Abstract
IGF-1R inhibitors arrived in the clinic accompanied by optimism based on preclinical activity of IGF-1R targeting, and recognition that low IGF bioactivity protects from cancer. This was tempered by concerns about toxicity to normal tissue IGF-1R and cross-reactivity with insulin receptor (InsR). In fact, toxicity is not a show-stopper; the key issue is efficacy. While IGF-1R inhibition induces responses as monotherapy in sarcomas and with chemotherapy or targeted agents in common cancers, negative Phase 2/3 trials in unselected patients prompted the cessation of several Pharma programs. Here, we review completed and on-going trials of IGF-1R antibodies, kinase inhibitors and ligand antibodies. We assess candidate bio-markers for patient selection, highlighting the potential predictive value of circulating IGFs/IGFBPs, the need for standardized assays for IGF-1R, and preclinical evidence that variant InsRs mediate resistance to IGF-1R antibodies. We review hypothesis-led and unbiased approaches to evaluate IGF-1R inhibitors with other agents, and stress the need to consider sequencing with chemotherapy. The last few years were a tough time for IGF-1R therapeutics, but also brought progress in understanding IGF biology. Even failed studies include patients who derived benefit; they should be investigated to identify features distinguishing the tumors and host environment of responders from non-responders. We emphasize the importance of incorporating biospecimen collection into trial design, and wording patient consents to allow post hoc analysis of trial material as new data become available. Such information represents the key to unlocking the potential of this approach, to inform the next generation of trials of IGF signalling inhibitors.
Keywords: IGF, Type 1 IGF receptor, IGF-1R cancer therapy, Therapeutic antibody, Tyrosine kinase inhibitor, Predictive biomarker
IGF-targeting: a promising start
The rationale for developing drugs to block IGF signalling has been reviewed elsewhere [1–3] and will not be repeated, except to highlight first the seminal work of Baserga in showing that type 1 IGF receptor (IGF-1R) is required for malignant transformation [4], and secondly the demonstration that low IGF bioactivity protects against development of clinical cancers [5,6]. A variety of experimental approaches was used to provide proof of principle [7–11]; strategies with clinical application fall into three categories: monoclonal anti-IGF1R antibodies, small molecule tyrosine kinase inhibitors (TKIs), and IGF ligand antibodies. Preclinical data indicate that these different classes of drug have different profiles of selectivity, efficacy and toxicity that may affect their use in clinical practice [12–22]. IGF-1R antibodies block ligand binding and induce receptor internalization and degradation [14], also resulting in insulin receptor (InsR) downregulation in cells expressing IGF-1R:InsR hybrid receptors [23]. The majority of IGF-1R TKIs described to date act by competing with ATP for binding in the IGF-1R kinase domain [18,20,24], and also inhibit InsR due to conservation of structure, particularly in the ATP-binding cleft of the kinase domain [25]. As a class effect, IGF-1R inhibitory drugs cause hyperglycemia, although this is usually mild and reversible [6,26]. IGF-1R TKIs induce hyperglycemia via direct inhibition of InsR kinase, while IGF-1R antibodies impair glucose tolerance by inducing elevated circulating levels of growth hormone and IGF binding proteins (IGFBPs), attributed to blockade of pituitary IGF-1Rs [2]. Several ATP non-competitive agents are in development, including one at an early stage of clinical testing [27,28]. IGF ligand antibodies act by neutralising IGF-1 and IGF-2, preventing receptor activation without affecting glucose tolerance [22,29]. All three classes of agent have anti-tumour activity in vitro and in vivo, although it should be acknowledged that some preclinical models were selected or designed to be IGF responsive, and relatively limited preclinical data led to an extremely large number of clinical trials [30].
Clinical trials: reality check
Given experimental data supporting a key role for IGF-1R in tumor biology, a substantial degree of optimism accompanied the commencement of clinical trials with anti-IGF-1R agents. This early enthusiasm was stimulated to a large degree by the near ubiquitous expression of IGF signalling components, including IGF-1R in most tumor types [30,31], and was reinforced by initial reports of apparently dramatic activity [32,33]. However, the data that emerged subsequently have been less impressive. Monotherapy trials of anti-IGF-1R antibodies and TKIs have generally reported disease stabilisation and minor responses, with few objective responses in common cancers [34–38]. There is, however, evidence of activity in heavily pre-treated Ewing sarcoma (EWS), other types of sarcomas, and neuroendocrine tumors, although data in the latter are conflicting [32,39–44]. In addition, IGF-1R antibodies were recently reported to have single agent activity in recurrent ovarian cancer and relapsed thymoma, although thymoma treatment was associated with the development of autoimmune conditions including red cell aplasia [45,46]. Activity in EWS and childhood sarcomas may be a consequence of functional interactions between oncogenic fusion proteins (EWS-FLI1 and related fusion partners) and the IGF pathway [47–50].
There is as yet little evidence regarding the efficacy of ligand antibodies and non-ATP binding IGF-1R inhibitors, although of note, initial reports of the latter suggest single agent activity in non-small cell lung cancer (NSCLC) [28,51]. Otherwise, it is clear that in common cancers there is insufficient clinical support for continued evaluation of IGF-1R antibodies and TKIs as monotherapy in unselected patients. In the context of expectations generated by preclinical data, these clinical results have been disappointing. It is unclear whether limited single agent activity in common cancers reflects IGF-independent growth in established tumors, adaptive resistance mechanisms (discussed below) or the inability to sustain target blockade. The latter issue is raised by preclinical data demonstrating that IGF-1R antibodies may inadequately maintain in vivo IGF-1R down-regulation or suppression of PI3K-AKT signalling [52,53]. One factor that could limit efficacy was highlighted by an immuno-SPECT study showing correlation between uptake of IGF-1R antibody R1507 in responsive but not resistant IGF-1R-positive bone sarcoma xenografts [54]. These findings suggest that microenvironmental factors may limit vascular access of therapeutic antibody to tumor tissue, an obstacle that may be circumvented by small molecule drugs.
Based on reports that IGF-1R mediates resistance to other modalities of treatment [55–58], IGF-1R targeting has been evaluated in combination with an extensive range of standard and/or novel cancer treatments, including cytotoxic drugs and inhibitors of EGFR, mammalian target of rapamycin (mTOR) and steroid hormone receptors [53,59–75]. This has also met with mixed results, with Phase 2/3 trial failures in unselected patients [76]. One thing is certain: some sort of signal is needed soon, and this will require patient selection.
Can we identify who will benefit?
While negative trials led to the high profile termination of several Pharma IGF programs, a number of IGF axis inhibitors are continuing active evaluation, including small molecule inhibitors and ligand antibodies. It is important for the success of these programs that information from completed trials is utilized in translational research, because even the negative studies include patients who derived benefit from IGF-1R inhibition, providing an opportunity to characterize responsive tumors. This information has the potential to provide a basis for stratification and selection, if future trials are to be successful. Without such an approach, the clinical utility of EGFR inhibitors would not have been recognized after initial negative Phase 3 evaluation in NSCLC [77].
Although IGF-1R is almost ubiquitously expressed by human tumors, sensitivity to anti-IGF-1R therapy varies widely between patients and cancer types [26]. Large-scale cancer genome sequencing projects have identified rare IGF1R gene mutations reported to influence basal (ligand-unstimulated) phosphorylation of IGF-1R substrates [78]. However, these or similar mutations, comparable to those in EGFR associated with EGFR inhibitor sensitivity, have not been reported in tumors of patients on IGF-1R inhibitor trials. Thus, it is unlikely that IGF-1R mutations will be relevant in selecting patients for IGF signalling inhibition. Therefore, there is an on-going search for predictive biomarkers, which fall generally into two groups: potential biomarkers in the IGF axis, and biomarkers in other pathways that influence response to IGF-1R inhibition (Table 1).
Table 1.
Candidate biomarker of sensitivity | Preclinical/clinical | Tumor type | Anti-IGF-1R strategy | Ref. | |
---|---|---|---|---|---|
Tumor expression of IGF axis components | High IGF-1R expression | PC – cell lines, xenografts | Rhabdomyosarcoma | MAb h7C10 | [52] |
High expression of IGF-1R, IRS-1, IRS-2 | PC – cell lines | Breast, colorectal | MAb h10H5 | [87] | |
IGF-1R upregulation, increased gene copy number | PC – cell lines | NSCLC | MAb R1507 | [91] | |
Circulating tumor cell (CTC) IGF-1R expression | Phase 1 CT | Hormone refractory prostate | MAb figitumumab | [93] | |
IGF gene signature | PC – cell lines, xenografts | Triple negative breast | TKI BMS-754807 | [117] | |
Nuclear IGF-1R localisation | Single-centre – tumor samples | Soft tissue, EWS, osteosarcoma | MAb, various | [101] | |
IGF-1R/IR heterodimerization, N-linked glycosylation | PC – cell lines, xenografts | Gastric, hepatocellular | MAb figitumumab | [96] | |
High tumor expression of IGF-1, low IGF-2 | Phase 2/3 CT | Metastatic colorectal (WT KRAS) | MAb dalotuzumab | [65] | |
IRS-2 amplification, low IGFBP-5 expression | PC – cell lines | SCLC, NSCLC, breast, colorectal | MAb figitumumab | [86] | |
Low IGFBP-5/IGFBP-4 ratio | PC – cell lines, tumor cohorts | Breast | TKI BMS-536924 | [89] | |
High tumour IGF-1 expression | Phase 2 CT | Advanced pancreatic | MAb dalotuzumab | [90] | |
High IGF-1R gene copy number, pathway expression | PC – cell lines | Colorectal | TKI linsitinib | [105] | |
High IGF-1, IGF-2, IGF-1R, low IGFBP-3 and -6 | PC – cell lines | Various sarcoma, neuroblastoma | TKI BMS-536924 | [92] | |
Host factors | High pre-treatment circulating free IGF-1 | Phase 2 CT | NSCLC | MAb figitumumab | [79] |
High pre-treatment circulating free IGF-1 | Phase 1/2 CT | NSCLC | MAb cixutumumab | [63] | |
High pre-treatment circulating free IGF-1 | Phase 1 CT | NSCLC | MAb figitumumab | [80] | |
High baseline IGF-1, IGF-2 & IGFBP-3, low IGFBP-2 | Phase 2 CT | Metastatic pancreatic | MAb ganitumab | [82] | |
High pre-treatment circulating free IGF-1 | Phase 2 CT | NSCLC | MAb R1507 | [81] | |
HbA1c <5.7% | Phase 2 CT | ER-positive breast | MAb figitumumab | [155] | |
Compensatory signalling by alternative kinases | Low level of EGFR pathway activation | PC – cell lines | Various sarcoma & neuroblastoma | TKI BMS-536924 | [92] |
Low HER2 expression, high AKT | PC – cell lines | HER2 positive breast | TKIs BMS-536924, NVP-AEW541 | [94] | |
Low level of PDGFRα pathway activation | PC – cell lines | Rhabdomyosarcoma | TKI BMS-754807 | [133] | |
Low insulin receptor expression | PC – cell lines, transgenic mice | Pancreatic NETs, breast cancer cells | MAb cixutumumab | [124] | |
Downstream molecules | RAS/MAPK activity reduced by IGF-1R targeting | PC – cell lines | Oesophageal | TKI NVP-AEW541 | [109] |
Tumour expression of WT EGFR and K-RAS | PC – cell lines | NSCLC | TKIs PQIP, linsitinib | [113] | |
Mutant K-RAS | PC – cell lines | NSCLC | TKIs, various | [115] | |
Mutant K-RAS | Phase 2 CT | NSCLC | MAb R1507 | [81] | |
WT K-RAS | PC – cell lines | Colorectal | TKI linsitinib | [105] | |
WT PTEN | PC – cell lines | Prostate | TKI NVP-AEW541 | [106] | |
Low baseline ERK phosphorylation | PC – cell lines, xenografts | SCLC | TKI linsitinib | [111] | |
High IRS-1 expression | PC – cell lines | Breast | TKI NVP-AEW541 | [88] | |
Cellular phenotype | Mesenchymal phenotype | Phase 2/3 CT | Metastatic colorectal (WT KRAS) | MAb dalotuzumab | [65] |
Screens for resistance/sensitivity mediators | RPS6, MST1R | PC – cell lines | Ewing family | TKI BMS-536924 | [104] |
MYB amplification | PC – cell lines | SCLC, NSCLC, breast, colorectal | MAb figitumumab | [86] | |
k-TSP classifier gene pair expression | PC – cell lines | Colorectal | TKI linsitinib | [105] | |
Insulin, MAPK, endocytic pathway gene expression | PC – cell lines | Ewing’s sarcoma | figitumumab, AVE-1642, NVP-AEW541 | [118] | |
CDK4/6 | PC – cell lines | Dedifferentiated liposarcoma | MAb R1507 | [152] | |
Imaging | Uptake of indium-111-labelled R1507 | PC – xenografts | Osteosarcoma | MAb R1507 | [54] |
Candidate predictive biomarkers in the IGF axis
The first candidate biomarker emerged from the Phase 2 figitumumab trial in NSCLC [59], with a report that high pre-treatment levels of circulating free IGF-1 were predictive of clinical benefit from addition of figitumumab to standard chemotherapy [79]. These findings were corroborated with respect to levels of free or total circulating IGFs in additional clinical trials [63,80–82]. Circulating markers present obvious advantages over tissue biomarkers: testing is minimally invasive and amenable to serial monitoring. There is novelty in the concept that tumor sensitivity to a targeted agent may be influenced by host factors; this idea is consistent with the known importance of circulating host-derived IGFs in normal and cancer biology, and the recognition that high circulating IGF-1 is associated with increased risk of developing common cancers [83,84]. However, this line of research suffered a setback when the original findings with respect to the figitumumab NSCLC data were retracted [85]. Nonetheless, more recent data support further investigation of IGF components as candidate biomarkers. Preclinical models find evidence of correlation between sensitivity to figitumumab or TKI NVP-AEW541 and expression of IRS2, IGFBP5 and MYB in colorectal cancer [86], and expression of IGF-1R, IGF-2 and IRS-1 and -2 in breast and colon models [87,88]. In breast cancer cells resistant to BMS 536924, the IGFBP-5/IGFBP-4 expression ratio was found to correlate with sensitivity to IGF-1R inhibition [89]. Clinical evidence supporting evaluation of IGF axis biomarkers comes from trials in pancreatic cancer, where sensitivity to dalotuzumab (IGF-1R antibody MK-0646) was linked with tumor IGF-1 expression, and to ganitumab with high circulating IGF-1, IGF-2 or IGFBP-3, or low IGFBP-2 [82,90].
In certain cancers, including NSCLC, breast, colorectal, EWS and rhabdomyosarcoma, tumor IGF-1R may be predictive of response [52,87,91,92]. IGF-1R expression by circulating tumor cells may correlate with tumor IGF-1R, providing a feasible approach for serial measurement [93]. This could be particularly relevant when monitoring response to IGF-1R antibody, where IGF-1R downregulation is an indicator of target engagement [14]. In rhabdomyosarcoma cell lines, there was significant correlation between elevated IGF-1R and anti-proliferative effects of an anti-IGF1R monoclonal antibody, enabling prediction of responses to IGF-1R-targeting in animal models [52]. This parameter and the finding of association with IGFs, IRS proteins and IGFBPs may indicate tumors that are more heavily dependent on IGF signalling, although evidence in this area is contradictory, with some preclinical and clinical studies reporting that IGF-1R expression or activation has no predictive value [72,94,95]. It is possible that the specific molecular context and the complexity of IGF-1R:insulin hybrid receptor biology may explain some of these contradictory results [26]. A well-designed Phase 2 study in patients with bone or soft tissue sarcomas tested the utility of IGF-1R as a predictive biomarker by measuring IGF-1R in fresh tumor biopsies pre-and post treatment [72]. The results suggested that PFS was similar in patients with ‘IGF-1R positive’ vs ‘IGF-1R negative’ tumors, apparently challenging the assumption that lack of IGF-1R has negative predictive value, although it was acknowledged that IGF-1R could be detected by western blotting of tumor lysates in tumors that were ‘IGF-1R negative’ by immunohistochemistry, casting doubt on the sensitivity of IHC detection [72]. These data highlight the importance of establishing standardized detection protocols for IGF-1R, as for estrogen receptor (ER) and HER2, and to develop robust assays for InsR and hybrid receptors in clinical tumors.
Subcellular IGF-1R localization may also be relevant to response to IGF-1R inhibition, although there is conflict in the literature on its significance. Aberrant IGF-1R glycosylation is reported to impair insertion of IGF-1R into the plasma membrane, conferring resistance to IGF-1R antibody [96], consistent with a model in which only receptors on the plasma membrane are accessible to antibody binding. IGF-1R is known to undergo ligand-induced translocation from the cell surface to the nucleus of human tumor cells, and nuclear IGF-1R is associated with resistance to EGFR inhibitor gefitinib [97–100], suggesting a role in IGF-1R-mediated resistance to targeted therapies. Exclusive nuclear IGF-1R localization was reported by Blay and colleagues to be associated with benefit from IGF-1R antibody in unresectable and metastatic sarcomas [101]. This may suggest that nuclear IGF-1R is a marker for dependence on IGF signalling, and sensitivity to its inhibition. It also raises the question of the location of IGF-1R:antibody binding; of note, quantum dot-labelled IGF-1R antibody has been found to undergo nuclear import [102].
Candidate predictive biomarkers outside the IGF axis
Given that the IGF axis engages in complex signalling interactions [103], it is predictable that candidate biomarkers will be identified outside the IGF–IGF-1R–IRS axis, and some such have been identified from tumor-specific screens. In childhood sarcoma cells, knockdowns of macrophage-stimulating 1 receptor (MST1R, also known as RON) and ribosomal protein S6 restored sensitivity to BMS-536924 in resistant cell lines [104]. Sensitivity to the dual IGF-1R/InsR inhibitor linsitinib (OSI-906) was associated with IGF1R gene copy number (but not IGF-1R protein) in colorectal cancer cell lines, and also with differential expression of 3 gene pairs and a trend to greater sensitivity in cells with wild-type KRAS [105]. The latter finding raises the issue of the extent to which activating mutations in IGF-1R effectors mediate resistance to IGF-1R blockade. In preclinical models, IGF-1R inhibition is capable of suppressing tumor cell growth in cells that lack functional PTEN, although there are conflicting reports of the consequences of PTEN loss in terms of the ability of IGF-1R targeting to enhance radiosensitivity [10,106,107]. While PTEN loss amplifies signalling to AKT, it does not induce constitutive pathway activation; logic dictates that constitutively activating mutations in the PI3K-AKT or RAS-RAF-ERK pathways should render cells refractory to IGF axis blockade. Indeed, preclinical data suggest that resistance to IGF-1R inhibition is observed in the context of constitutive AKT activation and high levels of activated ERKs [108–111], and sensitivity of KRAS mutant NSCLC was restored by MEK inhibition [112,113]. Consistent with this, IGF-1R antibody ganitumab was ineffective at sensitizing to FOLFIRI chemotherapy in patients with KRAS mutant colorectal cancer [114], although issues of chemotherapy scheduling may be a contributing factor (see below). In contrast, KRAS mutant NSCLC cells appear to be sensitive to IGF-1R inhibition: mutant KRAS was found to induce IGF-dependent PI3K activation that could be suppressed by targeting IGF-1R [115]. These findings suggest that KRAS mutation may have varying significance for response to IGF-1R blockade, depending upon the tumor type and/or molecular context.
Finally, predictive information may be derived by characterizing IGF-regulated gene expression. This approach was first applied by Lee and colleagues, with reports that an IGF gene signature induced in breast cancer cells in vitro is associated with poor prognosis in clinical cancers, and correlates with sensitivity to IGF-1R inhibitor BMS-754807 in triple negative breast cancer (TNBC) in vivo [116,117]. These data provide further support for the concept that tumors that are highly dependent on IGF signalling may also be sensitive to IGF-1R inhibition. A similar approach has been taken in EWS, to identify molecular signatures associated with resistance to IGF-1R antibody or TKI drugs [118].
Thus, there is evidence from several studies that levels of circulating IGFs and IGFBPs correlate with sensitivity to IGF axis inhibition. Otherwise, there is little consensus, with a range of candidate biomarkers identified in different tumors with different classes of agent. As a consequence, phase 2/3 trials have been undertaken without patient selection, although some are now reporting post hoc identification of markers that characterize responsive patient subgroups [65,82]. In order for this research to inform clinical practice, it is of critical importance to be able to test these factors in the tumors of additional trial patients. This in turn requires that trial consents should permit future ethically-approved research on archival and trial-specific material. For example, BATTLE-FL is building on previous NSCLC BATTLE trials [119], to incorporate biomarkers into a trial adding targeted agents including IGF-1R antibody cixutumumab to first-line carboplatin/pemetrexed chemotherapy (NCT01263782). Another example is the ongoing ISPY2 (NCT01042379) breast cancer clinical trial, which is incorporating ganitumab in combination with metformin, into neoadjuvant chemotherapy, and employing a rich biomarker driven, adaptive study design [120,121]. The analysis will be used to develop both predictive and prognostic biomarkers through correlation of tumor expression profiles by DNA microarray and reverse-phase protein arrays with tumor response at the time of surgery.
Approaches to design rational combination therapies
Given the large number of signalling pathways that converge on those activated by IGF-1R [103], it was in retrospect unlikely that IGF-1R targeting would have dramatic impact as monotherapy, except in specific cancers that are highly IGF-dependent. Predictive biomarkers serve an important purpose in identifying potentially responsive patients, but they do not make treatments more effective per se. To achieve that goal, it is necessary to understand how to combine novel drugs most effectively, and how to schedule targeted drugs with standard treatments. A large body of literature has highlighted the ability of IGF-1R to mediate resistance to other treatment modalities, and conversely has identified proteins capable of compensating for IGF-1R blockade. These findings provide the rationale for designing trials of IGF-1R inhibitory drugs in combination with other targeted agents, and with chemotherapy.
Trials of IGF-1R inhibitory drugs with targeted agents
IGF-1R signalling is involved in extensive cross-talk with other receptor tyrosine kinases (RTKs) and their downstream effectors, such that compensation from other pathways is likely to mediate resistance to inhibition of a single class of receptor [122]. For IGF-1R, the most closely-related candidate is InsR, with evidence that resistance to IGF-1R antibody therapy results from IGF-1R:InsR heterodimer formation, and IGF-2 signalling via the variant InsR isoform A (InsR-A) that is frequently expressed by human tumors [96,123–125]. Of note, InsR-A may be the more important IGF signalling receptor in hormone-resistant breast cancer [126–128]. This source of compensatory signalling may be blocked by IGF ligand antibodies and dual IGF-1R/InsR TKIs such as linsitinib and BMS-754807 [20,129]. Trials currently in progress will clarify whether this conceptual advantage translates to improved efficacy.
Cross-talk has been demonstrated between IGF-1R and other cell surface and nuclear receptors including epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor α (PDGFRα), Src and ER [130–136]. Preclinical studies implicate RTKs such as EGFR, HER2, PDGFRα and cMET in adaptive resistance to IGF-1R inhibition [133,137,138]. In particular, it is well-recognized that EGFR and IGF-1R are capable of providing reciprocal compensation, via receptor upregulation, enhanced PI3K-AKT signalling, and altered function of cell cycle regulators, patterns of chromatin modification and gene expression [55,87,130,139,140]. This recognition prompted trials of combined IGF-1R:EGFR inhibition, alone or with standard cancer treatments. Phase 1 evaluation suggests that such combinations are tolerable, with evidence of activity in non-adenocarcinoma NSCLC [61]. However, outcomes were not improved by the addition of IGF-1R antibodies ganitumab to EGFR antibody panitumumab in colorectal cancer, or cixutumumab to EGFR TKI erlotinib with gemcitabine in unselected patients with pancreatic cancer [67,73]. This and other IGF-1R:EGFR combination trials, such as the Phase 2 NSCLC trials of linsitinib and erlotinib (NCT01221077, NCT01186861) have been conducted in erlotinib naïve patients, whereas the ability of IGF-1R to mediate resistance to EGFR inhibition was identified preclinically in NSCLC cells induced to be erlotinib resistant [130]. Due to the paucity of trials that have to date incorporated post-treatment biopsies, it is unclear to what extent the mechanisms identified preclinically contribute to clinical resistance to IGF targeted therapy.
Components of downstream signalling pathways may also modulate response to IGF-1R inhibition [141]. While anti-IGF-1R antibodies down-regulate IGF-1R and inhibit activation of AKT, S6 and ERKs, at least acutely, it is recognized that AKT can be reactivated despite sustained IGF-1R down-regulation, leading to tumor progression [52,142]. Thus there is interest in combining IGF-1R inhibition with small molecule inhibitors of the PI3K/AKT pathway [143]. Given the redundancy in RTK signalling cascades, a logical therapeutic strategy would be to target the points at which these pathways converge. Inhibition of the mTOR complex 1 (mTORC1) by rapamycin and rapalogues everolimus and temsirolimus has been found to activate PI3K-Akt via a pro-oncogenic feedback loop that can be suppressed by IGF-1R blockade [21,144,145]. This could be an effective approach in tumors that harbor mutant LKB, in which expression of IGF axis components is upregulated [146], and in sarcomas where RPS6 has been shown to mediate resistance to IGF-1R inhibition [104]. Indeed, anti-tumor effects have been observed preclinically using rapamycin with figitumumab, ganitumab or ligand antibody BI 836845 in sarcoma models [21,147,148]. This strategy is being explored in clinical trials including NCT00927966, NCT00678769, NCT01016015, with promising evidence of durable disease control in metastatic adrenocortical cancers and sarcomas including EWS [70,72,149,150], findings that clearly merit further follow-up. IGFs regulate cell cycle progression by influencing the activity of cyclin-dependent kinases (CDKs), and IGF-1R inhibition has been shown to synergize with CDK4/6 inhibition in models of pancreatic ductal carcinoma and dedifferentiated liposarcoma [151,152].
Cross-talk between IGF and endocrine pathways has prompted clinical trials of IGF-1R inhibition with hormone therapy in breast and prostate cancer. IGF-1R was identified in a kinome-wide screen as a key mediator of resistance to endocrine therapy in hormone receptor positive breast cancer [153], but outcomes were not improved in this patient population by addition of ganitumab to the aromatase inhibitor exemestane or ER antagonist fulvestrant [154]. An earlier trial using figitumumab had similarly found no overall benefit from addition to exemestane, but a possible responsive subgroup based on glycemic control, with HbA1c <5.7% [155]. Additional preclinical data suggest that complete blockade of IGF signalling through IGF-1R and InsR A isoform is required for reversing resistance to endocrine therapy in ER+ breast cancer [156]. An ongoing study of BMS-754807 alone or with letrozole (a non-steroidal aromatase inhibitor) in patients with metastatic breast cancer is collecting post-treatment biopsies to evaluate potential mechanisms of resistance (NCT01225172). TNBC may also be an appropriate focus for this approach, given the adverse prognostic significance of IGF-1R expression in TNBC, and the sensitivity of TNBC cells to IGFs and IGF-1R inhibition [117,157,158]. In prostate cancer, preclinical data indicate that IGF-1R antibody cixutumumab caused significant delay in development of androgen resistance, and early trial results suggested benefit from IGF-1R blockade in men with this disease [93,159]. These data have prompted two neoadjuvant trials: figitumumab monotherapy [160], and cixutumumab with endocrine therapy (bicalutamide and goserelin) prior to radical prostatectomy [161].
Clinical trials combining IGF-1R inhibitory drugs with chemotherapy
The addition of IGF-1R inhibition to chemotherapy is supported by preclinical evidence that IGF-1R protects tumor cells from killing by cytotoxic drugs [68,162]. Further mechanisms that may be relevant include the ability of IGF-1R inhibition to suppress chemotherapy-induced IGF-1R activation and DNA damage repair [107,112,163–165]. Phase 1 trials suggested that IGF-1R inhibition is tolerable with chemotherapy, with evidence of clinical activity in heavily pre-treated patients [71,74,75,166–170]. The first reported Phase 2 trial with chemotherapy tested effects of figitumumab with paclitaxel and carboplatin in NSCLC. The initial results generated considerable enthusiasm when the trial found highly significant benefit in terms of objective responses and progression-free survival (PFS, hazard ratio 0.56) for patients receiving the higher dose of figitumumab [59]. However, it was subsequently determined that a substantial number of responses proved to be unconfirmed by RECIST criteria, and a later retraction stated that the differences in response rates were more modest, and without PFS prolongation [171]. Meanwhile, this trial had prompted a Phase 3 study that was prematurely discontinued after interim analysis favoured the paclitaxel/carboplatin only arm [172]. Unfortunately this pattern has been repeated, with failed trials being followed by subgroup analysis characterizing responsive patients. IGF-1R antibody ganitumab showed a trend to improved 6 month and overall survival when combined with gemcitabine in a Phase 2 trial in patients with pancreatic cancer [173]. This led to the Phase 3 GAMMA trial (NCT01231347) in unselected pancreatic cancer patients that was terminated prematurely due to lack of benefit [76]. Subsequently, as highlighted in the previous section, biomarker studies suggested that analysis of circulating IGFs and IGFBPs may identify subsets of ganitumab-responsive patients [82]. Two trials have evaluated addition of IGF-1R antibody to irinotecan-containing combination therapy in colorectal cancer: ganitumab with FOLFIRI chemotherapy in KRAS mutant cancers refractory to fluoropyrimidine- and oxaliplatin-based chemotherapy [114], and dalotuzumab with EGFR inhibitor cetuximab in chemorefractory KRAS wild-type or mutant cancers [64]. The latter suggested that addition of dalotuzumab actually worsened progression-free and overall survival. However, subsequent molecular analysis identified differential IGF expression and mesenchymal phenotype as potential markers of benefit from dalotuzumab [65,112]. It may also be relevant that these trials involved sustained antibody-mediated IGF-1R blockade, which could protect tumor cells from the toxicity of phase-specific cytotoxic drugs. Consistent with the concept that effects of IGF-1R inhibition on DNA repair play a major role in chemosensitization, no clinical activity has been seen in trials combining IGF-1R antibodies with taxanes, which do not damage DNA [174,175]. Furthermore, preclinical data suggest that the most effective inhibition of tumorigenesis occurs when the anti-IGF-1R agent is applied after DNA-damaging chemotherapy [176,177], which was not the design employed in the failed trials. These findings suggest that greater emphasis should be placed on the class of cytotoxic drug(s) with which IGF-1R agents are combined, and the scheduling of IGF-1R inhibition with chemotherapy; this will be more readily achieved using IGF-1R TKIs or short-half-life antibody therapeutics. Scheduling appears not to be critical for combination with radiation therapy, which causes DNA damage in all phases of the cell cycle, and to which IGF-1R inhibitory drugs have been shown to sensitize in head and neck, prostate and lung cancers [53,69,107,178], although this approach has not been tested clinically.
Conclusion
While many studies have not yet reported results, IGF-1R inhibitory antibodies and TKIs have not to date demonstrated significant benefit in unselected patients recruited to Phase 2/3 trials. This may be due, at least in part, to the lack of robust predictive biomarkers. Following the initial transition from bench to bedside, there is a clear need to look critically at the preclinical evidence, so that new trials are conducted in tumors with the appropriate molecular context. For example, IGF-1R inhibitory drugs may be more appropriately tested in triple negative rather than hormone receptor positive breast cancers [117,154,157,158], and in KRAS-mutant rather than wild-type colorectal and lung cancers [65,115]. Completed IGF-1R trials represent an opportunity to investigate the characteristics of responding and resistant subgroups, provided tumor tissue was collected. This in turn requires trial consents to be written with sufficiently broad scope to allow future translational research, so that newly identified biomarkers can be tested, and if validated, incorporated into future studies. There is as yet little consistent evidence regarding candidate tissue biomarkers for response to IGF-1R antibodies and TKIs, and it is not clear at this stage whether the same parameters will influence sensitivity to IGF ligand antibodies, which are still at a relatively early stage of development. However, enough trials have highlighted the association between response to IGF-1R inhibition and circulating IGFs, IGFBPs, and indicators of glycemic control [63,80–82,86,87,155] to support inclusion of these parameters in future trials of agents that remain under active investigation, for example [21,22,51,165]. Even amongst the failed trials, there are subsets of patients who have obtained durable benefit from IGF-1R inhibition. The challenge is to identify and characterize those patients, and use that information to increase the efficacy of this approach in cancer therapy.
Acknowledgments
The authors’ work is supported by NIHR Oxford Biomedical Research Centre, Prostate Cancer UK, Breast Cancer Campaign, Rosetrees Trust, UCARE-Oxford, Cancer Research UK (VMM), and by United States National Institutes of Health Grant CA136393, Mayo Clinic SPORE in Ovarian Cancer and the CA116201 Mayo Clinic Breast SPORE (PH).
Footnotes
Conflict of interest statement
Dr Haluska is the recipient of research funding from Bristol-Myers Squibb, Roche/Genentech and Merck, and is an unpaid consultant for BMS, Boehringer Ingelheim, and MedImmune. Dr. Macaulay has received a research Grant from AstraZeneca and is an unpaid consultant for Boehringer Ingelheim. The other authors have no conflict of interest.
Contributor Information
Helen King, Email: helen.king@stcatz.ox.ac.uk.
Tamara Aleksic, Email: taleksic13@googlemail.com.
Paul Haluska, Email: Haluska.Paul@mayo.edu.
Valentine M. Macaulay, Email: valentine.macaulay@oncology.ox.ac.uk.
References
- 1.Yuen JS, Macaulay VM. Targeting the type 1 insulin-like growth factor receptor as a treatment for cancer. Expert Opin Ther Targets. 2008;12:589–603. doi: 10.1517/14728222.12.5.589. [DOI] [PubMed] [Google Scholar]
- 2.Gualberto A, Pollak M. Emerging role of insulin-like growth factor receptor inhibitors in oncology: early clinical trial results and future directions. Oncogene. 2009;28:3009–21. doi: 10.1038/onc.2009.172. [DOI] [PubMed] [Google Scholar]
- 3.Tognon CE, Sorensen PH. Targeting the insulin-like growth factor 1 receptor (IGF1R) signaling pathway for cancer therapy. Expert Opin Ther Targets. 2012;16:33–48. doi: 10.1517/14728222.2011.638626. [DOI] [PubMed] [Google Scholar]
- 4.Sell C, Dumenil G, Deveaud C, et al. Effect of a null mutation of the insulin-like growth factor I receptor gene on growth and transformation of mouse embryo fibroblasts. Mol Cell Biol. 1994;14:3604–12. doi: 10.1128/mcb.14.6.3604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, et al. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med. 2011;3:70ra13. doi: 10.1126/scitranslmed.3001845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12:159–69. doi: 10.1038/nrc3215. [DOI] [PubMed] [Google Scholar]
- 7.Dunn SE, Ehrlich M, Sharp NJ, et al. A dominant negative mutant of the insulin-like growth factor-I receptor inhibits the adhesion, invasion, and metastasis of breast cancer. Cancer Res. 1998;58:3353–61. [PubMed] [Google Scholar]
- 8.Scotlandi K, Avnet S, Benini S, et al. Expression of an IGF-I receptor dominant negative mutant induces apoptosis, inhibits tumorigenesis and enhances chemosensitivity in Ewing’s sarcoma cells. Int J Cancer. 2002;101:11–6. doi: 10.1002/ijc.10537. [DOI] [PubMed] [Google Scholar]
- 9.Lee HY, Chun KH, Liu B, et al. Insulin-like growth factor binding protein-3 inhibits the growth of non-small cell lung cancer. Cancer Res. 2002;62:3530–7. [PubMed] [Google Scholar]
- 10.Rochester MA, Riedemann J, Hellawell GO, et al. Silencing of the IGF1R gene enhances sensitivity to DNA-damaging agents in both PTEN wild-type and mutant human prostate cancer. Cancer Gene Ther. 2005;12:90–100. doi: 10.1038/sj.cgt.7700775. [DOI] [PubMed] [Google Scholar]
- 11.Harper J, Burns JL, Foulstone EJ, et al. Soluble IGF2 receptor rescues Apc(Min/ +) intestinal adenoma progression induced by Igf2 loss of imprinting. Cancer Res. 2006;66:1940–8. doi: 10.1158/0008-5472.CAN-05-2036. [DOI] [PubMed] [Google Scholar]
- 12.Maloney EK, McLaughlin JL, Dagdigian NE, et al. An anti-insulin-like growth factor I receptor antibody that is a potent inhibitor of cancer cell proliferation. Cancer Res. 2003;63:5073–83. [PubMed] [Google Scholar]
- 13.Burtrum D, Zhu Z, Lu D, et al. A fully human monoclonal antibody to the insulin-like growth factor I receptor blocks ligand-dependent signaling and inhibits human tumor growth in vivo. Cancer Res. 2003;63:8912–21. [PubMed] [Google Scholar]
- 14.Sachdev D, Li SL, Hartell JS, et al. A chimeric humanized single-chain antibody against the type I insulin-like growth factor (IGF) receptor renders breast cancer cells refractory to the mitogenic effects of IGF-I. Cancer Res. 2003;63:627–35. [PubMed] [Google Scholar]
- 15.Mitsiades CS, Mitsiades NS, McMullan CJ, et al. Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer Cell. 2004;5:221–30. doi: 10.1016/s1535-6108(04)00050-9. [DOI] [PubMed] [Google Scholar]
- 16.Garcia-Echeverria C, Pearson MA, Marti A, et al. In vivo antitumor activity of NVP-AEW541-A novel, potent, and selective inhibitor of the IGF-IR kinase. Cancer Cell. 2004;5:231–9. doi: 10.1016/s1535-6108(04)00051-0. [DOI] [PubMed] [Google Scholar]
- 17.Cohen BD, Baker DA, Soderstrom C, et al. 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. Clin Cancer Res. 2005;11:2063–73. doi: 10.1158/1078-0432.CCR-04-1070. [DOI] [PubMed] [Google Scholar]
- 18.Ji QS, Mulvihill MJ, Rosenfeld-Franklin M, et al. 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. Mol Cancer Ther. 2007;6:2158–67. doi: 10.1158/1535-7163.MCT-07-0070. [DOI] [PubMed] [Google Scholar]
- 19.Rowinsky EK, Youssoufian H, Tonra JR, et al. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin Cancer Res. 2007;13:5549s–55s. doi: 10.1158/1078-0432.CCR-07-1109. [DOI] [PubMed] [Google Scholar]
- 20.Carboni JM, Wittman M, Yang Z, et al. BMS-754807, a small molecule inhibitor of insulin-like growth factor-1R/IR. Mol Cancer Ther. 2009;8:3341–9. doi: 10.1158/1535-7163.MCT-09-0499. [DOI] [PubMed] [Google Scholar]
- 21.Adam PF, Hofmann MH, Bogenrieder T, Borges E, Adolf GR. BI 836845 a fully human IGF ligand neutralizing antibody, to improve the efficacy of rapamycin by blocking rapamycin-induced AKT activation. J Clin Oncol. 2012;30(suppl):abstr. 3092. [Google Scholar]
- 22.Haluska P, Menefee ME, Plimack ER, Rosenberg JE, Northfelt DW, LaVallee T, et al. Safety, pharmacokinetics and antitumor activity of MEDI-573, an investigational monoclonal antibody that targets IGF-I and IGF-II, in adult patients with advanced solid tumors. J Clin Oncol. 2012;30(suppl):abstr. TPS2618. [Google Scholar]
- 23.Sachdev D, Singh R, Fujita-Yamaguchi Y, et al. 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 Res. 2006;66:2391–402. doi: 10.1158/0008-5472.CAN-05-3126. [DOI] [PubMed] [Google Scholar]
- 24.Wood ER, Shewchuk L, Hassel A, et al. Discovery of an inhibitor of insulin-like growth factor 1 receptor activation: implications for cellular potency and selectivity over insulin receptor. Biochem Pharmacol. 2009;78:1438–47. doi: 10.1016/j.bcp.2009.07.022. [DOI] [PubMed] [Google Scholar]
- 25.Favelyukis S, Till JH, Hubbard SR, et al. Structure and autoregulation of the insulin-like growth factor 1 receptor kinase. Nat Struct Biol. 2001;8:1058–63. doi: 10.1038/nsb721. [DOI] [PubMed] [Google Scholar]
- 26.Yee D. Insulin-like growth factor receptor inhibitors: baby or the bathwater? J Natl Cancer Inst. 2012;104:975–81. doi: 10.1093/jnci/djs258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Steiner L, Blum G, Friedmann Y, et al. ATP non-competitive IGF-1 receptor kinase inhibitors as lead anti-neoplastic and anti-papilloma agents. Eur J Pharmacol. 2007;562:1–11. doi: 10.1016/j.ejphar.2007.01.052. [DOI] [PubMed] [Google Scholar]
- 28.Ekman S, Frodin JE, Harmenberg J, et al. Clinical Phase I study with an Insulin-like growth factor-1 receptor inhibitor: experiences in patients with squamous non-small cell lung carcinoma. Acta Oncol. 2011;50:441–7. doi: 10.3109/0284186X.2010.499370. [DOI] [PubMed] [Google Scholar]
- 29.Friedbichler K, Hofmann MH, Kroez M, et al. Pharmacodynamic and antineoplastic activity of BI 836845, a fully human IGF ligand neutralizing antibody, and mechanistic rationale for combination with rapamycin. Mol Cancer Ther. 2013;13:399–409. doi: 10.1158/1535-7163.MCT-13-0598. [DOI] [PubMed] [Google Scholar]
- 30.Hartog H, Wesseling J, Boezen HM, et al. The insulin-like growth factor 1 receptor in cancer: old focus, new future. Eur J Cancer. 2007;43:1895–904. doi: 10.1016/j.ejca.2007.05.021. [DOI] [PubMed] [Google Scholar]
- 31.Macaulay VM. Insulin-like growth factors and cancer. Br J Cancer. 1992;65:311–20. doi: 10.1038/bjc.1992.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tolcher AW, Sarantopoulos J, Patnaik A, et al. Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. J Clin Oncol. 2009;27:5800–7. doi: 10.1200/JCO.2009.23.6745. [DOI] [PubMed] [Google Scholar]
- 33.Karp DD, Pollak MN, Cohen RB, et al. Safety, pharmacokinetics, and pharmacodynamics of the insulin-like growth factor type 1 receptor inhibitor figitumumab (CP-751,871) in combination with paclitaxel and carboplatin. J Thorac Oncol. 2009;4:1397–403. doi: 10.1097/JTO.0b013e3181ba2f1d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Carden CP, Kim ES, Jones RL, Alam SM, Johnson FM, Stephens AW, et al. Phase I study of intermittent dosing of OSI-906, a dual tyrosine kinase inhibitor of insulin-like growth factor-1 receptor (IGF-1R) and insulin receptor (IR) in patients with advanced solid tumors. J Clin Oncol. 2010;28(suppl):abstr. 2530. [Google Scholar]
- 35.Haluska P, Shaw HM, Batzel GN, et al. Phase I dose escalation study of the anti insulin-like growth factor-I receptor monoclonal antibody CP-751,871 in patients with refractory solid tumors. Clin Cancer Res. 2007;13:5834–40. doi: 10.1158/1078-0432.CCR-07-1118. [DOI] [PubMed] [Google Scholar]
- 36.Higano C, Alumkal J, Ryan CJ, et al. A phase II study evaluating the efficacy and safety of single agent IMC A12, a monoclonal antibody (MAb), against the insulin-like growth factor-1 receptor (IGF-IR), as monotherapy in patients with metastastic, asymptomatic castration-resistant prostate cancer (CRPC) J Clin Oncol. 2009;27(15s suppl):abstr. 5142. [Google Scholar]
- 37.Soria JC, Massard C, Lazar V, et al. A dose finding, safety and pharmacokinetic study of AVE1642, an anti-insulin-like growth factor-1 receptor (IGF-1R/ CD221) monoclonal antibody, administered as a single agent and in combination with docetaxel in patients with advanced solid tumours. Eur J Cancer. 2013;49:1799–807. doi: 10.1016/j.ejca.2013.01.003. [DOI] [PubMed] [Google Scholar]
- 38.Garcia JA, Elson P, Cooney MM, et al. Inhibition of the IGF pathway in metastatic castrate resistant prostate cancer (CRPC): results from a phase II study of OSI-906 (linsitinib) J Clin Oncol. 2013;31(suppl):abstr. e16022. [Google Scholar]
- 39.Olmos D, Postel-Vinay S, Molife LR, et al. Safety, pharmacokinetics, and preliminary activity of the anti-IGF-1R antibody figitumumab (CP-751,871) in patients with sarcoma and Ewing’s sarcoma: a phase 1 expansion cohort study. Lancet Oncol. 2010;11:129–35. doi: 10.1016/S1470-2045(09)70354-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kurzrock R, Patnaik A, Aisner J, et al. A phase I study of weekly R1507, a human monoclonal antibody insulin-like growth factor-I receptor antagonist, in patients with advanced solid tumors. Clin Cancer Res. 2010;16:2458–65. doi: 10.1158/1078-0432.CCR-09-3220. [DOI] [PubMed] [Google Scholar]
- 41.Tap WD, Demetri G, Barnette P, et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. J Clin Oncol. 2012;30:1849–56. doi: 10.1200/JCO.2011.37.2359. [DOI] [PubMed] [Google Scholar]
- 42.Reidy-Lagunes DL, Vakiani E, Segal MF, et al. A phase 2 study of the insulin-like growth factor-1 receptor inhibitor MK-0646 in patients with metastatic, well-differentiated neuroendocrine tumors. Cancer. 2012;118:4795–800. doi: 10.1002/cncr.27459. [DOI] [PubMed] [Google Scholar]
- 43.Schoffski P, Adkins D, Blay JY, et al. An open-label, phase 2 study evaluating the efficacy and safety of the anti-IGF-1R antibody cixutumumab in patients with previously treated advanced or metastatic soft-tissue sarcoma or Ewing family of tumours. Eur J Cancer. 2013;49:3219–28. doi: 10.1016/j.ejca.2013.06.010. [DOI] [PubMed] [Google Scholar]
- 44.Weigel B, Malempati S, Reid JM, et al. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2013;61:452–6. doi: 10.1002/pbc.24605. epub Aug 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ray-Coquard I, Haluska P, O’Reilly S, et al. A multicenter open-label phase II study of the efficacy and safety of ganitumab (AMG 479), a fully human monoclonal antibody against insulin-like growth factor type 1 receptor (IGF-1R) as second-line therapy in patients with recurrent platinum-sensitive ovarian cancer. J Clin Oncol. 2013;31(suppl):abstr. 5515. [Google Scholar]
- 46.Rajan A, Carter CA, Berman A, et al. Cixutumumab for patients with recurrent or refractory advanced thymic epithelial tumours: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2014;15:191–200. doi: 10.1016/S1470-2045(13)70596-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Toretsky JA, Kalebic T, Blakesley V, et al. The insulin-like growth factor-I receptor is required for EWS/FLI-1 transformation of fibroblasts. J Biol Chem. 1997;272:30822–7. doi: 10.1074/jbc.272.49.30822. [DOI] [PubMed] [Google Scholar]
- 48.Ayalon D, Glaser T, Werner H. Transcriptional regulation of IGF-I receptor gene expression by the PAX3-FKHR oncoprotein. Growth Horm IGF Res. 2001;11:289–97. doi: 10.1054/ghir.2001.0244. [DOI] [PubMed] [Google Scholar]
- 49.Martin MJ, Melnyk N, Pollard M, et al. The insulin-like growth factor I receptor is required for akt activation and suppression of anoikis in cells transformed by the ETV6-NTRK3 chimeric tyrosine kinase. Mol Cell Biol. 2006;26:1754–69. doi: 10.1128/MCB.26.5.1754-1769.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Huang HJ, Angelo LS, Rodon J, et al. R1507, an anti-insulin-like growth factor-1 receptor (IGF-1R) antibody, and EWS/FLI-1 siRNA in Ewing’s sarcoma: convergence at the IGF/IGFR/Akt axis. PLoS One. 2011;6:e26060. doi: 10.1371/journal.pone.0026060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ekman S, Harmenberg J, Frödin J-E, et al. A novel targeted oral insulin-like growth factor-1 receptor (IGF-1R) inhibitor and its implications for patients with non-small cell lung cancer (NSCLC): a phase I clinical trial. J Clin Oncol. 2013;30(suppl):abstr. 7539. [Google Scholar]
- 52.Cao L, Yu Y, Darko I, et al. Addiction to elevated insulin-like growth factor I receptor and initial modulation of the AKT pathway define the responsiveness of rhabdomyosarcoma to the targeting antibody. Cancer Res. 2008;68:8039–48. doi: 10.1158/0008-5472.CAN-08-1712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ferte C, Loriot Y, Clemenson C, et al. IGF-1R targeting increases the antitumor effects of DNA damaging agents in SCLC model: an opportunity to increase the efficacy of standard therapy. Mol Cancer Ther. 2013;12:1213–22. doi: 10.1158/1535-7163.MCT-12-1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Fleuren ED, Versleijen-Jonkers YM, van de Luijtgaarden AC, et al. Predicting IGF-1R therapy response in bone sarcomas: immuno-SPECT imaging with radiolabeled R1507. Clin Cancer Res. 2011;17:7693–703. doi: 10.1158/1078-0432.CCR-11-1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chakravarti A, Loeffler JS, Dyson NJ. Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res. 2002;62:200–7. [PubMed] [Google Scholar]
- 56.Hopkins A, Crowe PJ, Yang JL. Effect of type 1 insulin-like growth factor receptor targeted therapy on chemotherapy in human cancer and the mechanisms involved. J Cancer Res Clin Oncol. 2010;136:639–50. doi: 10.1007/s00432-010-0792-0. [DOI] [PubMed] [Google Scholar]
- 57.Chakraborty AK, Welsh A, Digiovanna MP. Co-targeting the insulin-like growth factor I receptor enhances growth-inhibitory and pro-apoptotic effects of anti-estrogens in human breast cancer cell lines. Breast Cancer Res Treat. 2010;120:327–35. doi: 10.1007/s10549-009-0382-5. [DOI] [PubMed] [Google Scholar]
- 58.Cortot AB, Repellin CE, Shimamura T, et al. Resistance to irreversible EGF receptor tyrosine kinase inhibitors through a multistep mechanism involving the IGF1R pathway. Cancer Res. 2013;73:834–43. doi: 10.1158/0008-5472.CAN-12-2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Karp DD, Paz-Ares LG, Novello S, et al. Phase II study of the anti-insulin-like growth factor type 1 receptor antibody CP-751,871 in combination with paclitaxel and carboplatin in previously untreated, locally advanced, or metastatic non-small-cell lung cancer. J Clin Oncol. 2009;27:2516–22. doi: 10.1200/JCO.2008.19.9331. [DOI] [PubMed] [Google Scholar]
- 60.Javale MM, Varadhachary GR, Fogelman DR, Shroff RT, Overman MJ, Ukegbu L, et al. Randomized phase II study of gemciabine (G) plus anti-IGF-1R antibody MK-0646, G plus erlotinib (E) plus MK-0646 and G plus E for advanced pancreatic cancer. J Clin Oncol. 2011;29(suppl):abstr. 4026. [Google Scholar]
- 61.Macaulay VM, Middleton MR, Eckhardt SG, et al. Phase I study of OSI-906, dual tyrosine kinase inhibitor of insulin-like growth factor-1 receptor (IGF-1R) and insulin receptor (IR) in combination with erlotinib (E) in patients with advanced solid tumors. J Clin Oncol. 2011;29(suppl):abstr. 3098. [Google Scholar]
- 62.Reidy DL, Vakiani E, Fakih MG, et al. Randomized, phase II study of the insulin-like growth factor-1 receptor inhibitor IMC-A12, with or without cetuximab, in patients with cetuximab- or panitumumab-refractory metastatic colorectal cancer. J Clin Oncol. 2010;28:4240–6. doi: 10.1200/JCO.2010.30.4154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Weickhardt A, Doebele R, Oton A, et al. A phase I/II study of erlotinib in combination with the anti-insulin-like growth factor-1 receptor monoclonal antibody IMC-A12 (cixutumumab) in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2012;7:419–26. doi: 10.1097/JTO.0b013e31823c5b11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Watkins D, Tabernero J, Schmoll H, et al. A randomized phase II/III study of the anti-IGF-1R antibody MK-0646 (dalotuzumab) in combination with cetuximab (Cx) and irinotecan (lr) in the treatment of chemofractory metastatic colorectal cancer (mCRC) with wild-type (wt) KRAS status. J Clin Oncol. 2011;29(suppl):abstr. 3501. [Google Scholar]
- 65.Watkins DJ, Ayers M, Cunningham D, Tabermero J, Telpar S, Kim T-Y, Kim TW, et al. Molecular analysis of the randomized phase II/III study of the anti-IGF-1R antibody dalotuzumab (MK-0646) in combination with cetuximab (Cx) and irinotecan (Ir) in the treatment of chemorefractory KRAS wild-type metastatic colorectal cancer (mCRC) J Clin Oncol. 2012;30(suppl):abstr. 3531. [Google Scholar]
- 66.Haluska P, Dhar A, Hou X, Huang F, Nuyten DSA, Park J. Phase II trial of the dual IGF-1R/IR inhibitor BMS-754807 with or without letrozole in aromatase inhibitor-resistant breast cancer. J Clin Oncol. 2011;29(suppl):abstr. TPS111. [Google Scholar]
- 67.Philip PA, Goldman B, Ramanathan RK, Lenz H-J, Lowy AM, Whitehead RP, et al. Dual blockade of epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor-1 (IGF-1R) signaling in metastatic pancreatic cancer: phase I/randomized II trial of gemcitabine, erlotinib, and cixutumumab versus gemcitabine plus erlotinib (SWOG-0727) J Clin Oncol. 2012;3(suppl):abstr. 4019. doi: 10.1002/cncr.28744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Awasthi N, Zhang C, Ruan W, et al. BMS-754807, a small-molecule inhibitor of insulin-like growth factor-1 receptor/insulin receptor, enhances gemcitabine response in pancreatic cancer. Mol Cancer Ther. 2012;11:2644–53. doi: 10.1158/1535-7163.MCT-12-0447. [DOI] [PubMed] [Google Scholar]
- 69.Matsumoto F, Valdecanas DN, Mason KA, et al. The impact of timing of EGFR and IGF-1R inhibition for sensitizing head and neck cancer to radiation. Anticancer Res. 2012;32:3029–35. [PubMed] [Google Scholar]
- 70.Naing A, LoRusso P, Fu S, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res. 2012;18:2625–31. doi: 10.1158/1078-0432.CCR-12-0061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Macaulay VM, Middleton MR, Protheroe AS, et al. Phase I study of humanized monoclonal antibody AVE1642 directed against the type 1 insulin-like growth factor receptor (IGF-1R), administered in combination with anticancer therapies to patients with advanced solid tumors. Ann Oncol. 2013;24:784–91. doi: 10.1093/annonc/mds511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Schwartz GK, Tap WD, Qin LX, et al. Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial. Lancet Oncol. 2013;14:371–82. doi: 10.1016/S1470-2045(13)70049-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Van Cutsem E, Eng C, Nowara E, et al. Randomized phase Ib/II trial of rilotumumab or ganitumab with panitumumab versus panitumumab alone in patients with wild-type KRAS metastatic colorectal cancer. Clin Cancer Res. 2014;4 doi: 10.1158/1078-0432.CCR-13-2752. epub June 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Doi T, Muro K, Yoshino T, et al. Phase 1 pharmacokinetic study of MK-0646 (dalotuzumab), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in combination with cetuximab and irinotecan in Japanese patients with advanced colorectal cancer. Cancer Chemother Pharmacol. 2013;72:643–52. doi: 10.1007/s00280-013-2240-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Mahadevan D, Sutton GR, Arteta-Bulos R, et al. 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 Chemother Pharmacol. 2014;73:467–73. doi: 10.1007/s00280-013-2372-x. [DOI] [PubMed] [Google Scholar]
- 76.Guha M. Anticancer IGF1R classes take more knocks. Nat Rev Drug Discov. 2013;12:250. doi: 10.1038/nrd3992. [DOI] [PubMed] [Google Scholar]
- 77.Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (iressa survival evaluation in lung cancer) Lancet. 2005;366:1527–37. doi: 10.1016/S0140-6736(05)67625-8. [DOI] [PubMed] [Google Scholar]
- 78.Craddock BP, Miller WT. Effects of somatic mutations in the C-terminus of insulin-like growth factor 1 receptor on activity and signaling. J Signal Transduct. 2012;2012:804801. doi: 10.1155/2012/804801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gualberto A, Hixon ML, Karp DD, et al. Pre-treatment levels of circulating free IGF-1 identify NSCLC patients who derive clinical benefit from figitumumab. Br J Cancer. 2011;104:68–74. doi: 10.1038/sj.bjc.6605972. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 80.Goto Y, Sekine I, Tanioka M, et al. Figitumumab combined with carboplatin and paclitaxel in treatment-naive Japanese patients with advanced non-small cell lung cancer. Invest New Drugs. 2012;30:1548–56. doi: 10.1007/s10637-011-9715-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ramalingam SS, Spigel DR, Chen D, et al. Randomized phase II study of erlotinib in combination with placebo or R1507, a monoclonal antibody to insulin-like growth factor-1 receptor, for advanced-stage non-small-cell lung cancer. J Clin Oncol. 2011;29:4574–80. doi: 10.1200/JCO.2011.36.6799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.McCaffery I, Tudor Y, Deng H, et al. Putative predictive biomarkers of survival in patients with metastatic pancreatic adenocarcinoma treated with gemcitabine and ganitumab, an IGF1R inhibitor. Clin Cancer Res. 2013;19:4282–9. doi: 10.1158/1078-0432.CCR-12-1840. [DOI] [PubMed] [Google Scholar]
- 83.Clemmons DR. Modifying IGF1 activity: an approach to treat endocrine disorders, atherosclerosis and cancer. Nat Rev Drug Discov. 2007;6:821–33. doi: 10.1038/nrd2359. [DOI] [PubMed] [Google Scholar]
- 84.Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer. 2008;8:915–28. doi: 10.1038/nrc2536. [DOI] [PubMed] [Google Scholar]
- 85.Gualberto A, Hixon ML, Karp DD, et al. Retraction of: pre-treatment levels of circulating free IGF-1 identify NSCLC patients who derive clinical benefit from figitumumab. Br J Cancer. 2012;107:2024. doi: 10.1038/bjc.2012.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Pavlicek A, Lira ME, Lee NV, et al. Molecular predictors of sensitivity to the insulin-like growth factor 1 receptor inhibitor, figitumumab (CP-751,871) Mol Cancer Ther. 2013;12:2929–39. doi: 10.1158/1535-7163.MCT-13-0442-T. [DOI] [PubMed] [Google Scholar]
- 87.Zha J, O’Brien C, Savage H, et al. Molecular predictors of response to a humanized anti-insulin-like growth factor-I receptor monoclonal antibody in breast and colorectal cancer. Mol Cancer Ther. 2009;8:2110–21. doi: 10.1158/1535-7163.MCT-09-0381. [DOI] [PubMed] [Google Scholar]
- 88.Mukohara T, Shimada H, Ogasawara N, et al. Sensitivity of breast cancer cell lines to the novel insulin-like growth factor-1 receptor (IGF-1R) inhibitor NVP-AEW541 is dependent on the level of IRS-1 expression. Cancer Lett. 2009;282:14–24. doi: 10.1016/j.canlet.2009.02.056. [DOI] [PubMed] [Google Scholar]
- 89.Becker MA, Hou X, Harrington SC, et al. IGFBP ratio confers resistance to IGF targeting and correlates with increased invasion and poor outcome in breast tumors. Clin Cancer Res. 2012;18:1808–17. doi: 10.1158/1078-0432.CCR-11-1806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Javle MM, Shroff RT, Varadhachary GR, et al. Tumor IGF-1 expression as a predictive biomarker for IGF1R-directed therapy in advanced pancreatic cancer (APC) J Clin Oncol. 2012;30(suppl):abstr. 4054. [Google Scholar]
- 91.Gong Y, Yao E, Shen R, et al. High expression levels of total IGF-1R and sensitivity of NSCLC cells in vitro to an anti-IGF-1R antibody (R1507) PLoS One. 2009;4:e7273. doi: 10.1371/journal.pone.0007273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Huang F, Greer A, Hurlburt W, et al. 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 Res. 2009;69:161–70. doi: 10.1158/0008-5472.CAN-08-0835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.de Bono JS, Attard G, Adjei A, et al. Potential applications for circulating tumor cells expressing the insulin-like growth factor-I receptor. Clin Cancer Res. 2007;13:3611–6. doi: 10.1158/1078-0432.CCR-07-0268. [DOI] [PubMed] [Google Scholar]
- 94.Browne BC, Eustace AJ, Kennedy S, et al. Evaluation of IGF1R and phosphorylated IGF1R as targets in HER2-positive breast cancer cell lines and tumours. Breast Cancer Res Treat. 2012;136:717–27. doi: 10.1007/s10549-012-2260-9. [DOI] [PubMed] [Google Scholar]
- 95.Wynes ME, Asuncion BR, Dziadziuszko R, Glisson BS, Wisuba II, Hirsch FR. Insulin-like growth factor (IGF-1R) protein expression (PE) and gene copy number 9GCN) for discrimination of response and outcome to figitumumab in NSCLC. J Clin Oncol. 2012;30(suppl):abstr. 7597. [Google Scholar]
- 96.Kim JG, Kang MJ, Yoon YK, et al. Heterodimerization of glycosylated insulin-like growth factor-1 receptors and insulin receptors in cancer cells sensitive to anti-IGF1R antibody. PLoS One. 2012;7:e33322. doi: 10.1371/journal.pone.0033322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Aleksic T, Chitnis MM, Perestenko OV, et al. Type 1 insulin-like growth factor receptor translocates to the nucleus of human tumor cells. Cancer Res. 2010;70:6412–9. doi: 10.1158/0008-5472.CAN-10-0052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sehat B, Tofigh A, Lin Y, et al. SUMOylation mediates the nuclear translocation and signaling of the IGF-1 receptor. Sci Signal. 2010;3:ra10. doi: 10.1126/scisignal.2000628. [DOI] [PubMed] [Google Scholar]
- 99.Sarfstein R, Pasmanik-Chor M, Yeheskel A, et al. Insulin-like growth factor-I receptor (IGF-IR) translocates to nucleus and autoregulates IGF-IR gene expression in breast cancer cells. J Biol Chem. 2012;287:2766–76. doi: 10.1074/jbc.M111.281782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bodzin AS, Wei Z, Hurtt R, et al. Gefitinib resistance in HCC mahlavu cells: upregulation of CD133 expression, activation of IGF-1R signaling pathway, and enhancement of IGF-1R nuclear translocation. J Cell Physiol. 2012;227:2947–52. doi: 10.1002/jcp.23041. [DOI] [PubMed] [Google Scholar]
- 101.Asmane I, Watkin E, Alberti L, et al. Insulin-like growth factor type 1 receptor (IGF-1R) exclusive nuclear staining: a predictive biomarker for IGF-1R monoclonal antibody (Ab) therapy in sarcomas. Eur J Cancer. 2012;48:3027–35. doi: 10.1016/j.ejca.2012.05.009. [DOI] [PubMed] [Google Scholar]
- 102.Zhang H, Sachdev D, Wang C, et al. Detection and downregulation of type I IGF receptor expression by antibody-conjugated quantum dots in breast cancer cells. Breast Cancer Res Treat. 2009;114:277–85. doi: 10.1007/s10549-008-0014-5. [DOI] [PubMed] [Google Scholar]
- 103.Chitnis MM, Yuen JS, Protheroe AS, et al. The type 1 insulin-like growth factor receptor pathway. Clin Cancer Res. 2008;14:6364–70. doi: 10.1158/1078-0432.CCR-07-4879. [DOI] [PubMed] [Google Scholar]
- 104.Potratz JC, Saunders DN, Wai DH, et al. Synthetic lethality screens reveal RPS6 and MST1R as modifiers of insulin-like growth factor-1 receptor inhibitor activity in childhood sarcomas. Cancer Res. 2010;70:8770–81. doi: 10.1158/0008-5472.CAN-10-1093. [DOI] [PubMed] [Google Scholar]
- 105.Pitts TM, Tan AC, Kulikowski GN, et al. Development of an integrated genomic classifier for a novel agent in colorectal cancer: approach to individualized therapy in early development. Clin Cancer Res. 2010;16:3193–204. doi: 10.1158/1078-0432.CCR-09-3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Isebaert SF, Swinnen JV, McBride WH, et al. Insulin-like growth factor-type 1 receptor inhibitor NVP-AEW541 enhances radiosensitivity of PTEN wild-type but not PTEN-deficient human prostate cancer cells. Int J Radiat Oncol Biol Phys. 2011;81:239–47. doi: 10.1016/j.ijrobp.2011.03.030. [DOI] [PubMed] [Google Scholar]
- 107.Chitnis MM, Lodhia KA, Aleksic T, et al. IGF-1R inhibition enhances radiosensitivity and delays double-strand break repair by both non-homologous end-joining and homologous recombination. Oncogene. 2013 doi: 10.1038/onc.2013.460. epub Nov 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Yeh J, Litz J, Hauck P, et al. Selective inhibition of SCLC growth by the A12 anti-IGF-1R monoclonal antibody correlates with inhibition of akt. Lung Cancer. 2008;60:166–74. doi: 10.1016/j.lungcan.2007.09.023. [DOI] [PubMed] [Google Scholar]
- 109.Bao XH, Takaoka M, Hao HF, et al. Esophageal cancer exhibits resistance to a novel IGF-1R inhibitor NVP-AEW541 with maintained RAS-MAPK activity. Anticancer Res. 2012;32:2827–34. [PubMed] [Google Scholar]
- 110.Flanigan SA, Pitts TM, Newton TP, et al. Overcoming IGF1R/IR resistance through inhibition of MEK signaling in colorectal cancer models. Clin Cancer Res. 2013;19:6219–29. doi: 10.1158/1078-0432.CCR-13-0145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zinn RL, Gardner EE, Marchionni L, et al. ERK phosphorylation is predictive of resistance to IGF-1R inhibition in small cell lung cancer. Mol Cancer Ther. 2013;12:1131–9. doi: 10.1158/1535-7163.MCT-12-0618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Sathyanarayanan S, Ayers M, Cunningham D, et al. Activity of the anti-IGF-1R antibody dalotuzumab (MK-0646) in KRAS-mutant colorectal cancer: preclinical and clinical data. J Clin Oncol. 2012;30(suppl):abstr. 3587. [Google Scholar]
- 113.Kim WY, Prudkin L, Feng L, et al. Epidermal growth factor receptor and K-Ras mutations and resistance of lung cancer to insulin-like growth factor 1 receptor tyrosine kinase inhibitors. Cancer. 2012;118:3993–4003. doi: 10.1002/cncr.26656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cohn AL, Tabernero J, Maurel J, et al. A randomized, placebo-controlled phase 2 study of ganitumab or conatumumab in combination with FOLFIRI for second-line treatment of mutant KRAS metastatic colorectal cancer. Ann Oncol. 2013;24:1777–85. doi: 10.1093/annonc/mdt057. [DOI] [PubMed] [Google Scholar]
- 115.Molina-Arcas M, Hancock DC, Sheridan C, et al. Coordinate direct input of both KRAS and IGF1 receptor to activation of PI3 kinase in KRAS-mutant lung cancer. Cancer Discov. 2013;3:548–63. doi: 10.1158/2159-8290.CD-12-0446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Creighton CJ, Casa A, Lazard Z, et al. Insulin-like growth factor-I activates gene transcription programs strongly associated with poor breast cancer prognosis. J Clin Oncol. 2008;26:4078–85. doi: 10.1200/JCO.2007.13.4429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Litzenburger BC, Creighton CJ, Tsimelzon A, et al. High IGF-IR activity in triple-negative breast cancer cell lines and tumorgrafts correlates with sensitivity to anti-IGF-IR therapy. Clin Cancer Res. 2011;17:2314–27. doi: 10.1158/1078-0432.CCR-10-1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Garofalo C, Mancarella C, Grilli A, et al. Identification of common and distinctive mechanisms of resistance to different anti-IGF-IR agents in Ewing’s sarcoma. Mol Endocrinol. 2012;26:1603–16. doi: 10.1210/me.2012-1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kim ES, Herbst RS, Wistuba II, et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 2011;1:44–53. doi: 10.1158/2159-8274.CD-10-0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yee D, Haddad T, Albain K, et al. Adaptive trials in the neoadjuvant setting: a model to safely tailor care while accelerating drug development. J Clin Oncol. 2012;30:4584–6. doi: 10.1200/JCO.2012.44.1022. author reply 4588–9. [DOI] [PubMed] [Google Scholar]
- 121.DeMichele A, Berry DA, Zujewski J, et al. Developing safety criteria for introducing new agents into neoadjuvant trials. Clin Cancer Res. 2013;19:2817–23. doi: 10.1158/1078-0432.CCR-12-2620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wilson TR, Fridlyand J, Yan Y, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–9. doi: 10.1038/nature11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Belfiore A, Frasca F, Pandini G, et al. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586–623. doi: 10.1210/er.2008-0047. [DOI] [PubMed] [Google Scholar]
- 124.Ulanet DB, Ludwig DL, Kahn CR, et al. Insulin receptor functionally enhances multistage tumor progression and conveys intrinsic resistance to IGF-1R targeted therapy. Proc Natl Acad Sci USA. 2010;107:10791–8. doi: 10.1073/pnas.0914076107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Bid HK, Zhan J, Phelps DA, et al. Potent inhibition of angiogenesis by the IGF-1 receptor-targeting antibody SCH717454 is reversed by IGF-2. Mol Cancer Ther. 2012;11:649–59. doi: 10.1158/1535-7163.MCT-11-0575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Fagan DH, Uselman RR, Sachdev D, et al. Acquired resistance to tamoxifen is associated with loss of the type I insulin-like growth factor receptor: implications for breast cancer treatment. Cancer Res. 2012;72:3372–80. doi: 10.1158/0008-5472.CAN-12-0684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Weroha SJ, Haluska P. The insulin-like growth factor system in cancer. Endocrinol Metab Clin North Am. 2012;41:335–50. vi. doi: 10.1016/j.ecl.2012.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Harrington SC, Weroha SJ, Reynolds C, et al. Quantifying insulin receptor isoform expression in FFPE breast tumors. Growth Horm IGF Res. 2012;22:108–15. doi: 10.1016/j.ghir.2012.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Buck E, Gokhale PC, Koujak S, et al. Compensatory insulin receptor (IR) activation on inhibition of insulin-like growth factor-1 receptor (IGF-1R): rationale for cotargeting IGF-1R and IR in cancer. Mol Cancer Ther. 2010;9:2652–64. doi: 10.1158/1535-7163.MCT-10-0318. [DOI] [PubMed] [Google Scholar]
- 130.Sharma SV, Lee DY, Li B, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell. 2010;141:69–80. doi: 10.1016/j.cell.2010.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Thariat J, Bensadoun RJ, Etienne-Grimaldi MC, et al. Contrasted outcomes to gefitinib on tumoral IGF1R expression in head and neck cancer patients receiving postoperative chemoradiation (GORTEC trial 2004–02) Clin Cancer Res. 2012;18:5123–33. doi: 10.1158/1078-0432.CCR-12-1518. [DOI] [PubMed] [Google Scholar]
- 132.Gallardo A, Lerma E, Escuin D, et al. Increased signalling of EGFR and IGF1R, and deregulation of PTEN/PI3K/Akt pathway are related with trastuzumab resistance in HER2 breast carcinomas. Br J Cancer. 2012;106:1367–73. doi: 10.1038/bjc.2012.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Huang F, Hurlburt W, Greer A, et al. Differential mechanisms of acquired resistance to insulin-like growth factor-i receptor antibody therapy or to a small-molecule inhibitor, BMS-754807, in a human rhabdomyosarcoma model. Cancer Res. 2010;70:7221–31. doi: 10.1158/0008-5472.CAN-10-0391. [DOI] [PubMed] [Google Scholar]
- 134.Song RX, Chen Y, Zhang Z, et al. Estrogen utilization of IGF-1-R and EGF-R to signal in breast cancer cells. J Steroid Biochem Mol Biol. 2010;118:219–30. doi: 10.1016/j.jsbmb.2009.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dayyani F, Parikh NU, Varkaris AS, et al. Combined Inhibition of IGF-1R/IR and Src family kinases enhances antitumor effects in prostate cancer by decreasing activated survival pathways. PLoS One. 2012;7:e51189. doi: 10.1371/journal.pone.0051189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Shin DH, Lee HJ, Min HY, et al. Combating resistance to anti-IGFR antibody by targeting the integrin beta3-src pathway. J Natl Cancer Inst. 2013;105:1558–70. doi: 10.1093/jnci/djt263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hou X, Huang F, Macedo LF, et al. Dual IGF-1R/InsR inhibitor BMS-754807 synergizes with hormonal agents in treatment of estrogen-dependent breast cancer. Cancer Res. 2011;71:7597–607. doi: 10.1158/0008-5472.CAN-11-1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Haluska P, Carboni JM, TenEyck C, et al. HER receptor signaling confers resistance to the insulin-like growth factor-I receptor inhibitor, BMS-536924. Mol Cancer Ther. 2008;7:2589–98. doi: 10.1158/1535-7163.MCT-08-0493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Buck E, Eyzaguirre A, Rosenfeld-Franklin M, et al. Feedback mechanisms promote cooperativity for small molecule inhibitors of epidermal and insulin-like growth factor receptors. Cancer Res. 2008;68:8322–32. doi: 10.1158/0008-5472.CAN-07-6720. [DOI] [PubMed] [Google Scholar]
- 140.Jameson MJ, Taniguchi LE, Vankoevering KK, et al. Activation of the insulin-like growth factor-1 receptor alters p27 regulation by the epidermal growth factor receptor in oral squamous carcinoma cells. J Oral Pathol Med. 2012;42:332–8. doi: 10.1111/jop.12014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Bertrand FE, Steelman LS, Chappell WH, et al. Synergy between an IGF-1R antibody and Raf/MEK/ERK and PI3K/Akt/mTOR pathway inhibitors in suppressing IGF-1R-mediated growth in hematopoietic cells. Leukemia. 2006;20:1254–60. doi: 10.1038/sj.leu.2404217. [DOI] [PubMed] [Google Scholar]
- 142.Atzori F, Tabernero J, Cervantes A, et al. A phase I pharmacokinetic and pharmacodynamic study of dalotuzumab (MK-0646), an anti-insulin-like growth factor-1 receptor monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res. 2011;17:6304–12. doi: 10.1158/1078-0432.CCR-10-3336. [DOI] [PubMed] [Google Scholar]
- 143.Fox EM, Kuba MG, Miller TW, et al. Autocrine IGF-I/insulin receptor axis compensates for inhibition of AKT in ER-positive breast cancer cells with resistance to estrogen deprivation. Breast Cancer Res. 2013;15:R55. doi: 10.1186/bcr3449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.O’Reilly KE, Rojo F, She QB, et al. MTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006;66:1500–8. doi: 10.1158/0008-5472.CAN-05-2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Yuen JS, Akkaya E, Wang Y, et al. Validation of the type 1 insulin-like growth factor receptor as a therapeutic target in renal cancer. Mol Cancer Ther. 2009;8:1448–59. doi: 10.1158/1535-7163.MCT-09-0101. [DOI] [PubMed] [Google Scholar]
- 146.Yilmaz E, Byers LA, Diao L, Giri U, Gudikote J, Fan YH, Wang J, et al. Use of proteomic analysis of LKB/1AMPK/mTOR pathways to identify IGF-1R pathway upregulation with LKB 1 loss or mTOR inhibition in NSCLC: implications for targeted combinations. J Clin Oncol. 2012;30(suppl):abstr. 10612. [Google Scholar]
- 147.Kurmasheva RT, Dudkin L, Billups C, et al. The insulin-like growth factor-1 receptor-targeting antibody, CP-751,871, suppresses tumor-derived VEGF and synergizes with rapamycin in models of childhood sarcoma. Cancer Res. 2009;69:7662–71. doi: 10.1158/0008-5472.CAN-09-1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Beltran PJ, Chung YA, Moody G, et al. Efficacy of ganitumab (AMG 479), alone and in combination with rapamycin, in Ewing’s and osteogenic sarcoma models. J Pharmacol Exp Ther. 2011;337:644–54. doi: 10.1124/jpet.110.178400. [DOI] [PubMed] [Google Scholar]
- 149.Quek R, Wang Q, Morgan JA, et al. Combination mTOR and IGF-1R inhibition: phase I trial of everolimus and figitumumab in patients with advanced sarcomas and other solid tumors. Clin Cancer Res. 2011;17:871–9. doi: 10.1158/1078-0432.CCR-10-2621. [DOI] [PubMed] [Google Scholar]
- 150.Naing A, Lorusso P, Fu S, et al. Insulin growth factor receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with metastatic adrenocortical carcinoma. Br J Cancer. 2013;108:826–30. doi: 10.1038/bjc.2013.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Heilmann AM, Perera RM, Ecker V, et al. CDK4/6 and IGF1 receptor inhibitors synergize to Suppress the growth of p16INK4A-deficient pancreatic cancers. Cancer Res. 2014;74:3947–58. doi: 10.1158/0008-5472.CAN-13-2923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Miller ML, Molinelli EJ, Nair JS, et al. Drug synergy screen and network modeling in dedifferentiated liposarcoma identifies CDK4 and IGF1R as synergistic drug targets. Sci Signal. 2013;6:ra85. doi: 10.1126/scisignal.2004014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Fox EM, Miller TW, Balko JM, et al. A kinome-wide screen identifies the insulin/IGF-I receptor pathway as a mechanism of escape from hormone dependence in breast cancer. Cancer Res. 2011;71:6773–84. doi: 10.1158/0008-5472.CAN-11-1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Robertson JF, Ferrero JM, Bourgeois H, et al. Ganitumab with either exemestane or fulvestrant for postmenopausal women with advanced, hormone-receptor-positive breast cancer: a randomised, controlled, double-blind, phase 2 trial. Lancet Oncol. 2013;14:228–35. doi: 10.1016/S1470-2045(13)70026-3. [DOI] [PubMed] [Google Scholar]
- 155.Ryan PD, Neven P, Blackwell KL, et al. Figitumumab plus exemestane versus exemestane as first-line treatment of postmenopausal hormone receptor-positive advanced breast cancer: a randomized open-label phase II trial. Cancer Res. 2011;71:Abstract P1–17-01. [Google Scholar]
- 156.Haluska P, Hou X, Huang F, et al. Complete IGF signaling blockade by the dual-kinase inhibitor, BMS-754807, is sufficient to overcome tamoxifen and letrozole resistance in vitro and in vivo. Cancer Res. 2009;69:Abstract nr 402. [Google Scholar]
- 157.Hartog H, Horlings HM, van der Vegt B, et al. 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 Res Treat. 2011;129:725–36. doi: 10.1007/s10549-010-1256-6. [DOI] [PubMed] [Google Scholar]
- 158.Davison Z, de Blacquiere GE, Westley BR, et al. Insulin-like growth factor-dependent proliferation and survival of triple-negative breast cancer cells: implications for therapy. Neoplasia. 2011;13:504–15. doi: 10.1593/neo.101590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Plymate SR, Haugk K, Coleman I, et al. An antibody targeting the type I insulin-like growth factor receptor enhances the castration-induced response in androgen-dependent prostate cancer. Clin Cancer Res. 2007;13:6429–39. doi: 10.1158/1078-0432.CCR-07-0648. [DOI] [PubMed] [Google Scholar]
- 160.Chi KN, Gleave ME, Fazli L, et al. A phase II pharmacodynamic study of preoperative figitumumab in patients with localized prostate cancer. Clin Cancer Res. 2012;18:3407–13. doi: 10.1158/1078-0432.CCR-12-0482. [DOI] [PubMed] [Google Scholar]
- 161.Dean JP, Sprenger CC, Wan J, et al. Response of the insulin-like growth factor (IGF) system to IGF-IR inhibition and androgen deprivation in a neoadjuvant prostate cancer trial: effects of obesity and androgen deprivation. J Clin Endocrinol Metab. 2013;98:E820–8. doi: 10.1210/jc.2012-3856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Dunn SE, Hardman RA, Kari FW, et al. Insulin-like growth factor 1 (IGF-1) alters drug sensitivity of HBL100 human breast cancer cells by inhibition of apoptosis induced by diverse anticancer drugs. Cancer Res. 1997;57:2687–93. [PubMed] [Google Scholar]
- 163.Trojanek J, Ho T, Del Valle L, et al. Role of the insulin-like growth factor I/ insulin receptor substrate 1 axis in Rad51 trafficking and DNA repair by homologous recombination. Mol Cell Biol. 2003;23:7510–24. doi: 10.1128/MCB.23.21.7510-7524.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Turney BW, Kerr M, Chitnis MM, et al. Depletion of the type 1 IGF receptor delays repair of radiation-induced DNA double strand breaks. Radiother Oncol. 2012;103:402–9. doi: 10.1016/j.radonc.2012.03.009. [DOI] [PubMed] [Google Scholar]
- 165.Harb WA, Sessa C, Hirte HW, et al. A phase I study evaluating the combination of OSI-906, a dual inhibitor of insulin growth factor-1 receptor (IGF-1R) and insulin receptor (IR) with weekly paclitaxel (PAC) in patients with advanced solid tumors. J Clin Oncol. 2011;29(suppl):abstr. 3099. [Google Scholar]
- 166.Molife LR, Fong PC, Paccagnella L, et al. The insulin-like growth factor-I receptor inhibitor figitumumab (CP-751,871) in combination with docetaxel in patients with advanced solid tumours: results of a phase Ib dose-escalation, open-label study. Br J Cancer. 2010;103:332–9. doi: 10.1038/sj.bjc.6605767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Harb WA, Sessa C, Hirte HW, et al. Final results of a phase I study evaluating the combination of linsitinib, a dual inhibitor of insulin-like growth factor-1 receptor (IGF-1R), and insulin receptor (IR) with weekly paclitaxel (PAC) in patients (Pts) with advanced solid tumors. J Clin Oncol. 2013;31(suppl):abstr. e13502. [Google Scholar]
- 168.Rosen LS, Puzanov I, Friberg G, et al. Safety and pharmacokinetics of ganitumab (AMG 479) combined with sorafenib, panitumumab, erlotinib, or gemcitabine in patients with advanced solid tumors. Clin Cancer Res. 2012;18:3414–27. doi: 10.1158/1078-0432.CCR-11-3369. [DOI] [PubMed] [Google Scholar]
- 169.Ellis PM, Shepherd FA, Laurie SA, et al. NCIC CTG IND. 190 phase I trial of dalotuzumab (MK-0646) in combination with cisplatin and etoposide in extensive-stage small-cell lung cancer. J Thorac Oncol. 2014;9:410–3. doi: 10.1097/JTO.0000000000000058. [DOI] [PubMed] [Google Scholar]
- 170.Okusaka T, Ikeda M, Fukutomi A, et al. Safety, tolerability, pharmacokinetics and antitumor activity of ganitumab, an investigational fully human monoclonal antibody to insulin-like growth factor type 1 receptor, combined with gemcitabine as first-line therapy in patients with metastatic pancreatic cancer: a phase 1b study. Jpn J Clin Oncol. 2014;44:442–7. doi: 10.1093/jjco/hyu034. [DOI] [PubMed] [Google Scholar]
- 171.Anonymous. Retraction. J Clin Oncol. 2012;30:4179. doi: 10.1200/jco.2012.47.3447. [DOI] [PubMed] [Google Scholar]
- 172.Langer CJ, Novello S, Park K, et al. Randomized, phase III trial of first-line figitumumab in combination with paclitaxel and carboplatin versus paclitaxel and carboplatin alone in patients with advanced non-small-cell lung cancer. J Clin Oncol. 2014;32:2059–66. doi: 10.1200/JCO.2013.54.4932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Kindler HL, Richards DA, Garbo LE, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol. 2012;23:2834–42. doi: 10.1093/annonc/mds142. [DOI] [PubMed] [Google Scholar]
- 174.de Bono JS, Piulats JM, Pandha HS, et al. Phase II randomized study of figitumumab plus docetaxel and docetaxel alone with crossover for metastatic castration-resistant prostate cancer. Clin Cancer Res. 2014;20:1925–34. doi: 10.1158/1078-0432.CCR-13-1869. [DOI] [PubMed] [Google Scholar]
- 175.Cohen SJ, Feng Y, Catalano PJ, et al. Randomized phase II study of paclitaxel with or without the anti-IGF-IR antibody cixutumumab (IMC-A12) as second-line treatment for patients with metastatic esophageal or GE junction cancer. J Clin Oncol. 2014;32(5s suppl):abstr. 4020. [Google Scholar]
- 176.Zeng X, Sachdev D, Zhang H, et al. Sequencing of type I insulin-like growth factor receptor inhibition affects chemotherapy response in vitro and in vivo. Clin Cancer Res. 2009;15:2840–9. doi: 10.1158/1078-0432.CCR-08-1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zeng X, Zhang H, Oh A, et al. Enhancement of doxorubicin cytotoxicity of human cancer cells by tyrosine kinase inhibition of insulin receptor and type I IGF receptor. Breast Cancer Res Treat. 2012;133:117–26. doi: 10.1007/s10549-011-1713-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Riesterer O, Yang Q, Raju U, et al. Combination of anti-IGF-1R antibody A12 and ionizing radiation in upper respiratory tract cancers. Int J Radiat Oncol Biol Phys. 2011;79:1179–87. doi: 10.1016/j.ijrobp.2010.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]