The insulin-like growth factor (IGF) system consists of two ligands (IGF-I and IGF-II), which both signal through IGF-I receptor (IGF-IR) to stimulate proliferation and inhibit apoptosis, with activity contributing to malignant growth of many types of human cancers. We have developed a humanized, affinity-matured anti-human IGF-IR monoclonal antibody (h10H5), which binds with high affinity and specificity to the extracellular domain. h10H5 inhibits IGF-IR-mediated signaling by blocking IGF-I and IGF-II binding and by inducing cell surface receptor down-regulation via internalization and degradation, with the extracellular and intracellular domains of IGF-IR being differentially affected by the proteasomal and lysosomal inhibitors. In vitro, h10H5 exhibits antiproliferative effects on cancer cell lines. In vivo, h10H5 shows single-agent antitumor efficacy in human SK-N-AS neuroblastoma and SW527 breast cancer xenograft models and even greater efficacy in combination with the chemotherapeutic agent docetaxel or an anti–vascular endothelial growth factor antibody. Antitumor activity of h10H5 is associated with decreased AKT activation and glucose uptake and a 316-gene transcription profile with significant changes involving DNA metabolic and cell cycle machineries. These data support the clinical testing of h10H5 as a biotherapeutic for IGF-IR-dependent human tumors and furthermore illustrate a new method of monitoring its activity noninvasively in vivo via 2-fluoro-2-deoxy-d-glucose-positron emission tomography imaging. [Mol Cancer Ther 2008;7(9):2599–608]

The insulin-like growth factor (IGF) system plays crucial roles in regulating cellular proliferation, survival, and transformation (13). The central node of this system is IGF-I receptor (IGF-IR), a homodimeric receptor tyrosine kinase linked by disulfide bonds, with each monomer consisting of an extracellular α-subunit and a membrane-spanning β-subunit (4). The extracellular domain (ECD) of IGF-IR mediates ligand binding, which induces the activation of intracellular tyrosine kinase activity. Phosphatidylinositol 3-kinase, mitogen-activated protein kinase/extracellular signal-regulated kinase, and other downstream pathways are further recruited to amplify and execute prosurvival and proliferative signals that lead to stimulated growth (1).

Tumor cells can use the IGF system for their abnormal growth. Overexpression of IGF-I or IGF-II and/or IGF-IR and activation of this pathway are often observed in several prevalent cancer types (1). In particular, epigenetic changes, such as loss-of-imprinting at the IGF-II locus, frequently occur in colon and ovarian cancers as well as in several pediatric malignancies (5). High serum IGF-I concentrations correlate with increased risk for breast, prostate, and colon cancers in humans (6). Gain-of-function studies show that the IGF system can drive tumorigenesis in animal models (7, 8), whereas IGF-IR deficiency conversely inhibits cellular transformation (9). Taken together, these data provide a strong scientific rationale for developing targeted therapies to inhibit this pathway in human cancer.

Multiple approaches, such as anti-IGF-IR monoclonal antibodies and small-molecule receptor tyrosine kinase inhibitors, have been developed to inhibit the activity of this pathway in cancer (1021), and many of them are being actively tested in phase I or II clinical trials (22). Because small-molecule inhibitors of the IGF-IR kinase often cross-inhibit insulin receptor, we have developed a novel humanized, affinity-matured IgG1κ monoclonal antibody (h10H5) against the IGF-IR ECD. We report that h10H5 effectively inhibits IGF-IR mediated signaling by blocking both IGF-I and IGF-II binding to IGF-IR and by inducing IGF-IR down-regulation through internalization and degradation while leaving insulin receptor unaffected. Furthermore, we show that degradation of the ECD and intracellular domain of IGF-IR is differentially affected by the lysosomal and proteasomal inhibitors, consistent with immunofluorescence data showing h10H5 uptake by clathrin-mediated endocytosis followed by transport to late endosomes and lysosomes for degradation. Functionally, h10H5 exhibits antiproliferative activity against cancer cell lines in vitro and in vivo, which can be enhanced by not only chemotherapy but also anti–vascular endothelial growth factor (VEGF) antibodies. Notably, the antitumor activity of h10H5 is associated with decreased AKT activation, a composite transcription profile with significant changes involving DNA metabolic and cell cycle machineries, and can also be monitored in vivo by 2-fluoro-2-deoxy-d-glucose (FDG)-positron emission tomography (PET) imaging. Taken together, these data not only support the clinical testing of h10H5 as a biotherapeutic for human tumors that rely on IGF-IR for proliferation and survival but also show the utility of a novel endpoint assay for h10H5 activity that can be used to assess antitumor activity noninvasively in vivo.

Generation and Characterization of Anti-IGF-IR Antibody h10H5

BALB/c mice (Charles River Laboratories) were hyperimmunized with human IGF-IR ECD (amino acids 1-902) in Ribi adjuvant (Ribi Immunochem Research). B cells from these mice, all of which showed high anti-IGF-IR antibody titers by direct ELISA and by fluorescence-activated cell sorting of IGF-IR-expressing MCF7 cells, were fused with mouse myeloma cells (X63.Ag8.653; American Type Culture Collection) based on described protocols (23). After 10-12 days, the supernatants were harvested and screened for antibody production by direct ELISA and fluorescence-activated cell sorting. Murine 10H5 was chosen from a series of positive clones for its strong binding to the purified IGF-IR ECD and cell-surface IGF-IR as well as its effective blockade of ligand-receptor interaction. Humanization of 10H5 was achieved by direct CDR grafts onto the acceptor human consensus (subtype III) framework of trastuzumab (24). Mutant h10H5 carrying a D265A mutation (based on Kabat numbering), which has diminished binding to all Fcγ receptors, was produced for antibody-dependent cell-mediated cytotoxicity studies. Biacore analysis, using the F(ab) fragment of h10H5 to avoid avidity effects, showed a dissociation equilibrium constant (Kd) of 217 and 159 pmol/L for binding to the recombinant human and cynomolgus monkey IGF-IR ECD, respectively (data not shown). h10H5 did not react with the closely related human insulin receptor or murine IGF-IR, thus showing strong binding selectivity (data not shown).

Reagents and Cell Lines

Human breast cancer cell lines MCF7 and SW527 and neuroblastoma cell line SK-N-AS were obtained from the American Type Culture Collection. Antibodies to phosphorylated and total IGF-IR β-chain, AKT, and extracellular signal-regulated kinase 1/2 were purchased from Cell Signaling Technology. Anti-IGF-IR α-chain monoclonal antibody 10F5 was generated in-house as for murine 10H5. Lysosomal protease inhibitors leupeptin and pepstatin A were from Roche Applied Science. Bortezomib was produced in-house and is the active ingredient of the proteasome inhibitor Velcade that has been approved for treating multiple myeloma (25). Recombinant IGF-I and IGF-II were purchased from R&D Systems.

Solid-Phase Binding Assay

Recombinant human IGF-IR ECD was coated onto 96-well plates at 200 ng/well by overnight incubation at 4°C. All subsequent steps were at room temperature: after washing with 0.05% Tween 20 in PBS, plates were blocked for 1 h with 5% bovine serum albumin in PBS, incubated for 1 h with serially diluted h10H5, anti-IGF-IR antibody αIR3 (Calbiochem), or control antibody anti-gp120 (to herpes simplex virus glycoprotein D). Biotinylated IGF-I or IGF-II (80 ng/mL) was subsequently added for 1 h, washed, and detected with streptavidin-horseradish peroxidase (1:6,000) for 20 min. The signals were developed with TMB substrate, and the reactions were stopped with 100 μL/well of 1 mol/L phosphoric acid before reading the absorbance at A450 nm.

Microscopy

SK-N-AS and MCF7 cells grown on LabTek II slides were incubated with 5 μg/mL h10H5 for 5 min to 4 h at 37°C, 5% CO2 in complete (10% fetal bovine serum) growth medium containing lysosomal protease inhibitors (5 μmol/L pepstatin A and 10 μg/mL leupeptin) and 10 μg/mL Alexa 488-transferrin. Cells were then chilled, washed five times in cell medium, fixed for 20 min with 3% paraformaldehyde, quenched for 10 min with 50 mmol/L NH4Cl in PBS, and permeabilized with saponin buffer [0.4% (w/v) saponin, 2% fetal bovine serum, 1% bovine serum albumin]. Uptaken h10H5 was detected with Cy3 anti-human and where indicated cells were also stained with 1:1,000 mouse anti-LAMP1 (BD Biosciences) or 1:100 rabbit anti-IGF-IR β-subunit followed by FITC-conjugated secondaries (Jackson ImmunoResearch). Slides were coverslipped with 4′,6-diamidino-2-phenylindole–containing VectaShield (Vector Labs) and imaged by epifluorescence microscopy using the ×100 objective of a DeltaVision deconvolution microscope (Applied Precision) powered by SoftWorx (version 3.4.4). Figures were compiled using Adobe Photoshop CS.

Immunoprecipitation and Western Blotting

Cells were lysed in lysis buffer [20 mmol/L Tris-HCl (pH 7.5), 150 mmol/L NaCl, 1 mmol/L EDTA, 1 mmol/L EGTA, 1% Triton X-100] with protease inhibitors and/or phosphatase inhibitor cocktail (Sigma) on ice for 20 min and cleared by centrifugation at 14,000 rpm for 15 min. Equal amount of cell lysates measured by the BCA assay (Pierce Biotechnology) were immunoprecipitated with anti-IGF-IR antibody 10F5 or directly Western blotted.

Cell Proliferation/Viability and Glucose Uptake Assays

For cell viability assays, MCF7 cells were seeded at 2,000 per well and incubated overnight at 37°C in RPMI 1640 with 10% fetal bovine serum (Sigma) and 1% Glutamax (Promega). Cells were then starved in serum-free RPMI 1640 (phenol red–free) for 5 h followed by addition of serially diluted h10H5 for 1 h and then supplemented with 1% fetal bovine serum and 1 ng/mL IGF-I for 5 days. Cell viability was determined using the CellTiter-Glo Luminescent Cell Viability kit (Promega) according to the manufacturer's instructions.

For the FDG uptake assay, SK-N-AS cells were plated in 96-well plates at 10,000 per well. The following day, medium was removed and replaced with glucose-free DMEM containing 0.1% or 0% fetal bovine serum and a range of h10H5 concentrations for a further 48 h. To determine FDG uptake, 2 μCi/well [3H]FDG ([5,6-3H]; 0.74-2.22 TBq/mmol; 20-60 Ci/mmol; American Radiolabeled Chemicals) was added for the last 24 h and harvested onto Unifilter GF/C filter plates using a FilterMate Harvester (Perkin-Elmer). Microscint 20 scintillation fluid was added to all wells and radioactivity incorporated per well (counts/min) was determined using a Packard TopCount NXT microplate scintillation counter.

Microarray and Quantitative Real-time Reverse Transcription-PCR

Total RNA was prepared from SK-N-AS xenograft tumors using the RNeasy kit (Qiagen). Affymetrix human genome U133 Plus 2.0 arrays were employed for expression profiling according to the manufacturer's instructions. Probe set analysis was done using Affymetrix MAS5.0. Quantitative real-time reverse transcription-PCR was done in triplicates for each sample using the TaqMan Gold reagent kit (Applied Biosystems) and Stratagene Mx4000 according to the manufacturer's instructions (standard curve method: user bulletin no. 2, ABI PRISM 7700 SDS). For normalization, the mean expression level of target genes was divided by the mean of the expression level of the housekeeping gene RPL19. The sequences of the primer/probe sets (5′-FAM reporter dye and 3′-TAMRA quencher labeled) are listed in Supplementary Table S1.1

1

Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

Student's t test was used to perform statistical analysis of differentially expressed genes, and ESAE scores, which are modified Fisher's exact probability P values, were generated. Functional annotation of the genes was done using the Database for Annotation, Visualization and Integrated Discovery.2

Tumor Xenograft Studies

Female nu/nu and C.B-17 SCID beige mice were inoculated s.c. with 10 × 106 SK-N-AS and 5 × 106 SW527 tumor cells. Once tumors reached a mean volume of 130 to 260 mm3, mice were then randomized into groups of 8 to 10 mice and treated with antibody and/or chemotherapy. Percent tumor growth inhibition (%TGI) was calculated as 100 × [(CT) / C], where C and T are the mean tumor volume of the control and treated groups, respectively. Data analysis and generation of P values were done using the Dunnett's t test for tumor volumes, or a log-rank test for doubling time, defined as the number of days for a tumor to double its size measured at the day of randomization, with JMP software version 6.0 (SAS Institute).

FDG-PET Imaging Study

A separate SK-N-AS xenograft study was conducted to evaluate FDG-PET imaging as a measure of drug response. Dynamic PET scans were done on tumors following caliper measurements on day 0 before treatment and again on days 3, 7, 10, and 14. At the beginning of a 30-min dynamic PET scan, ∼250 μCi F18-FDG was injected into the lateral tail vein of each mouse. PET data were processed into a time series of images to allow quantification of the tumor relative to the blood pool using the liver signal as a proxy for blood (26) and the region of interest analysis was done (Siemens Preclinical Solutions). Numerical integration and calculations for the conversion of the raw data into Patlak plots were done with the PET Parser program. The slope of the linear portion of the Patlak plot is equal to Ki, the FDG uptake rate constant (units/s). Treatment responses were assessed as the percentage change in Ki relative to the pretreatment value, with a negative change representing a decrease in FDG uptake. These difference data were used for the t tests comparing vehicle and treatment groups at the imaging time points. Data collation and statistical analysis were done using Microsoft Excel using the built-in Student's t test in a two-tailed comparison.

h10H5 Blocks IGF Binding to IGF-IR and Inhibits the Receptor-Mediated Signaling

h10H5 is a humanized anti-IGF-IR monoclonal antibody with high binding affinity and specificity as described in Materials and Methods. In competitive solid-phase binding assays, h10H5 inhibited IGF-I and IGF-II binding to IGF-IR with an IC50 of 3.4 nmol/L (Supplementary Figs. S1A and B).1 To investigate the effects of h10H5 on IGF-IR signaling, serum-starved human breast cancer MCF7 cells were treated with antibody for 20 min before IGF-I or IGF-II stimulation. In comparison with untreated cells (Supplementary Fig. S2A,1 lane 1), IGF-I induced robust and sustained phosphorylation of IGF-IR and AKT from 10 to 120 min, but only transient extracellular signal-regulated kinase 1/2 phosphorylation, which peaked at 10 min and reduced at later time points (Supplementary Fig. S2A,1 lanes 2-6). Exposure of cells to h10H5 strongly inhibited these effects of IGF-I throughout the entire time course (Supplementary Fig. S2A,1 lanes 7-11), with the greatest effect at 1 to 10 μg/mL h10H5 (Supplementary Fig. S2B and C,1 lanes 4-6). In contrast, the control (anti-gp120) antibody failed to inhibit IGF-I- or IGF-II-mediated signaling at the same concentrations (Supplementary Fig. S2A,1 lanes 12-14; Supplementary Fig. S2B and C,1 lane 3). In the absence of ligand stimulation, h10H5 (up to 100 μg/mL) did not induce any observable phosphorylation of IGF-IR, AKT, or extracellular signal-regulated kinase 1/2 (data not shown). Taken together, these data indicate that h10H5 is a potent antagonist of IGF-I- and IGF-II-mediated IGF-IR signaling.

Proteasomal and Lysosomal Inhibitors Differentially Affect h10H5-Induced IGF-IR α- and β-Subunit Down-Regulation

One common mechanism of action of anti-IGF-IR antibodies is the induction of receptor down-regulation (1214, 1719). Similarly, h10H5 led to IGF-IR depletion in MCF7 cells after 1 h (Supplementary Fig. S2A,1 lanes 9-11). We expanded our analysis to a different cell line, SK-N-AS, by examining IGF-IR levels in a more prolonged time course with various doses of h10H5 treatment (Fig. 1A). As in MCF7 cells, IGF-IR was already partially down-regulated at 1 h, reaching maximal reduction at 4 to 8 h and not diminishing further at 24 h with ≥1 μg/mL h10H5 (data not shown). Down-regulation also occurred to a similar extent at 0.1 μg/mL but was slower at early time points. In contrast, neither IGF-I nor anti-gp120 antibody treatment affected IGF-1R levels (Fig. 1A), consistent with prior observation showing minimal effects of IGF-I on IGF-IR degradation (14). Although both h10H5 and IGF-ligands bind IGF-IR with high affinity, only h10H5 possesses bivalent binding capacity. Down-regulation of IGF-IR likely involves antibody-mediated cross-linking, because F(ab) fragments do not elicit this effect as shown previously using anti-IGF-IR antibody 19D12 (19). h10H5-mediated IGF-IR down-regulation was specific, because insulin receptor levels were unaffected by h10H5 treatment (Fig. 1A).

Figure 1.

h10H5-induced IGF-IR down-regulation is mediated by both proteasomal and lysosomal pathways. A, SK-N-AS cells were treated with serially diluted h10H5, and the cell lysates were analyzed for IGF-IR and insulin receptor levels using anti-IGF-IR β-subunit and anti-insulin receptor β-chain antibodies following 1, 2, 4, and 8 h of continuous treatment. B, SK-N-AS cells were pretreated with combination of 5 μmol/L pepstatin A and 10 μg/mL leupeptin for 1 h and subsequently exposed to h10H5 for the indicated times in the continued presence of the inhibitors. Cell lysates were analyzed for IGF-IR α- and β-subunits by Western blotting. C, SK-N-AS cells were pretreated with 30 μmol/L of the proteasome inhibitor bortezomib and analyzed as in B. β-Actin was used as a loading control.

Figure 1.

h10H5-induced IGF-IR down-regulation is mediated by both proteasomal and lysosomal pathways. A, SK-N-AS cells were treated with serially diluted h10H5, and the cell lysates were analyzed for IGF-IR and insulin receptor levels using anti-IGF-IR β-subunit and anti-insulin receptor β-chain antibodies following 1, 2, 4, and 8 h of continuous treatment. B, SK-N-AS cells were pretreated with combination of 5 μmol/L pepstatin A and 10 μg/mL leupeptin for 1 h and subsequently exposed to h10H5 for the indicated times in the continued presence of the inhibitors. Cell lysates were analyzed for IGF-IR α- and β-subunits by Western blotting. C, SK-N-AS cells were pretreated with 30 μmol/L of the proteasome inhibitor bortezomib and analyzed as in B. β-Actin was used as a loading control.

Close modal

Previously, the lysosomal inhibitor methylamine but not the proteasome inhibitor MG115 was shown to reduce the down-regulation of the β-subunit of IGF-IR induced by the anti-IGF-IR antibody A12 (Imclone; ref. 14). However, IGF-IR degradation was still evident at 24 h after methylamine treatment, suggesting that additional pathways are likely employed in A12-mediated IGF-IR down-regulation. We therefore investigated whether similar mechanisms are involved in h10H5-induced IGF-IR down-regulation. Because the ECD and intracellular domain of IGF-IR are exposed to different cellular environments during the internalization and trafficking processes, we examined whether they are differently regulated. Antibodies specific to the ECD α-subunit and the β-subunit intracellular domain were used to follow their levels before and after h10H5 treatment using bortezomib to inhibit the proteasome (25) and leupeptin and pepstatin A to suppress lysosomal protease activities. The α- and β-subunits of IGF-IR exhibited similar kinetics of h10H5-induced down-regulation in SK-N-AS cells in the absence of these inhibitors (DMSO control; Fig. 1B and C). Treatment with both classes of inhibitors delayed h10H5-induced IGF-IR down-regulation (Fig. 1B and C), but bortezomib was more effective at preventing β-subunit down-regulation (Fig. 1C), whereas the lysosomal inhibitors preferentially inhibited α-subunit degradation (Fig. 1B). These results are consistent with the possibility that the ECD (α-subunit) is degraded in the lysosomes, whereas the intracellular domain is more readily accessible to the cytoplasmic proteasome, in accordance with their membrane topologies. The difference between our data and previously published results (14) could be due to that bortezomib is a more potent inhibitor of proteasome than MG115.

h10H5 Colocalizes with Lysosomal Markers following h10H5-Mediated Endocytosis

Anti-IGF-IR antibodies have been reported to induce efficient IGF-IR internalization (13, 18), but the detailed intracellular trafficking processes are not well characterized. We therefore did immunofluorescence microscopy to monitor the kinetics and localization of h10H5 internalization. IGF-IR is normally localized to the plasma membrane of MCF7 cells (data not shown) but rapidly internalizes within 5 min on h10H5 addition most likely via clathrin-coated vesicles as shown by colocalization with transferrin (Fig. 2A-C), which is known to internalize via this pathway. By 20 min, much less of the h10H5 signals remained colocalized with transferrin, indicating divergence from the transferrin recycling pathway (Fig. 2D-F). After 60 min, it was detectable within a subset of late endosomes and lysosomes as shown by colocalization with LAMP1 (Fig. 2H-I). The total h10H5 signal was weaker by the 4-h time point, indicating some degradation of the antibody, despite of the presence of lysosomal protease inhibitors, but was still detectable within the lysosomal lumen, surrounded by LAMP1 on the limiting membrane (Fig. 2J-L). Similar findings were observed in SK-N-AS cells (data not shown). The trafficking route of h10H5 confirms the above lysosomal inhibitor data by showing that h10H5 directs IGF-IR to lysosomes, where degradation of luminally exposed ECD occurs. Indeed, further immunofluorescence analysis revealed that the α-subunit is more stable than the β-subunit proteasomally degraded intracellular domain following h10H5 treatment in the presence of lysosomal inhibitors (data not shown).

Figure 2.

h10H5-induced IGF-IR internalization and trafficking. MCF7 cells were incubated with 5 μg/mL h10H5 in the presence of lysosomal protease inhibitors for 5 min (A-C), 20 min (D-F), 1 h (G-I), or 4 h (J-L) and then fixed, permeabilized, and stained with Cy3-conjugated anti-human antibody, thereby indicating the localization of the h10H5-IGF-IR complex in the red channel (left). Middle, localization of Alexa 488-transferrin (B and E) or LAMP1 staining (H and K) in the green channel; right, merged images from the respective red and green channels, with colocalization exhibiting a yellow/orange color. Arrows, examples of colocalization. Bar, 20 μm in main panels. Insets, boxed regions (magnification, ×3).

Figure 2.

h10H5-induced IGF-IR internalization and trafficking. MCF7 cells were incubated with 5 μg/mL h10H5 in the presence of lysosomal protease inhibitors for 5 min (A-C), 20 min (D-F), 1 h (G-I), or 4 h (J-L) and then fixed, permeabilized, and stained with Cy3-conjugated anti-human antibody, thereby indicating the localization of the h10H5-IGF-IR complex in the red channel (left). Middle, localization of Alexa 488-transferrin (B and E) or LAMP1 staining (H and K) in the green channel; right, merged images from the respective red and green channels, with colocalization exhibiting a yellow/orange color. Arrows, examples of colocalization. Bar, 20 μm in main panels. Insets, boxed regions (magnification, ×3).

Close modal

h10H5 Inhibits IGF-I-Dependent Proliferation of Human Cancer Cell Lines

The observed inhibitory effect of h10H5 on IGF-IR signaling prompted us to examine whether this antibody could also affect cell growth in vitro. Although MCF7 cells grew 4- to 5-fold in the presence of a control (B-cell-specific) antibody, h10H5 significantly inhibited their IGF-I-dependent growth during the 5-day continuous treatment (Fig. 3). This inhibition was dose dependent, with an IC50 between 34 and 57 ng/mL, and was similarly observed in human SK-N-AS neuroblastoma and SW527 breast cancer cells (data not shown).

Figure 3.

h10H5 inhibits tumor cell growth in vitro. MCF7 cells were serum starved for 5 h and then incubated in the presence of 1% serum and 1 ng/mL IGF-I, with serially diluted h10H5 (solid circles) or control anti-BR3 antibody (open circles) for 5 d. The CellTiter-Glo luminescent cell viability assay kit was used to assess cell viability. Dotted line, luminescence level of the MCF7 cells on h10H5 addition (day 0).

Figure 3.

h10H5 inhibits tumor cell growth in vitro. MCF7 cells were serum starved for 5 h and then incubated in the presence of 1% serum and 1 ng/mL IGF-I, with serially diluted h10H5 (solid circles) or control anti-BR3 antibody (open circles) for 5 d. The CellTiter-Glo luminescent cell viability assay kit was used to assess cell viability. Dotted line, luminescence level of the MCF7 cells on h10H5 addition (day 0).

Close modal

h10H5 Displays Antitumor Activity In vivo in Association with Decreased AKT Activation and Glucose Uptake

To assess the effect of h10H5 on the growth of tumor xenografts, nude mice bearing s.c. SK-N-AS tumors were given a single i.v. dose from 0.5 to 200 mg/kg (Fig. 4A). TGI was observed in all h10H5 dose groups compared with the vehicle control (Fig. 4A), with 51% TGI (P = 0.0003) as low as 0.5 mg/kg and 66% to 74% (P < 0.0001) at 2 to 200 mg/kg, suggesting that maximal efficacy in this model could be reached at relatively low doses of h10H5. Time to the first tumor doubling was also significantly longer (P = 0.03-0.0009) in all h10H5 groups with a range of 4.5 to 6.4 days compared with 2.7 days for the vehicle group. Thus, h10H5 shows strong single-agent activity in this model. Furthermore, a Fc-mutated (D256A) version of h10H5 that is no longer capable of binding to Fcγ receptors (data not shown) resulted in identical TGI (Supplementary Fig. S3),1 showing for the first time in vivo that antibody-dependent cell-mediated cytotoxicity is not part of the mechanism of action of h10H5.

Figure 4.

h10H5 inhibits the growth of SK-N-AS human neuroblastoma xenograft tumors at low doses. A, maximal efficacy is reached at a relatively low dose. h10H5 from 0.5 to 200 mg/kg was given as a single i.v. injection on day 0, and SK-N-AS tumor growth was followed for 14 d. Bars, SE (n = 9 per group). Slash marks, animals that were euthanized due to large tumor size, with all animals in the vehicle group being euthanized on day 9 due to excessive tumor burden (hence, %TGI for the h10H5 groups was calculated on this day). B, h10H5 treatment resulted in down-regulation of IGF-IR as well as inhibition of AKT activation in vivo. h10H5 was administrated i.p. at 5 or 20 mg/kg, and SK-N-AS xenograft tumors were collected at 6, 24, and 48 h after treatment. Tumors were homogenized and analyzed by Western blotting using antibodies against the IGF-IR β-subunit, AKT, phosphorylated AKT, and β-actin. C, SK-N-AS cells were incubated with serially diluted 10H5 in vitro for 48 h in either serum-free (open squares) or 0.1% serum-containing (solid squares) medium. [3H]FDG was added during the last 24 h of the incubation, and radioactivity incorporated (counts/min) was measured by scintillation counting. D, h10H5 treatment results in decreased tumor FDG uptake. Dynamic PET scans were done on tumors on day 0 before treatment and again on days 3, 7, and 10. Treatment responses on a given post-treatment day were assessed as the percentage change in the FDG uptake rate constant Ki (units/s) relative to the pretreatment value. Dotted and solid lines, vehicle and 10 mg/kg h10H5 groups, respectively. Bars, SE.

Figure 4.

h10H5 inhibits the growth of SK-N-AS human neuroblastoma xenograft tumors at low doses. A, maximal efficacy is reached at a relatively low dose. h10H5 from 0.5 to 200 mg/kg was given as a single i.v. injection on day 0, and SK-N-AS tumor growth was followed for 14 d. Bars, SE (n = 9 per group). Slash marks, animals that were euthanized due to large tumor size, with all animals in the vehicle group being euthanized on day 9 due to excessive tumor burden (hence, %TGI for the h10H5 groups was calculated on this day). B, h10H5 treatment resulted in down-regulation of IGF-IR as well as inhibition of AKT activation in vivo. h10H5 was administrated i.p. at 5 or 20 mg/kg, and SK-N-AS xenograft tumors were collected at 6, 24, and 48 h after treatment. Tumors were homogenized and analyzed by Western blotting using antibodies against the IGF-IR β-subunit, AKT, phosphorylated AKT, and β-actin. C, SK-N-AS cells were incubated with serially diluted 10H5 in vitro for 48 h in either serum-free (open squares) or 0.1% serum-containing (solid squares) medium. [3H]FDG was added during the last 24 h of the incubation, and radioactivity incorporated (counts/min) was measured by scintillation counting. D, h10H5 treatment results in decreased tumor FDG uptake. Dynamic PET scans were done on tumors on day 0 before treatment and again on days 3, 7, and 10. Treatment responses on a given post-treatment day were assessed as the percentage change in the FDG uptake rate constant Ki (units/s) relative to the pretreatment value. Dotted and solid lines, vehicle and 10 mg/kg h10H5 groups, respectively. Bars, SE.

Close modal

In a separate xenograft experiment, we examined whether h10H5 treatment in vivo affected tumor-associated IGF-IR signaling. SK-N-AS tumor samples treated at 5 or 20 mg/kg h10H5 were collected at 6, 24, and 48 h after dosing and analyzed by Western blotting. IGF-IR down-regulation was observed as early as 6 h and maintained at least up to 48 h (Fig. 4B). Furthermore, phosphorylated AKT levels decreased significantly, whereas total AKT levels remained unchanged in these tumors (Fig. 4B), in agreement with prior observations that the small-molecule inhibitor of IGF-IR kinase NVP-ADW742 and anti-IGF-IR antibody A12 can inhibit AKT activation in the xenograft tumors (16, 27). Thus, IGF-IR-mediated signaling in the SK-N-AS tumors was effectively inhibited by h10H5.

Although it is feasible to collect xenograft tumors to measure drug activity in preclinical studies, obtaining tumor biopsies from metastatic cancer patients poses a significant challenge. Noninvasive imaging techniques, such as measurement of glucose uptake in tumors, would be a highly desirable alternative for monitoring drug responses in clinical trials. We therefore first examined any possible effect of h10H5 on glucose uptake in SK-N-AS cells in vitro. Indeed, [5,6-3H]FDG uptake into SK-N-AS cells in the presence of h10H5 was inhibited with an IC50 of 3 μg/mL (Fig. 4C), independent of serum, encouraging us to assess the utility of FDG-PET in an additional SK-N-AS xenograft study. As expected, a single injection of h10H5 at 10 mg/kg resulted in significant inhibition of SK-N-AS tumor growth compared with vehicle treatment (data not shown). TGI on days 3, 7, 10, and 14 was 42.2% (P = 0.004), 63.2% (P < 0.0001), 72.6% (P < 0.0001), and 64.1% (P < 0.0001), respectively. FDG-PET imaging of the SK-N-AS tumors was done in parallel and revealed decreases of 37%, 28%, and 37% in the rate of FDG uptake on days 3, 7, and 14, respectively (Fig. 4D), relative to the baseline rate on day 0 (P < 0.05 for all time points of h10H5 treatment). These results show that FDG-PET changes accompany h10H5 treatment and correlate with the inhibition of SK-N-AS xenograft tumor growth. Therefore, FDG-PET, as an indicator of tumor cell density/function (28), is useful for monitoring the h10H5 antitumor activities in vivo, similar to its applicability to other cancer therapeutics (29). Although FDG-PET provides a viable alternative to routine imaging techniques, its utility is limited to FDG-avid tumors, which varies in frequency depending on tumor types.

h10H5 Induces Dynamic Changes of Tumor Transcriptional Profiles

In addition to the h10H5 effect on IGF-IR signaling and glucose uptake, we also investigated global regulation of gene expression induced by the antibody. Previously, differential gene expression, particularly those involved in cell cycle regulation and apoptosis, was noted by analyzing LuCap 35V human prostate xenograft tumors treated with the anti-IGF-IR antibody A12 (30). However, those tumors were harvested from a single time point after treatment. To compare and better characterize the major processes affected by blockade of IGF-IR signaling during h10H5 treatment, we followed the kinetic alterations of gene expression as early as 24 h as well as at a later time point of 2 weeks in SK-N-AS tumors receiving weekly 5 mg/kg h10H5 treatment. Interestingly, a total of 316 genes exhibited differential expression with a false discovery rate of <5% comparing vehicle and h10H5 treatment at 24 h (Supplementary Table S2),1 with 200 genes being down-regulated (classes I and II) and 116 being up-regulated (classes III and IV). DNA metabolism and cell cycle were identified as the top ranked functional categories (Supplementary Table S3),1 with highly significant ESAE scores (2.6E-19 to 3.8E-23). This gene expression signature emerged 24 h after treatment before obvious TGI and correlated with significant efficacy observed several days later (Fig. 4A). Interestingly, the majority of the genes that were affected after 24 h were no longer affected after 2 weeks: classes I and IV showed recovery to vehicle levels by this time point (Fig. 5A), However, this recovery was incomplete, because a much smaller set of genes remained down-regulated (class II) or up-regulated (class III) at 2 weeks (Fig. 5A). These data suggest that parallel and/or alternative signaling pathways are used for tumor adaptation to growth in the absence of IGF-IR signaling, consistent with the observed cytostatic but not cytotoxic effect of h10H5 on tumor growth. We further validated the expression patterns of selected genes using quantitative real-time PCR analysis (Fig. 5B). PCNA, E2F1, and CDC6 recapitulated the pattern of the class I genes expression, with statistically significantly reduced expression at 24 h and full recovery at 2 weeks. In contrast, the p55 γ-regulatory subunit of phosphoinositide 3-kinase (PIK3R3) and Aurora kinase A (AURKA) are examples of the class II genes that remain significantly down-regulated after 2 weeks. PIK3R3 is frequently up-regulated in ovarian, prostate, breast, and liver cancers (31), and its down-regulation of PIK3R3 has been associated with decreased IGF-II-dependent growth of glioblastoma-derived neurospheres (32) and increased apoptosis in ovarian cancer cells (31). Thus, these microarray data implicate intermediate molecular links between IGF-IR pathway inhibition and decreased tumor growth and highlight the dynamic and complex effects induced by a therapeutic antibody. Such a gene expression signature could also be useful for monitoring the transient and/or persistent responses to h10H5 therapy. However, independent verification with other tumor types would be necessary to further establish the significance and broader applicability of this gene signature.

Figure 5.

A composite transcription signature is induced by h10H5 treatment. A, expression profile of 336 Affymetrix probe sets that were differentially expressed between h10H5 and vehicle-treated tumors at 24 h with a false discovery rate of <5%. Four independent tumors, each represented by a column on the heat map, were analyzed from the vehicle, h10H5 24-h, and h10H5 2-week groups. Classes I and IV genes exhibited decreased and increased expression at 24 h, respectively, but recovered to comparable levels at 2 wk to those in the vehicle groups. In contrast, classes II and III showed consistent down-regulation or up-regulation at both time points, respectively. B, quantitative real-time PCR validation of selected differentially expressed genes. All the data were normalized to a housekeeping gene RPL19. Mean and SD were calculated for each group, with the mean mRNA level in the vehicle group being set to 1. Statistical analysis using pair-wise t tests were done by comparing the treatment and vehicle groups (*, P < 0.05).

Figure 5.

A composite transcription signature is induced by h10H5 treatment. A, expression profile of 336 Affymetrix probe sets that were differentially expressed between h10H5 and vehicle-treated tumors at 24 h with a false discovery rate of <5%. Four independent tumors, each represented by a column on the heat map, were analyzed from the vehicle, h10H5 24-h, and h10H5 2-week groups. Classes I and IV genes exhibited decreased and increased expression at 24 h, respectively, but recovered to comparable levels at 2 wk to those in the vehicle groups. In contrast, classes II and III showed consistent down-regulation or up-regulation at both time points, respectively. B, quantitative real-time PCR validation of selected differentially expressed genes. All the data were normalized to a housekeeping gene RPL19. Mean and SD were calculated for each group, with the mean mRNA level in the vehicle group being set to 1. Statistical analysis using pair-wise t tests were done by comparing the treatment and vehicle groups (*, P < 0.05).

Close modal

h10H5 Cooperates with Other Therapies

Although the IGF-IR pathway is frequently exploited by cancer cells for dysregulated growth, the microarray data (Fig. 5) indicate that other compensating or parallel pathways may allow tumor cells to evade IGF-IR inhibition. Combination of anti-IGF-IR antibodies with anti-EGFR antibodies, tamoxifen, radiation therapy, or some chemotherapeutic agents have shown improved efficacy over several forms of monotherapy (17, 18, 33). We therefore investigated whether h10H5 can be combined with docetaxel, a commonly used chemotherapeutic agent, in the SW527 human breast cancer xenograft model. h10H5 alone resulted in significant inhibition of SW527 tumor growth compared with vehicle treatment (P < 0.01; Fig. 6), with respective TGI values on days 11 and 14 of 52% and 36% and increased time to the first tumor doubling from 5.6 to 10.2 days (P < 0.002). Docetaxel alone also significantly inhibited tumor growth, with TGI of 54% on day 14 (P < 0.01, relative to the vehicle treatment), whereas docetaxel in combination with h10H5 further increase TGI to 82% (P < 0.01, relative to docetaxel treatment). Time to the first tumor doubling was significantly prolonged in the combination group (docetaxel + h10H5) compared with either h10H5 or docetaxel group (P < 0.01). The combination of h10H5 and docetaxel was therefore significantly more effective at inhibiting tumor growth than either monotherapy.

Figure 6.

h10H5 effectively cooperates with docetaxel and anti-VEGF antibody to inhibit the growth of SW527 breast cancer xenograft tumors. Solid arrows (for vehicle, h10H5, and B20-4.1) and open arrows (docetaxel), the days on which test materials were administered through i.p. and i.v. injections, respectively. Whereas h10H5 and B20.1 were given weekly doses at 5 and 10 mg/kg, respectively, with a loading dose of h10H5 at 10 mg/kg, docetaxel was administrated at 12.5 mg/kg on days 0, 4, and 8. Tumor volume changes were monitored for 14 d. Bars, SE. Slash marks, animals that were euthanized due to large tumor size, ulcerated tumor, or >20% weight loss. Inset, various treatment regimens.

Figure 6.

h10H5 effectively cooperates with docetaxel and anti-VEGF antibody to inhibit the growth of SW527 breast cancer xenograft tumors. Solid arrows (for vehicle, h10H5, and B20-4.1) and open arrows (docetaxel), the days on which test materials were administered through i.p. and i.v. injections, respectively. Whereas h10H5 and B20.1 were given weekly doses at 5 and 10 mg/kg, respectively, with a loading dose of h10H5 at 10 mg/kg, docetaxel was administrated at 12.5 mg/kg on days 0, 4, and 8. Tumor volume changes were monitored for 14 d. Bars, SE. Slash marks, animals that were euthanized due to large tumor size, ulcerated tumor, or >20% weight loss. Inset, various treatment regimens.

Close modal

We also determined whether h10H5 could also cooperate with another class of antitumor agents, the antiangiogenic therapy via VEGF blockade (34). The anti-VEGF antibody B20-4.1, which binds both human and murine VEGF with affinity similar to that of bevacizumab (Avastin; Genentech) to human VEGF (35), was therefore tested in the SW527 breast tumor model alone or in combination with h10H5 as above. At weekly doses of 10 mg/kg, B20-4.1 significantly inhibited tumor growth, with TGI of 60% on day 14 (P < 0.0001, relative to vehicle group). Combining B20-4.1 with h10H5 increased the level of TGI to 68% (P = 0.02, relative to B20-4.1 group) but did not result in a statistically significant increase in time to the first tumor doubling. These results show that although h10H5 or B20-4.1 delays SW527 tumor growth as single agents, the combination of h10H5 with B20-4.1 increases the levels of tumor inhibition. Inhibition of IGF-IR signaling has been shown to reduce VEGF production in tumor cells (36, 37) and to interfere with retinal neovascularization through its effect on endothelial cells (38). Our data provide further support that these two pathways likely also interact in the SW527 tumor model.

In summary, we show that h10H5 is a potent inhibitor of IGF-IR signaling. In an 8-week repeat-dose toxicity study, h10H5 was well tolerated on administration at doses from 5 to 50 mg/kg in cynomolgus monkeys, significantly above the minimum efficacious dose of 2 mg/kg established here. Our data provide support for investigation of this antibody as a single agent or in combination with other treatments in cancer patients. Furthermore, we show that FDG-PET could be a useful approach for monitoring the activities of anti-IGF-IR antibodies in FDG-avid tumors. Multiple anti-IGF-IR antibodies are being pursued in clinical trials. Although some of them are of human origin (14, 18, 19), others (13, 17), including h10H5, are humanized. In addition, they sometimes differ in antibody isotype, binding affinity/epitope, and antibody-dependent cell-mediated cytotoxicity activity (22). Whether or how these combinatorial attributes might contribute to the differences in drug efficacy, safety, and immunogenicity remains to be determined. Critical variables defining the best-in-class anti-IGF-IR antibody would shed light on developing more effective antibody-based cancer therapeutics in the future.

All authors are employees of Genentech.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Jo-Anne Hongo, Kurt Schroeder, and Farzam Farahi for assistance with hybridoma production and purification; Phil Hass, Mark Nagel, and the CMC subteam for producing and purifying h10H5; Yongmei Chen for generating various 10H5 expression vectors; Mikeal Paquet, Stacy Frankovitz, Carol O'Brien, and Heidi Savage for assistance with in vitro cell-based assays; Robert Soriano for microarray analysis; Karissa Peth for assistance with the FDG-PET study; and John Hanson for developing the PET Parser Program.

1
Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia.
Nat Rev Cancer
2004
;
4
:
505
–18.
2
Baserga R, Peruzzi F, Reiss K. The IGF-1 receptor in cancer biology.
Int J Cancer
2003
;
107
:
873
–7.
3
LeRoith D, Roberts CT, Jr. The insulin-like growth factor system and cancer.
Cancer Lett
2003
;
195
:
127
–37.
4
Adams TE, Epa VC, Garrett TP, Ward CW. Structure and function of the type 1 insulin-like growth factor receptor.
Cell Mol Life Sci
2000
;
57
:
1050
–93.
5
Feinberg AP. The epigenetics of cancer etiology.
Semin Cancer Biol
2004
;
14
:
427
–32.
6
Renehan AG, Zwahlen M, Minder C, O'Dwyer ST, Shalet SM, Egger M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: systematic review and meta-regression analysis.
Lancet
2004
;
363
:
1346
–53.
7
DiGiovanni J, Kiguchi K, Frijhoff A, et al. Deregulated expression of insulin-like growth factor 1 in prostate epithelium leads to neoplasia in transgenic mice.
Proc Natl Acad Sci U S A
2000
;
97
:
3455
–60.
8
Pravtcheva DD, Wise TL. Metastasizing mammary carcinomas in H19 enhancers-Igf2 transgenic mice.
J Exp Zool
1998
;
281
:
43
–57.
9
Sell C, Rubini M, Rubin R, Liu JP, Efstratiadis A, Baserga R. Simian virus 40 large tumor antigen is unable to transform mouse embryonic fibroblasts lacking type 1 insulin-like growth factor receptor.
Proc Natl Acad Sci U S A
1993
;
90
:
11217
–21.
10
Arteaga CL, Kitten LJ, Coronado EB, et al. Blockade of the type I somatomedin receptor inhibits growth of human breast cancer cells in athymic mice.
J Clin Invest
1989
;
84
:
1418
–23.
11
Li SL, Liang SJ, Guo N, Wu AM, Fujita-Yamaguchi Y. Single-chain antibodies against human insulin-like growth factor I receptor: expression, purification, and effect on tumor growth.
Cancer Immunol Immunother
2000
;
49
:
243
–52.
12
Hailey J, Maxwell E, Koukouras K, Bishop WR, Pachter JA, Wang Y. 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
–53.
13
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.
14
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.
15
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.
16
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.
17
Goetsch L, Gonzalez A, Leger O, et al. A recombinant humanized anti-insulin-like growth factor receptor type I antibody (h7C10) enhances the antitumor activity of vinorelbine and anti-epidermal growth factor receptor therapy against human cancer xenografts.
Int J Cancer
2005
;
113
:
316
–28.
18
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.
19
Wang Y, Hailey J, Williams D, et al. Inhibition of insulin-like growth factor-I receptor (IGF-IR) signaling and tumor cell growth by a fully human neutralizing anti-IGF-IR antibody.
Mol Cancer Ther
2005
;
4
:
1214
–21.
20
Haluska P, Carboni JM, Loegering DA, et al. In vitro and in vivo antitumor effects of the dual insulin-like growth factor-I/insulin receptor inhibitor, BMS-554417.
Cancer Res
2006
;
66
:
362
–71.
21
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.
22
Yee D. Targeting insulin-like growth factor pathways.
Br J Cancer
2006
;
94
:
465
–8.
23
Hongo JA, Mora-Worms M, Lucas C, Fendly BM. Development and characterization of murine monoclonal antibodies to the latency-associated peptide of transforming growth factor β1.
Hybridoma
1995
;
14
:
253
–60.
24
Carter P, Presta L, Gorman CM, et al. Humanization of an anti-p185HER2 antibody for human cancer therapy.
Proc Natl Acad Sci U S A
1992
;
89
:
4285
–9.
25
Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class of potent and effective antitumor agents.
Cancer Res
1999
;
59
:
2615
–22.
26
Green LA, Gambhir SS, Srinivasan A, et al. Noninvasive methods for quantitating blood time-activity curves from mouse PET images obtained with fluorine-18-fluorodeoxyglucose.
J Nucl Med
1998
;
39
:
729
–34.
27
Wu JD, Odman A, Higgins LM, et al. In vivo effects of the human type I insulin-like growth factor receptor antibody A12 on androgen-dependent and androgen-independent xenograft human prostate tumors.
Clin Cancer Res
2005
;
11
:
3065
–74.
28
Zhou SM, Wong TZ, Marks LB. Using FDG-PET activity as a surrogate for tumor cell density and its effect on equivalent uniform dose calculation.
Med Phys
2004
;
31
:
2577
–83.
29
Eschmann SM, Friedel G, Paulsen F, et al. 18F-FDG PET for assessment of therapy response and preoperative re-evaluation after neoadjuvant radio-chemotherapy in stage III non-small cell lung cancer.
Eur J Nucl Med Mol Imaging
2007
;
34
:
463
–71.
30
Wu JD, Haugk K, Coleman I, et al. Combined in vivo effect of A12, a type 1 insulin-like growth factor receptor antibody, and docetaxel against prostate cancer tumors.
Clin Cancer Res
2006
;
12
:
6153
–60.
31
Zhang L, Huang J, Yang N, et al. Integrative genomic analysis of phosphatidylinositol 3′-kinase family identifies PIK3R3 as a potential therapeutic target in epithelial ovarian cancer.
Clin Cancer Res
2007
;
13
:
5314
–21.
32
Soroceanu L, Kharbanda S, Chen R, et al. Identification of IGF2 signaling through phosphoinositide-3-kinase regulatory subunit 3 as a growth-promoting axis in glioblastoma.
Proc Natl Acad Sci U S A
2007
;
104
:
3466
–71.
33
Allen GW, Saba C, Armstrong EA, et al. Insulin-like growth factor-I receptor signaling blockade combined with radiation.
Cancer Res
2007
;
67
:
1155
–62.
34
Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer.
Nat Rev Drug Discov
2004
;
3
:
391
–400.
35
Liang WC, Wu X, Peale FV, et al. Cross-species vascular endothelial growth factor (VEGF)-blocking antibodies completely inhibit the growth of human tumor xenografts and measure the contribution of stromal VEGF.
J Biol Chem
2006
;
281
:
951
–61.
36
Reinmuth N, Liu W, Fan F, et al. Blockade of insulin-like growth factor I receptor function inhibits growth and angiogenesis of colon cancer.
Clin Cancer Res
2002
;
8
:
3259
–69.
37
Wu KD, Zhou L, Burtrum D, Ludwig DL, Moore MA. Antibody targeting of the insulin-like growth factor I receptor enhances the anti-tumor response of multiple myeloma to chemotherapy through inhibition of tumor proliferation and angiogenesis.
Cancer Immunol Immunother
2007
;
56
:
343
–57.
38
Kondo T, Vicent D, Suzuma K, et al. Knockout of insulin and IGF-1 receptors on vascular endothelial cells protects against retinal neovascularization.
J Clin Invest
2003
;
111
:
1835
–42.

Supplementary data