Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 12.
Published in final edited form as: Pediatr Blood Cancer. 2010 Dec 22;56(4):595–603. doi: 10.1002/pbc.22741

Initial Testing (Stage 1) of the IGF-1 Receptor Inhibitor BMS-754807 by the Pediatric Preclinical Testing Program

E Anders Kolb 1,*, Richard Gorlick 2, Richard Lock 3, Hernan Carol 3, Christopher L Morton 4, Stephen T Keir 5, C Patrick Reynolds 6, Min H Kang 6, John M Maris 7, Catherine Billups 4, Malcolm A Smith 8, Peter J Houghton 9
PMCID: PMC4263954  NIHMSID: NIHMS639061  PMID: 21298745

Abstract

Background

BMS-754807 is a small molecule ATP-competitive inhibitor of the type-1 insulin-like growth factor receptor currently in phase 1 clinical trials.

Procedures

BMS-754807 was tested against the Pediatric Preclinical Testing Program (PPTP) in vitro panel at concentrations ranging from 1.0 nM to 10 μM and was tested against the PPTP in vivo panels at a dose of 25 mg/kg administered orally BID for 6 days, repeated for 6 weeks.

Results

In vitro BMS-754807 showed a median EC50 value of 0.62 μM against the PPTP cell lines. The median EC50 for the four Ewing sarcoma cell lines was less than that for the remaining PPTP cell lines (0.19 μM vs. 0.78 μM, P = 0.0470). In vivo BMS-754807 induced significant differences in EFS distribution compared to controls in 18 of 32 evaluable solid tumor xenografts (56%) tested, but in none of the ALL xenografts studied. Criteria for intermediate activity for the time to event activity measure (EFS T/C >2) were met in 7 of 27 solid tumor xenografts evaluable for this measure. The best response was PD2 (progressive disease with growth delay), which was observed in 18 of 32 solid tumor xenografts. PD2 responses were most commonly observed in the rhabdomyosarcoma, neuroblastoma, osteosarcoma, Ewing sarcoma, and Wilms tumor panels.

Conclusions

BMS-754807 activity in vitro is consistent with a specific IGF-1R effect that has half-maximal response in the 0.1 μM range and that is observed in a minority of the PPTP cell lines. In vivo intermediate activity was most commonly observed in the neuroblastoma and rhabdomyosarcoma panels.

Keywords: developmental therapeutics, IGF-1 receptor inhibitor, preclinical testing

INTRODUCTION

For approximately two decades, the insulin like growth factor has been implicated in the pathogenesis of pediatric malignancies. Increasingly specific small molecules and antibodies directed to the insulin-like growth factor 1 receptor (IGF-1R) have renewed interest in this therapeutic strategy [1].

IGF-1R may bind both IGF-1 and IGF-2 resulting in autophosphorylation of the receptor. Phosphorylated IGF-1R initiates intracellular signaling through insulin receptor substrates 1–4 (IRS1–4) and the Src homology collagen-like adaptor protein (Shc). Phosphorylation of IRS-1 activates the p85 regulatory subunit of phosphatidylinositol 3-kinase (PI3K) ultimately leading to conversion of phosphatidylinositol-4,5-biphoshate (PIP2) to phosphatidylinositol-3,4,5-biphoshate (PIP3). PIP3 activates phosphoinositide-dependent kinase-1 (PDK1), resulting in phosphorylation and activation of the serine/threonine kinase Akt. Akt enhances intracellular metabolism through glycogen synthase kinase-3 (GSK3); protein synthesis and cell growth through the mammalian target of rapamycin (mTOR); cell survival through BAD (a Bcl-2 family member); and cell proliferation through p28Kip1 (reviewed in Refs. [2,3]). In parallel to the signaling activated though PI-3K, IGF-1R also activates Ras through Shc. Once activated, Ras initiates signaling through Raf, which ultimately leads to activation and phosphorylation of the mitogen-activated protein kinase-1 and -2 (MAPK) through the MAPK/ERK kinases (MEK1 and MEK2) [4]. Activation of MAPK through Ras results in enhanced proliferation, migration, and survival [4,5].

Currently in various stages of clinical development are several human or fully humanized anti-IGF-1R monoclonal antibodies, all requiring intravenous administration [1]. Clinical development of these agents is moving forward rapidly in sarcomas [6]. Small molecule inhibitors of IGF-1R have the advantage of oral bioavailability, and unlike the anti-IGF-1R antibodies, small molecule inhibitors of IGF-1R may also target the insulin receptor (IR) and the hybrid IGF-1R/IR receptor. In terms of blocking tumor growth and survival, there may be an advantage to inhibition of both IGF-1R and IR [7,8]. However, by targeting the IR, there is a possibility for symptomatic hyperglycemia in patients, thus limiting the clinical efficacy of the compounds.

BMS-754807 is an oral, reversible ATP-competitive antagonist of IGF-1R that blocks the activity of both IGF-1R and IR in in vitro kinase assays [9,10]. The agent inhibits the growth of a broad range of cancer cell lines, including mesenchymal, epithelial, and hematopoietic tumor cell lines [9]. BMS-754807 showed growth inhibitory activity against xenograft tumor models, and synergy was observed for combinations of the agent with both targeted agents and standard cytotoxic agents [9]. In the current report we have evaluated BMS-754807 against a panel of pediatric tumors, in vitro and in vivo, through the NCI-supported Pediatric Preclinical Testing Program (PPTP).

MATERIALS AND METHODS

In Vitro Testing

In vitro testing was performed using DIMSCAN, a semi-automatic fluorescence-based digital image microscopy system that quantifies viable (using fluorescein diacetate [FDA]) cell numbers in tissue culture multiwell plates [11]. Cells were incubated in the presence of BMS-754807 for 96 hr at concentrations from 1 nM to 10 μM and analyzed as previously described [12]. The activity of BMS-754807 was compared to that of a murine monoclonal antibody (mAb391) directed against the human IGF-1 receptor tested at a saturating concentration (50 μg/ml) [13].

In Vivo Tumor Growth Inhibition Studies

CB17SC-M scid−/− female mice (Taconic Farms, Germantown, NY), were used to propagate subcutaneously implanted kidney/rhabdoid tumors, sarcomas (Ewing, osteosarcoma, rhabdomyosarcoma), neuroblastoma, and non-glioblastoma brain tumors, while BALB/c nu/nu mice were used for glioma models, as previously described [14]. Human leukemia cells were propagated by intravenous inoculation in female non-obese diabetic (NOD)/scid−/− mice as described previously [15]. Female mice were used irrespective of the patient gender from which the original tumor was derived. All mice were maintained under barrier conditions and experiments were conducted using protocols and conditions approved by the institutional animal care and use committee of the appropriate consortium member. Ten mice (solid tumors) or eight mice (leukemias) were used in each control or treatment group. Tumor volumes (cm3) (solid tumor xenografts) or percentages of human CD45-positive (hCD45) cells (ALL xenografts) were determined as previously described [16] and responses were determined using three activity measures as previously described [16]. An in-depth description of the analysis methods is included in the Supplemental Response Definitions Section.

Statistical Methods

The exact log-rank test, as implemented using Proc StatXact for SAS®, was used to compare event-free survival distributions between treatment and control groups. P-values were two-sided and were not adjusted for multiple comparisons given the exploratory nature of the studies. The Mann–Whitney test was used to test the difference of medians of EC50 values between the groups of lines with similar tumor types to the remaining lines of the panel.

Drugs and Formulation

BMS-754807 was provided to the PPTP by Bristol Myers Squibb, through the Cancer Therapy Evaluation Program (NCI). BMS-754807 was dissolved in PEG400/water (80:20), and was administered PO twice daily (BID) for 6 days per week for 6 consecutive weeks at 25 mg/kg. mAb391 was provided to the PPTP by Bristol Myers Squibb.

RESULTS

BMS-754807 In Vitro Testing

BMS-754807 was evaluated against the 23 cell lines in the PPTP in vitro panel using 96-hr exposures to concentrations ranging from 1.0 nM to 10.0 μM. The median EC50 for the in vitro panel was 0.62 μM. There was more than a 70-fold range in EC50 values, with the most sensitive cell line being the rhabdomyosarcoma cell line Rh41 which had an EC50 of 0.07 μM and with the least sensitive cell line (Rh18) having an EC50 of 4.96 μM. The median EC50 for the 4 Ewing sarcoma cell lines was less than that for the remaining 19 PPTP cell lines (0.19 μM vs. 0.78 μM, P = 0.0470) (Table I). The median EC50 value for BMS-754807 for the five cell lines with the greatest response to the anti-IGF-1R monoclonal antibody mAb391 (all with inhibition >30%) was 0.12 μM, while the median EC50 for the 10 cell lines with the least evidence of mAb391 treatment effect was approximately 10-fold higher at 1.0 μM (P = 0.0017). This observation is consistent with a specific IGF-1R effect for BMS-754807 that has half-maximal response in the 0.1 μM range and that is observed in a minority of the PPTP cell lines, and with a non-IGF-1R effect that occurs in all of the cell lines and that shows half-maximal response at approximately 1 μM.

TABLE I.

Activity of BMS-754807 and mAb391 Against the PPTP In Vitro Panel

Cell line EC50 (μM)a Median EC50 ratiob Max inhibition (100—T/C) mAb391 inhibition at 50 μg/ml (100—T/C)
RD Rhabdomyosarcoma 1.12 0.56 100 17.7
Rh41 Rhabdomyosarcoma 0.07 9.20 98.4 86.4
Rh18 Rhabdomyosarcoma 4.96 0.13 100 20.8
Rh30 Rhabdomyosarcoma 0.19 3.32 98.8 39.0
BT-12 Rhabdoid (CNS) 0.78 0.79 74.7 10.9
CHLA-266 Rhabdoid (CNS) 0.89 0.70 98.0 8.2
TC-71 Ewing family tumor 0.11 5.86 98.5 69.4
CHLA-9 Ewing family tumor 0.12 5.31 96.9 28.0
CHLA-10 Ewing family tumor 0.62 1.00 98.6 0.0
CHLA-258 Ewing family tumor 0.27 2.31 99.4 50.9
GBM2 Glioblastoma 1.47 0.42 98.3 0.0
NB-1643 Neuroblastoma 0.12 5.15 93.3 47.1
NB-EBc1 Neuroblastoma 0.35 1.76 96.0 19.4
CHLA-90 Neuroblastoma 0.77 0.81 98.5 22.7
CHLA-136 Neuroblastoma 0.52 1.18 97.7 29.8
NALM-6 Pre-B cell ALL 0.49 1.26 92.7 0.0
COG-LL-317 T-cell ALL 1.38 0.45 99.6 0.0
RS4;11 Pre-B cell ALL 0.38 1.65 95.9 20.2
MOLT-4 T-cell ALL 0.53 1.18 94.9 5.4
CCRF-CEM T-cell ALL 1.13 0.55 91.3 0.0
Kasumi-1 AML 1.20 0.52 100 14.3
Karpas-299 Anaplastic large cell lymphoma 1.64 0.38 99.7 9.8
Ramos-RA1 Burkitt’s lymphoma 1.31 0.47 100 0.0
Median 0.62 1.00 98.4 17.7
a

The EC50 is the drug concentration achieving half maximal biological effect;

b

The median EC50 ratio is the relative EC50 values for the cell lines of the PPTP panel.

BMS-754807 In Vivo Testing

BMS-754807 was evaluated in 45 xenograft models. Thirty-five of 856 mice died during the study (4.1%), with 7 of 427 in the control arms (1.6%), and 28 of 429 in the BMS-754807 treatment arms (6.5%). Four solid tumor xenografts were inevaluable because of toxicity (GBM2, BT-39, and D456 from the GBM panel; CHLA-258 from the Ewing sarcoma panel) and a medulloblastoma xenograft (BT-50) was inevaluable because of inadequate growth of tumors in control animals. One of the eight ALL xenografts (ALL-4) was excluded from efficacy reporting because of excessive toxicity. A complete summary of results is provided in Supplemental Table I, including total numbers of mice, number of mice that died (or were otherwise excluded), numbers of mice with events and average times to event, tumor growth delay, as well as numbers of responses and T/C values.

Antitumor effects were evaluated using the PPTP activity measures for time to event (EFS T/C), tumor growth delay (tumor volume T/C), and objective response. BMS-754807 induced significant differences in EFS distribution compared to controls in 18 of 32 evaluable solid tumor xenografts (56%) tested as shown (Table II). Significant growth delay was observed in most of the solid tumor panels, including panels for rhabdoid tumors (3 of 3), Wilms tumor (3 of 3), rhabdomyosarcoma (2 of 6), Ewing sarcoma (2 of 4), neuroblastoma (4 of 6), and osteosarcoma (4 of 6). None of the seven evaluable ALL xenografts showed significant differences in EFS distribution between treated and control animals.

TABLE II.

Activity of BMS-754807 Against the PPTP In Vivo Tumor Panel

Xenograft line Histology Median time to event P-value EFS T/C Median final RTV Tumor volume T/C P-value T/C volume activity EFS activity Response activity
BT-29 Rhabdoid 21.0 0.027 1.5 >4 0.64 0.035 Low Low Low
KT-14 Rhabdoid >EP <0.001 >1.6 1.9 0.35 <0.001 Int NE Int
KT-12 Rhabdoid 11.4 0.008 1.5 >4 0.75 0.035 Low Low Low
KT-11 Wilms 17.3 0.004 1.8 >4 0.51 0.001 Low Low Int
KT-13 Wilms 13.4 <0.001 1.5 >4 0.39 <0.001 Int Low Low
KT-5 Wilms 34.3 <0.001 2.1 >4 0.60 0.003 Low Int Int
SK-NEP-1 Ewing 7.0 0.231 1.1 >4 0.86 0.218 Low Low Low
EW5 Ewing 13.4 0.042 2.1 >4 0.48 0.017 Low Int Int
EW8 Ewing 12.6 0.006 1.8 >4 0.73 0.035 Low Low Int
TC-71 Ewing 7.6 0.126 0.9 >4 1.15 0.353 Low Low Low
Rh10 ALV RMS 25.5 0.979 1.7 >4 0.50 0.043 Low Low Int
Rh28 ALV RMS 25.8 0.203 2.6 >4 0.51 0.009 Low Low Int
Rh30 ALV RMS 13.9 0.429 1.1 >4 0.81 0.105 Low Low Low
Rh30R ALV RMS 24.8 <0.001 2.3 >4 0.34 <0.001 Int Int Int
Rh41 ALV RMS 20.4 0.121 1.5 >4 0.59 0.011 Low Low Low
Rh18 EMB RMS 26.7 <0.001 2.1 >4 0.38 <0.001 Int Int Int
BT-28 Medulloblastoma 8.0 0.504 0.9 >4 0.96 0.912 Low Low Low
BT-45 Medulloblastoma 13.4 0.174 0.9 >4 1.10 0.280 Low Low Low
BT-41 Ependymoma >EP 1.000 2.4 0.71 0.089 Low NE Int
BT-44 Ependymoma 18.2 0.301 1.1 >4 0.71 0.029 Low Low Low
NB-SD Neuroblastoma 10.9 0.934 0.9 >4 1.10 0.574 Low Low Low
NB-1771 Neuroblastoma 11.4 <0.001 2.5 >4 0.30 0.002 Int Int Int
NB-1691 Neuroblastoma 9.9 0.426 1.0 >4 0.88 0.481 Low Low Low
NB-EBc1 Neuroblastoma 13.7 <0.001 2.7 >4 0.27 <0.001 Int Int Int
NB-1643 Neuroblastoma 27.1 0.012 3.4 >4 0.52 0.200 Low Int Int
SK-N-AS Neuroblastoma 7.7 0.004 1.6 >4 0.59 0.007 Low Low Int
OS-1 Osteosarcoma >EP <0.001 >1.3 1.3 0.75 0.035 Low NE Int
OS-2 Osteosarcoma >EP 0.055 >1.2 3.0 0.76 0.079 Low NE Int
OS-17 Osteosarcoma >EP 0.011 >1.4 3.1 0.77 0.074 Low NE Int
OS-9 Osteosarcoma 35.2 <0.001 1.6 >4 0.64 <0.001 Low Low Int
OS-33 Osteosarcoma 16.8 0.002 1.3 >4 0.74 0.003 Low Low Low
OS-31 Osteosarcoma 21.0 0.477 1.1 >4 0.94 0.353 Low Low Low
ALL-2 ALL B-precursor 10.9 0.612 0.7 >25 Low Low
ALL-3 ALL B-precursor 4.8 0.167 0.5 >25 Low Low
ALL-7 ALL B-precursor 4.2 0.932 1.0 >25 Low Low
ALL-8 ALL T-cell 4.6 0.627 0.9 >25 Low Low
ALL-16 ALL T-cell 4.4 0.141 0.5 >25 Low Low
ALL-17 ALL B-precursor 5.5 0.100 0.6 >25 Low Low
ALL-19 ALL B-precursor 4.6 0.097 0.7 >25 Low Low

Criteria for intermediate activity for the time to event activity measure (EFS T/C >2) were met in 7 of 27 (26%) solid tumor xenografts evaluable for this measure. Intermediate activity was most commonly observed in the neuroblastoma panel (3 of 6) and rhabdomyosarcoma panel (2 of 6). Single xenografts in the Wilms tumor and Ewing sarcoma panels also showed intermediate activity. Five models were inevaluable for the EFS T/C activity measure due to slow tumor growth in control animals, including three xenografts from the osteosarcoma panel.

Objective responses (i.e., tumor regression) were not observed for any of the solid tumor or ALL xenografts. The best response was PD2 (progressive disease with growth delay), which was observed in 18 of 32 solid tumor xenografts. PD2 responses were most commonly observed in the rhabdomyosarcoma (4 of 6), neuroblastoma (4 of 6), osteosarcoma (4 of 6), Ewing sarcoma (2 of 4), and Wilms tumor (2 of 3) panels. Each of the ALL xenografts showed PD1 (progressive disease without growth delay) (Fig. 1).

Fig. 1.

Fig. 1

BMS-754807 in vitro activity. Top panel: The median EC50 ratio graph shows the relative EC50 values for the cell lines of the PPTP panel. Each bar represents the ratio of the panel EC50 to the EC50 value of the indicated cell line. Bars to the right represent cell lines with higher sensitivity, while bars to the left indicate cell lines with lesser sensitivity. Bars highlighted with an asterisk (*) represent the three cell lines with greatest sensitivity to mAb391. Bottom panels: Representative dose response curves for Rh41 rhabdomyosarcoma and TC-71 Ewing sarcoma cell lines.

The in vivo testing results for the objective response measure of activity are presented in Figure 2 in a “heat-map” format as well as a “COMPARE”-like format, based on the scoring criteria described in the Supplemental Response Definitions Section. The latter analysis demonstrates relative tumor sensitivities around the midpoint score of 5 (stable disease). Typical tumor responses are shown in Figure 3 (KT14 (rhabdoid), KT5 (Wilms), Rh28 (rhabdomyosarcoma), OS1 (osteosarcoma)).

Fig. 2.

Fig. 2

BMS-754807 in vivo objective response activity. Left: The colored heat map depicts group response scores. A high level of activity is indicated by a score of 6 or more, intermediate activity by a score of ≥2 but <6, and low activity by a score of <2. Right: Representation of tumor sensitivity based on the difference of individual tumor lines from the midpoint response (stable disease). Bars to the right of the median represent lines that are more sensitive, and to the left are tumor models that are less sensitive. Red bars indicate lines with a significant difference in EFS distribution between treatment and control groups, while blue bars indicate lines for which the EFS distributions were not significantly different.

Fig. 3.

Fig. 3

BMS-754807 activity against individual solid tumor xenografts. Kaplan–Meier curves for EFS, median relative tumor volume graphs, and individual tumor volume graphs are shown for selected lines KT-5 (Wilms), KT-14 (rhabdoid), Rh28 (rhabdomyosarcoma), and OS-1 osteosarcoma xenografts. Controls (gray lines); Treated (black lines).

DISCUSSION

IGF-1 is a known mitogen for osteosarcoma [1719], neuroblastoma [20,21], brain tumors (including glioblastoma [22,23], astrocytoma [24], medulloblastoma [25]), Wilms tumor [26], and hepatocellular carcinoma [27]. In alveolar rhabdomyosarcoma, the PAX3-FKHR or PAX7-FKHR chimeric transcription factors associated with t(2;13)(q35;q14) and t(1;13)(q36;q14), enhance transcription IGF-1R [28,29]. In embryonal rhabdomyosarcoma, there is loss of imprinting at the IGF-2 locus, which may be a primary genetic event for embryonal rhabdomyosarcoma [30]. In Ewing sarcoma IGF-1R is a potent mediator of autocrine growth [31,32]. Cases of Ewing sarcoma with the type-1 EWS-FLI1 chimeric transcription factor are associated with an improved prognosis and with lower IGF-1R expression compared to cases with non-type 1 translocations [33]. EWS-FLI1 silencing leads to increased levels of insulin-like growth factor binding protein 3 gene (IGFBP-3), a major regulator of IGF-1 [34]. Expression of IGF-1R, IGF-1, and IGF-2 is consistently reported in osteosarcoma cell lines and patient samples [35]. Further evidence of IGF-mediated growth in osteosarcoma can be inferred from experiments demonstrating inhibition of growth and metastases of murine osteosarcoma following hypophysectomy [36], and inhibition of tumor growth in xenografts following treatment with anti-IGF-1R antibody [37]. The role of the IGF-1 axis in acute lymphoblastic leukemia is less clearly defined [3739].

In pediatric cancers, there are several IGF-1R targeted therapies currently in clinical trials, and an increasing body of preclinical data suggesting that IGF-1R is a high priority molecular target [6]. BMS-754807 is a small molecule inhibitor of IGF-1R with good oral bioavailability that is currently in clinical testing in adults with solid tumors. In the current report, in vitro activity of BMS-754807 was most pronounced in the Ewing sarcoma cell lines. Carboni et al. [9] also observed greater in vitro sensitivity for Ewing sarcoma cell lines compared to cell lines derived from other cancer types. Results for the murine anti-IGF-1R monoclonal antibody mAb391 show consistency with those for BMS-754807, as the EC50 for BMS-754807 is 10-fold lower in the mAb391-sensitive lines compared to the mAb391-resistant lines. The BMS-754807 micromolar range activity against cell lines non-responsive to mAb391 likely reflects non-IGF-1R/non-IR inhibitory activities of BMS-754807. For example, using biochemical assays of kinase inhibition, BMS-754807 shows low nanomolar activity against Met, TrkA, TrkB, and AurA, with IC50 values two- to fivefold greater than those observed for IGF-1R and IR [9].

BMS-754807 was well tolerated (6.3% mortality), so was tested at close to the maximal dose tolerated on this schedule. Toxicity was seen most frequently in the glioblastoma models that are propagated in athymic nude mice, although was no pattern of toxicity that could be associated with the mechanism of drug action. The in vivo response to previously tested anti-IGF-1R antibodies also predicts response to BMS-754807. The KT-14 rhabdoid tumor line, the EW5 Ewing tumor line, the RH10, RH28, RH30R rhabdomyosarcoma lines, the NB-1771, NB-EBc1, and NB1643 neuroblastoma lines, and the OS1, OS1, OS9, and OS17 osteosarcoma lines have consistently demonstrated intermediate to high responses to IGF-1R inhibitors [40,41]. As previously published by the PPTP, IGF-1R expression alone does not predict response to IGF-1R directed monoclonal antibodies [40]. In published studies antibodies that block ligand binding to IGF-1R induced regressions of tumors whereas BMS-754807 induced growth inhibition. The reason for this difference is not understood. However, unlike small molecule inhibitors of IGF-1R, the antibodies induce internalization and degradation of the IGF-1 receptor. The antibodies may also have superior pharmacokinetics in mice compared with the small molecule inhibitors.

A high priority for future research is to identify predictive biomarkers of response to agents targeted against IGF-1R [42]. In response to another small molecule inhibitor of IGF-1R (BMS-536924), Huang et al. recently demonstrated that IGF-1R, IGF-1, and IGF-2 are highly expressed in sensitive cell lines, while IGF binding proteins (IGFBP-3 and IGFBP-6) are highly expressed in resistant cell lines. Overexpression of EGFR and downstream components of EGFR signaling were also noted in BMS-536924 resistant tumor lines [43]. In hepatocellular carcinoma cell lines, HER3 activation by EGFR in response to IGR-1R inhibition seems to be one potential mechanism of resistance [44]. Tumor cell death is enhanced in rhabdomyosarcoma [43] and hepatocellular carcinoma cell lines [44] when both IGF-1R and EGFR signaling are inhibited. Clinical applications of IGF-1R inhibitors should focus on defining biomarkers of resistance, as well as combination strategies to inhibit alternate signaling though salvage pathways like EGFR. Cross talk between IGF-1R and EGFR make combination strategies targeting both receptors potentially more efficacious than single agent strategies.

The broad activity of BMS-754807 in pediatric sarcomas and neuroblastoma xenografts suggests that this agent may be effective in pediatric tumors. The fact that the activity of BMS-754807 is, for the most part, predicted by previously tested IGF-1R inhibitors in the PPTP panel supports the idea that a subset of pediatric tumors are susceptible to IGF-1R inhibition. Thus, it is likely that BMS-754807 may have limited single agent activity, similar to other kinase inhibitors. However, it is important to acknowledge that the efficacy of therapies targeting receptor tyrosine kinases may be maximized when used in rational combinations targeting multiple, related signaling pathways. Priority for clinical development of IGF-1R inhibitors should not be assigned based solely on results of single agent trial results. Priority should also be assigned to rational combinations in which an IGF-1R inhibitor is combined with inhibitors of targets such as mTOR [40], SRC family kinases [9], EGFR [9,43,44], or HER2 [9,45]. Further preclinical testing by the PPTP evaluating these and other rational combinations as well as combination with standard cytotoxic agents used in treatment of childhood malignancies may facilitate pediatric development of BMS-754807.

Supplementary Material

Supp Material
Supp TableS1

Acknowledgments

Additional Supporting Information may be found in the online version of this article.

Grant sponsor: National Cancer Institute; Grant numbers: NO1-CM-42216, CA21765, CA108786.

This work was supported by NO1-CM-42216, CA21765, and CA108786 from the National Cancer Institute and used BMS-754807 supplied by Bristol Myers Sqibb. In addition to the authors this paper represents work contributed by the following: Sherry Ansher, Joshua Courtright, Edward Favours, Henry S. Friedman, Debbie Payne-Turner, Charles Stopford, Chandra Tucker, Amy E. Watkins, Jianrong Wu, Joe Zeidner, Ellen Zhang, and Jian Zhang. Children’s Cancer Institute Australia for Medical Research is affiliated with the University of New South Wales and Sydney Children’s Hospital.

Footnotes

Conflict of interest: Nothing to declare.

References

  • 1.Gualberto A, Pollak M. Emerging role of insulin-like growth factor receptor inhibitors in oncology: Early clinical trial results and future directions. Oncogene. 2009;28:3009–3021. doi: 10.1038/onc.2009.172. [DOI] [PubMed] [Google Scholar]
  • 2.Rikhof B, de Jong S, Suurmeijer AJ, et al. The insulin-like growth factor system and sarcomas. J Pathol. 2009;217:469–482. doi: 10.1002/path.2499. [DOI] [PubMed] [Google Scholar]
  • 3.Kolb EA, Gorlick R. Development of IGF-IR Inhibitors in Pediatric Sarcomas. Curr Oncol Rep. 2009;11:307–313. doi: 10.1007/s11912-009-0043-1. [DOI] [PubMed] [Google Scholar]
  • 4.Hermanto U, Zong CS, Wang LH. Inhibition of mitogen-activated protein kinase kinase selectively inhibits cell proliferation in human breast cancer cells displaying enhanced insulin-like growth factor I-mediated mitogen-activated protein kinase activation. Cell Growth Differ. 2000;11:655–664. [PubMed] [Google Scholar]
  • 5.Kurmasheva RT, Houghton PJ. IGF-I mediated survival pathways in normal and malignant cells. Biochim Biophys Acta. 2006;1766:1–22. doi: 10.1016/j.bbcan.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 6.Kim SY, Toretsky JA, Scher D, et al. The role of IGF-1R in pediatric malignancies. Oncologist. 2009;14:83–91. doi: 10.1634/theoncologist.2008-0189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Avnet S, Sciacca L, Salerno M, et al. Insulin receptor isoform A and insulin-like growth factor II as additional treatment targets in human osteosarcoma. Cancer Res. 2009;69:2443–2452. doi: 10.1158/0008-5472.CAN-08-2645. [DOI] [PubMed] [Google Scholar]
  • 8.Zhang H, Pelzer AM, Kiang DT, et al. Down-regulation of type I insulin-like growth factor receptor increases sensitivity of breast cancer cells to insulin. Cancer Res. 2007;67:391–397. doi: 10.1158/0008-5472.CAN-06-1712. [DOI] [PubMed] [Google Scholar]
  • 9.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;8:3341–3349. doi: 10.1158/1535-7163.MCT-09-0499. [DOI] [PubMed] [Google Scholar]
  • 10.Wittman MD, Carboni JM, Yang Z, et al. Discovery of a 2,4-disubstituted pyrrolo[1,2-f][1,2,4]triazine inhibitor (BMS-754807) of insulin-like growth factor receptor (IGF-1R) kinase in clinical development. J Med Chem. 2009;52:7360–7363. doi: 10.1021/jm900786r. [DOI] [PubMed] [Google Scholar]
  • 11.Frgala T, Kalous O, Proffitt RT, et al. A fluorescence microplate cytotoxicity assay with a 4-log dynamic range that identifies synergistic drug combinations. Mol Cancer Ther. 2007;6:886–897. doi: 10.1158/1535-7163.MCT-04-0331. [DOI] [PubMed] [Google Scholar]
  • 12.Houghton PJ, Morton CL, Kolb EA, et al. Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;50:37–45. doi: 10.1002/pbc.21214. [DOI] [PubMed] [Google Scholar]
  • 13.Hailey J, Maxwell E, Koukouras K, et al. Neutralizing anti-insulin-like growth factor receptor 1 antibodies inhibit receptor function and induce receptor degradation in tumor cells. Mol Cancer Ther. 2002;1:1349–1353. [PubMed] [Google Scholar]
  • 14.Friedman HS, Colvin OM, Skapek SX, et al. Experimental chemotherapy of human medulloblastoma cell lines and transplantable xenografts with bifunctional alkylating agents. Cancer Res. 1988;48:4189–4195. [PubMed] [Google Scholar]
  • 15.Liem NL, Papa RA, Milross CG, et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood. 2004;103:3905–3914. doi: 10.1182/blood-2003-08-2911. [DOI] [PubMed] [Google Scholar]
  • 16.Houghton PJ, Morton CL, Tucker C, et al. The pediatric preclinical testing program: Description of models and early testing results. Pediatr Blood Cancer. 2007;49:928–940. doi: 10.1002/pbc.21078. [DOI] [PubMed] [Google Scholar]
  • 17.Pollak M, Richard M. Suramin blockade of insulinlike growth factor I-stimulated proliferation of human osteosarcoma cells. J Natl Cancer Inst. 1990;82:1349–1352. doi: 10.1093/jnci/82.16.1349. [DOI] [PubMed] [Google Scholar]
  • 18.Bostedt KT, Schmid C, Ghirlanda-Keller C, et al. Insulin-like growth factor (IGF) I down-regulates type 1 IGF receptor (IGF 1R) and reduces the IGF I response in A549 non-small-cell lung cancer and Saos-2/B-10 osteoblastic osteosarcoma cells. Exp Cell Res. 2001;271:368–377. doi: 10.1006/excr.2001.5388. [DOI] [PubMed] [Google Scholar]
  • 19.MacEwen EG, Pastor J, Kutzke J, et al. IGF-1 receptor contributes to the malignant phenotype in human and canine osteosarcoma. J Cell Biochem. 2004;92:77–91. doi: 10.1002/jcb.20046. [DOI] [PubMed] [Google Scholar]
  • 20.El-Badry OM, Helman LJ, Chatten J, et al. Insulin-like growth factor II-mediated proliferation of human neuroblastoma. J Clin Invest. 1991;87:648–657. doi: 10.1172/JCI115042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Weber A, Huesken C, Bergmann E, et al. Coexpression of insulin receptor-related receptor and insulin-like growth factor 1 receptor correlates with enhanced apoptosis and dedifferentiation in human neuroblastomas. Clin Cancer Res. 2003;9:5683–5692. [PubMed] [Google Scholar]
  • 22.Sandberg AC, Engberg C, Lake M, et al. The expression of insulin-like growth factor I and insulin-like growth factor II genes in the human fetal and adult brain and in glioma. Neurosci Lett. 1988;93:114–119. doi: 10.1016/0304-3940(88)90022-5. [DOI] [PubMed] [Google Scholar]
  • 23.Resnicoff M, Li W, Basak S, et al. Inhibition of rat C6 glioblastoma tumor growth by expression of insulin-like growth factor I receptor antisense mRNA. Cancer Immunol Immunother. 1996;42:64–68. doi: 10.1007/s002620050252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Antoniades HN, Galanopoulos T, Neville-Golden J, et al. Expression of insulin-like growth factors I and II and their receptor mRNAs in primary human astrocytomas and meningiomas; in vivo studies using in situ hybridization and immunocytochemistry. Int J Cancer. 1992;50:215–222. doi: 10.1002/ijc.2910500210. [DOI] [PubMed] [Google Scholar]
  • 25.Del Valle L, Enam S, Lassak A, et al. Insulin-like growth factor I receptor activity in human medulloblastomas. Clin Cancer Res. 2002;8:1822–1830. [PubMed] [Google Scholar]
  • 26.Hirschfeld S, Helman L. Diverse roles of insulin-like growth factors in pediatric solid tumors. In Vivo. 1994;8:81–90. [PubMed] [Google Scholar]
  • 27.Scharf JG, Braulke T. The role of the IGF axis in hepatocarcinogenesis. Horm Metab Res. 2003;35:685–693. doi: 10.1055/s-2004-814151. [DOI] [PubMed] [Google Scholar]
  • 28.Gidding CE, Germain GS, Dilling MB, et al. The influence of recombinant human insulin-like growth factor-I (rhIGF-I) on cell growth and cytotoxicity of drugs in childhood rhabdomyosarcoma cell lines and xenograft models. Cancer Chemother Pharmacol. 2000;45:21–30. doi: 10.1007/PL00006738. [DOI] [PubMed] [Google Scholar]
  • 29.Thimmaiah KN, Easton J, Huang S, et al. Insulin-like growth factor I-mediated protection from rapamycin-induced apoptosis is independent of Ras-Erk1-Erk2 and phosphatidylinositol 3−-kinase-Akt signaling pathways. Cancer Res. 2003;63:364–374. [PubMed] [Google Scholar]
  • 30.Fukuzawa R, Umezawa A, Ochi K, et al. High frequency of inactivation of the imprinted H19 gene in “sporadic” hepatoblastoma. Int J Cancer. 1999;82:490–497. doi: 10.1002/(sici)1097-0215(19990812)82:4<490::aid-ijc4>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
  • 31.Scotlandi K, Benini S, Nanni P, et al. Blockage of insulin-like growth factor-I receptor inhibits the growth of Ewing’s sarcoma in athymic mice. Cancer Res. 1998;58:4127–4131. [PubMed] [Google Scholar]
  • 32.Scotlandi K, Benini S, Sarti M, et al. Insulin-like growth factor I receptor-mediated circuit in Ewing’s sarcoma/peripheral neuroectodermal tumor: A possible therapeutic target. Cancer Res. 1996;56:4570–4574. [PubMed] [Google Scholar]
  • 33.de Alava E, Panizo A, Antonescu CR, et al. Association of EWS-FLI1 type 1 fusion with lower proliferative rate in Ewing’s sarcoma. Am J Pathol. 2000;156:849–855. doi: 10.1016/S0002-9440(10)64953-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Prieur A, Tirode F, Cohen P, et al. EWS/FLI-1 silencing and gene profiling of Ewing cells reveal downstream oncogenic pathways and a crucial role for repression of insulin-like growth factor binding protein 3. Mol Cell Biol. 2004;24:7275–7283. doi: 10.1128/MCB.24.16.7275-7283.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Burrow S, Andrulis IL, Pollak M, et al. Expression of insulin-like growth factor receptor, IGF-1, and IGF-2 in primary and metastatic osteosarcoma. J Surg Oncol. 1998;69:21–27. doi: 10.1002/(sici)1096-9098(199809)69:1<21::aid-jso5>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
  • 36.Pollak M, Sem AW, Richard M, et al. Inhibition of metastatic behavior of murine osteosarcoma by hypophysectomy. J Natl Cancer Inst. 1992;84:966–971. doi: 10.1093/jnci/84.12.966. [DOI] [PubMed] [Google Scholar]
  • 37.Kolb EA, Gorlick R, Houghton PJ, et al. Initial testing (stage 1) of a monoclonal antibody (SCH 717454) against the IGF-1 receptor by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;50:1190–1197. doi: 10.1002/pbc.21450. [DOI] [PubMed] [Google Scholar]
  • 38.Vorwerk P, Wex H, Hohmann B, et al. Expression of components of the IGF signalling system in childhood acute lymphoblastic leukaemia. Mol Pathol. 2002;55:40–45. doi: 10.1136/mp.55.1.40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kolb EA, Morton C, Houghton PJ, et al. Pediatric Preclinical Testing Program (PPTP) evaluation of the fully human anti-IGF-1R antibody IMC-A12. Eur J Cancer Suppl. 2008;6:176, Abstr #558. [Google Scholar]
  • 40.Kolb EA, Gorlick R, Houghton PJ, et al. Initial testing (stage 1) of a monoclonal antibody (SCH 717454) against the IGF-1 receptor by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;50:1190–1197. doi: 10.1002/pbc.21450. [DOI] [PubMed] [Google Scholar]
  • 41.Houghton PJ, Morton CL, Gorlick R, et al. Initial testing of a monoclonal antibody (IMC-A12) against IGF-1R by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer. 2010;54:921–926. doi: 10.1002/pbc.22367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Carden CP, Molife LR, de Bono JS. Predictive biomarkers for targeting insulin-like growth factor-I (IGF-I) receptor. Mol Cancer Ther. 2009;8:2077–2078. doi: 10.1158/1535-7163.MCT-09-0641. [DOI] [PubMed] [Google Scholar]
  • 43.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;69:161–170. doi: 10.1158/0008-5472.CAN-08-0835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Desbois-Mouthon C, Baron A, Blivet-Van Eggelpoel MJ, et al. Insulin-like growth factor-1 receptor inhibition induces a resistance mechanism via the epidermal growth factor receptor/HER3/AKT signaling pathway: Rational basis for cotargeting insulin-like growth factor-1 receptor and epidermal growth factor receptor in hepatocellular carcinoma. Clin Cancer Res. 2009;15:5445–5456. doi: 10.1158/1078-0432.CCR-08-2980. [DOI] [PubMed] [Google Scholar]
  • 45.Chakraborty AK, Liang K, DiGiovanna MP. Co-targeting insulin-like growth factor I receptor and HER2: Dramatic effects of HER2 inhibitors on nonoverexpressing breast cancer. Cancer Res. 2008;68:1538–1545. doi: 10.1158/0008-5472.CAN-07-5935. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Material
Supp TableS1

RESOURCES