compared with single therapies (Table 1: Ref #12, Nbib728949) (Gradishar et al. 2015). 
BMS-754807 has been given in combination with the aromatase inhibitor, letrozole, in ER-positive breast cancers with acquired resistance to non-steroidal aromatase inhibitors in a phase 2 trial completed during late 2014 (Tables 1 and 2; Ref #13, Nbib1225172). Furthermore, a phase 1/2 trial, for an estimated completion in 2017, has been designed to test MEDI-573 in combination with an aromatase inhibitor (AI) vs AI alone in relation to its safety, tolerability and anti-tumor ability in metastatic breast cancers (Tables 1 and 2: Ref #2, Nbib1446159).

A phase 2 clinical trial to investigate the effects of the endocrine therapy letrozole plus OSI-906 with and without erlotinib in breast cancer was terminated in 2011 as all the patients experienced severe toxicities and tumor progression (Table 1: Ref #21, Nbib1205685). Moreover, a phase 2 clinical trial to test a combination of exemestane with CP-751,871 (AVE1642) for women diagnosed with ER-positive advanced breast cancer was terminated in 2011 as the manufacturer discontinued the production of AVE1642 (Table 1: Ref #22, Nbib372996). A similar study was also terminated based on the discontinuation of AVE1642 (Table 1: Ref #24, Nbib774878). These terminated studies serve to highlight that there are toxicity issues associated with some IGF-directed targeted combination therapies and demonstrate the intrinsic problems with translation of preclinical findings into effective clinical practice.

Triple-negative breast cancers

The need to develop new therapies for TNBC currently reliant on chemotherapy and radiotherapy, and to better identify prognostic markers and therapeutic targets, is an ongoing clinical issue. IGF signaling has been shown to be a direct therapeutic target in TNBC (Creighton et al. 2008, Bhargava et al. 2011, Davison et al. 2011, Taliaferro-Smith et al. 2015). Briefly, IGF1R was shown to be expressed and activated by phosphorylation in TNBC (Lerma et al. 2007, Law et al. 2008), high IGF1R mRNA expression leads to poorer overall survival in basal/triple-negative groups (Peiro et al. 2011) and rare high-level amplification of IGF1R has been detected in some basal-like breast cancers (Adelaide et al. 2007). Collectively, these studies highlight that IGF1R is an oncogenic driver and therapeutic target in TNBC. It has been suggested that the lack of clinical trials testing IGF1R therapies in TNBC has been attributed to insufficient preclinical data demonstrating effectiveness. The following sections give an overview of the preclinical evidence of IGF-targeted therapies in TNBC and combinatorial approaches that show clinical potential.

IGF1R and chemotherapy

Owing to the lack of effective targeted therapies, chemotherapy is the most widely used treatment for TNBC and based on evidence that IGF1R decreases sensitivity and response to chemotherapy (Wang et al. 2010b, Beckwith & Yee 2014), chemo- and IGF1R-targeted combination therapies have been of clinical interest. Specifically, the timing and sequence of administering an IGF1R therapy with chemotherapy have been shown to be important in its effectiveness (Zeng et al. 2009). A preclinical study aimed at investigating the combined effects of the anti-IGF1R/InsR inhibitor, BMS-754807 in conjunction with chemotherapy in TNBC, has shown a complete regression response in vivo using the MC1 tumor-graft model of TNBC compared with either agent alone, in parallel with reversal in an IGF gene expression signature (Table 2) (Litzenburger et al. 2011). Given this evidence in the literature that IGF1R inhibitors and chemotherapy may be beneficial, further preclinical and clinical testing with other breast cancer therapies is of interest.

IGF1R and EGFR

EGFR inhibitors have been widely tested in various malignancies such as breast, lung, pancreatic and head and neck cancers (Mendelsohn & Baselga 2006, Subramaniam et al. 2015). EGFR is overexpressed in up to 70% of basal-like TNBC (Livasy et al. 2006), but EGFR monotherapies such as gefitinib and erlotinib have to date not been successful for breast cancer (Masuda et al. 2012), potentially owing to modulation and crosstalk with other growth factors including IGF1R (i.e. EGFR/IGF1R dimer formation) (Morgillo et al. 2006, Guix et al. 2008). There are several studies now suggesting that IGF1R and EGFR combinatorial therapies are worth investigating further for cancer and given there is crosstalk between the two growth factors, combinations have the potential to be more effective than targeting each factor in isolation (Guix et al. 2008, Saxena et al. 2008, Castano et al. 2013, Wong et al. 2014).

A recent study identified that levels of the IGF1 and EGF ligands were significantly higher in the stromal micro-environment and bone marrow in TNBC tumors, which were otherwise inactive before being exposed to the active stimuli (Castano et al. 2013). The study demonstrated that high IGF-1 and EGF expression levels lead to an induction of the transcription factors Oct4, c-Myc and Zeb1 as a trigger for subsequent tumor recurrence and metastasis. Interestingly, IGF1R and EGFR were shown to be expressed in the tumor cells suggesting that the paracrine interplay between stromal and epithelial cells is essential for the maintenance of tumor growth. Importantly, the authors demonstrated that IGF1R activation itself was not sufficient enough to initiate tumor growth and that EGFR activation was also required.

Inhibition of HER-1 (EGFR) and IGF1R using HER-1-418 and IGF-1R-56 peptide mimics has been shown to promote increased anti-tumor responses in the TNBC MDA-MB-231 cell line (Overholser et al. 2015). An in vitro study using MDA-MB-231 and MDA-MB-468 TNBC cells also reported that co-treatment with gefitinib and the IGF1R inhibitor, AG1024, was effective in producing additive-to-synergistic effects on tumor cell growth (Table 2) (Camirand et al. 2005). Lastly, bispecific antibodies such as XGFR2, XGFR3, XGFR4 and EI-04 have been developed, which target both IGF1R and EGFR activity and have been shown to have potent anti-tumor activity that may be a more effective alternative approach to dual-targeting IGF1R and EGFR in breast cancer than using single-targeted therapies (Table 2) (Dong et al. 2011, Croasdale et al. 2012).

A phase 1 clinical trial completed in 2011 reports that co-treatment with OSI-906 and erlotinib is tolerable leading to effective IGF-1R/InsR phosphorylation reduction in advanced solid cancers such as non-small-cell lung cancer, pancreatic, colorectal and prostate (Tables 1 and 2; Ref #17, Nbib739453) (Macaulay et al. 2016). Based on the indirect effects of metformin on IGF1R/EGFR activity (Rozengurt et al. 2010, Liu et al. 2011), there may be value in testing novel metformin combination therapies. In line with this therapeutic approach, a phase 1 trial that commenced in 2012 has been designed to test metformin in combination with erlotinib for TNBC (Tables 1 and 2: Ref #6, Nbib1650506).

In breast cancer, a number of trials for ER-negative/TNBC cancer have tested EGFR inhibitors in combination with chemotherapy and other targeted therapies (Clinical Trials Gov. Identifiers: Nbib744408, Nbib834678, Nbib239343, Nbib491816 and Nbib998036). 
In light of the fact that EGFR- and IGF1R-directed combination therapies have shown some preclinical success, and EGFR inhibitors have entered into clinical trials in combination with other drug therapies such as chemotherapy, VEGF-inhibitors and metformin in TNBC, the further addition of IGF1R-inhibitors may also be beneficial to these treatment regimens. In line with this thinking, the I-SPY 2 Trial includes the testing of AMG479 plus metformin for locally advanced breast cancers in addition to a standard neoadjuvant chemotherapy regimen with or without trastuzumab as determined by the HER2 status (Tables 1 and 2: Ref #1, Nbib1042379) (Barker et al. 2009). A triple combination therapy, comprising cixutumumab, gemcitabine and erlotinib has also been tested in a phase 1 trial as a first-line therapy for pancreatic cancer and may also be an effective approach for TNBC (Tables 1 and 2: Ref #28, Nbib617708).

IGF1R and VEGF

In the search for better prognostic biomarkers in TNBC, a recent study has demonstrated that VEGF-A, IGF-1 and IGF1R serum levels were significantly higher in women with TNBC than those with other breast cancer types and were also associated with poorer prognosis (Bahhnassy et al. 2015a). In a separate study, elevated tissue expression of IGF1R, IGF-1 and VEGF-A mRNA and protein was identified in TNBC vs non-TNBC; however, only IGF-1 was found to be an independent prognostic factor (Bahnassy et al. 2015b). IGF1R and VEGF-A are both client proteins of heat-shock protein (Hsp)90, a molecular chaperone involved in the maturation and protein folding of many oncogenic factors, and therapies inhibiting Hsp90 activity in breast cancer cell lines have shown preclinical potential (Jensen et al. 2008, Terwisscha van Scheltinga et al. 2014). Interestingly, downregulation of IGF1R and VEGF-A secretion after Hsp90 inhibition by NVP-AUY922 in vivo was evident in both the MCF7 and MDA-MB-231 cell lines. Importantly, the study provides new evidence that loss of IGF1R and VEGF-A is involved in mediating an anti-tumor response, and they are potential biomarkers of effective Hsp90 therapeutic inhibition (Terwisscha van Scheltinga et al. 2014).

In a phase 1 trial, R1507, a humanized IGF1R mAb, has been shown to be tolerable in combination with chemotherapy plus the VEGF-A inhibitor bevacizumab (Table 2) (Mahadevan et al. 2014). Similarly, a phase 2 trial tested the safety and PK of AMG479 in conjunction with bevacizumab for advanced solid tumors (Tables 1 and 2: Ref #20, Nbib974896). Currently, there are no specific clinical trials testing IGF1R and VEGF combination therapies in TNBC, but a phase 2 trial completed in 2015 tested chemotherapy in conjunction with cixutumumab with and without bevacizumab for recurrent non-small-cell lung carcinoma (Tables 1 and 2: Ref #25, Nbib955305). Clearly, there is scope for the use of IGF1R and VEGF dual therapies and future testing is likely to have value for the improvement of available TNBC therapies.

IGF1R and IGF2

There is evidence that non-genomic activation of MAPK/ERK/ phosphoinositide 3-kinase (PI3K) and mTOR pathways by estrogen receptor beta (ERβ) in conjunction with IGF-2 is involved in regulating cancer growth in TNBC (Hamilton et al. 2015). This is proposed to occur via the activation of growth factor pathways such as VEGF and EGFR in addition to metabolic pathways. Specifically, a study involving shRNA knockdown of ERβ and treatment with the ERβ antagonist (PHTPP and ERβ agonist (DPN)) demonstrated that ERβ is involved in stimulating the proliferation of the MDA-MB-231 cell line (Hamilton et al. 2015). It has also been shown that high ERβ is expressed in TNBC and associated with worse prognostic outcomes (Hamilton et al. 2015). A combination therapy comprising the IGF-2 sequestering antibody MAB292 and topotecan to inhibit the hypoxia-inducible factor-1 enhances the anti-migratory effect of NVP-AEW541 in the TNBC MDA-MB-231 cell line (Mancini et al. 2014).

HER2 amplified breast cancer

In recent years, IGF1R activity has been shown to play a role in driving HER2 therapy trastuzumab resistance, and co-targeting HER2 and IGF1R is thought to be a viable therapeutic approach to overcome resistance (Browne et al. 2012). Mechanisms associated with IGF1R-driven HER2 therapy resistance are not well defined; however, there are studies suggesting that it is mediated via de-regulation of the PTEN/PI3K/AKT signaling pathway (Gallardo et al. 2012). Although PI3K activity is involved in HER2-therapy resistance, it has been proposed that HER2-overexpressing cancers treated with PI3K inhibitors should also be combined with HER2-targeted therapies such as trastuzumab or lapatinib to overcome the AKT-mediated activation of other tyrosine kinase growth factor receptors including IGF1R, InsR and HER3 as a result of PI3K inhibitor treatment (Table 2) (Chakrabarty et al. 2012). In addition, it has been shown that IGF1R-dependent cell motility in trastuzumab resistance occurs via the stimulation of FAK signaling and Forkhead box protein M1 (FoxM1), and co-targeting HER2 and IGF1R is seen to be beneficial mostly in trastuzumab-resistant cells (Sanabria-Figueroa et al. 2015).

A combination of figitumumab plus the HER2-targeted therapies, trastuzumab and neratinib (HER2 TKI), was shown to elicit a synergistic effect on enhancing cell apoptosis and reducing cell proliferation in the BT474 and MCF7 breast cancer cells lines (Table 2) (Chakraborty et al. 2015). Indirectly targeting IGF1R overexpression in HER2-amplified breast cancers by the introduction of miR-630 into the cell can restore the sensitivity of the cell to different HER2 inhibitors and may have clinical benefit to prevent IGF1R-driven HER2-therapy resistance (Corcoran et al. 2014). A separate study has similarly demonstrated that overexpression of miR-375 can restore the sensitivity of SKBR3 HER2-amplified cells to targeted HER2 therapy via an IGF1R-mediated mechanism (Ye et al. 2014).

Identifying better clinical biomarkers of HER2 therapy response is essential due to the high rate of HER2 therapy-resistant breast cancers that develop in individuals. Micro-RNAs (miRNAs) have been identified as short (18–25) nucleotide non-coding RNAs, which in effect can act as tumor suppressors in the cell by inhibiting gene expression. Most importantly, some miRNAs are known to be decreased in cancer vs normal cells and therefore re-introduction of miRNA back into the cell has therapeutic implications (Zhang et al. 2007). Specifically, overexpression of miR-630 has been shown to increase the degradation of both IGF1R mRNA and protein in pancreatic cancer cells (Farhana et al. 2013). In relation to IGF1R activity in HER2-expressing breast cancers, indirectly targeting IGF1R overexpression in HER2-amplified breast cancers by the introduction of miR-630 into the cell can restore the sensitivity of the cell to different HER2 inhibitors and may have clinical benefit to prevent IGF1R-driven HER2 therapy resistance (Corcoran et al. 2014). A separate study has similarly demonstrated that overexpression of miR-375 can restore the sensitivity of SKBR3 HER2-amplified cells to targeted HER2 therapy via an IGF1R-mediated mechanism (Ye et al. 2014). miR-630 is also reduced in the HER2 therapy-resistant MDA-MB-435 cell line compared with that in the HER2 therapy-sensitive SKBR3 cell line and in HER2-expressing tumors compared with matched peritumors, suggesting that miRNAs can be used as diagnostic and predictive markers of HER2 therapy response (Corcoran et al. 2014).

Similar to the bispecific antibodies that target both IGF1R and EGFR (Dong et al. 2011, Croasdale et al. 2012), a tetravalent bispecific antibody, MM-141, has been developed that targets both IGF1R and erbB3 (HER3), preventing IGF-1 and IGF-2 ligand binding to IGF1R and heregulin binding to erbB3, subsequent ligand-induced receptor phosphorylation, and p-AKT downstream signaling (Fitzgerald et al. 2014). MM-141 is more effective in reducing p-AKT than antibodies that target IGF1R and erbB3 individually and can enhance the anti-tumor effects of the chemotherapeutic agent, docetaxel, in DU145 prostate cancer xenografts (Table 2) (Fitzgerald et al. 2014). Given the potential clinical benefits of using MM-141 as a dual-targeting drug, a phase 1 clinical trial was completed in 2015, which tested the dose levels and frequency of MM-141 in patients with advanced solid cancers (Table 1: Ref #9, Nbib1733004). Using this type of therapy approach in combination with chemotherapy and/or other targeted therapies may be an effective strategy in preventing IGF1R-driven trastuzumab resistance in breast cancer and requires further clinical testing. A phase 2 trial completed in 2014, in women with HER2-positive breast cancers tested capecitabine and lapatinib with or without cixutumumab (Table 1: 
Ref #15, Nbib684983).

One study has demonstrated that high overexpression of the IGF1R-driven tyrosine kinase activated the phosphorylated form of Src (pY416), and is correlated with poorer overall survival (OS) and disease-free survival (DFS) in adjuvant trastuzumab therapy treated HER2-positive/hormone receptor negative breast cancers (Peiro et al. 2014). Thus, co-targeting strategies using Src inhibitors in conjunction with HER2-targeted therapies may be clinically effective for HER2-amplified cancers to eliminate IGF1R-mediated HER2 therapy resistance (Browne et al. 2012). At present, there are no clinical trials testing this type of combination therapy for breast cancer; however, a phase 2 trial testing the safety of IMC-A12 and the Src inhibitor, sorafenib, in combination for advanced liver cancer may give some further insight into the potential of combination therapy (Tables 1 and 2: Ref #27, Nbib906373). Given that there is some progress in targeting Src activity in cancer, combination therapies such as IMC-A12 and sorafenib in addition to trastuzumab may be effective as a prevention of IGF1R-driven HER2 therapy resistance. There is certainly future potential for investigating the involvement of IGF1R in HER2-driven cancers, and this is reflected in the progression of clinical trials in this therapeutic area of interest.

BRCA-mutated breast cancer

IGF1R and DNA-damaging agents

BRCA-mutated breast cancers are susceptible to the impairment of DNA damage repair by homologous recombination and interestingly are known to have high IGF-1 protein expression (Hudelist et al. 2007). Specific therapies aimed at targeting DNA repair factors include poly ADP-ribose polymerase (PARP) inhibitors (e.g. olaparib). Most important to the development of more effective IGF1R-targeted therapies, PARP regulation in the ER-positive MCF7 and HER2-positive BT474 cell lines is mediated via IGF1R (Kim et al. 2015). Preclinical support for the use of dual IGF1R and PARP inhibitors is highlighted by a study that showed an increased sensitivity to olaparib by the addition of the IGF1R inhibitor, BMS-536924, in BRCA1-mutated (MDA-MB-436 and HCC1937) compared with BRCA1 wild-type (BT20 and MDA-MB-231) TNBC cell lines (Table 2) (Amin et al. 2015). There is also compelling evidence that IGFBP-3 is involved in DNA damage repair, highlighting the need for further investigation of the role of IGF-1-signaling in the DNA repair process in breast cancer (Lin et al. 2014). There are no current clinical trials testing PARP inhibitors in conjunction with IGF1R-inhibitors; however, there is a phase 2 trial that commenced in 2016 designed to test the PARP inhibitor, veliparib, with chemotherapy agents cisplatin and vinorelbine ditartrate in TNBC, BRCA-mutated breast cancers (Clinical Trials Gov. Identifier; Nbib2595905, Table 2) and a phase 1 trial that was completed in 2015 for recurrent and/or metastatic breast cancer (Clinical Trials Gov. Identifier; Nbib1104259).

Downstream mediators of IGF1R signaling: potential therapeutic targets

Targeting IGF1R signaling is intrinsically difficult because it is not a singular pathway but comprises various downstream mediators that promote oncogenesis. Additionally, a number of other signaling receptors can also regulate many IGF1R downstream mediators, which make it difficult to attribute all the tumorigenic effects to IGF1R. The disappointing success of IGF1R-directed therapies is most certainly in part attributable to the complex interplay of growth factor signaling that exists in the cancer cell to provide for sustainable growth, migration and survival. As a number of growth factors can activate AKT/PI3K and MAPK signaling pathways, inhibition of IGF1R signaling may in fact be more effective by targeting its ‘activity’ (i.e. downstream mediator-targeted therapies inhibition) rather than IGF1R ‘expression’ or both as discussed in the following sections (Fig. 1).

Figure 1
Figure 1

Schematic representation of therapeutic targeting of IGF1R expression and activity in breast cancer. Insulin-like growth factor receptor (IGF1R) signaling is mediated after the formation of homo- and heterodimers of IGF1R and the insulin receptor (InsR) in response to the binding of their respective ligands (i.e. IGF1 and IGF2 to IGF1R and insulin to InsR. In the cytoplasm, IGF1/IGF2 form a complex with insulin growth factor-binding proteins (IGFBPs) to reduce the pool of available ligand that can bind to IGF1R. IGF1R activation leads to downstream cell signaling of effector molecules that regulate PI3K/AKT, FAK and MAPK/MEK intracellular signaling pathways. SphK1 is phosphorylated and activated by ERK leading to both intracellular and extracellular activity of the bioactive lipid molecular sphingosine 1 phosphate (SIP) (Pitson et al. 2003, Granata et al. 2007). IGF1R can be activated after SIP1 and SIP3 receptor-mediated phosphorylation (Martin et al. 2009). Targeted therapies have been developed to inhibit IGF1R expression including IGF1R monoclonal antibodies (mAbs), IGF1/2 mAbs and IGF1R/InsR tyrosine kinase inhibitors. IGF1R-directed cancer therapies that inhibit IGF1R downstream effector molecules have also been developed including PI3K; AKT; mTOR; mRNA translation; FAK; MEK and SphK1. Novel combination therapies aimed at targeting IGF1R expression and activity are developing this area of breast cancer therapeutics. Reprinted by permission from the American Association for Cancer Research: Iams WT & Lovly CM, Molecular pathways: clinical applications and future direction of insulin-like growth factor-1 receptor pathway blockade, Clinical Cancer Research, 30 Sep 2015, volume 21 issue 19, pages 4270 to 4277, doi:10.1158/1078-0432.ccr-14-2518.

Citation: Endocrine-Related Cancer 23, 11; 10.1530/ERC-16-0218

IGF1R signaling is directly involved in the regulation of downstream mediators involved in protein synthesis via activation of AKT/PI3K signaling (e.g. mammalian target of rapamycin complex 1 (mTORC1) and 4E-binding protein 1 (4E-BP1)), as well as apoptosis (e.g. BAD), cellular proliferation (i.e. Ras, MEK and ERK), altered integrin expression/cell motility (i.e. Src-focal adhesion kinase (FAK), RhoA/Rho-associated kinase (ROCK) and metabolic pathways (i.e. xCT (SLC7A11) and pyruvate kinase M2 (Yang & Yee 2014, Salani et al. 2015)). A variety of combinatorial therapies targeting IGF1R (NVP-AEW541), PI3K (NVP-BKM120), mTORC (KU0063794) and MEK (PD0325901) in the MDA-MB-231 cell line have shown synergistic effects in vitro by a reduction in cell growth (i.e. combination index (CI) < 1) in parallel with an increase in cell apoptosis and G0/G1 cell cycle arrest (Ayub et al. 2015). Importantly, these combinations have a potential to prevent the re-activation of these tyrosine kinase pathways and reduce toxicity by the administration of lower drug doses.

PI3K inhibitors

PI3K signaling is upregulated in an estimated 70% of breast cancers (Leroy et al. 2016), whether as a consequence of increased IGF1R (or EGFR) pathway activation, or secondary to oncogenic mutations in PIK3CA, reported to occur in about 25% of all breast cancers (Zardavas et al. 2014). IGF1R pathway activation is known to confer resistance to PI3K inhibition (Leroy et al. 2016), suggesting the possible utility of combined IGF1R/PI3K inhibition. A phase 1/2 clinical trial that commenced in 2012 has tested the effectiveness of a dual therapy comprising AMG479 and the 
PI3K-inhibitor, BYL719, in PIK3CA-mutated or -amplified solid cancers (Tables 1 and 2: Ref #7, Nbib1708161).

Using the MDA-MB-231 and MDA-MB-436 TNBC cell lines, a combination therapy comprising the PI3K inhibitor, SF1126 plus gefitinib, has shown some promising preclinical results (Table 2) (Deng et al. 2015). Dual inhibition of 
PI3K/AKT activity using PI-103 and EGFR activity by gefitinib or erlotinib showed enhanced combined drug effects on reducing cell viability and expression of anti-apoptotic proteins in basal-like SUM149PT and MDA-MB-468 breast cancer cell lines, but not for mesenchymal stem-like Hs578T and MDA-MB-231 cell lines (Table 2) (Yi et al. 2013). Further complexity in understanding how to therapeutically target IGF1R and its downstream mediators is highlighted by the ability of IGF1R to induce basal and estrogen-induced phosphorylation of the stress-activated protein kinase c-Jun (JNK) via PI3K activation (Fan et al. 2015). Whether co-treatment with IGF1R plus JNK-inhibitors such as SP600125 would be effective in overcoming anti-estrogen therapy resistance remains to be further tested (Fan et al. 2015).

AKT inhibitors

Given that PI3K is integral to downstream IGF1R-mediated AKT activity, targeting AKT as an alternative to PI3K has logically been pursued as a therapeutic target. A phase 1b study has demonstrated the clinical potential of the AKT inhibitor, MM-2206, combined with paclitaxal and trastuzumab in HER2-driven cancers (Table 2) (Chien et al. 2016). Moreover, the I-SPY 2 Trial has included the testing of the AKT inhibitor, MK-2206, with and without the HER-2 inhibitor, trastuzumab (Tables 1 and 2: Ref #1, Nbib1042379). Further, a phase 2 trial commenced in 2013 to test whether the co-therapies comprising MK-2206, anastrozole (post-menopausal women) and goserelin acetate (a gonadotropin-releasing hormone agonist; premenopausal women) is more effective in suppressing estrogen production in PI3KA mutant, ER-positive or HER2-negative breast cancers (Tables 1 and 2: Ref #4, Nbib1776008).

Completed clinical trials to test various AKT inhibitor combination therapies in breast cancer include phase 1 trials testing the maximum tolerated doses (MTD) and dose-limiting toxicities (DLT) of MK-2206 combined with the dual HER2/EGFR inhibitor, lapatinib ditosylate (Tables 1 and 2: Ref #14, Nbib1245205), or trastuzumab which was shown to be associated with good safety and clinical outcomes (Tables 1 and 2: Ref #18, Nbib963547) (Hudis et al. 2013). At present, there are no clinical trials testing IGF1R and AKT dual inhibition, which may be more effective than just targeting the downstream effects of IGF1R activity.

FAK inhibitors

The IGF signaling pathway is well established to regulate cell migration in breast cancer via the activation of FAK and RhoA/Rho-associated kinase (ROCK), as further discussed in subsequent sections; therefore, co-targeting IGF1R and FAK activation is potentially a viable therapeutic approach (Zhang et al. 2005, Taliaferro-Smith et al. 2015). As with many other oncogenic signaling pathways, there is often crosstalk and/or reciprocal regulation that has been reported for IGF1R and FAK in TNBC (Taliaferro-Smith et al. 2015). Interestingly, inhibiting both IGF1R and FAK has shown effective anti-tumor effects in a number of different cancers including esophageal adenocarcinoma and glioma (Liu et al. 2007, Watanabe et al. 2008). The specific 
ATP-competitive tyrosine kinase small molecule inhibitor TAE226 that targets both IGF1R and FAK has been used to inhibit MDA-MB-231 breast cancer bone metastasis (Kurio et al. 2011). There is evidence from MDA-MB-231 cells that IGF-1 stimulation leads to the cellular re-distribution of FAK from focal adhesion proteins to regulate cell motility, which can be blocked using the ROCK inhibitor, Y-27632 (Zhang et al. 2005). Moreover, a recent study supports the potential benefit of using IGF1R and FAK co-targeted therapies in TNBC by the effective suppression of epithelial-to-mesenchymal transition, cell migration and invasion (Table 2) (Taliaferro-Smith et al. 2015). Currently, there are no clinical trials testing IGF1R and FAK inhibitor dual therapies for breast cancer, yet given the literature, there is certainly some worth in pursuing this therapy approach.

mTOR inhibitors/mRNA translation inhibitors

Targeting IGF1R downstream activators involved in the regulation of mRNA translation using therapies such as the anti-viral drug ribavirin, the antisense oligonucleotide 4E-ASO, the small molecule inhibitor, 4EGI-1, and the natural inhibitor, silvestrol (Silvera et al. 2010, Pettersson et al. 2011) is an emerging therapeutic window of opportunity, which can interrogate many converging oncogenic signaling pathways and is undergoing extensive preclinical and clinical testing (Clinical Trials Gov. Identifier; Nbib1309490) (Drygin et al. 2011, Devlin et al. 2015).

IGF-1 signaling downstream mediators contribute to cancer growth via the regulation of cap-dependent mRNA translation primarily by the activation of mTOR and S6K1 to initiate the activation of the translational modulators 4EBP1 and eIF4E (Sonenberg & Hinnebusch 2009).

Recently published novel findings highlight that the IGF-1 downstream signaling human oncogene, amplified in breast cancer 1 (AIB1), possesses cancer-promoting effects potentially via positively regulating global polyribosome mRNA recruitment in ER-positive MCF7L and ER-negative, MDA-MB-231 and MDA-MB-435/LCC6 breast cancer cells (Anzick et al. 1997, Ochnik et al. 2016). Importantly, these findings highlight the need for a better identification of potential therapeutic targets in ‘IGF-expressing’ breast cancers. In relation to therapeutically inhibiting mTOR and suppressing IGF-induced mRNA translation, it has been shown that basal-like TNBC breast cancers respond synergistically (i.e. CI < 1) to co-treatment of the mTOR inhibitor, temsirolimus and gefitinib by reduced phosphorylation of the translational mediator, eIF4B (Table 2) (Madden et al. 2014). Briefly, the in vitro study demonstrated, using a luciferase-based reporter assay system, that cap-dependent mRNA translation was reduced by the co-treatment with temsirolimus and gefitinib compared with the individual drugs (Madden et al. 2014).

A phase 1/2 clinical trial aims to test the associated side effects and MTD of temsirolimus plus cixutumumab in metastatic or locally recurring breast cancer (Table 1: Ref #8, Nbib699491) in addition to survival outcomes of temsirolimus plus the tyrosine kinase inhibitor (neratinib) in triple-negative, HER2-amplified breast cancers (Tables 1 and 2: Ref #5, Nbib1111825). A phase 1 breast cancer trial has been completed testing ridaforolimus in conjunction with either MK-2206 or the notch inhibitor, MK-0752 (Tables 1 and 2: Ref #11, Nbib1295632) (Gupta et al. 2015, Piha-Paul et al. 2015). The development of these dual-targeting therapies is advancing the knowledge required to eventually underpin therapies that are working and therapies that, despite the promising basic science findings, do not show the same clinical benefit.

MEK inhibitors

Targeting the anti-apoptotic actions of the IGF-1 downstream effector, MEK-1, using small molecule inhibitors including PD98059 and U0126 in breast cancer in conjunction with anti-hormonal therapies has been shown to be effective in blocking IGF-dependent survival in ER-positive cells (Table 2) (Periyasamy-Thandavan et al. 2012). Dual blockade of MEK and EGFR using the MEK inhibitor, selumetinib and gefitinib, has been shown to be potentially of value for a subset of TNBC (Table 2) (Maiello et al. 2015). Future studies investigating these types of dual therapies have merit; yet, the authors note that without sufficient biomarkers of efficacy, there will be many individuals who will not respond. Conversely, the WNT pathway element, DVL3, can potentially suppress the response of anti-IGF1R-targeted drugs via MEK-ERK regulation in cancer (Gao et al. 2014). The study reports improved the clinical outcomes for individuals expressing low–absent DVL3 expression after figitumumab or AVE164 therapy and highlights that there is a need for positive and negative IGF1R therapy response biomarkers to be identified (Gao et al. 2014).

All breast cancer molecular subtypes

The safety and tolerability of the IGF1R monoclonal antibody R1507 in combination with several standard oncology regimens have been shown to be manageable in a variety of advanced solid malignancies including breast (Table 2) (Mahadevan et al. 2014). Different combination therapies including cixutumumab have been studied in a phase 1b trial to test the tolerability in solid cancers with the following therapies: (1) gemcitabine plus erlotinib; (2) paclitaxel plus bevacizumab; (3) carboplatin plus etoposide; (4) mFOLFOX6 (combination chemotherapy) plus bevacizumab; (5) capecitabine plus trastuzumab and (6) sorafenib. Based on the findings that they were well tolerated, with some individuals presenting with a lasting response for a median of 29 weeks, the combination treatments are potentially viable for a subset of cancers (Mahadevan et al. 2014).

For advanced, previously treated cancers that are known to overexpress IGF1R, a phase 1 study that commenced in 2014 has been designed to evaluate the disease response and toxicities of the chemotherapy drug IGF-methotrexate, which binds specifically to the IGF1R (Table 1: Ref #3, Nbib2045368) (McTavish et al. 2009). mTOR inhibition using ridaforolimus in conjunction with the IGF1R-inhibitor MK0646 in a phase 1 trial for advanced cancer has also shown promising results and again highlights the need for further novel approaches to target IGF1R expression and/or its activity (Tables 1 and 2: Ref #23, Nbib730379) (Di Cosimo et al. 2015).

Emerging IGF1R co-related oncogenic pathway: sphingosine kinase signaling

IGFBP-3 in breast tumors has the potential to influence IGF1R-dependent survival pathways because it has been shown to potentiate IGF1R signaling, through a mechanism involving the activation of the oncogenic lipid kinase, sphingosine kinase 1 (SphK1) and generation of the intracellular second messenger prosurvival lipid, sphingosine 1-phosphate (SIP) (Martin et al. 2009). SIP has both intracellular activity, the targets of which are not well defined, and extracellular oncogenic activity after binding to SIP receptors 1–5 in the cell membrane to activate various G-protein-responsive pathways involved in cell proliferation, migration and survival (Hannun & Obeid 2008). It has been reported that activation of ERK by growth factors such as IGF-1 leads to SphK1 phosphorylation on residue Ser225 and in effect enhances intracellular production of SIP (Pitson et al. 2003, Granata et al. 2007). As IGF-1 signaling can regulate SphK1 via phosphorylation, it may even be considered as a downstream mediator of IGF1R activity (Fig. 1).

Both sphingolipid signaling (Wang et al. 2010b) and IGF1R activation (Beckwith & Yee 2014) have been shown in many studies to contribute to the resistance of a variety of cancer chemotherapies and increase sensitivity/response to doxorubicin (Fu et al. 2015, de Groot et al. 2016), yet in-depth studies identifying co-dependence of IGF1R and sphingolipid signaling to therapy responsiveness are lacking. In contrast, co-treatment strategies targeting SphK1 and EGFR have shown exciting preclinical success and are currently being investigated for TNBC (Martin et al. 2014). Recently, SphK1 mRNA and protein expression have been shown to be elevated in TNBC compared with non-adjacent non-cancerous breast tissue as well as in the MDA-MB-231, Hs578T and MDA-MB-436 TNBC compared with non-TNBC cell lines (Li et al. 2016). Not only was SphK1 expression higher in TNBC; the increased serum levels of SIP were also evident in breast cancer patients compared with healthy donor individuals (Li et al. 2016). SphK1 siRNA knockdown in the MDA-MB-231 and Hs578T cell lines is effective in reducing cell proliferation, migration and apoptosis in vitro, potentially via the downregulation of PI3K/AKT signaling (Li et al. 2016). In terms of the prognostic significance of IGF1R and SphK1, poorer DFS and OS are evident in women with both ER-positive and ER-negative breast cancers expressing high IGF1R and SphK1 mRNA compared with single-gene analysis, highlighting these kinases as dual therapeutic targets (Fig. 2 and Table 2).

Figure 2
Figure 2

Prognostic significance of IGF1R and SphK1 co-expression in ER-positive and ER-negative breast cancers. Kaplan–Meier (KM) mRNA survival analysis was performed as follows in: (A) estrogen receptor (ER)-positive and (B) ER-negative, lymph node-negative breast cancers after the stratification for high IGF1R and SphK1 mRNA co-expression to determine disease-free survival (DFS) and overall survival (OS) probability (http://glados.ucd.ie/BreastMark/). (C) Table includes hazard ratio (HR) and log-rank P values for DFS and OS of IGF1R and SphK1 individual gene KM analysis.

Citation: Endocrine-Related Cancer 23, 11; 10.1530/ERC-16-0218

A S1P receptor modulator that also shows SphK1 inhibitory activity (Novartis; fingolimod, FTY-720) has been FDA approved as an immunosuppressive agent for multiple sclerosis, and has also been suggested to exhibit anticancer activity (Pitman et al. 2012). Specifically in relation to cancer, a number of different SphK inhibitors have been developed and shown to elicit anticancer effects including SKI-II, ABC294640 and MP-A08 (Pitman & Pitson 2010, Pitman et al. 2015, McNaughton et al. 2016). Currently, SphK1 inhibitors are not clinically in testing for breast cancer, but the preclinical and prognostic findings of a relationship between IGF1R and SphK1 expression provide evidence that there is a need to learn more about the potential clinical benefits of IGF1R and SphK1 as novel dual therapeutic targets (Fig. 1).

Future IGF-targeted therapies: where to now

This review has aimed to provide a comprehensive assessment of the current testing of IGF1R-directed therapies for breast cancer based on the different molecular subtypes. In some instances, preclinical studies and clinical trials that have tested IGF1R-therapies in other solid cancers and cell lines have been discussed as a platform to potentially translate through as therapies that may also be beneficial to IGF1R-expressing breast cancers. Since IGF-targeted therapies have led to a large percentage of negative clinical trial results, there is now a greater need for the next wave of IGF1R-directed clinical trials to be better designed based on preclinical findings and current clinical trials.

It is increasingly clear that IGF-signaling plays a cancer-promoting role not only in the most widely tested ER-positive breast cancers but also in some TNBC, HER2-positive and BRCA-mutated breast cancers, and all are potentially important in terms of anti-IGF1R combination therapy clinical testing. As there has been a very low success rate for anti-IGF1R monotherapies, it is difficult to further predict the success of many of the preclinical and current clinical trials. However, a list of the preclinical and clinical IGF1R-targeted combination therapies discussed in this review based on breast cancer molecular subtype has now been summarized as a guide and suggestion for future clinical testing directions based on breast cancer subtype (Table 2). In terms of combination therapies that should be prioritized, we have made note of the preclinical studies that are of particular interest and worth for possible future clinical testing.

Conclusion

The vast array of potential ways of combining IGF1R-targeted therapies with other existing and emerging therapies is developing rapidly and likely to be clinically important. In light of the high degree of heterogeneity that exists in breast cancer, between and within molecular subtypes, it is becoming increasingly difficult to design effective targeted therapies with broad applicability. The complexity of cancer cell signaling pathways, and how their re-activation can occur to overcome the inhibitory actions of breast cancer therapies, has in effect led to new molecular forms of treatment-resistant breast cancer. Given that we are now learning more about the potential pitfalls associated with many therapeutic approaches for breast cancer that lead to therapy resistance, the next wave of research aims to consolidate this information to ultimately underpin more effective ways of targeting key oncogenic signaling pathways to prevent and/or treat therapy resistance.

Multiple lines of evidence indicate that IGF1R-directed therapies are important and the vast amount of literature supporting the integral role of IGF-1 signaling in breast cancer should not be ignored despite the lack of clinical success of IGF1R-targeted monotherapies. In essence, this lack of success only serves to highlight that IGF1R signaling is more complicated than initially understood, and most certainly requires concurrent combination therapies targeted to other growth factor pathways (e.g. HER2/HER3, EGFR, VEGF, and InsR), its downstream mediators (e.g. PI3K, mTOR, eIF4E, FAK) and/or co-related signaling pathways (e.g. SphK1), to be clinically effective. Current and future clinical trials will work toward providing more effective IGF-directed targeting strategies.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

Aleksandra Ochnik was supported by research fellowship funding from Cancer Institute NSW through the Sydney Vital Translational Cancer Research Centre. Grant funding to Robert Baxter from Cancer Council NSW is gratefully acknowledged.

References

  • Adelaide J, Finetti P, Bekhouche I, Repellini L, Geneix J, Sircoulomb F, Charafe-Jauffret E, Cervera N, Desplans J & Parzy D et al. 2007 Integrated profiling of basal and luminal breast cancers. Cancer Research 67 1156511575. (doi:10.1158/0008-5472.CAN-07-2536)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Amin O, Beauchamp MC, Nader PA, Laskov I, Iqbal S, Philip CA, Yasmeen A & Gotlieb WH 2015 Suppression of Homologous Recombination by insulin-like growth factor-1 inhibition sensitizes cancer cells to PARP inhibitors. BMC Cancer 15 817. (doi:10.1186/s12885-015-1803-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Andre F, Slimane K, Bachelot T, Dunant A, Namer M, Barrelier A, Kabbaj O, Spano JP, Marsiglia H & Rouzier R et al. 2004 Breast cancer with synchronous metastases: trends in survival during a 14-year period. Journal of Clinical Oncology 22 33023308. (doi:10.1200/JCO.2004.08.095)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anzick SL, Kononen J, Walker RL, Azorsa DO, Tanner MM, Guan XY, Sauter G, Kallioniemi OP, Trent JM & Meltzer PS 1997 AIB1, a steroid receptor coactivator amplified in breast and ovarian cancer. Science 277 965968. (doi:10.1126/science.277.5328.965)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Awasthi N, Zhang C, Ruan W, Schwarz MA & Schwarz RE 2012 BMS-754807, a small-molecule inhibitor of insulin-like growth factor-1 receptor/insulin receptor, enhances gemcitabine response in pancreatic cancer. Molecular Cancer Therapeutics 11 26442653. (doi:10.1158/1535-7163.MCT-12-0447)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ayub A, Yip WK & Seow HF 2015 Dual treatments targeting IGF-1R, PI3K, mTORC or MEK synergize to inhibit cell growth, induce apoptosis, and arrest cell cycle at G1 phase in MDA-MB-231 cell line. Biomedicine and Pharmacotherapy 75 4050. (doi:10.1016/j.biopha.2015.08.031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bahhnassy A, Mohanad M, Shaarawy S, Ismail MF, El-Bastawisy A, Ashmawy AM & Zekri AR 2015a Transforming growth factor-beta, insulin-like growth factor I/insulin-like growth factor I receptor and vascular endothelial growth factor-A: prognostic and predictive markers in triple-negative and non-triple-negative breast cancer. Molecular Medicine Reports 12 851864. (doi:10.3892/mmr.2015.3560)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bahnassy A, Mohanad M, Ismail MF, Shaarawy S, El-Bastawisy A & Zekri AN 2015b Molecular biomarkers for prediction of response to treatment and survival in triple negative breast cancer patients from Egypt. Experimental and Molecular Pathology 99 303311. (doi:10.1016/j.yexmp.2015.07.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA & Esserman LJ 2009 I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clinical Pharmacology & Therapeutics 86 97100. (doi:10.1038/clpt.2009.68)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baxter RC 2014 IGF binding proteins in cancer: mechanistic and clinical insights. Nature Reviews Cancer 14 329341. (doi:10.1038/nrc3720)

  • Becker MA, Ibrahim YH, Cui X, Lee AV & Yee D 2011 The IGF pathway regulates ERalpha through a S6K1-dependent mechanism in breast cancer cells. Molecular Endocrinology 25 516528. (doi:10.1210/me.2010-0373)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Beckwith H & Yee D 2014 Insulin-like growth factors, insulin, and growth hormone signaling in breast cancer: implications for targeted therapy. Endocrine Practice 20 12141221. (doi:10.4158/EP14208.RA)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Belani CP, Dahlberg SE, Rudin CM, Fleisher M, Chen HX, Takebe N, Ramalingam SS & Schiller JH 2013 Three-arm randomized phase II study of cisplatin and etoposide (CE) versus CE with either vismodegib (V) or cixutumumab (Cx) for patients with extensive stage-small cell lung cancer(ES-SCLC) (ECOG 1508). Journal of Clinical Oncology 31 abstract 7508.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Belfiore A, Frasca F, Pandini G, Sciacca L & Vigneri R 2009 Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocrine Reviews 30 586623. (doi:10.1210/er.2008-0047)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhargava R, Beriwal S, McManus K & Dabbs DJ 2011 Insulin-like growth factor receptor-1 (IGF-1R) expression in normal breast, proliferative breast lesions, and breast carcinoma. Applied Immunohistochemistry & Molecular Morphology 19 218225. (doi:10.1097/pai.0b013e3181ffc58c)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bohula EA, Playford MP & Macaulay VM 2003 Targeting the type 1 insulin-like growth factor receptor as anti-cancer treatment. Anticancer Drugs 14 669682. (doi:10.1097/00001813-200310000-00001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brockhoff M, Hau JC, Fontana P, Zimmermann C, Pover AD, Erdmann D & Chene P 2012 Factors influencing the inhibition of protein kinases. Journal of Enzyme Inhibition and Medicinal Chemistry 27194200. (doi:10.3109/14756366.2011.583922)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Browne BC, Eustace AJ, Kennedy S, O’Brien NA, Pedersen K, McDermott MS, Larkin A, Ballot J, Mahgoub T & Sclafani F et al. 2012 Evaluation of IGF1R and phosphorylated IGF1R as targets in HER2-positive breast cancer cell lines and tumours. Breast Cancer Research and Treatment 136 717727. (doi:10.1007/s10549-012-2260-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Burtrum D, Zhu Z, Lu D, Anderson DM, Prewett M, Pereira DS, Bassi R, Abdullah R, Hooper AT & Koo H et al. 2003 A fully human monoclonal antibody to the insulin-like growth factor I receptor blocks ligand-dependent signaling and inhibits human tumor growth in vivo. Cancer Research 63 89128921.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Camirand A, Zakikhani M, Young F & Pollak M 2005 Inhibition of insulin-like growth factor-1 receptor signaling enhances growth-inhibitory and proapoptotic effects of gefitinib (Iressa) in human breast cancer cells. Breast Cancer Research 7 R570R579.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carboni JM, Wittman M, Yang Z, Lee F, Greer A, Hurlburt W, Hillerman S, Cao C, Cantor GH & Dell-John J et al. 2009 BMS-754807, a small molecule inhibitor of insulin-like growth factor-1R/IR. Molecular Cancer Therapeutics 8 33413349. (doi:10.1158/1535-7163.MCT-09-0499)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Castano Z, Marsh T, Tadipatri R, Kuznetsov HS, Al-Shahrour F, Paktinat M, Greene-Colozzi A, Nilsson B, Richardson AL & McAllister SS 2013 Stromal EGF and igf-I together modulate plasticity of disseminated triple-negative breast tumors. Cancer Discovery 3 922935. (doi:10.1158/2159-8290.CD-13-0041)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chakrabarty A, Sanchez V, Kuba MG, Rinehart C & Arteaga CL 2012 Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. PNAS 109 27182723. (doi:10.1073/pnas.1018001108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chakraborty AK, Zerillo C & DiGiovanna MP 2015 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 152 533544. (doi:10.1007/s10549-015-3504-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chien AJ, Cockerill A, Fancourt C, Schmidt E, Moasser MM, Rugo HS, Melisko ME, Ko AH, Kelley RK & Korn WM et al. 2016 A phase 1b study of the Akt-inhibitor MK-2206 in combination with weekly paclitaxel and trastuzumab in patients with advanced HER2-amplified solid tumor malignancies. Breast Cancer Research and Treatment 155 521530. (doi:10.1007/s10549-016-3701-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cohen BD, Baker DA, Soderstrom C, Tkalcevic G, Rossi AM, Miller PE, Tengowski MW, Wang F, Gualberto A & Beebe JS et al. 2005 Combination therapy enhances the inhibition of tumor growth with the fully human anti-type 1 insulin-like growth factor receptor monoclonal antibody CP-751,871. Clinical Cancer Research 1120632073. (doi:10.1158/1078-0432.CCR-04-1070)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Corcoran C, Rani S, Breslin S, Gogarty M, Ghobrial IM, Crown J & O’Driscoll L 2014 miR-630 targets IGF1R to regulate response to HER-targeting drugs and overall cancer cell progression in HER2 over-expressing breast cancer. Molecular Cancer 13 71. (doi:10.1186/1476-4598-13-71)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Creighton CJ, Casa A, Lazard Z, Huang S, Tsimelzon A, Hilsenbeck SG, Osborne CK & Lee AV 2008 Insulin-like growth factor-I activates gene transcription programs strongly associated with poor breast cancer prognosis. Journal of Clinical Oncology 26 40784085. (doi:10.1200/JCO.2007.13.4429)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Croasdale R, Wartha K, Schanzer JM, Kuenkele KP, Ries C, Mayer K, Gassner C, Wagner M, Dimoudis N & Herter S et al. 2012 Development of tetravalent IgG1 dual targeting IGF-1R-EGFR antibodies with potent tumor inhibition. Archives of Biochemistry and Biophysics 526 206218. (doi:10.1016/j.abb.2012.03.016)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Davison Z, de Blacquiere GE, Westley BR & May FE 2011 Insulin-like growth factor-dependent proliferation and survival of triple-negative breast cancer cells: implications for therapy. Neoplasia 13 504515. (doi:10.1593/neo.101590)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dawood S, Broglio K, Buzdar AU, Hortobagyi GN & Giordano SH 2010 Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: an institutional-based review. Journal of Clinical Oncology 28 9298. (doi:10.1200/JCO.2008.19.9844)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Bono JS, Attard G, Adjei A, Pollak MN, Fong PC, Haluska P, Roberts L, Melvin C, Repollet M & Chianese D et al. 2007 Potential applications for circulating tumor cells expressing the insulin-like growth factor-I receptor. Clinical Cancer Research 13 36113616. (doi:10.1158/1078-0432.CCR-07-0268)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • de Groot S, Charehbili A, van Laarhoven HW, Mooyaart AL, Dekker-Ensink NG, van de Ven S, Janssen LG, Swen JJ, Smit VT & Heijns JB et al. 2016 Insulin-like growth factor 1 receptor expression and IGF1R 3129G > T polymorphism are associated with response to neoadjuvant chemotherapy in breast cancer patients: results from the NEOZOTAC trial (BOOG 2010-01). Breast Cancer Research 18 3. (doi:10.1186/s13058-015-0663-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deng M, Wang J, Chen Y, Zhang L & Liu D 2015 Combination of SF1126 and gefitinib induces apoptosis of triple-negative breast cancer cells through the PI3K/AKT-mTOR pathway. Anticancer Drugs 26 422427. (doi:10.1097/CAD.0000000000000202)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Devlin JR, Hannan KM, Hein N, Cullinane C, Kusnadi E, Ng PY, George AJ, Shortt J, Bywater MJ & Poortinga G et al. 2015 Combination therapy targeting ribosome biogenesis and mRNA translation synergistically extends survival in MYC-driven lymphoma. Cancer Discovery 6 5970. (doi:10.1158/2159-8290.CD-14-0673)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Di Cosimo S, Sathyanarayanan S, Bendell JC, Cervantes A, Stein MN, Brana I, Roda D, Haines BB, Zhang T & Winter CG et al. 2015 Combination of the mTOR inhibitor ridaforolimus and the anti-IGF1R monoclonal antibody dalotuzumab: preclinical characterization and phase I clinical trial. Clinical Cancer Research 21 4959. (doi:10.1158/1078-0432.CCR-14-0940)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dong J, Sereno A, Aivazian D, Langley E, Miller BR, Snyder WB, Chan E, Cantele M, Morena R & Joseph IB et al. 2011 A stable IgG-like bispecific antibody targeting the epidermal growth factor receptor and the type I insulin-like growth factor receptor demonstrates superior anti-tumor activity. mAbs 3 273288. (doi:10.4161/mabs.3.3.15188)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Drygin D, Lin A, Bliesath J, Ho CB, O’Brien SE, Proffitt C, Omori M, Haddach M, Schwaebe MK & Siddiqui-Jain A et al. 2011 Targeting RNA polymerase I with an oral small molecule CX-5461 inhibits ribosomal RNA synthesis and solid tumor growth. Cancer Research 71 14181430. (doi:10.1158/0008-5472.CAN-10-1728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fagan DH & Yee D 2008 Crosstalk between IGF1R and estrogen receptor signaling in breast cancer. Journal of Mammary Gland Biology and Neoplasia 13 423429. (doi:10.1007/s10911-008-9098-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fan P, Cunliffe HE, Maximov PY, Agboke FA, McDaniel RE, Zou X, Ramos P, Russell ML & Jordan VC 2015 Integration of downstream signals of insulin-like growth factor-1 receptor by endoplasmic reticulum stress for estrogen-induced growth or apoptosis in breast cancer cells. Molecular Cancer Research 13 13671376. (doi:10.1158/1541-7786.MCR-14-0494)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Farhana L, Dawson MI, Murshed F, Das JK, Rishi AK & Fontana JA 2013 Upregulation of miR-150* and miR-630 induces apoptosis in pancreatic cancer cells by targeting IGF-1R. PLoS ONE 8 e61015. (doi:10.1371/journal.pone.0061015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fitzgerald JB, Johnson BW, Baum J, Adams S, Iadevaia S, Tang J, Rimkunas V, Xu L, Kohli N & Rennard R et al. 2014 MM-141, an IGF-IR- and ErbB3-directed bispecific antibody, overcomes network adaptations that limit activity of IGF-IR inhibitors. Molecular Cancer Therapeutics 13 410425. (doi:10.1158/1535-7163.MCT-13-0255)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Friedbichler K, Hofmann MH, Kroez M, Ostermann E, Lamche HR, Koessl C, Borges E, Pollak MN, Adolf G & Adam PJ 2014 Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand-neutralizing antibody, and mechanistic rationale for combination with rapamycin. Molecular Cancer Therapeutics 13 399409. (doi:10.1158/1535-7163.MCT-13-0598)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fu P, Du F, Liu Y, Hong Y, Yao M & Zheng S 2015 Wogonin increases doxorubicin sensitivity by down-regulation of IGF-1R/AKT signaling pathway in human breast cancer. Cellular and Molecular Biology 61 123127.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gallardo A, Lerma E, Escuin D, Tibau A, Munoz J, Ojeda B, Barnadas A, Adrover E, Sanchez-Tejada L & Giner D et al. 2012 Increased signalling of EGFR and IGF1R, and deregulation of PTEN/PI3K/Akt pathway are related with trastuzumab resistance in HER2 breast carcinomas. British Journal of Cancer 106 13671373. (doi:10.1038/bjc.2012.85)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gao S, Bajrami I, Verrill C, Kigozi A, Ouaret D, Aleksic T, Asher R, Han C, Allen P & Bailey D et al. 2014 Dsh homolog DVL3 mediates resistance to IGFIR inhibition by regulating IGF-RAS signaling. Cancer Research 74 58665877. (doi:10.1158/0008-5472.CAN-14-0806)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Garcia-Echeverria C, Pearson MA, Marti A, Meyer T, Mestan J, Zimmermann J, Gao J, Brueggen J, Capraro HG & Cozens R et al. 2004 In vivo antitumor activity of NVP-AEW541-A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell 5 231239. (doi:10.1016/S1535-6108(04)00051-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Girnita A, Girnita L, del Prete F, Bartolazzi A, Larsson O & Axelson M 2004 Cyclolignans as inhibitors of the insulin-like growth factor-1 receptor and malignant cell growth. Cancer Research 64 236242. (doi:10.1158/0008-5472.CAN-03-2522)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goetsch L, Gonzalez A, Leger O, Beck A, Pauwels PJ, Haeuw JF & Corvaia N 2005 A recombinant humanized anti-insulin-like growth factor receptor type I antibody (h7C10) enhances the antitumor activity of vinorelbine and anti-epidermal growth factor receptor therapy against human cancer xenografts. International Journal of Cancer 113 316328. (doi:10.1002/ijc.20543)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gong Y, Yao E, Shen R, Goel A, Arcila M, Teruya-Feldstein J, Zakowski MF, Frankel S, Peifer M & Thomas RK et al. 2009 High expression levels of total IGF-1R and sensitivity of NSCLC cells in vitro to an anti-IGF-1R antibody (R1507). PLoS ONE 4 e7273. (doi:10.1371/journal.pone.0007273)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gradishar WJ, Yardley D, Layman RM, Sparano JA, Chuang E, Northfelt DW, Schwartz GN, Youssoufian H, Tang S & Novosiadly R et al. 2015 Clinical and translational results of a phase 2, randomized trial of an anti-IGF-1R (cixutumumab) in women with breast cancer that progressed on endocrine therapy. Clinical Cancer Research 22 301309. (doi:10.1158/1078-0432.CCR-15-0588)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Granata R, Trovato L, Lupia E, Sala G, Settanni F, Camussi G, Ghidoni R & Ghigo E 2007 Insulin-like growth factor binding protein-3 induces angiogenesis through IGF-I- and SphK1-dependent mechanisms. Journal of Thrombosis and Haemostasis 5 835845. (doi:10.1111/j.1538-7836.2007.02431.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greenall SA, Bentley JD, Pearce LA, Scoble JA, Sparrow LG, Bartone NA, Xiao X, Baxter RC, Cosgrove LJ & Adams TE 2013 Biochemical characterization of individual human glycosylated pro-insulin-like growth factor (IGF)-II and big-IGF-II isoforms associated with cancer. Journal of Biological Chemistry 288 5968. (doi:10.1074/jbc.M112.432013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Guix M, Faber AC, Wang SE, Olivares MG, Song Y, Qu S, Rinehart C, Seidel B, Yee D & Arteaga CL et al. 2008 Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cells is mediated by loss of IGF-binding proteins. Journal of Clinical Investigation 118 26092619. (doi:10.1172/jci34588)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gupta S, Argiles G, Munster PN, Hollebecque A, Dajani O, Cheng JD, Wang R, Swift A, Tosolini A & Piha-Paul SA 2015 A phase I trial of combined ridaforolimus and MK-2206 in patients with advanced malignancies. Clinical Cancer Research 21 52355244. (doi:10.1158/1078-0432.CCR-15-0180)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haluska P, Menefee M, Plimack ER, Rosenberg J, Northfelt D, LaVallee T, Shi L, Yu XQ, Burke P & Huang J et al. 2014 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 20 47474757. (doi:10.1158/1078-0432.CCR-14-0114)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hamilton N, Marquez-Garban D, Mah V, Fernando G, Elshimali Y, Garban H, Elashoff D, Vadgama J, Goodglick L & Pietras R 2015 Biologic roles of estrogen receptor-beta and insulin-like growth factor-2 in triple-negative breast cancer. BioMed Research International 2015 925703. (doi:10.1155/2015/925703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hannun YA & Obeid LM 2008 Principles of bioactive lipid signalling: lessons from sphingolipids. Nature Reviews Molecular Cell Biology 9 139150. (doi:10.1038/nrm2329)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hartog H, Horlings HM, van der Vegt B, Kreike B, Ajouaou A, van de Vijver MJ, Marike Boezen H, de Bock GH, van der Graaf WT & Wesseling J 2011 Divergent effects of insulin-like growth factor-1 receptor expression on prognosis of estrogen receptor positive versus triple negative invasive ductal breast carcinoma. Breast Cancer Research and Treatment 129 725736. (doi:10.1007/s10549-010-1256-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hou X, Huang F, Macedo LF, Harrington SC, Reeves KA, Greer A, Finckenstein FG, Brodie A, Gottardis MM & Carboni JM et al. 2011 Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hormonal agents in treatment of estrogen-dependent breast cancer. Cancer Research 71 75977607. (doi:10.1158/0008-5472.CAN-11-1080)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang F, Greer A, Hurlburt W, Han X, Hafezi R, Wittenberg GM, Reeves K, Chen J, Robinson D & Li A et al. 2009 The mechanisms of differential sensitivity to an insulin-like growth factor-1 receptor inhibitor (BMS-536924) and rationale for combining with EGFR/HER2 inhibitors. Cancer Research 69 161170. (doi:10.1158/0008-5472.CAN-08-0835)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hudelist G, Wagner T, Rosner M, Fink-Retter A, Gschwantler-Kaulich D, Czerwenka K, Kroiss R, Tea M, Pischinger K & Kostler WJ et al. 2007 Intratumoral IGF-I protein expression is selectively upregulated in breast cancer patients with BRCA1/2 mutations. Endocrine-Related Cancer 14 10531062. (doi:10.1677/ERC-06-0075)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hudis C, Swanton C, Janjigian YY, Lee R, Sutherland S, Lehman R, Chandarlapaty S, Hamilton N, Gajria D & Knowles J et al. 2013 A phase 1 study evaluating the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors. Breast Cancer Research 15 R110. (doi:10.1186/bcr3577)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iams WT & Lovly CM 2015 Molecular pathways: clinical applications and future direction of insulin-like growth factor-1 receptor pathway blockade. Clinical Cancer Research 21 42704277. (doi:10.1158/1078-0432.ccr-14-2518)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jensen MR, Schoepfer J, Radimerski T, Massey A, Guy CT, Brueggen J, Quadt C, Buckler A, Cozens R & Drysdale MJ et al. 2008 NVP-AUY922: a small molecule HSP90 inhibitor with potent antitumor activity in preclinical breast cancer models. Breast Cancer Research 10 R33. (doi:10.1186/bcr1996)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jeselsohn R, Buchwalter G, De Angelis C, Brown M & Schiff R 2015 ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer. Nature Reviews Clinical Oncology 12 573583. (doi:10.1038/nrclinonc.2015.117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ji QS, Mulvihill MJ, Rosenfeld-Franklin M, Cooke A, Feng L, Mak G, O’Connor M, Yao Y, Pirritt C & Buck E et al. 2007 A novel, potent, and selective insulin-like growth factor-I receptor kinase inhibitor blocks insulin-like growth factor-I receptor signaling in vitro and inhibits insulin-like growth factor-I receptor dependent tumor growth in vivo. Molecular Cancer Therapeutics 6 21582167. (doi:10.1158/1535-7163.MCT-07-0070)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kalimutho M, Parsons K, Mittal D, Lopez JA, Srihari S & Khanna KK 2015 Targeted therapies for triple-negative breast cancer: combating a stubborn disease. Trends in Pharmacological Sciences 36 822846. (doi:10.1016/j.tips.2015.08.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khatri A, Brundage RC, Hull JM, Williams BW, Yee D & Kirstein MN 2012 Pharmacodynamic modeling of sequence-dependent antitumor activity of insulin-like growth factor blockade and gemcitabine. AAPS Journal 14 19. (doi:10.1208/s12248-011-9308-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim JG, Kang MJ, Yoon YK, Kim HP, Park J, Song SH, Han SW, Park JW, Kang GH & Kang KW et al. 2012 Heterodimerization of glycosylated insulin-like growth factor-1 receptors and insulin receptors in cancer cells sensitive to anti-IGF1R antibody. PLoS ONE 7 e33322. (doi:10.1371/journal.pone.0033322)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim H, Tarhuni A, Abd Elmageed ZY & Boulares AH 2015 Poly(ADP-ribose) polymerase as a novel regulator of 17beta-estradiol-induced cell growth through a control of the estrogen receptor/IGF-1 receptor/PDZK1 axis. Journal of Translational Medicine 13 233. (doi:10.1186/s12967-015-0589-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson JJ Jr, Rocha-Lima CM, Safran H, Chan D, Kocs DM & Galimi F et al. 2012 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 23 28342842. (doi:10.1093/annonc/mds142)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • King H, Aleksic T, Haluska P & Macaulay VM 2014 Can we unlock the potential of IGF-1R inhibition in cancer therapy? Cancer Treatment Reviews 40 10961105. (doi:10.1016/j.ctrv.2014.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kleinberg DL, Ameri P & Singh B 2011 Pasireotide, an IGF-I action inhibitor, prevents growth hormone and estradiol-induced mammary hyperplasia. Pituitary 14 4452. (doi:10.1007/s11102-010-0257-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kurio N, Shimo T, Fukazawa T, Takaoka M, Okui T, Hassan NM, Honami T, Hatakeyama S, Ikeda M & Naomoto Y et al. 2011 Anti-tumor effect in human breast cancer by TAE226, a dual inhibitor for FAK and IGF-IR in vitro and in vivo. Experimental Cell Research 317 11341146. (doi:10.1016/j.yexcr.2011.02.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Law JH, Habibi G, Hu K, Masoudi H, Wang MY, Stratford AL, Park E, Gee JM, Finlay P & Jones HE et al. 2008 Phosphorylated insulin-like growth factor-i/insulin receptor is present in all breast cancer subtypes and is related to poor survival. Cancer Research 681023810246. (doi:10.1158/0008-5472.CAN-08-2755)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lawrence MC, McKern NM & Ward CW 2007 Insulin receptor structure and its implications for the IGF-1 receptor. Current Opinion in Structural Biology 17 699705. (doi:10.1016/j.sbi.2007.07.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y & Pietenpol JA 2011 Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. Journal of Clinical Investigation 121 27502767. (doi:10.1172/JCI45014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lerma E, Peiro G, Ramon T, Fernandez S, Martinez D, Pons C, Munoz F, Sabate JM, Alonso C & Ojeda B et al. 2007 Immunohistochemical heterogeneity of breast carcinomas negative for estrogen receptors, progesterone receptors and Her2/neu (basal-like breast carcinomas). Modern Pathology 20 12001207. (doi:10.1038/modpathol.3800961)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leroy C, Ramos P, Cornille K, Bonenfant D, Fritsch C, Voshol H & Bentires-Alj M 2016 Activation of IGF1R/p110beta/AKT/mTOR confers resistance to alpha-specific PI3K inhibition. Breast Cancer Research 18 41. (doi:10.1186/s13058-016-0697-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lewis I, Schmid HA, Kneuer R, Hoyer D, Silva AP, Weckbecker G, Bruns C & Pless J 2014 SOM230: a new therapeutic modality for Cushing’s disease. Chimia 68 483484. (doi:10.2533/chimia.2014.483)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li J, Song Z, Wang Y, Yin Y, Liu Y, Yuan R & Nan X 2016 Overexpression of SphK1 enhances cell proliferation and invasion in triple-negative breast cancer via the PI3K/AKT signaling pathway. Tumor Biology 37 1058710593. (doi:10.1007/s13277-016-4954-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lin MZ, Marzec KA, Martin JL & Baxter RC 2014 The role of insulin-like growth factor binding protein-3 in the breast cancer cell response to DNA-damaging agents. Oncogene 33 8596. (doi:10.1038/onc.2012.538)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lisztwan J, Pornon A, Chen B, Chen S & Evans DB 2008 The aromatase inhibitor letrozole and inhibitors of insulin-like growth factor I receptor synergistically induce apoptosis in in vitro models of estrogen-dependent breast cancer. Breast Cancer Research 10 R56. (doi:10.1186/bcr2113)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Litzenburger BC, Creighton CJ, Tsimelzon A, Chan BT, Hilsenbeck SG, Wang T, Carboni JM, Gottardis MM, Huang F & Chang JC et al. 2011 High IGF-IR activity in triple-negative breast cancer cell lines and tumorgrafts correlates with sensitivity to anti-IGF-IR therapy. Clinical Cancer Research 17 23142327. (doi:10.1158/1078-0432.CCR-10-1903)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu TJ, LaFortune T, Honda T, Ohmori O, Hatakeyama S, Meyer T, Jackson D, de Groot J & Yung WK 2007 Inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor kinase suppresses glioma proliferation in vitro and in vivo. Molecular Cancer Therapeutics 6 13571367. (doi:10.1158/1535-7163.MCT-06-0476)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu B, Fan Z, Edgerton SM, Yang X, Lind SE & Thor AD 2011 Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle 10 29592966. (doi:10.4161/cc.10.17.16359)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT & Perou CM 2006 Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Modern Pathology 19 264271. (doi:10.1038/modpathol.3800528)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Macaulay VM, Middleton MR, Eckhardt SG, Rudin CM, Juergens RA, Gedrich R, Gogov S, McCarthy S, Poondru S & Stephens A et al. 2016 Phase I dose escalation study of linsitinib (OSI-906) and erlotinib in patients with advanced solid tumors. Clinical Cancer Research 22 28972907. (doi:10.1158/1078-0432.CCR-15-2218)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Madden JM, Mueller KL, Bollig-Fischer A, Stemmer P, Mattingly RR & Boerner JL 2014 Abrogating phosphorylation of eIF4B is required for EGFR and mTOR inhibitor synergy in triple-negative breast cancer. Breast Cancer Research and Treatment 147 283293. (doi:10.1007/s10549-014-3102-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mahadevan D, Sutton GR, Arteta-Bulos R, Bowden CJ, Miller PJ, Swart RE, Walker MS, Haluska P, Munster PN & Marshall J et al. 2014 Phase 1b study of safety, tolerability and efficacy of R1507, a monoclonal antibody to IGF-1R in combination with multiple standard oncology regimens in patients with advanced solid malignancies. Cancer Chemotherapy and Pharmacology 73 467473. (doi:10.1007/s00280-013-2372-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maiello MR, D’Alessio A, Bevilacqua S, Gallo M, Normanno N & De Luca A 2015 EGFR and MEK blockade in triple negative breast cancer cells. Journal of Cellular Biochemistry 116 27782785. (doi:10.1002/jcb.25220)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maloney EK, McLaughlin JL, Dagdigian NE, Garrett LM, Connors KM, Zhou XM, Blattler WA, Chittenden T & Singh R 2003 An anti-insulin-like growth factor I receptor antibody that is a potent inhibitor of cancer cell proliferation. Cancer Research 63 50735083.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mancini M, Gariboldi MB, Taiana E, Bonzi MC, Craparotta I, Pagin M & Monti E 2014 Co-targeting the IGF system and HIF-1 inhibits migration and invasion by (triple-negative) breast cancer cells. British Journal of Cancer 110 28652873. (doi:10.1038/bjc.2014.269)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martin JL, Lin MZ, McGowan EM & Baxter RC 2009 Potentiation of growth factor signaling by insulin-like growth factor-binding protein-3 in breast epithelial cells requires sphingosine kinase activity. Journal of Biological Chemistry 284 2554225552. (doi:10.1074/jbc.M109.007120)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martin JL, de Silva HC, Lin MZ, Scott CD & Baxter RC 2014 Inhibition of insulin-like growth factor-binding protein-3 signaling through sphingosine kinase-1 sensitizes triple-negative breast cancer cells to EGF receptor blockade. Molecular Cancer Therapeutics 13 316328. (doi:10.1158/1535-7163.MCT-13-0367)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Masuda H, Zhang D, Bartholomeusz C, Doihara H, Hortobagyi GN & Ueno NT 2012 Role of epidermal growth factor receptor in breast cancer. Breast Cancer Research and Treatment 136 331345. (doi:10.1007/s10549-012-2289-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matsubara J, Yamada Y, Hirashima Y, Takahari D, Okita NT, Kato K, Hamaguchi T, Shirao K, Shimada Y & Shimoda T 2008 Impact of insulin-like growth factor type 1 receptor, epidermal growth factor receptor, and HER2 expressions on outcomes of patients with gastric cancer. Clinical Cancer Research 14 30223029. (doi:10.1158/1078-0432.CCR-07-1898)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McCaffery I, Tudor Y, Deng H, Tang R, Suzuki S, Badola S, Kindler HL, Fuchs CS, Loh E & Patterson SD et al. 2013 Putative predictive biomarkers of survival in patients with metastatic pancreatic adenocarcinoma treated with gemcitabine and ganitumab, an IGF1R inhibitor. Clinical Cancer Research 19 42824289. (doi:10.1158/1078-0432.CCR-12-1840)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McNaughton M, Pitman M, Pitson SM, Pyne NJ & Pyne S 2016 Proteasomal degradation of sphingosine kinase 1 and inhibition of dihydroceramide desaturase by the sphingosine kinase inhibitors, SKi or ABC294640, induces growth arrest in androgen-independent LNCaP-AI prostate cancer cells. Oncotarget 7 1666316675. (doi:10.18632/oncotarget.7693)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McTavish H, Griffin RJ, Terai K & Dudek AZ 2009 Novel insulin-like growth factor-methotrexate covalent conjugate inhibits tumor growth in vivo at lower dosage than methotrexate alone. Translational Research 153 275282. (doi:10.1016/j.trsl.2009.02.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mendelsohn J & Baselga J 2006 Epidermal growth factor receptor targeting in cancer. Seminars in Oncology 33 369385. (doi:10.1053/j.seminoncol.2006.04.003)

  • Mendes D, Alves C, Afonso N, Cardoso F, Passos-Coelho JL, Costa L, Andrade S & Batel-Marques F 2015 The benefit of HER2-targeted therapies on overall survival of patients with metastatic HER2-positive breast cancer – a systematic review. Breast Cancer Research 17 140. (doi:10.1186/s13058-015-0648-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mireuta M, Birman E, Barmash M & Pollak M 2014 Quantification of binding of IGF-1 to BI 836845, a candidate therapeutic antibody against IGF-1 and IGF-2, and effects of this antibody on IGF-1:IGFBP-3 complexes in vitro and in male C57BL/6 mice. Endocrinology 155 703715. (doi:10.1210/en.2013-1791)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morcavallo A, Stefanello M, Iozzo RV, Belfiore A & Morrione A 2014 Ligand-mediated endocytosis and trafficking of the insulin-like growth factor receptor I and insulin receptor modulate receptor function. Frontiers in Endocrinology 5 220. (doi:10.3389/fendo.2014.00220)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morgillo F, Woo JK, Kim ES, Hong WK & Lee HY 2006 Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Research 66 1010010111. (doi:10.1158/0008-5472.CAN-06-1684)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Landfair D, O’Connor M, Pirritt C, Sun Y & Yao Y et al. 2009 Discovery of OSI-906: a selective and orally efficacious dual inhibitor of the IGF-1 receptor and insulin receptor. Future Medicinal Chemistry 1 11531171. (doi:10.4155/fmc.09.89)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ochnik AM, Peterson MS, Avdulov SV, Oh AS, Bitterman PB & Yee D 2016 Amplified in breast cancer regulates transcription and translation in breast cancer cells. Neoplasia 18 100110. (doi:10.1016/j.neo.2016.01.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Overholser J, Ambegaokar KH, Eze SM, Sanabria-Figueroa E, Nahta R, Bekaii-Saab T & Kaumaya PT 2015 Anti-tumor effects of peptide therapeutic and peptide vaccine antibody co-targeting HER-1 and HER-2 in esophageal cancer (EC) and HER-1 and IGF-1R in triple-negative breast cancer (TNBC). Vaccines 3 519543. (doi:10.3390/vaccines3030519)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pal SK, Childs BH & Pegram M 2011 Triple negative breast cancer: unmet medical needs. Breast Cancer Research and Treatment 125627636. (doi:10.1007/s10549-010-1293-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pavlicek A, Lira ME, Lee NV, Ching KA, Ye J, Cao J, Garza SJ, Hook KE, Ozeck M & Shi ST et al. 2013 Molecular predictors of sensitivity to the insulin-like growth factor 1 receptor inhibitor Figitumumab (CP-751,871). Molecular Cancer Therapeutics 12 29292939. (doi:10.1158/1535-7163.MCT-13-0442-T)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peiro G, Adrover E, Sanchez-Tejada L, Lerma E, Planelles M, Sanchez-Paya J, Aranda FI, Giner D & Gutierrez-Avino FJ 2011 Increased insulin-like growth factor-1 receptor mRNA expression predicts poor survival in immunophenotypes of early breast carcinoma. Modern Pathology 24 201208. (doi:10.1038/modpathol.2010.191)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peiro G, Ortiz-Martinez F, Gallardo A, Perez-Balaguer A, Sanchez-Paya J, Ponce JJ, Tibau A, Lopez-Vilaro L, Escuin D & Adrover E et al. 2014 Src, a potential target for overcoming trastuzumab resistance in HER2-positive breast carcinoma. British Journal of Cancer 111689695. (doi:10.1038/bjc.2014.327)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Periyasamy-Thandavan S, Takhar S, Singer A, Dohn MR, Jackson WH, Welborn AE, LeRoith D, Marrero M, Thangaraju M & Huang S et al. 2012 Insulin-like growth factor 1 attenuates antiestrogen- and antiprogestin-induced apoptosis in ER+ breast cancer cells by MEK1 regulation of the BH3-only pro-apoptotic protein Bim. Breast Cancer Research 14 R52. (doi:10.1186/bcr3153)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Perou CM 2011 Molecular stratification of triple-negative breast cancers. Oncologist 15 (Supplement 5) 3948. (doi:10.1634/theoncologist.2010-S5-39)

  • Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H & Akslen LA et al. 2000 Molecular portraits of human breast tumours. Nature 406 747752. (doi:10.1038/35021093)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pettersson F, Yau C, Dobocan MC, Culjkovic-Kraljacic B, Retrouvey H, Puckett R, Flores LM, Krop IE, Rousseau C & Cocolakis E et al. 2011 Ribavirin treatment effects on breast cancers overexpressing eIF4E, a biomarker with prognostic specificity for luminal B-type breast cancer. Clinical Cancer Research 17 28742884. (doi:10.1158/1078-0432.CCR-10-2334)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Piha-Paul SA, Munster PN, Hollebecque A, Argiles G, Dajani O, Cheng JD, Wang R, Swift A, Tosolini A & Gupta S 2015 Results of a phase 1 trial combining ridaforolimus and MK-0752 in patients with advanced solid tumours. European Journal of Cancer 51 18651873. (doi:10.1016/j.ejca.2015.06.115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pitman MR & Pitson SM 2010 Inhibitors of the sphingosine kinase pathway as potential therapeutics. Current Cancer Drug Targets 10 354367. (doi:10.2174/156800910791208599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pitman MR, Woodcock JM, Lopez AF & Pitson SM 2012 Molecular targets of FTY720 (fingolimod). Current Molecular Medicine 1212071219. (doi:10.2174/156652412803833599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pitman MR, Powell JA, Coolen C, Moretti PA, Zebol JR, Pham DH, Finnie JW, Don AS, Ebert LM & Bonder CS et al. 2015 A selective ATP-competitive sphingosine kinase inhibitor demonstrates anti-cancer properties. Oncotarget 6 70657083. (doi:10.18632/oncotarget.3178)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pitson SM, Moretti PA, Zebol JR, Lynn HE, Xia P, Vadas MA & Wattenberg BW 2003 Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO Journal 22 54915500. (doi:10.1093/emboj/cdg540)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pitts TM, Tan AC, Kulikowski GN, Tentler JJ, Brown AM, Flanigan SA, Leong S, Coldren CD, Hirsch FR & Varella-Garcia M et al. 2010 Development of an integrated genomic classifier for a novel agent in colorectal cancer: approach to individualized therapy in early development. Clinical Cancer Research 16 31933204. (doi:10.1158/1078-0432.CCR-09-3191)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pollak M 2012 The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nature Reviews Cancer 12 159169. (doi:10.1038/nrc3215)

  • Rozengurt E, Sinnett-Smith J & Kisfalvi K 2010 Crosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clinical Cancer Research 1625052511. (doi:10.1158/1078-0432.CCR-09-2229)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sachdev D, Singh R, Fujita-Yamaguchi Y & Yee D 2006 Down-regulation of insulin receptor by antibodies against the type I insulin-like growth factor receptor: implications for anti-insulin-like growth factor therapy in breast cancer. Cancer Research 66 23912402. (doi:10.1158/0008-5472.CAN-05-3126)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Salani B, Ravera S, Amaro A, Salis A, Passalacqua M, Millo E, Damonte G, Marini C, Pfeffer U & Sambuceti G et al. 2015 IGF1 regulates PKM2 function through Akt phosphorylation. Cell Cycle 14 15591567. (doi:10.1080/15384101.2015.1026490)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sanabria-Figueroa E, Donnelly SM, Foy KC, Buss MC, Castellino RC, Paplomata E, Taliaferro-Smith L, Kaumaya PT & Nahta R 2015 Insulin-like growth factor-1 receptor signaling increases the invasive potential of human epidermal growth factor receptor 2-overexpressing breast cancer cells via Src-focal adhesion kinase 
and forkhead box protein M1. Molecular Pharmacology 87 150161. (doi:10.1124/mol.114.095380)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sanderson MP, Apgar J, Garin-Chesa P, Hofmann MH, Kessler D, Quant J, Savchenko A, Schaaf O, Treu M & Tye H et al. 2015 BI 885578, a novel IGF1R/INSR tyrosine kinase inhibitor with pharmacokinetic properties that dissociate antitumor efficacy and perturbation of glucose homeostasis. Molecular Cancer Therapeutics 14 27622772. (doi:10.1158/1535-7163.MCT-15-0539)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saxena NK, Taliaferro-Smith L, Knight BB, Merlin D, Anania FA, O’Regan RM & Sharma D 2008 Bidirectional crosstalk between leptin and insulin-like growth factor-I signaling promotes invasion and migration of breast cancer cells via transactivation of epidermal growth factor receptor. Cancer Research 68 97129722. (doi:10.1158/0008-5472.CAN-08-1952)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schwartz GK, Tap WD, Qin LX, Livingston MB, Undevia SD, Chmielowski B, Agulnik M, Schuetze SM, Reed DR & Okuno SH et al. 2013 Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial. Lancet Oncology 14 371382. (doi:10.1016/S1470-2045(13)70049-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sichani MM, Yazdi FS, Moghaddam NA, Chehrei A, Kabiri M, Naeimi A & Taheri D 2010 Prognostic value of insulin- like growth factor-I receptor expression in renal cell carcinoma. Saudi Journal of Kidney Diseases and Transplantation 21 6974.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Silvera D, Formenti SC & Schneider RJ 2010 Translational control in cancer. Nature Reviews Cancer 10 254266. (doi:10.1038/nrc2824)

  • Singh B, Smith JA, Axelrod DM, Ameri P, Levitt H, Danoff A, Lesser M, de Angelis C, Illa-Bochaca I & Lubitz S et al. 2014a Insulin-like growth factor-I inhibition with pasireotide decreases cell proliferation and increases apoptosis in pre-malignant lesions of the breast: a phase 1 proof of principle trial. Breast Cancer Research 16 463. (doi:10.1186/s13058-014-0463-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Singh RK, Gaikwad SM, Jinager A, Chaudhury S, Maheshwari A & Ray P 2014b IGF-1R inhibition potentiates cytotoxic effects of chemotherapeutic agents in early stages of chemoresistant ovarian cancer cells. Cancer Letters 354 254262. (doi:10.1016/j.canlet.2014.08.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sonenberg N & Hinnebusch AG 2009 Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136 731745. (doi:10.1016/j.cell.2009.01.042)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subramaniam D, He AR, Hwang J, Deeken J, Pishvaian M, Hartley ML & Marshall JL 2015 Irreversible multitargeted ErbB family inhibitors for therapy of lung and breast cancer. Current Cancer Drug Targets 14 775793. (doi:10.2174/1568009614666141111104643)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Takahari D, Yamada Y, Okita NT, Honda T, Hirashima Y, Matsubara J, Takashima A, Kato K, Hamaguchi T & Shirao K et al. 2009 Relationships of insulin-like growth factor-1 receptor and epidermal growth factor receptor expression to clinical outcomes in patients with colorectal cancer. Oncology 76 4248. (doi:10.1159/000178164)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taliaferro-Smith L, Oberlick E, Liu T, McGlothen T, Alcaide T, Tobin R, Donnelly S, Commander R, Kline E & Nagaraju GP et al. 2015 FAK activation is required for IGF1R-mediated regulation of EMT, migration, and invasion in mesenchymal triple negative breast cancer cells. Oncotarget 6 47574772. (doi:10.18632/oncotarget.3023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tao Z, Shi A, Lu C, Song T, Zhang Z & Zhao J 2014 Breast cancer: epidemiology and etiology. Cell Biochemistry and Biophysics 72333338. (doi:10.1007/s12013-014-0459-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taunk NK, Goyal S, Moran MS, Yang Q, Parikh R & Haffty BG 2010 Prognostic significance of IGF-1R expression in patients treated with breast-conserving surgery and radiation therapy. Radiotherapy and Oncology 96 204208. (doi:10.1016/j.radonc.2010.03.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Terwisscha van Scheltinga AG, Berghuis P, Nienhuis HH, Timmer-Bosscha H, Pot L, Gaykema SB, Lub-de Hooge MN, Kosterink JG, de Vries EG & Schroder CP 2014 Visualising dual downregulation of insulin-like growth factor receptor-1 and vascular endothelial growth factor-A by heat shock protein 90 inhibition effect in triple negative breast cancer. European Journal of Cancer 50 25082516. (doi:10.1016/j.ejca.2014.06.008)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vasilcanu R, Vasilcanu D, Rosengren L, Natalishvili N, Sehat B, Yin S, Girnita A, Axelson M, Girnita L & Larsson O 2008 Picropodophyllin induces downregulation of the insulin-like growth factor 1 receptor: potential mechanistic involvement of Mdm2 and beta-arrestin1. Oncogene 27 16291638. (doi:10.1038/sj.onc.1210797)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, Pegram M, Oh DY, Dieras V & Guardino E et al. 2012 Trastuzumab emtansine for HER2-positive advanced breast cancer. New England Journal of Medicine 367 17831791. (doi:10.1056/NEJMoa1209124)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Y, Lipari P, Wang X, Hailey J, Liang L, Ramos R, Liu M, Pachter JA, Bishop WR & Wang Y 2010a A fully human insulin-like growth factor-I receptor antibody SCH 717454 (Robatumumab) has antitumor activity as a single agent and in combination with cytotoxics in pediatric tumor xenografts. Molecular Cancer Therapeutics 9 410418. (doi:10.1158/1535-7163.MCT-09-0555)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang YH, Xiong J, Wang SF, Yu Y, Wang B, Chen YX, Shi HF & Qiu Y 2010b Lentivirus-mediated shRNA targeting insulin-like growth factor-1 receptor (IGF-1R) enhances chemosensitivity of osteosarcoma cells in vitro and in vivo. Molecular and Cellular Biochemistry 341 225233. (doi:10.1007/s11010-010-0453-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Watanabe N, Takaoka M, Sakurama K, Tomono Y, Hatakeyama S, Ohmori O, Motoki T, Shirakawa Y, Yamatsuji T & Haisa M et al. 2008 Dual tyrosine kinase inhibitor for focal adhesion kinase and insulin-like growth factor-I receptor exhibits anticancer effect in esophageal adenocarcinoma in vitro and in vivo. Clinical Cancer Research 14 46314639. (doi:10.1158/1078-0432.CCR-07-4755)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wong MH, Xue A, Julovi SM, Pavlakis N, Samra JS, Hugh TJ, Gill AJ, Peters L, Baxter RC & Smith RC 2014 Cotargeting of epidermal growth factor receptor and PI3K overcomes PI3K-Akt oncogenic dependence in pancreatic ductal adenocarcinoma. Clinical Cancer Research 20 40474058. (doi:10.1158/1078-0432.CCR-13-3377)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang Y & Yee D 2012 Targeting insulin and insulin-like growth factor signaling in breast cancer. Journal of Mammary Gland Biology and Neoplasia 17 251261. (doi:10.1007/s10911-012-9268-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yang Y & Yee D 2014 IGF-I regulates redox status in breast cancer cells by activating the amino acid transport molecule xC. Cancer Research 74 22952305. (doi:10.1158/0008-5472.CAN-13-1803)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ye XM, Zhu HY, Bai WD, Wang T, Wang L, Chen Y, Yang AG & Jia LT 2014 Epigenetic silencing of miR-375 induces trastuzumab resistance in HER2-positive breast cancer by targeting IGF1R. BMC Cancer 14 134. (doi:10.1186/1471-2407-14-134)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yee D 2012 Insulin-like growth factor receptor inhibitors: baby or the bathwater? Journal of the National Cancer Institute 104 975981. (doi:10.1093/jnci/djs258)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yi YW, Hong W, Kang HJ, Kim HJ, Zhao W, Wang A, Seong YS & Bae I 2013 Inhibition of the PI3K/AKT pathway potentiates cytotoxicity of EGFR kinase inhibitors in triple-negative breast cancer cells. Journal of Cellular and Molecular Medicine 17 648656. (doi:10.1111/jcmm.12046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yin D, Sleight B, Alvey C, Hansson AG & Bello A 2013 Pharmacokinetics and pharmacodynamics of figitumumab, a monoclonal antibody targeting the insulin-like growth factor 1 receptor, in healthy participants. Journal of Clinical Pharmacology 53 2128. (doi:10.1177/0091270011432934)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zanardi E, Bregni G, de Braud F & Di Cosimo S 2015 Better together: targeted combination therapies in breast cancer. Seminars in Oncology 42 887895. (doi:10.1053/j.seminoncol.2015.09.029)

    • PubMed