Purpose: Recent studies have highlighted a role of HER3 in HER2-driven cancers (e.g., breast cancer), implicating the upregulation of the receptor in resistance to HER-targeted therapies and Hsp90 inhibitors (e.g., AUY922). Therefore, we have developed an affibody-based PET radioconjugate that quantitatively assesses HER3 changes induced by Hsp90 inhibition in vivo.

Experimental Design: ZHER3:8698 affibody molecules were conjugated via the C-terminus cysteine to DFO-maleimide for 89Zr radiolabeling. The probe was characterized in vitro and in vivo in a panel of human breast cell lines and xenograft models with varying HER3 receptor levels. In addition, the radioconjugate was investigated as a tool to monitor the outcome of AUY922, an Hsp90 inhibitor, in an MCF-7 xenograft model.

Results: We demonstrated that 89Zr-DFO-ZHER3:8698 can track changes in receptor expression in HER3-positive xenograft models and monitor the outcome of AUY922 treatment. Our in vitro findings showed that MCF-7 cells, which are phenotypically different from BT474, develop resistance to treatment with AUY922 through HER3/IGF-1Rβ–mediated signaling. Of note, the lack of response in vitro due to HER3 recovery was confirmed in vivo using 89Zr-DFO-ZHER3:8698–based imaging. Upon AUY922 treatment, higher radioconjugate uptake was detected in treated MCF-7 xenografts, correlating with an AUY922-induced HER3 upregulation concomitant with an increase in IGF-1Rβ expression.

Conclusions: These data underline the potential of HER3-based PET imaging to noninvasively provide information about HER3 expression and to identify patients not responding to targeted therapies due to HER3 recovery. Clin Cancer Res; 24(8); 1853–65. ©2018 AACR.

This article is featured in Highlights of This Issue, p. 1771

Translational Relevance

Currently, inter- and intratumor heterogeneity is a clinical challenge, as histologic techniques can fail to provide a representative indication of molecular variation, due to dependence on the section of tumor that is chosen for sampling. This underscores the need to introduce novel imaging biomarkers that allow the examination of the whole tumor mass and may significantly help to better understand and treat cancer. Therefore, we have developed a novel PET radiotracer that provides information on heterogeneous HER3 expression and receptor expression changes due to activation of compensatory pathways through feedback induction of HER3, which is increasingly being recognized as a key player in therapeutic resistance. This would aid in the selection of patients for novel HER3-targeted therapies and potentially enable patients to be spared ineffective therapies, and, if necessary, switched to more effective therapeutic regimens sooner.

HER3, a member of the HER family of receptor tyrosine kinases, is a key driver of carcinogenesis (1, 2). Relative to other family members, HER3 has diminished kinase activity and its phosphorylation depends on dimerization with other receptors. The HER2–HER3 heterodimer is the most potent HER family dimer, but HER3 also dimerizes with EGFR (HER1) and cross-talks with other non-HER family members, such as c-Met and IGF-1Rβ. Recent studies have highlighted HER3 critical role in tumor progression and have defined its importance in the development of resistance to targeted therapies. In fact, EGFR- and HER2-driven cancers frequently become unresponsive to anti-HER–targeted therapeutics and inhibitors of the downstream PI3K/AKT/mTOR pathway through compensatory activation of HER3-mediated signaling which bypasses the original therapeutic blockade (3). The same may be true for Hsp90 inhibitors (e.g., AUY922), which are known to interfere with the posttranslational folding of several oncogenic client proteins (e.g., HER2, HER3, and AKT) and have shown moderate therapeutic efficacy in breast cancer (4–6). For example, a strong association between sensitivity to anti-HER2 inhibitors, including trastuzumab and lapatinib, and HER3 protein upregulation has been demonstrated using both breast cancer cell lines and core biopsies of HER2+ tumors from breast cancer patients (7, 8). In addition, combining an AKT inhibitor with either lapatinib or PF-04929113 (Hsp90 inhibitor) prevented the feedback-mediated induction of phosphorylated HER3, highlighting the fact that targeted therapies against HER3 are synergistic with multiple PI3K inhibitors (9).

Given the importance of HER3 signaling, several mAbs targeting the receptor ectodomain have recently been developed, with some already in phase I and II clinical trials (i.e., MM-121, patritumab, LJM716, and KTN3379; refs. 10, 11). Investigations conducted with LJM716 highlighted significant growth-inhibitory effects in HER2-positive cells and breast cancer xenograft models when combined with trastuzumab (12, 13). Moreover, KTN3379 combined with trastuzumab yielded greater and more durable tumor regression in both ligand-dependent and -independent breast cancer models with different levels of HER2 expression and sensitivity to HER2-targeted drugs (14). Despite promising results from clinical trials, the success of HER3-targeting therapies is likely to depend on the development of reliable HER3-specific biomarkers (15, 16). In this context, consideration has been given to natural ligands, which activate HER3 including NRG-1/HRG (17). However, even though NRG-1 is the most potent inducer of HER3 phosphorylation, it may be of limited utility as a biomarker given the fact that there are multiple routes of HER3 activation including mutations, ligand-independent dimerization and interactions with other HER and non-HER family ligands (2). Currently, the activation status of HER3 is analyzed by methods such as IHC, proteomics, and RNA-based technologies that rely on tissue obtained at biopsy or at surgical resection. These invasive procedures might not accurately address inter- and intratumor heterogeneity that may exist within the same tumor deposit or within different lesions in the same patient (18).

These considerations make a strong case for developing an imaging biomarker for monitoring receptor changes in response to therapies affecting HER3 expression that could help physicians tailor treatment regimens to patient-specific biology. Recently, anti-HER3 mAbs and an anti-HER3-F(ab')2 were radiolabeled with 64Cu and 89Zr for molecular imaging of HER3 expression (19–22). 89Zr-radiolabeled mAb (GSK2849330) is currently being evaluated in patients with advanced solid tumors (NCT02345174). However, the clinical use of full-length mAbs as imaging agents is not optimal, due to their long biological half-life and relatively slow tumor penetration, both of which affect tumor-to-background contrast at early time points (23). To circumvent these issues, we, and others, have recently developed low molecular weight (∼7 kDa) affibody-based positron emission tomography (PET) imaging agents that recognize HER3 on the cellular membrane (24, 25).

Here, we demonstrate using breast adenocarcinoma cell lines that HER3 expression changes induced by Hsp90 inhibition in vitro can also be quantitatively measured following AUY922 treatment in breast cancer xenograft models by 89Zr-DFO-ZHER3:8698 imaging. Texture analysis of PET images permitted a clear distinction between control and treated mice providing evidence for alternative and more robust image analysis than simple uptake metrics. In the future, clinical application of ZHER3:8698-based radioconjugates can be of use to guide the treatment decisions for breast cancer patients by providing specific information on therapy-induced HER3 receptor changes in a noninvasive manner.

Cell lines

Human breast adenocarcinoma (MCF-7, BT-474, MDA-MB-468, and MDA-MB-231) cell lines were obtained from the ATCC. The MCF-7-Tet-Off-HER2 cell line was kindly donated by Professor Joaquin Arribas and maintained as described previously (26). All cell lines were tested and authenticated by short tandem repeat (STR) DNA profiling analysis. The cells were cultured in DMEM (MCF-7, MDA-MB-231), RPMI1640 (BT-474, MDA-MB-468), or DMEM/Ham/F12 (MCF-7-Tet-Off-HER2) with high glucose (Gibco, Life Technologies) and supplemented with 10% FBS (Gibco, Life Technologies). In addition, DMEM/Ham/F12 contained 1 μg/mL of doxycycline (Sigma), 0.2 mg/mL of neomycin (Sigma), and 0.1 mg/mL of hygromycin (Life Technologies). All cell lines were maintained at 37°C in a humidified atmosphere supplied with 5% CO2.

Drug treatments

AUY922 and picropodophyllin (PPP) were purchased from LC Laboratories and Stratech Scientific, respectively. Both compounds were dissolved in 100% DMSO (Sigma-Aldrich) and diluted with culture medium to a desired concentration, with a final DMSO concentration <0.1% (v/v). Unless otherwise stated, the cell lines were exposed to 32 nmol/L of AUY922 and/or 10 μmol/L of PPP for 48 hours (27, 28).

Preparation of 89Zr-labeled DFO–affibody conjugates

The conjugation of deferoxamine-maleimide (DFO-mal, Macrocyclics) to the ZHER3:8698-Cys and ZTAQ-Cys affibody molecules (Affibody), 89Zr-radiolabeling procedures as well as the in vitro serum stability of 89Zr-DFO-ZHER3:8698 are described in detail in the Supplementary Information.

89Zr-DFO-ZHER3:8698in vitro binding affinity and specificity

The dissociation constant (Kd) and the specificity of binding of 89Zr-DFO-ZHER3:8698 were assessed as described previously (24). A detailed description of the procedure is given in the Supplementary Information.

Animal studies

The detailed methods are described in the Supplementary Information. Briefly, animal experiments were performed on 6- to 8-week-old female NCr athymic mice (Charles River) or NSG (NOD-Scid Gamma, The Jackson Laboratory) in compliance with license issued under the UK Animals (Scientific Procedures) Act 1986 and approved by the institutional ethical review according to the United Kingdom National Cancer Research Institute Guidelines for Animal Welfare in Cancer Research (29). Mice were inoculated subcutaneously on the shoulder with MCF-7, BT-474, MDA-MB-468, or MDA-MB-231 cells (6.5 × 106) resuspended in PBS and mixed with BD Matrigel Matrix (7:3 v/v, BD Matrigel Matrix, BD Biosciences).

PET imaging studies

PET/CT imaging studies were conducted using an Albira PET/SPECT/CT imaging system performed at 3 hours postinjection (Bruker). The detailed imaging and data analysis protocols are given in the Supplementary Information.

Ex vivo biodistribution

Mice were euthanized by cervical dislocation for biodistribution studies, 3 and 24 hours post-tracer injection. The major organs/tissues were dissected, weighed, and their associated radioactivity was measured in a gamma counter. The %ID/g was determined for each organ/tissue. Unless otherwise stated, the data were expressed as the mean of n = 3 mice ± SD.

During AUY922 treatment study, the overall tumor volumes in some mice were significantly reduced which limited the use of all xenografts for ex vivo biodistribution studies.

IHC

Formalin-fixed tumors (10%, v/v) were embedded in paraffin, sectioned into 3-μm-thick slices, and mounted on microscope slides. The detailed staining procedures with the various antibodies are described in the Supplementary Information.

Tumor ex vivo autoradiography

Tumors were dissected and immediately embedded in an optimal cutting temperature compound (Tissue-Tek), frozen on dry ice, and subsequently processed as described previously (30). Further experimental details are given in the Supplementary Information.

Immunoblotting analysis

Western blotting (WB) was carried out as described previously (31). A complete list of used antibodies and the detailed methods are provided in the Supplementary Information.

Immunoprecipitation

Tissue lysates (300 μg) and whole-cell lysates (200 μg) were incubated overnight in a thermomixer at 4°C and 650 rpm, with 10 μg of anti-HER3 mouse mAb (Merck Millipore). Following formation of the antibody–antigen complex, the immunocomplexes were purified using Dynabeads ProteinA (Life Technologies) according to the manufacturer's instructions. The immunoprecipitates were analyzed by Western blot analysis.

Proliferation and scratch-wound assays

MCF-7 and BT-474 cells were used for proliferation and scratch-wound assays with an Incucyte Zoom imaging system (Essen BioScience) according to the protocols provided by the manufacturer. Detailed descriptions of the procedures and image analysis are described in the Supplementary Information.

Statistical analyses

Unless otherwise stated, data were expressed as the mean ± SD. Statistical significance, sample size calculations, and correlation analysis are described in detail in the Supplementary Information.

Hsp90 inhibition promotes HER3/IGF-1Rβ signaling in breast cancer cells with low HER2 expression

To identify HER3-mediated drug resistance, we focused on the role of Hsp90 inhibition on HER3 signaling using two breast cancer cell lines with different HER2 levels, BT-474 (HER2+++, HER3+++) and MCF-7 cells (HER2+, HER3+++). Both cell lines were treated with AUY922 for up to 48 hours and subsequently, the expression levels of HER receptors and PI3K/AKT pathway activity were assessed by Western blot analysis. Increased Hsp70/72 expression was used as a marker of efficient Hsp90 inhibition (32). In BT-474 cells, Hsp90 inhibition induced a downregulation of both total and phosphorylated HER2 and HER3 (pHER2-Y1248 and pHER3-Y1289, respectively) as early as 8 hours after treatment, concomitantly with the loss of PI3K/AKT signaling, as shown by a decrease in phosphorylated (pAKT-S473) and total AKT (Fig. 1A). Furthermore, EGFR and phosphorylated EGFR (pEGFR-Y1068), IGF-1Rβ, and c-Met were downregulated 48 hours following AUY922 treatment (Supplementary Fig. S1). Densitometric analysis of two independent experiments revealed a decrease 48 hours after AUY922 treatment in the expression of both HER3 and IGF-1Rβ, of 0.38 ± 0.12 and 0.62 ± 0.01, respectively. Similarly, in MCF-7 cells, which have low HER2 expression, Hsp90 inhibition caused a downregulation of c-Met. The total and phosphorylated HER2 and EGFR levels were undetectable and only a transient downregulation of HER3/pHER3-Y1289 was found, followed by a HER3 recovery and activation 48 hours after treatment initiation. In addition, increased PI3K and pAKT-S473 levels at 48 hours posttreatment indicated HER3-mediated activation of this pathway (Fig. 1A). Furthermore, HER3 and IGF-1Rβ expression levels in response to Hsp90 inhibition were determined by densitometry to be 0.99 ± 0.19 and 1.20 ± 0.52, respectively. The prevalence of IGF-1Rβ levels suggested this receptor as the potential HER3 dimerization partner in MCF-7 cells following AUY922-mediated HER2 downregulation. To study this process, BT-474 and MCF-7 cells were treated with AUY922 alone or in combination with the IGF-1Rβ inhibitor PPP for 48 hours with the expectation of reversing the HER3-mediated resistance. As shown in Fig. 1B and Supplementary Fig. S2, downregulation of HER3, HER2, AKT, MAPK, IGF-1Rβ, and c-Met was observed when BT-474 cells were treated with AUY922 alone or in combination with PPP. IGF-1Rβ inhibition alone was not effective in abrogating PI3K/AKT signaling, confirming the greater dependency of BT-474 cells on HER2-HER3 dimerization. Conversely, in MCF-7 cells, total and phosphorylated HER3 were not downregulated to the same extent in response to Hsp90 inhibition, maintaining residual levels of phosphorylated AKT and MAPK. Importantly, in the presence or absence of HRG stimulation, HER3 levels were the same (Fig. 1B; Supplementary Fig. S3). However, inhibition of IGF-1Rβ reversed this phenotype suggesting a direct interaction between both receptors and the role of IGF-1Rβ in HER3-mediated signaling in response to Hsp90 blockade. Densitometric analysis of HER3 and IGF-1Rβ expression per treatment and per cell line is presented in Supplementary Table S1.

Figure 1.

AUY922 potentiates HER3-mediated downstream signaling and invasive phenotype in low-HER2–expressing cells. A, Time course of AUY922 treatment (32 nmol/L) in BT-474 and MCF-7 breast cancer cells up to 48 hours. B, BT-474, MCF-7, MCF-7-HER2 (-dox, induced HER2 expression), and MCF-7-HER2 (+dox, noninduced HER2 expression) cells were treated with AUY922 (32 nmol/L) and/or PPP (10 mmol/L) for 48 hours. A and B, HER receptors, IGF-1Rb, MAPK, and PI3K/AKT pathway activation were monitored by Western blot analysis using whole-cell lysates. Hsp70/72 expression was used as a surrogate for AUY922 treatment efficacy, and GAPDH as a loading control. C and D, BT-474 and MCF-7 cells proliferation assessment for 72 hours in response to AUY922 (32 nmol/L) and/or PPP (10 mmol/L). Confluence was monitored by phase contrast microscopy and expressed as the mean normalized confluence values ± SEM (n = 3 independent experiments) per time point. E and F, Invasive phenotype monitoring of BT-474 and MCF-7 cells by scratch-wound assay, in response to AUY922 (32 nmol/L) and/or PPP (10 mmol/L) treatment for 72 hours. Data are expressed as the mean relative wound density values ± SEM (n = 3 independent experiments) per time point.

Figure 1.

AUY922 potentiates HER3-mediated downstream signaling and invasive phenotype in low-HER2–expressing cells. A, Time course of AUY922 treatment (32 nmol/L) in BT-474 and MCF-7 breast cancer cells up to 48 hours. B, BT-474, MCF-7, MCF-7-HER2 (-dox, induced HER2 expression), and MCF-7-HER2 (+dox, noninduced HER2 expression) cells were treated with AUY922 (32 nmol/L) and/or PPP (10 mmol/L) for 48 hours. A and B, HER receptors, IGF-1Rb, MAPK, and PI3K/AKT pathway activation were monitored by Western blot analysis using whole-cell lysates. Hsp70/72 expression was used as a surrogate for AUY922 treatment efficacy, and GAPDH as a loading control. C and D, BT-474 and MCF-7 cells proliferation assessment for 72 hours in response to AUY922 (32 nmol/L) and/or PPP (10 mmol/L). Confluence was monitored by phase contrast microscopy and expressed as the mean normalized confluence values ± SEM (n = 3 independent experiments) per time point. E and F, Invasive phenotype monitoring of BT-474 and MCF-7 cells by scratch-wound assay, in response to AUY922 (32 nmol/L) and/or PPP (10 mmol/L) treatment for 72 hours. Data are expressed as the mean relative wound density values ± SEM (n = 3 independent experiments) per time point.

Close modal

To further verify the role of HER2 expression in inducing a HER3-mediated unresponsiveness to AUY922 treatment, MCF-7 cells overexpressing HER2 under the control of tetracycline (Tet-Off) were exposed to AUY922 and PPP for 48 hours. HER2 was induced by the removal of doxycycline from the culture medium (MCF-7-HER2, -dox; Fig. 1B). The effect of both inhibitors on MCF-7-HER2 (-dox, HER2+++) was similar to that observed in BT-474 cells. Moreover, MCF-7-HER2 (+dox, HER2+) responded similarly to the parental MCF-7 (HER2+) cell line (Fig. 1B; Supplementary Fig. S2), confirming that high HER2 expression is required for efficient HER3 downregulation in response to Hsp90 inhibition.

Given the role of HER3 in inducing PI3K/AKT signaling, the proliferative and invasive potential was assessed in response to AUY922 treatment and IGF-1Rβ inhibition. In BT-474 cells, proliferation was impaired in response to both treatment regimens, with a normalized confluence at 72 hours posttreatment estimated to be 24%–40% of that exhibited by the control cells (Fig. 1C; Supplementary Fig. S4A). On the other hand, AUY922 treatment alone did not affect the confluence of MCF-7 cells (Fig. 1D). The proliferative potential of these cells was counteracted by IGF-1Rβ inhibition, leading to normalized confluences of 22% (PPP alone) and 28% (AUY922 + PPP) at 72 hours posttreatment (Fig. 1D; Supplementary Fig. S4A). In addition, both Hsp90 and IGF-1Rβ inhibition showed an impact on wound invasion rates in BT-474 cells, with the relative wound densities (RWD) determined to be 19% (AUY922), 16% (PPP), and 15% (AUY922 + PPP), compared with 25% in control cells (Fig. 1E; Supplementary Fig. S4B). However, in MCF-7 cells, Hsp90 inhibition resulted in a faster wound healing rate compared with control cells (95% and 85% RWDs, respectively) at 48 hours. IGF-1Rβ blocking reduced the native and AUY922-induced invasive potential with RWDs of ∼30% in both PPP alone and AUY922 combined with PPP, at 72 hours posttreatment (Fig. 1F; Supplementary Fig. S4B). These results highlight a greater impact of IGF-1Rβ inhibition on reducing the invasiveness of both cell lines. Importantly, these data were in agreement with the increased levels of N-cadherin observed in MCF-7 and MCF-7-HER2 (+dox) in response to AUY922 (Supplementary Fig. S2), suggesting the level of invasiveness was maintained following Hsp90 inhibition.

89Zr-DFO-ZHER3:8698 uptake correlates with HER3 expression in breast adenocarcinoma cells

With the aim of monitoring changes in HER3 expression levels in vivo, HER3-specific ZHER3:8698 and nonspecific ZTAQ affibody molecules were radiolabeled with the long-lived positron emitter 89Zr (t1/2 = 78.4 hours) as it enabled investigation of the pharmacokinetics of the conjugate at later time points. Of note, the ZHER3:8698 molecules cross-react with HER3 of mouse origin making the pharmacokinetic studies more representative of the clinical setting. The DFO–affibody conjugates were characterized by high-performance liquid chromatography (HPLC) and mass spectrometry (Supplementary Fig. S5 and S6). The radiolabeling reaction of the conjugates with 89Zr resulted in a >95% radiometal incorporation, as determined by instant thin-layer chromatography (ITLC; Supplementary Fig. S7), and a specific activity of 2.5–2.7 MBq/μg (0.067–0.073 mCi/μg). The schematic structure of the 89Zr-DFO-ZHER3:8698 is shown in Fig. 2A. Stability studies showed that less than 3% demetalation occurred after incubating 89Zr-DFO-ZHER3:8698 in mouse serum at 37°C for 3 hours. The degree of demetalation increased to approximately 15% after 24-hour incubation (Supplementary Fig. S7 and S8). Receptor saturation binding analysis revealed the saturable binding of the radioconjugate to HER3 highly expressing MCF-7 cells, with a mean dissociation constant (Kd) of 0.55 ± 0.05 nmol/L and receptor density (Bmax) of 21,896 ± 764 sites/cell (Fig. 2B). The specificity of 89Zr-DFO-ZHER3:8698 binding was assessed in a panel of breast cancer cells, with MDA-MB-231 and BT-474 cells having the lowest and the highest HER3 expression levels, respectively (Fig. 2C and D). Preincubating the cells with either 100-fold molar excess of nonradiolabeled ZHER3:8698 or HRG (competing for the same binding site), significantly reduced 89Zr-DFO-ZHER3:8698 binding (Fig. 2D). Importantly, the measured cell-associated radioactivity was in agreement with the HER3 protein levels per cell line (Fig. 2C and D).

Figure 2.

In vitro specificity of 89Zr-DFO-ZHER3:8698 for HER3 detection in breast cancer cells. A, Schematic illustrating the radiolabeling reaction of DFO-conjugated ZHER3:8698 affibody molecule with the positron emitter 89Zr. B, Saturation binding of 89Zr-DFO-ZHER3:8698 to MCF-7 cells. The data are expressed as the mean values ± SEM (n = 3 independent experiments). C, HER3 expression in a panel of breast cancer cell lines. Representative Western blot from whole-cell lysates, with GAPDH used as the loading control. D,In vitro binding specificity of 89Zr-DFO-ZHER3:8698 in breast cancer cells and specific blocking using 100-fold molar excess of either unlabeled ZHER3:8698 or the natural HER3 ligand HRG. The data are expressed as the mean values ± SEM (n = 3 independent experiments). *, P = 0.0357; *, P = 0.0446 for MDA-MB468 cells. **, P = 0.0087; *, P = 0.015 for MCF-7 cells. **, P = 0.009; **, P = 0.0097 for BT-474 cells.

Figure 2.

In vitro specificity of 89Zr-DFO-ZHER3:8698 for HER3 detection in breast cancer cells. A, Schematic illustrating the radiolabeling reaction of DFO-conjugated ZHER3:8698 affibody molecule with the positron emitter 89Zr. B, Saturation binding of 89Zr-DFO-ZHER3:8698 to MCF-7 cells. The data are expressed as the mean values ± SEM (n = 3 independent experiments). C, HER3 expression in a panel of breast cancer cell lines. Representative Western blot from whole-cell lysates, with GAPDH used as the loading control. D,In vitro binding specificity of 89Zr-DFO-ZHER3:8698 in breast cancer cells and specific blocking using 100-fold molar excess of either unlabeled ZHER3:8698 or the natural HER3 ligand HRG. The data are expressed as the mean values ± SEM (n = 3 independent experiments). *, P = 0.0357; *, P = 0.0446 for MDA-MB468 cells. **, P = 0.0087; *, P = 0.015 for MCF-7 cells. **, P = 0.009; **, P = 0.0097 for BT-474 cells.

Close modal

89Zr-DFO-ZHER3:8698 accumulates in HER3-expressing breast cancer xenografts

The specific uptake of 89Zr-DFO-ZHER3:8698 (3 μg of protein; 7.2–8.1 MBq/mouse) was evaluated 3 hours postradioconjugate administration using mice bearing subcutaneous breast cancer xenografts with different HER3 expression levels. The obtained static PET/CT images allowed an accurate delineation of volumes of interest (VOI) in the case of both MCF-7 and MDA-MB-468 xenografts compared with MDA-MB-231 (Fig. 3A). In addition, MCF-7 xenografts imaged with 89Zr-DFO-ZTAQ, exhibited negligible tumor uptake, further confirming the in vivo specificity of 89Zr-DFO-ZHER3:8698 (Fig. 3A). The PET imaging data were corroborated by ex vivo biodistribution studies (Fig. 3B; Supplementary Fig. S9; Supplementary Table S2), with MCF-7 xenografts showing a tumor uptake of 2.67 ± 0.32 percent of the injected dose per gram of tissue (%ID/g ± SD), with tumor-to-blood and tumor-to-muscle ratios of 4.75 and 20.4, respectively. Conversely, lower tumor uptakes were measured in MDA-MB-468 (1.28 ± 0.13 %ID/g) and MDA-MB-231 (0.55 ± 0.25 %ID/g) xenograft models (Fig. 3B; Supplementary Table S2). As anticipated, low tumor uptake was also observed in MCF-7 xenografts injected with 89Zr-DFO-ZTAQ (0.17 ± 0.04 %ID/g; Fig. 3A and B). Among the nontarget-expressing organs, the kidneys exhibited the highest accumulation of 89Zr-DFO-ZHER3:8698 with the uptake ranging from approximately 80 to 165 %ID/g across different xenografts. This uptake occurs due to the glomerular filtration of the affibody molecules followed by radioconjugate reabsorption, degradation, and retention in proximal tubular cells (33). In addition, 89Zr-DFO-ZHER3:8698 was detected in other HER3-expressing normal organs such as lungs, liver, and intestines (25).

Figure 3.

PET/CT imaging and ex vivo analysis of 89Zr-DFO-ZHER3:8698 in mice bearing subcutaneous breast cancer xenografts. A, Representative 15-minute coronal fused PET/CT images of mice bearing MCF-7, MDA-MB-468, or MDA-MB-231 xenografts. The mice received approximately 8 MBq of either 89Zr-DFO-ZHER3:8698 or 89Zr-DFO-ZTAQ via tail vein injection, with image acquisition taking place 3 hours after injection. The arrows indicate the tumors and the kidneys. B,Ex vivo biodistribution at 3 hours after injection of the radioconjugates. Data are expressed as the mean values ± SD (n = 3 animals). *, P = 0.0136; ***, P = 0.0002; ****, P < 0.0001. C, Representative Western blot of whole tumor tissue lysates evaluating HER3, HER2, and EGFR expression in the indicated xenograft models. D, Histopathologic analysis of HER3 expression in MCF-7, MDA-MB-468, and MDA-MB-231 xenografts displaying the highest HER3 expression in MCF-7 xenografts and the lowest in MDA-MB-231. E, Representative ex vivo autoradiography sections taken 3 hours after injection of 89Zr-DFO-ZHER3:8698. F, Representative segmented xenografts following PET/CT image acquisition 3 hours after 89Zr-DFO-ZHER3:8698 injection. These images highlight the greater radioactivity accumulation observed in MCF-7 xenografts, and the heterogeneity of uptake across the tumor burden. Color map defined within the tumor volume only.

Figure 3.

PET/CT imaging and ex vivo analysis of 89Zr-DFO-ZHER3:8698 in mice bearing subcutaneous breast cancer xenografts. A, Representative 15-minute coronal fused PET/CT images of mice bearing MCF-7, MDA-MB-468, or MDA-MB-231 xenografts. The mice received approximately 8 MBq of either 89Zr-DFO-ZHER3:8698 or 89Zr-DFO-ZTAQ via tail vein injection, with image acquisition taking place 3 hours after injection. The arrows indicate the tumors and the kidneys. B,Ex vivo biodistribution at 3 hours after injection of the radioconjugates. Data are expressed as the mean values ± SD (n = 3 animals). *, P = 0.0136; ***, P = 0.0002; ****, P < 0.0001. C, Representative Western blot of whole tumor tissue lysates evaluating HER3, HER2, and EGFR expression in the indicated xenograft models. D, Histopathologic analysis of HER3 expression in MCF-7, MDA-MB-468, and MDA-MB-231 xenografts displaying the highest HER3 expression in MCF-7 xenografts and the lowest in MDA-MB-231. E, Representative ex vivo autoradiography sections taken 3 hours after injection of 89Zr-DFO-ZHER3:8698. F, Representative segmented xenografts following PET/CT image acquisition 3 hours after 89Zr-DFO-ZHER3:8698 injection. These images highlight the greater radioactivity accumulation observed in MCF-7 xenografts, and the heterogeneity of uptake across the tumor burden. Color map defined within the tumor volume only.

Close modal

The biodistribution data collected 24 hours postinjection of 89Zr-DFO-ZHER3:8698 showed a decrease in the MCF-7 tumor signal which may be due to the release of the radionuclide from the conjugate in vivo (the stability measured in the serum was approximately 84% at 24 hours). Consequently, as free zirconium-89 is known to accumulate in the mineral components of the bone, an increase in bone uptake was detected (34). The tumor-to-organ ratios were lower for the majority of the nontarget tissues in comparison with the results obtained at 3 hours (Supplementary Tables S3 and S4).

The tumor targeting by the radioactive agent observed in the various models correlated with the HER3 expression per xenograft assessed by Western blot analysis of HER receptors, and HER3 IHC staining (Fig. 3C and D). Furthermore, autoradiography of tumor tissue slices highlighted a difference in radioactivity accumulation in MCF-7 tumors when compared with MDA-MB-468 and MDA-MB-231 (Fig. 3E). Three-dimensional (3D) segmentation of the tumor volumes further highlighted heterogeneous uptake, particularly in MCF-7 xenografts (Fig. 3F; the corresponding 3D movie is displayed as Supplementary Movie S1).

89Zr-DFO-ZHER3:8698 imaging can identify HER3-mediated resistance to Hsp90 inhibition in breast cancer xenografts

In light of the effects of AUY922 treatment on HER3 expression observed in vitro (Fig. 1; Supplementary Fig. S1–S4), it was hypothesized that 89Zr-DFO-ZHER3:8698 could monitor HER3 recovery in response to Hsp90 inhibition in vivo. A pilot study was designed to test this hypothesis using MCF-7 and BT-474 xenografts. At the end of the AUY922 treatment, the image-derived radioactivity concentration (%ID/g normalized to the initial values obtained on day 0) for MCF-7 tumor bearing mice (Fig. 4A; the corresponding 3D movie is displayed as Supplementary Movie S2) showed a statistically significant difference between control and treated groups (P = 0.0131), with the treatment group displaying approximately 54% greater %ID/g ratio (1.23 ± 0.33) than the control group (0.80 ± 0.06; Fig. 4B).

Figure 4.

AUY922 treatment response monitoring in MCF-7 xenografts by 89Zr-DFO-ZHER3:8698 PET/CT imaging. MCF-7 xenografts were randomized into two groups: control and treatment. The treatment group received 40 mg/kg of AUY922 i.p. every second day for a period of 2 weeks. A, Representative axial fused PET/CT images of mice bearing MCF-7 tumors. Each mouse received 7.2–8.1 MBq of 89Zr-DFO-ZHER3:8698 via tail vein injection, with image acquisition taking place at 3 hours after injection. The mice were imaged before initiating AUY922 treatment (day 0), and following administration of the last treatment dose (day 14). The %ID/g ratios were determined by dividing the %ID/g on day 14 by that obtained on day 0. B, Scatter plot of the %ID/g ratios for both control (n = 6) and AUY922-treated mice (n = 7). The horizontal lines indicate the mean for each group. *, P = 0.0131. C, Scatter plot of the ex vivo tumor biodistribution at 3 hours after injection of the radioconjugate on day 14, for both control (n = 4) and AUY922-treated mice (n = 6). The horizontal lines indicate the mean per group. **P = 0.0036. D, Histopathologic analysis of control and AUY922-treated MCF-7 xenografts. Tumor sections were stained with hematoxylin and eosin (H&E), HER3, or CD31.

Figure 4.

AUY922 treatment response monitoring in MCF-7 xenografts by 89Zr-DFO-ZHER3:8698 PET/CT imaging. MCF-7 xenografts were randomized into two groups: control and treatment. The treatment group received 40 mg/kg of AUY922 i.p. every second day for a period of 2 weeks. A, Representative axial fused PET/CT images of mice bearing MCF-7 tumors. Each mouse received 7.2–8.1 MBq of 89Zr-DFO-ZHER3:8698 via tail vein injection, with image acquisition taking place at 3 hours after injection. The mice were imaged before initiating AUY922 treatment (day 0), and following administration of the last treatment dose (day 14). The %ID/g ratios were determined by dividing the %ID/g on day 14 by that obtained on day 0. B, Scatter plot of the %ID/g ratios for both control (n = 6) and AUY922-treated mice (n = 7). The horizontal lines indicate the mean for each group. *, P = 0.0131. C, Scatter plot of the ex vivo tumor biodistribution at 3 hours after injection of the radioconjugate on day 14, for both control (n = 4) and AUY922-treated mice (n = 6). The horizontal lines indicate the mean per group. **P = 0.0036. D, Histopathologic analysis of control and AUY922-treated MCF-7 xenografts. Tumor sections were stained with hematoxylin and eosin (H&E), HER3, or CD31.

Close modal

These results were in agreement with the ex vivo measurements, which showed a more prominent tumor targeting of the radioconjugate in the treatment group (1.56 ± 0.10 %ID/g) compared with the control animals (1.15 ± 0.14 %ID/g) with a P = 0.0036 (Fig. 4C; Supplementary Fig. S10). The more intense HER3 staining of tumor sections in the treated xenografts further confirmed that greater %ID/g ratios were due to HER3 restoration following Hsp90 inhibition (Fig. 4D). Moreover, CD31 staining showed no discernible difference in vessel density between control and treated mice, indicating the observed effects were unlikely to relate to AUY922-induced alterations of the vasculature (Fig. 4D; Supplementary Fig. S11), as previously reported in BT-474 xenografts (27). Furthermore, no significant differences were observed in terms of tumor volume between control and treated mice (Supplementary Fig. S12). Importantly, the same treatment using BT-474 xenografts resulted in a significantly lower (P = 0.0009) 89Zr-DFO-ZHER3:8698 uptake in the treated mice (Supplementary Fig. S13).

89Zr-DFO-ZHER3:8698 texture analysis identifies AUY922-treated xenografts and correlates with ex vivo treatment response

Recently, several studies have demonstrated that texture analysis in cancer imaging provides a high-throughput extraction of quantitative imaging biomarkers and assists in the design of descriptive and predictive models of tumor phenotypes (35, 36). We postulated that multiple texture features extracted from 89Zr-DFO-ZHER3:8698 images would provide a more comprehensive analysis of AUY922 treatment response, rather than a single, gross measure such as [%ID/g]max or [%ID/g]mean (Supplementary Fig. S9). In addition, a predictive classification model to distinguish between control and treated mice was built using Linear Discriminant Analysis (LDA), a method that aims to minimize intraclass and maximize interclass scatter, thus providing a linear combination of features that characterizes or separates the two classes, that is, control and treated. The study workflow is described in Fig. 5A. A total of 89 parameters were extracted, out of which, 63 were more evidently different between control and treated groups. The ratios of post-/pretreatment texture parameters are depicted in Fig. 5B and Supplementary Fig. S14A, presenting a distinction between control and treated xenografts. Moreover, the LDA models were assessed by examining their ability in successfully classifying the test mouse, as treated or control. For instance, assigning control mouse 1 (C1) as the test subject in the %ID/g-based LDA model, resulted in a borderline separation between control and treated mice (Fig. 5C). Interestingly, using the texture-based LDA model with the same test subject (C1) provided a more accurate and robust separation between the two classes (Fig. 5D). Of note, our analysis was unbiased given that all extracted texture parameters were included in the model design, circumventing the main issue related to false discovery rates in texture analysis reported by Chalkidou and colleagues (37).

Figure 5.

89Zr-DFO-ZHER3:8698 texture analysis identifies AUY922-treated MCF-7 xenografts. A, Radiomic workflow from PET imaging data acquisition to LDA model testing. B, Texture analysis heatmap comprising the ratios of post/pretreatment for extracted texture parameters per mouse (Control C1-6, and AUY922 A1-7). C, Representative leave-one-out cross-validation of the %ID/g LDA model, using control mouse C1 as the test subject. D, Representative leave-one-out cross-validation of the Texture LDA model, using control mouse C1 as the test subject. C and D, The dashed lines represent a crude separation point between the two groups determined as the mean of the maximum and the minimum relative ratios of the control and AUY922 treatment groups, respectively.

Figure 5.

89Zr-DFO-ZHER3:8698 texture analysis identifies AUY922-treated MCF-7 xenografts. A, Radiomic workflow from PET imaging data acquisition to LDA model testing. B, Texture analysis heatmap comprising the ratios of post/pretreatment for extracted texture parameters per mouse (Control C1-6, and AUY922 A1-7). C, Representative leave-one-out cross-validation of the %ID/g LDA model, using control mouse C1 as the test subject. D, Representative leave-one-out cross-validation of the Texture LDA model, using control mouse C1 as the test subject. C and D, The dashed lines represent a crude separation point between the two groups determined as the mean of the maximum and the minimum relative ratios of the control and AUY922 treatment groups, respectively.

Close modal

Densitometric analysis of Western blot data (Fig. 6A) denoted a significantly greater expression of Hsp70/72 (P < 0.0001) in the treated xenografts, clearly confirming efficient Hsp90 inhibition. As expected, HER2 expression was low in both groups. Significant upregulation of both HER3 (P = 0.031) and IGF-1Rβ (P = 0.002) were detected (Fig. 6A and B). Furthermore, no discernible differences in terms of phosphorylated or total AKT were observed between control and treated mice (Fig. 6A and B). HER3 expression variability assessed by Western blot analysis is relatively high, most likely due to the fact this technique yields semiquantitative data limited to a snapshot of the tumor microenvironment that is sampled (Supplementary Fig. S15). Thus, it does not represent the heterogeneous nature of the receptor expression in the entire tumor mass (Supplementary Fig. S16). Therefore, even though no obvious correlation was detected between the %ID/g ratios and HER3 expression as determined by Western blot analysis (Supplementary Fig. S15). Such findings suggest a role for molecular imaging probes (e.g., radiolabeled HER3-specific affibody molecules) as surrogate metrics to monitor the spatial distribution of heterogeneous receptor expression. Strong correlations were observed between HER3 and Hsp70/72 protein levels (Pearson r = 0.61; P = 0.0265; Fig. 6C), as well as between IGF-1Rβ and Hsp70/72 (Pearson r = 0.73; P = 0.0049; Fig. 6D), confirming the induction of HER3-IGF-1Rβ signaling in response to AUY922 treatment. In addition, the strong correlation between %ID/g ratios and Hsp70/72 protein levels (Pearson r = 0.81; P = 0.0008) confirmed the capacity to assess Hsp90 inhibition efficiency via 89Zr-DFO-ZHER3:8698 imaging (Fig. 6E). Various texture parameters were in agreement with HER3, IGF-1Rβ, and Hsp70/72 expression, with 41.6%, 58.4%, and 77.5% of the parameters having a correlation of ≥ 0.35, respectively (Supplementary Fig. S14B).

Figure 6.

AUY922 treatment promotes HER3-IGF-1Rβ–mediated signaling in MCF-7 xenografts. A, HER receptors, IGF-1Rβ, and PI3K/AKT pathway activation were monitored by Western blot analysis using whole-tissue lysates from all control and AUY922-treated mice. Hsp70/72 expression was used as a surrogate for AUY922 treatment efficacy, and GAPDH as a loading control. B, Minimum-to-maximum box-and-whisker plot of the quantified protein expression represented in A. Data are expressed as the normalized protein expression per antibody for control and AUY922 groups. The black lines represent the median value. *P = 0.0306; **P = 0.0022; ****P < 0.0001. C and D, Correlation between HER3/IGF-1Rβ and Hsp70/72 protein expression per mouse. The dashed lines represent the 95% confidence levels. E, Correlation between %ID/g ratios obtained from 89Zr-DFO-ZHER3:8698 PET images and Hsp70/72 protein expression per mouse. The dashed lines represent the 95% confidence levels. F, BT-474 and MCF-7 cells were treated with 32 nmol/L of AUY922 for 48 hours. Equal amounts of whole-cell lysates were immunoprecipitated with a mouse anti-HER3 antibody followed by Western blot analysis of HER3 and IGF-1Rβ. Whole-tissue lysates from control mouse C5 and AUY922-treated mouse A6 were also immunoprecipitated against HER3 and analyzed by Western blot analysis. Ten percent of the input lysates was used as a loading control.

Figure 6.

AUY922 treatment promotes HER3-IGF-1Rβ–mediated signaling in MCF-7 xenografts. A, HER receptors, IGF-1Rβ, and PI3K/AKT pathway activation were monitored by Western blot analysis using whole-tissue lysates from all control and AUY922-treated mice. Hsp70/72 expression was used as a surrogate for AUY922 treatment efficacy, and GAPDH as a loading control. B, Minimum-to-maximum box-and-whisker plot of the quantified protein expression represented in A. Data are expressed as the normalized protein expression per antibody for control and AUY922 groups. The black lines represent the median value. *P = 0.0306; **P = 0.0022; ****P < 0.0001. C and D, Correlation between HER3/IGF-1Rβ and Hsp70/72 protein expression per mouse. The dashed lines represent the 95% confidence levels. E, Correlation between %ID/g ratios obtained from 89Zr-DFO-ZHER3:8698 PET images and Hsp70/72 protein expression per mouse. The dashed lines represent the 95% confidence levels. F, BT-474 and MCF-7 cells were treated with 32 nmol/L of AUY922 for 48 hours. Equal amounts of whole-cell lysates were immunoprecipitated with a mouse anti-HER3 antibody followed by Western blot analysis of HER3 and IGF-1Rβ. Whole-tissue lysates from control mouse C5 and AUY922-treated mouse A6 were also immunoprecipitated against HER3 and analyzed by Western blot analysis. Ten percent of the input lysates was used as a loading control.

Close modal

Conversely to MCF-7, in BT-474 xenografts AUY922 treatment led to a decrease in HER2, HER3, and IGF-1Rβ (Supplementary Fig. S13C).

HER3 immunoprecipitation studies with MCF-7 and BT-474 cells revealed a direct interaction between IGF-1Rβ and HER3 following AUY922 treatment in MCF-7 but not in BT-474 cells (Fig. 6F; Supplementary Fig. S17 and S18), highlighting a potential role for HER3-IGF-1Rβ complexes in the therapeutic efficacy of AUY922. The same interaction was observed following HER3 immunoprecipitation using MCF-7 control and AUY922-treated tumor tissues (Fig. 6F), thus supporting the formation of HER3-IGF-1Rβ dimers in response to Hsp90 inhibition in xenograft models with low HER2 background (Fig. 6F; Supplementary Fig. S17 and S18).

Aberrant regulation of HER receptor tyrosine kinases (RTK) is common in breast cancer and, even though the introduction of molecularly targeted drugs has reduced relapses among breast cancer patients (38, 39), acquired or de novo resistance is still observed in advanced and adjuvant disease settings (40). Several mechanisms have been reported and linked to anti-HER-mediated drug resistance including the upregulation of HER3 receptors via a negative feedback loop of the downstream PI3K/AKT/mTOR pathway (41). Early identification of patients likely to resist therapy could be facilitated by the development of imaging biomarkers capable of stratifying patients based on their individual HER3 expression changes in response to anti-HER inhibitors and could have enormous implications on clinical practice.

Currently, 18F-FDG is the most widely used radiopharmaceutical for PET, which has also shown utility to monitor response to different treatment regimens (42). However, 18F-FDG accumulates in tissues with increased glucose consumption regardless of the specific molecular signature of the tumor (43). Therefore, it has limited use when selecting patients for targeted drugs where prior knowledge of a given receptor status is essential for treatment initiation. Given such limitations, HER3-specific mAbs, affibody molecules, nanobody constructs and peptides have been radiolabeled to visualize different levels of HER3 in vivo (21, 44–46). The studies presented herein further strengthen a contribution of PET-based molecular imaging to noninvasively assess increased abundance of HER3 in response to therapeutic intervention. To test our hypothesis, a novel affibody-based PET radioconjugate (89Zr-DFO-ZHER3:8698), which specifically binds to the extracellular domain of HER3, was developed and investigated in breast cancer xenografts. From a clinical standpoint (e.g., radionuclide characteristics and availability) labeling with fluorine-18 would be more suitable for patient administration due to a lower radiation exposure burden and radioactive half-life (t1/2 = 108 minutes) compatibility with the pharmacokinetics of the affibody molecules. Therefore, in the meantime, we have also developed a F-18-labeled analogue of the 89Zr-DFO-ZHER3:8698 that is a promising avenue for future investigations (24).

We tested the Hsp90 inhibitor, AUY922 known to induce proteosomal degradation of HER2:HER3 heterodimers (4–6, 47, 48) in vitro using HER3-expressing cells (BT-474 and MCF-7). Interestingly, MCF-7 cells developed HER3-mediated resistance to AUY922 and the unresponsiveness to the inhibitor was correlated with HER2 baseline expression, given the greater therapeutic efficacy in BT-474 cells. Furthermore, in MCF-7 cells, 48 hours post-AUY922 treatment, the drug triggered an alternative survival pathway promoting HER3/IGF-1Rβ heterodimerization and the lack of response to AUY922 was reversed by IGF-1Rβ suppression. Notably, the prominent activation of HER3/IGF-1Rβ complexes enhanced the invasive potential of MCF-7 cells without affecting cellular proliferation.

These in vitro findings demonstrating that residual HER3 levels may attenuate the antiproliferative effects of AUY922 in cells with low HER2 expression led us to investigate whether 89Zr-DFO-ZHER3:8698 imaging could monitor the lack of response to Hsp90 inhibition in vivo. As expected, accumulation of the conjugate in vivo correlated with HER3 receptor levels present in different xenograft models, which was in line with recently published data (25). Furthermore, several groups, including ours, have previously shown that HER2-specific imaging agents labeled with a wide range of radionuclides can measure the effects of administrating Hsp90 inhibitors on HER2 downregulation (31, 49, 50).

Herein, we report an important observation that following AUY922 treatment, a clear distinction in 89Zr-DFO-ZHER3:8698 accumulation between control and AUY922-treated mice is due to HER3 down- and upregulation in BT-474 and MCF-7 xenografts, respectively.

These results were corroborated by texture analysis, whereby the applied algorithm resulted in a better separability between these two groups than the conventionally used PET quantification %ID/g-based treatment response values (corresponding to the standardized uptake value (SUV) in clinical studies). These data highlight the potential of using basic texture features extracted from PET images to establish links between spatial variability in a given tumor's architecture and mapping them with tumor-specific pathway activation profiles. The authors recognize that additional investigations are required to better understand the association between these different texture features and tumor and/or gene/protein expression, but such an approach clearly has the potential to assist in future clinical decision-making.

In addition, the higher radioconjugate uptake detected in the treated xenografts correlated with Hsp90-induced upregulation of HER3 expression. The evident increase in IGF-1Rβ levels in response to AUY922 treatment, together with immunoprecipitation studies, further confirmed a role for HER3-IGF-1Rβ heterodimerization in acquired resistance to Hsp90 inhibition. These effects observed in MCF-7 xenografts highlight the potential AUY922 resistance mechanism that could be associated to low HER2 and high HER3 expression.

Interestingly, Whitesell and colleagues have recently reported that MCF-7 cells treated with ganetespib (Hsp90 inhibitor) proliferate to a greater extent and shRNA knockdown of Hsp90 leads to upregulation of IGF-1Rβ. Furthermore, MCF-7 xenografts treated with this inhibitor showed no statistically significant difference compared to control mice in terms of tumor growth and event-free survival (51). Together, these data are in agreement with the lack of effect of Hsp90 inhibition in MCF-7 cells and xenografts reported herein, and with a prominent role for IGF-1Rβ in HER3 signaling.

These results are also in agreement with gene expression analysis of the TCGA Provisional dataset with the largest number of breast invasive carcinoma samples (n = 1,105), whereby a statistically significant cooccurrence of the expression of HER3 and IGF-1R genes was found (P = 0.020; Supplementary Table S5; 52). Moreover, alterations in these genes correlated with shorter overall and disease-free survival rates (Supplementary Tables S6 and S7). In contrast, using the same dataset, no statistically significant cooccurrence of HER3 and IGF-1R genes was found in samples with upregulated HER2 expression (Supplementary Table S8), further supporting a dependency on HER3-IGF-1Rβ–mediated signaling in the presence of low HER2 expression. Further work will be carried out in multiple breast cancer xenograft models, to study the potential of HER3 and IGF-1Rβ therapeutics in overcoming the discussed resistance mechanism in response to Hsp90 inhibition and anti-HER2–targeted therapies.

In conclusion, consistent with the findings reported herein, several recent Hsp90 inhibitor–based clinical trials have shown that the use of such agents might not be an effective therapeutic strategy in breast cancer (7–9, 44, 45). Therefore, studies comprising combinatorial therapeutic approaches with agents targeting enriched resistance-conferring receptors (e.g., HER3, IGF-1Rβ) may be required for a more effective clinical use of Hsp90 inhibitors. We believe that HER3 PET imaging may prove to be an important measure in quantifying changes in HER3 expression resulting from acquired resistance to Hsp90 inhibition and thus serve as a valuable tool in facilitating treatment personalization.

No potential conflicts of interest were disclosed.

Conception and design: C.D. Martins, K.J. Harrington, G. Kramer-Marek

Development of methodology: C.D. Martins, C. Da Pieve, R. Smith, G. Smith, G. Kramer-Marek

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C.D. Martins, T.A. Burley, D.M. Ciobota, L. Allott

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C.D. Martins, R. Smith, K.J. Harrington, G. Kramer-Marek

Writing, review, and/or revision of the manuscript: C.D. Martins, C. Da Pieve, T.A. Burley, R. Smith, K.J. Harrington, W.J.G. Oyen, G. Smith, G. Kramer-Marek

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.D. Martins

Study supervision: G. Kramer-Marek

This research was supported in part by the Cancer Research UK-Cancer Imaging Centre (C1060/A16464) and NIHR Clinical Research Facility at the Royal Marsden and the ICR. This report is an independent research funded by the National Institute for Health Research. The views expressed in this publication are those of the authors and not necessarily those of the NHS, the National Institute for Health Research, or the Department of Health. The authors thank AffibodyAB for supplying the affibody molecules. The authors acknowledge Professor Joaquin Arribas for donating the MCF-7-Tet-Off-HER2 cell line. The authors especially thank Dr. Terry Spinks for continuous help with PET/SPECT/CT imaging system. The authors thank Frances Daley and the Pathology Core Facility for IHC assistance. The authors appreciate the constructive comments from Prof. Martin Leach. In addition, the authors thank the EPSRC UK National Mass Spectrometry Facility at Swansea University for providing technical services.

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.

1.
Baselga
J
,
Swain
SM
. 
Novel anticancer targets: revisiting ERBB2 and discovering ERBB3
.
Nat Rev Cancer
2009
;
9
:
463
75
.
2.
Gala
K
,
Chandarlapaty
S
. 
Molecular pathways: HER3 targeted therapy
.
Clin Cancer Res
2014
;
20
:
1410
6
.
3.
Amin
DN
,
Campbell
MR
,
Moasser
MM
. 
The role of HER3, the unpretentious member of the HER family, in cancer biology and cancer therapeutics
.
Semin Cell Develop Biol
2010
;
21
:
944
50
.
4.
Kamal
A
,
Thao
L
,
Sensintaffar
J
,
Zhang
L
,
Boehm
MF
,
Fritz
LC
, et al
A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors
.
Nature
2003
;
425
:
407
10
.
5.
Wainberg
ZA
,
Anghel
A
,
Rogers
AM
,
Desai
AJ
,
Kalous
O
,
Conklin
D
, et al
Inhibition of HSP90 with AUY922 induces synergy in HER2-amplified trastuzumab-resistant breast and gastric cancer
.
Mol Cancer Ther
2013
;
12
:
509
19
.
6.
Safavi
S
,
Jarnum
S
,
Vannas
C
,
Udhane
S
,
Jonasson
E
,
Tomic
TT
, et al
HSP90 inhibition blocks ERBB3 and RET phosphorylation in myxoid/round cell liposarcoma and causes massive cell death in vitro and in vivo
.
Oncotarget
2016
;
7
:
433
45
.
7.
Garrett
JT
,
Olivares
MG
,
Rinehart
C
,
Granja-Ingram
ND
,
Sanchez
V
,
Chakrabarty
A
, et al
Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase
.
Proc Natl Acad Sci USA
2011
;
108
:
5021
6
.
8.
Narayan
M
,
Wilken
JA
,
Harris
LN
,
Baron
AT
,
Kimbler
KD
,
Maihle
NJ
. 
Trastuzumab-induced HER reprogramming in "resistant" breast carcinoma cells
.
Cancer Res
2009
;
69
:
2191
4
.
9.
Chandarlapaty
S
,
Sawai
A
,
Scaltriti
M
,
Rodrik-Outmezguine
V
,
Grbovic-Huezo
O
,
Serra
V
, et al
AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity
.
Cancer Cell
2011
;
19
:
58
71
.
10.
Aurisicchio
L
,
Marra
E
,
Roscilli
G
,
Mancini
R
,
Ciliberto
G
. 
The promise of anti-ErbB3 monoclonals as new cancer therapeutics
.
Oncotarget
2012
;
3
:
744
58
.
11.
Garner
AP
,
Bialucha
CU
,
Sprague
ER
,
Garrett
JT
,
Sheng
Q
,
Li
S
, et al
An antibody that locks HER3 in the inactive conformation inhibits tumor growth driven by HER2 or neuregulin
.
Cancer Res
2013
;
73
:
6024
35
.
12.
Garrett
JT
,
Sutton
CR
,
Kurupi
R
,
Bialucha
CU
,
Ettenberg
SA
,
Collins
SD
, et al
Combination of antibody that inhibits ligand-independent HER3 dimerization and a p110alpha inhibitor potently blocks PI3K signaling and growth of HER2+ breast cancers
.
Cancer Res
2013
;
73
:
6013
23
.
13.
Garner
A
,
Sheng
Q
,
Bialucha
U
,
Chen
DS
,
Chen
Y
,
Das
R
, et al
LJM716: an anti-HER3 antibody that inhibits both HER2 and NRG driven tumor growth by trapping HER3 in the inactive conformation [abstract]
.
In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31–Apr 4; Chicago, IL. Philadelphia (PA): AACR; 2012
.
Abstract nr 2733
.
14.
Xiao
Z
,
Carrasco
RA
,
Schifferli
K
,
Kinneer
K
,
Tammali
R
,
Chen
H
, et al
A Potent HER3 monoclonal antibody that blocks both ligand-dependent and -independent activities: differential impacts of PTEN status on tumor response
.
Mol Cancer Ther
2016
;
15
:
689
701
.
15.
Dey
N
,
Williams
C
,
Leyland-Jones
B
,
De
P
. 
A critical role for HER3 in HER2-amplified and non-amplified breast cancers: function of a kinase-dead RTK
.
Am J Translat Res
2015
;
7
:
733
50
.
16.
Kol
A
,
Terwisscha van Scheltinga
AG
,
Timmer-Bosscha
H
,
Lamberts
LE
,
Bensch
F
,
de Vries
EG
, et al
HER3, serious partner in crime: therapeutic approaches and potential biomarkers for effect of HER3-targeting
.
Pharmacol Ther
2014
;
143
:
1
11
.
17.
Schoeberl
B
,
Faber
AC
,
Li
D
,
Liang
MC
,
Crosby
K
,
Onsum
M
, et al
An ErbB3 antibody, MM-121, is active in cancers with ligand-dependent activation
.
Cancer Res
2010
;
70
:
2485
94
.
18.
Kurozumi
S
,
Padilla
M
,
Kurosumi
M
,
Matsumoto
H
,
Inoue
K
,
Horiguchi
J
, et al
HER2 intratumoral heterogeneity analyses by concurrent HER2 gene and protein assessment for the prognosis of HER2 negative invasive breast cancer patients
.
Breast Cancer Res Treat
2016
;
158
:
99
111
.
19.
Lockhart
AC
,
Liu
Y
,
Dehdashti
F
,
Laforest
R
,
Picus
J
,
Frye
J
, et al
Phase 1 Evaluation of [(64)Cu]DOTA-patritumab to assess dosimetry, apparent receptor occupancy, and safety in subjects with advanced solid tumors
.
Mol Imaging Biol
2016
;
18
:
446
53
.
20.
Wehrenberg-Klee
E
,
Turker
NS
,
Heidari
P
,
Larimer
B
,
Juric
D
,
Baselga
J
, et al
Differential receptor tyrosine kinase PET imaging for therapeutic guidance
.
J Nucl Med
2016
;
57
:
1413
9
.
21.
Warnders
FJ
,
Terwisscha van Scheltinga
AG
,
Knuehl
C
,
van Roy
M
,
de Vries
EF
,
Kosterink
JG
, et al
HER3 specific biodistribution and tumor uptake of 89Zr-MSB0010853 visualized by real-time and non-invasive PET imaging
.
J Nucl Med
2017
;
58
:
1210
15
.
22.
Bensch
F
,
Lamberts
LE
,
Srneenk
MM
,
Jorritsma-Smit
A
,
Lub-de Hooge
MN
,
Van Scheltinga
AGTT
. 
89Zr-lumretuzumab PET imaging before and during HER3 antibody lumretuzumab treatment in patients with solid tumors
.
Cancer Res
2017
;
23
:
6128
37
.
23.
van Dongen
GA
,
Visser
GW
,
Lub-de Hooge
MN
,
de Vries
EG
,
Perk
LR
. 
Immuno-PET: a navigator in monoclonal antibody development and applications
.
Oncologist
2007
;
12
:
1379
89
.
24.
Da Pieve
C
,
Allott
L
,
Martins
CD
,
Vardon
A
,
Ciobota
DM
,
Kramer-Marek
G
, et al
Efficient [18F]AlF radiolabeling of ZHER3:8698 affibody molecule for imaging of HER3 positive tumors
.
Bioconjug Chem
2016
;
27
:
1839
49
.
25.
Rosestedt
M
,
Andersson
KG
,
Mitran
B
,
Tolmachev
V
,
Lofblom
J
,
Orlova
A
, et al
Affibody-mediated PET imaging of HER3 expression in malignant tumours
.
Sci Rep
2015
;
5
:
15226
.
26.
Pedersen
K
,
Angelini
PD
,
Laos
S
,
Bach-Faig
A
,
Cunningham
MP
,
Ferrer-Ramon
C
, et al
A naturally occurring HER2 carboxy-terminal fragment promotes mammary tumor growth and metastasis
.
Mol Cell Biol
2009
;
29
:
3319
31
.
27.
Eccles
SA
,
Massey
A
,
Raynaud
FI
,
Sharp
SY
,
Box
G
,
Valenti
M
, et al
NVP-AUY922: a novel heat shock protein 90 inhibitor active against xenograft tumor growth, angiogenesis, and metastasis
.
Cancer Res
2008
;
68
:
2850
60
.
28.
Girnita
A
,
Girnita
L
,
del Prete
F
,
Bartolazzi
A
,
Larsson
O
,
Axelson
M
. 
Cyclolignans as inhibitors of the insulin-like growth factor-1 receptor and malignant cell growth
.
Cancer Res
2004
;
64
:
236
42
.
29.
Workman
P
,
Aboagye
EO
,
Balkwill
F
,
Balmain
A
,
Bruder
G
,
Chaplin
DJ
, et al
Guidelines for the welfare and use of animals in cancer research
.
Br J Cancer
2010
;
102
:
1555
77
.
30.
Kramer-Marek
G
,
Bernardo
M
,
Kiesewetter
DO
,
Bagci
U
,
Kuban
M
,
Aras
O
, et al
PET of HER2-positive pulmonary metastases with 18F-ZHER2:342 affibody in a murine model of breast cancer: comparison with 18F-FDG
.
J Nucl Med
2012
;
53
:
939
46
.
31.
Kramer-Marek
G
,
Kiesewetter
DO
,
Capala
J
. 
Changes in HER2 expression in breast cancer xenografts after therapy can be quantified using PET and (18)F-labeled affibody molecules
.
J Nucl Med
2009
;
50
:
1131
9
.
32.
Garcia-Carbonero
R
,
Carnero
A
,
Paz-Ares
L
. 
Inhibition of HSP90 molecular chaperones: moving into the clinic
.
Lancet Oncol
2013
;
14
:
e358
69
.
33.
Altai
M
,
Varasteh
Z
,
Andersson
K
,
Eek
A
,
Boerman
O
,
Orlova
A
. 
In vivo and in vitro studies on renal uptake of radiolabeled affibody molecules for imaging of HER2 expression in tumors
.
Cancer Biother Radiopharm
2013
;
28
:
187
95
.
34.
Deri
MA
,
Zeglis
BM
,
Francesconi
LC
,
Lewis
JS
. 
PET imaging with (8)(9)Zr: from radiochemistry to the clinic
.
Nucl Med Biol
2013
;
40
:
3
14
.
35.
Kumar
V
,
Gu
Y
,
Basu
S
,
Berglund
A
,
Eschrich
SA
,
Schabath
MB
, et al
Radiomics: the process and the challenges
.
Magn Reson Imaging
2012
;
30
:
1234
48
.
36.
Parmar
C
,
Grossmann
P
,
Bussink
J
,
Lambin
P
,
Aerts
HJ
. 
Machine learning methods for quantitative radiomic biomarkers
.
Sci Rep
2015
;
5
:
13087
.
37.
Chalkidou
A
,
O'Doherty
MJ
,
Marsden
PK
. 
False Discovery rates in PET and CT studies with texture features: a systematic review
.
PLoS One
2015
;
10
:
e0124165
.
38.
Slamon
DJ
,
Leyland-Jones
B
,
Shak
S
,
Fuchs
H
,
Paton
V
,
Bajamonde
A
, et al
Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2
.
New Engl J Med
2001
;
344
:
783
92
.
39.
Piccart-Gebhart
MJ
,
Procter
M
,
Leyland-Jones
B
,
Goldhirsch
A
,
Untch
M
,
Smith
I
, et al
Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer
.
New Engl J Med
2005
;
353
:
1659
72
.
40.
Vogel
CL
,
Cobleigh
MA
,
Tripathy
D
,
Gutheil
JC
,
Harris
LN
,
Fehrenbacher
L
, et al
Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer
.
J Clin Oncol
2002
;
20
:
719
26
.
41.
Sergina
NV
,
Rausch
M
,
Wang
DH
,
Blair
J
,
Hann
B
,
Shokat
KM
, et al
Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3
.
Nature
2007
;
445
:
437
41
.
42.
Avril
S
,
Muzic
RF
 Jr.
,
Plecha
D
,
Traughber
BJ
,
Vinayak
S
,
Avril
N
. 
(1)(8)F-FDG PET/CT for monitoring of treatment response in breast cancer
.
J Nucl Med
2016
;
57
:
34S
9S
.
43.
Kramer-Marek
G
,
Oyen
WJ
. 
Targeting the human epidermal growth factor receptors with immuno-PET: imaging biomarkers from bench to bedside
.
J Nucl Med
2016
;
57
:
996
1001
.
44.
Terwisscha van Scheltinga
AG
,
Lub-de Hooge
MN
,
Abiraj
K
,
Schroder
CP
,
Pot
L
,
Bossenmaier
B
, et al
ImmunoPET and biodistribution with human epidermal growth factor receptor 3 targeting antibody (8)(9)Zr-RG7116
.
mAbs
2014
;
6
:
1051
8
.
45.
Orlova
A
,
Malm
M
,
Rosestedt
M
,
Varasteh
Z
,
Andersson
K
,
Selvaraju
RK
, et al
Imaging of HER3-expressing xenografts in mice using a Tc-99m(CO)(3)-HEHEHE-Z(HER3:08699) affibody molecule
.
Eur J Nucl Med Mol I
2014
;
41
:
1450
9
.
46.
Larimer
BM
,
Phelan
N
,
Wehrenberg-Klee
E
,
Mahmood
U
. 
Phage display selection, in vitro characterization, and correlative PET imaging of a novel HER3 Peptide
.
Mol Imag Biol.
2017 Jul 21
. [Epub ahead of print].
47.
Kong
A
,
Rea
D
,
Ahmed
S
,
Beck
JT
,
Lopez Lopez
R
,
Biganzoli
L
, et al
Phase 1B/2 study of the HSP90 inhibitor AUY922 plus trastuzumab in metastatic HER2-positive breast cancer patients who have progressed on trastuzumab-based regimen
.
Oncotarget
2016
;
7
:
37680
92
.
48.
Scaltriti
M
,
Dawood
S
,
Cortes
J
. 
Molecular pathways: targeting hsp90–who benefits and who does not
.
Clin Cancer Res
2012
;
18
:
4508
13
.
49.
Holland
JP
,
Caldas-Lopes
E
,
Divilov
V
,
Longo
VA
,
Taldone
T
,
Zatorska
D
, et al
Measuring the pharmacodynamic effects of a novel Hsp90 inhibitor on HER2/neu expression in mice using Zr-89-DFO-Trastuzumab
.
PLoS One
2010
;
5
:
e8859
.
50.
Oude Munnink
TH
,
de Vries
EG
,
Vedelaar
SR
,
Timmer-Bosscha
H
,
Schroder
CP
,
Brouwers
AH
, et al
Lapatinib and 17AAG reduce 89Zr-trastuzumab-F(ab')2 uptake in SKBR3 tumor xenografts
.
Mol Pharm
2012
;
9
:
2995
3002
.
51.
Whitesell
L
,
Santagata
S
,
Mendillo
ML
,
Lin
NU
,
Proia
DA
,
Lindquist
S
. 
HSP90 empowers evolution of resistance to hormonal therapy in human breast cancer models
.
Proc Natl Acad Sci U S A
2014
;
111
:
18297
302
.
52.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.

Supplementary data