<<

Review Molecular Therapeutics PET Imaging of Receptor Tyrosine Kinases in Cancer Weijun Wei1,2,DalongNi2, Emily B. Ehlerding3, Quan-Yong Luo1,andWeiboCai2,3,4

Abstract

Overexpression and/or mutations of the receptor tyrosine and visualize RTKs with noninvasive diagnostic tools will kinase (RTK) subfamilies, such as epidermal greatly refine clinical treatment strategies for cancer patients, receptors (EGFR) and vascular endothelial growth factor facilitate precise response prediction, and improve drug devel- receptors (VEGFR), are closely associated with tumor cell opment. Positron emission tomography (PET) agents using growth, differentiation, proliferation, apoptosis, and cellular targeted radioactively labeled antibodies have been developed invasiveness. Monoclonal antibodies (mAb) and tyrosine to visualize tumor RTKs and are changing clinical decisions for kinase inhibitors (TKI) specifically inhibiting these RTKs have certain cancer types. In the present review, we primarily focus shown remarkable success in improving patient survival in on PET imaging of RTKs using radiolabeled antibodies with an many cancer types. However, poor response and even drug emphasis on the clinical applications of these immunoPET resistance inevitably occur. In this setting, the ability to detect probes. Mol Cancer Ther; 17(8); 1625–36. 2018 AACR.

Introduction epidermal (EGFR) belongs to another family of RTKs and includes three other members (erbB2/HER- Due to their complex heterogeneity and tendency to spread 2, erbB3/HER-3, and erbB4/HER-4). We previously showed throughout the body, treating is very challenging. Over that HER-kinase–targeted imaging agents would enable maxi- the past quarter century, progress in cancer biology has led to mum benefit(i.e.,patientstratification, therapeutic response the discovery and identification of receptor tyrosine kinases monitoring, and new drug development) in cancer patient (RTK), which control many fundamental cell behaviors and management (4). c-Met, another RTK for hepatocyte growth drive tumor initiation, maintenance, and progression (1). factor (HGF), has been found overexpressed or aberrantly These discoveries have fueled the development of effective activated in a variety of cancers (5). targeted therapeutics such as monoclonal antibodies (mAb) Although mAbs targeting the above-mentioned RTKs have and inhibitors (TKI), shifting cancer patients' become a standard of care for patients with certain mutation- care from traditional empirical treatments to an era of person- positive tumors, the efficacy of the currently approved mAbs as alized treatment. single agents is very limited; therefore, synergistic regimens Human RTKs contain 20 subfamilies and only approximately containing conventional chemotherapeutic agents and the halfoftheRTKfamiliesarewellunderstood(2).RTKsare mAbs are applied (6). In addition, drug resistance almost anchored in the cytoplasmic membrane, and gain-of-function invariably occurs in cancer patients treated with these thera- mutations and/or overexpression of these RTKs are closely peutic antibodies. Traditionally, biopsy and immunohis- related to growth and proliferation in malignant tissues (2). tochemistry are performed to determine the RTK status of Of them, vascular endothelial growth factor A and VEGFRs have cancer tissues and to guide subsequent treatment. However, implicated roles in tumor angiogenesis, and visualization of spatial expression levels of RTKs can vary over time and among VEGFR expression in vivo with a radiopharmaceutical may be lesions (1), indicating the urgent need for novel noninvasive a viable clinical option for imaging angiogenesis (3). The approaches to visualize RTKs' plasticity throughout the whole body. Moreover, noninvasive methods that can select patients who will potentially benefit from combinational therapy and 1Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth predict treatment response will greatly optimize management People's Hospital, Shanghai, China. 2Department of Radiology, University of strategies for cancer patients. Wisconsin–Madison, Wisconsin. 3Department of Medical Physics, University of In recent years, molecular imaging has rapidly developed and 4 Wisconsin–Madison, Wisconsin. University of Wisconsin Carbone Cancer Cen- has been widely used both in clinical setting and in preclinical ter, Madison, Wisconsin. arenas (7–9). Single-photon emission computed tomography W. Wei and D. Ni contributed equally to this article. (SPECT) and positron emission tomography (PET) are radionu- Corresponding Authors: Weibo Cai, University of Wisconsin–Madison, Room 7137, clide molecular imaging techniques that enable noninvasive 1111 Highland Avenue, Madison, WI 53705. Phone: 608-262-1749; Fax: 608-265- evaluation of biochemical changes and expression of molecular 0614; E-mail: [email protected]; and Quan-Yong Luo, Department of Nuclear targets within living subjects. Although a SPECT probe, 111In- fi Medicine, Shanghai Jiao Tong University Af liated Sixth People's Hospital, 600# labeled , has been used in clinical trials to detect Yishan Road, Shanghai 200233, China. E-mail: [email protected] HER2-positive metastatic breast cancer (10), the sensitivity of doi: 10.1158/1535-7163.MCT-18-0087 SPECT is several orders of magnitude lower than that of PET (11). 2018 American Association for Cancer Research. In comparison, PET imaging is of high sensitivity and can be used

www.aacrjournals.org 1625

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

to investigate physiological and molecular mechanisms of human ward strategy for generating RTK-specific PET radiotracers has diseases (11). PET imaging of RTKs with radiolabeled mAbs, mostly, but not entirely, involved the radiolabeling of therapeutic denoted as immunoPET, may provide a noninvasive method for mAbs. Antibody fragments, nanobodies, and smaller molecule assessing the dynamics of RTKs, selecting patients for personal- inhibitors have also been investigated for in vivo visualization of ized treatment, predicting response to RTK inhibition therapy, RTKs. With shorter biological half-lives, these probes are well and facilitating drug development (7). suited for fast imaging protocols when labeled with rapidly 68 18 Most PET imaging clinical trials have been focused on using decaying radioisotopes such as Ga (t1/2, 68 minutes) and F radiolabeled FDA-approved antibodies (12), such as trastuzumab (t1/2, 110 minutes). Our review focuses mainly on the current for breast cancer and esophagogastric adenocarcinoma (EGA; research in the field of antibody-based immunoPET probes and refs. 13–17), for colorectal and lung cancers (18– highlights potential clinical implications of immunoPET in visu- 20), and for several indications (21–26). PET alizing RTKs and in guiding clinical decisions (Fig. 1). 64 imaging using the positron emitters Cu (t1/2, 12.7 hours) and 89 Zr (t1/2, 78.4 hours) for antibody labeling has been extensively studied over the last decade (8, 27). 89Zr has a longer half-life, PET Imaging of the HGF/c-Met Pathway which generally matches the serum half-life of most mAbs and Dysregulation of HGF/c-Met signaling is implicated in a num- therefore is suitable for antibody imaging (28, 29). A straightfor- ber of malignancies (30), and synergistic effects of EGF and HGF

Figure 1. RTKs are deregulated in most human cancers and control many fundamental cell behaviors by regulating biochemical signals. Representative antibody-based PET probes targeting corresponding RTKs (or pathways) are shown. MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol-3-kinase; JAK, Janus kinase; PKC, protein kinase C; PLCg, phospholipase C-g; STAT, signal transducer and activator of transcription.

1626 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of RTKs

were noted in non–small cell lung cancer (NSCLC; ref. 31). probe showed high affinity for human c-Met in 15 subjects with Moreover, c-Met amplification can drive HER3-dependent PI3K high risk of colorectal cancer (49). In addition to optical imaging signaling, thereby mediating NSCLC resistance to EGFR inhibitors in assessing c-Met (49, 50), 18F-AH113804 is a peptide-based (32, 33). Currently, c-Met targeted therapy involves small- c-Met–specific PET imaging probe that has been used to visualize molecule inhibitors (, , etc.; refs. 31, 34), locoregional recurrence of breast cancer in a clinical trial (51). and antibodies (, ficlatuzumab, rilotuzumab, These results suggest that HGF/c-Met–specific PET imaging may , etc.; refs. 35, 36). Phase II trials in patients with correctly identify patients most likely to benefit from c-Met– advanced NSCLC demonstrated that the addition of onartuzu- targeted therapies or from EGFR inhibition therapy, as it has been mab to the EGFR inhibitor resulted in an improvement reported that c-Met is implicated in acquired resistance to EGFR in progression-free and overall survival for c-Met–positive inhibitors in NSCLC (31, 33). patients (37); however, a phase III study failed to observe the additive therapeutic effect of onartuzumab to erlotinib (38). PET Imaging of the VEGF/VEGFR Pathway Despite these disappointing results, suppressing the HGF/c-Met pathway may still have an antitumor effect in other primary The VEGF/VEGFR signaling pathway plays an important role in tumors and/or overcome the resistance of molecularly targeted the regulation of angiogenesis. VEGFs bind to three associated therapies (30, 39). Noninvasive PET visualization of c-Met transmembrane RTKs known as VEGFR-1, VEGFR-2, and VEGFR- dynamics could therefore potentially facilitate patient stratifica- 3. Among them, VEGFR-2 is a key receptor involved in the devel- tion and guide c-Met–directed therapies. opment of blood vasculature and is an attractive target for anti- Initial attempts to develop nuclear medicine imaging probes angiogenic tumor therapy (52). Approved antiangiogenic drugs for detecting c-Met expression used murine mAbs (40, 41). Based such as bevacizumab, , , , and pazo- on these studies, taking the step to evaluate immunoPET probes panib target this pathway and are associated with modest survival using humanized or fully human anti-c-Met mAbs is relatively advantages in certain kinds of cancers (53). Bevacizumab and straightforward. Using the humanized therapeutic mAb onartu- are IgG1 antibodies directed against vascular endo- zumab, which inhibits HGF binding and therefore the HGF/MET thelial growth factor A (VEGF-A) and VEGFR2, respectively. To date, signaling pathway (35), Jagoda and colleagues initially synthe- clinical PET studies using 89Zr-Df-bevacizumab were performed in sized 89Zr-DFO-onartuzumab and 76Br-onartuzumab and found patients with breast cancer, neuroendocrine tumors, renal cell that the former probe exhibited higher uptake and tumor-to- carcinoma (RCC), NSCLC, and glioma (21, 22, 26, 54, 55). muscle ratios in MKN-45 gastric carcinoma models (42). Li and Based on fundamental preclinical studies (3, 9, 56, 57), in a colleagues developed c-Met–targeting bivalent cys-diabodies clinical feasibility study, Gaykema and colleagues demonstrated and radiolabeled one of the candidates with 89Zr. The authors that 89Zr-Df-bevacizumab could be used to detect primary breast found that uptake of 89Zr-labeled H2 cys-diabody was higher in cancer (22). Another pilot clinical study in patients with meta- the gefitinib-resistant NSCLC model than in the low c-MET– static RCC demonstrated that 89Zr-Df-bevacizumab PET visual- expressing NSCLC model, indicating that immunoPET could be ized renal tumor lesions with striking heterogeneity between used to assess c-MET expression levels for both therapeutic and lesions, therefore reflecting differences in vascular characteristics diagnostic purposes (43). Pool and colleagues then reported that (24). Everolimus, an inhibitor of mammalian target of rapamycin the readily translatable probe 89Zr-Df-onartuzumab could effec- (mTOR), also inhibits VEGF-A expression, but there are not tively discriminate erlotinib-induced c-Met upregulation in reliable biomarkers to predict efficacy of everolimus in patients NSCLC models (44). This study herein highlighted the potential with metastatic RCC at the moment. van Es and colleagues value of 89Zr-Df-onartuzumab PET in assessing c-Met upregula- investigated the value of 89Zr-Df-bevacizumab as a tool to identify tion-mediated erlotinib resistance in patients with NSCLC. More everolimus efficacy in 13 patients with metastatic RCC, and importantly, c-Met–directed treatment using NVP-AUY-922, a reported that everolimus decreased 89Zr-Df-bevacizumab tumor heat shock protein 90 (HSP90) inhibitor (45), could reduce uptake in 10 patients who received continuous everolimus treat- 89Zr-Df-onartuzumab uptake in HCC827 models (Fig. 2A). Rilo- ment and achieved stable disease at 3 months (Fig. 2D; ref. 26). tumumab (AMG102) is an HGF-binding antibody and can bind Jansen and colleagues reported that 89Zr-Df-bevacizumab had to and neutralize HGF, thus preventing its binding to c-Met. poor uptake in mice bearing diffuse intrinsic pontine glioma However, a phase III trial failed to observe the therapeutic effects (DIPG; ref. 23), implying that treatment with bevacizumab in of in patients with c-Met–positive gastric or gastro- DIPG patients is justified only after 89Zr-Df-bevacizumab immu- esophageal adenocarcinoma (46). Previously, there were no tools noPET demonstrates positive VEGF expression in tumor tissue. In to noninvasively determine the levels of HGF present in the local spite of these disappointing preclinical data, the same team tumor microenvironment. To this end, 89Zr-DFO-AMG102 was further reported that, in children with DIPG, five of seven primary developed by Price and colleagues, and this probe could selec- tumors showed focal 89Zr-Df-bevacizumab uptake while no sig- tively accumulate in tumors with high levels of HGF protein (Fig. nificant uptake was seen in the healthy brain (58). A further 2B and C; ref. 47). study by Veldhuijzen van Zanten and colleagues reported that Our team produced a recombinant human HGF and labeled 89Zr-Df-bevacizumab uptake was correlated with microvascular the agent with 64Cu. PET imaging revealed specific and prominent proliferation in a patient with DIPG and highlighted that 89Zr-Df- uptake of the tracer in c-Met–positive U87MG tumors but signif- bevacizumab PET could be used to delineate intralesional het- icantly lower uptake in c-Met–negative MDA-MB-231 tumors erogeneity (Fig. 2E; ref. 59). These results indicate that 89Zr-Df- (48). One concern for these tracers based on the HGF ligand is bevacizumab immunoPET is feasible in children with DIPG and their potential to stimulate tumor growth by activating c-Met may help select patients with the greatest chance of benefiting (48). Burggraaf and colleagues initially developed a fluorescently from bevacizumab treatment. Notably, 89Zr-Df-bevacizumab labeled peptide (GE-137) for optical imaging of c-Met, and this could also offer a tool to refine clinical management of patients

www.aacrjournals.org Mol Cancer Ther; 17(8) August 2018 1627

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

Figure 2. Representative c-Met and VEGF pathway–specific PET probes. A,89Zr-Df-onartuzumab is a c-Met targeting PET probe. While in vehicle-treated HCC827 xenografts uptake of 89Zr-Df-onartuzumab did not differ before treatment and after treatment (left), the corresponding uptake of the radiotracer in the NVP-AUY- 922–treated mice decreased more than 30% after treatment (right), indicating that 89Zr-Df-onartuzumab PET may provide a powerful tool for visualizing c-Met dynamics. B, 89Zr-DFO-AMG102 is another HGF/c-Met signaling pathway–specific PET probe. PET maximum intensity projection (MIP) images in HGF and MET double–positive U87MG tumor models at 24 and 120 hours after injection of 89Zr-DFO-AMG102. C, Corresponding MIP images in MKN45 tumor models that had negative HGF and positive MET in the local tumor microenvironment. These results imply that 89Zr-DFO-AMG102 may act as a companion diagnostic tool for selection of patients more likely to respond to any HGF-targeted therapy. D, 89Zr-Df-bevacizumab PET MIP image before everolimus treatment showed multiple local and distant metastases from a left kidney tumor. 89Zr-Df-bevacizumab PET demonstrated that tumor uptake decreased significantly after 6 weeks of treatment using everolimus. E, In vivo performance of 89Zr-Df-bevacizumab PET in the diagnosis and localization of DIPG. Gadolinium-enhanced T1 MR images (top) showed the enhanced primary pontine glioma and metastatic gliomas (white arrows) in the right ventricular trigone and cervicomedullary junction, and all these glioma lesions were clearly visualized by 89Zr-Df-bevacizumab PET (bottom). Panels reproduced with permission from ref. 26, 47, 59, SNMMI and ref. 44, Springer.

with von Hippel–Lindau disease (25), and neuroendocrine is a mAb Fab derivative of bevacizumab and has a tumors (55). From a clinical perspective, a patient's baseline higher affinity for all soluble and matrix-bound human VEGF-A plasma VEGF-A level is an independent prognostic factor for isoforms than bevacizumab (62). Nagengast and colleagues ini- certain cancer types (60), and resistance to antiangiogenic ther- tially created 89Zr-Df-ranibizumab and reported that VEGF PET apies ultimately leads to patients' relapse and poor survival (53, imaging using 89Zr-Df-ranibizumab allowed serial analysis of 61). Therefore, it would be significant to develop personalized angiogenic changes in ovarian tumor models following treatment therapeutic strategies through immunoPET imaging of angiogen- with the kinase inhibitor sunitinib (63). In addition, several esis biomarkers that might help select proper patients and reflect studies have been performed to image tumor vasculature by the response of tumor cells to antiangiogenic drugs. visualizing VEGFR-2. Meyer and colleagues engineered and

1628 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of RTKs

radiolabeled single-chain VEGF and further validated the feasi- help select proper patients for subsequent therapy and elucidate bility of imaging VEGFR-1 and VEGFR-2 in an orthotopic murine the underlying resistance mechanisms (4, 67–69). tumor model (64). We synthesized and characterized a ramucir- Trastuzumab was the first clinically approved anti-HER2 anti- umab-based PET imaging agent, 64Cu-NOTA-RamAb, for map- body for breast cancer patients. In preclinical studies, trastuzu- ping VEGFR-2 expression in vivo (65), and found that this tech- mab has been used as a targeting agent to map HER2 expression nology could visualize VEGFR-2 in nude mice bearing HCC4006 and distribution either by PET imaging or by SPECT imaging and A549 NSCLC tumor models. Based on our results, a study (4, 13, 70, 71). In addition, 89Zr-Df-trastuzumab PET, rather from Eric and colleagues further suggested that a three-time- than 18F-FDG PET, successfully assessed the pharmacodynamic point method could be used to assess the in vivo performance of effects of (an EGFR-HER2 dual inhibitor) in HER2- 64Cu-NOTA-RamAb (66). positive gastric cancer models (Fig. 3A and B; ref. 72). In clinical practice, HER2 status has been determined by immunohis- tochemistry, and fluorescence in situ hybridization has been PET Imaging of HER2 validated to predict the efficacy of the HER2-targeting anti- The human receptor 2 (HER2) trans- body–drug conjugate . Because SPECT membrane oncoprotein is overexpressed in many human tumors, imaging using 111In-trastuzumab discovered new HER2-positive and several HER2-targeting agents have entered clinical practice lesions in 13 of 15 patients with breast cancer (10), Dijkers and (4). However, despite this success, responses to these antibodies colleagues developed 89Zr-Df-trastuzumab (13), and reported and small molecules have been hampered by resistance or poor that PET imaging using this probe in breast cancer patients was responses. In this setting, molecular PET imaging techniques can able to detect both previously known metastatic lesions and

Figure 3. Representative HER2-, EGFR-, and HER3-targeting PET probes and their potential clinical applications. A, 89Zr-Df-trastuzumab PET specifically imaged HER2-positive NCI-N87 tumors (red circle). On the contrary, 18F-FDG and 18F-FLT PET nonspecifically detected both the HER2-positive NCI-N87 tumor (red circle) and HER2-negative MKN-74 tumor (black circle). B, Afatinib therapy in HER2-positive xenograft models reduced uptake of 89Zr-Df-trastuzumab in a time- dependent manner, justifying the potential value of 89Zr-trastuzumab PET in measuring the pharmacodynamic effects of afatinib in the clinical setting. C, 64Cu-PCTA- cetuximab PET imaging clearly visualized EGFR-positive TE-8 xenograft models (white dotted circle). D, 177Lu-cetuximab radioimmunotherapy, rather than saline or cetuximab treatment, markedly reduced 18F-FDG uptake in TE-8 models. E, 89Zr-Df- is a HER3-specific immunoPET probe. PET/CT scanning performed 4 days after injection of 89Zr-lumretuzumab detected one lung metastasis (white arrow) from ampullary cancer. Panels reproduced with permission from ref. 72, 99, SNMMI and ref. 115, AACR.

www.aacrjournals.org Mol Cancer Ther; 17(8) August 2018 1629

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

lesions which had been previously undetected (73). Recently, 64Cu-NOTA-/89Zr-DFO-pertuzumab are effective 89Zr-Df-trastuzumab PET has been investigated as a useful tool tools for imaging HER2 expression. More recently, a first-in- to predict response in breast cancer patients treated with trastu- human study in patients with HER2-positive breast cancer zumab emtansine, and to improve the understanding of tumor showed that 89Zr-DFO-pertuzumab PET/CT was safe and dem- heterogeneity in breast cancers (14). Indeed, the heterogeneity onstrated optimal imaging 5 to 8 days after administration of within and across tumor lesions in a single patient or among the tracer. Additionally, 89Zr-DFO-pertuzumab PET/CT was different patients is increasingly being recognized, with signif- able to image multiple sites of HER2-positive malignancy icant therapeutic implications (74). In support of this concept, including HER2-positive brain metastases (86). Further clinical Ulaner and colleagues demonstrated that 89Zr-DFO-trastuzu- studies are still needed to validate the efficacies of these two mab PET/CT detected unsuspected HER2-positive metastases in probes, especially in improving patient stratification. patients with HER2-negative primary breast cancer (15, 75). It is Compared with intact antibodies, antibody fragments and quite difficult for conventional biopsy techniques, which gen- single-domain antibodies (sdAb) have superior imaging charac- erally obtain samples from a limited number of lesions, to teristics, such as rapid clearance, high target-to-background ratios, identify these unique cohorts. This exciting finding may facilitate reduced radiation dose, and engineered sites for site-specific better management of HER2-negative breast cancer patients, as conjugation (87, 88). However, compared with clinically avail- about 10% to 15% of patients with HER2-negative primary able antibodies, the immune responses, safety profiles, and effi- breast cancer may still benefit from HER2-targeted therapy cacy of these novel sdAbs in humans are largely unknown. (76). Furthermore, early changes in 89Zr-Df-trastuzumab uptake Positron emitter–labeled nanobodies as probes for evaluating in breast cancer metastases following treatment with heat HER2 status have been reported (89–92). One strength of such shock protein 90 inhibitor NVP-AUY922 correlated with probes, for example, [18F]RL-I-5F7 and [18F]RL-I-2Rs15d (89, 90), changes of lesion size measured on CT images (77). In addition is their ability to rapidly cross the intact blood–brain barrier and to 89Zr-labeled trastuzumab, 64Cu-DOTA-trastuzumab is anoth- detect brain metastases from breast cancers 1 hour after injection er alternative for optimizing trastuzumab treatment (16). of the tracers. In comparison, the best time points for detecting HER2 is overexpressed in EGA and trastuzumab has been brain metastases using either 89Zr-Df-trastuzumab or 89Zr-DFO- approved by the FDA to treat EGA patients with positive HER2 pertuzumab were 4 to 5 days for the former tracer and 5 to 8 days expression (78). Considering the fact that only a subset of patients for the latter following tracer administration (73, 86). Another with HER2-positive EGA respond to trastuzumab (79), O'Dono- strength is the higher tumor-to-blood and tumor-to-muscle ratios ghue and colleagues studied the value of 89Zr-DFO-trastuzumab which will result in higher contrast PET imaging (89, 93). A phase I in assessing HER2 status in primary and metastatic EGA (17), and study from Keyaerts and colleagues showed PET imaging using a reported that this imaging method detected local and metastatic 68Ga-HER2-nanobody is a safe procedure and tracer accumula- lesions in 80% of the imaged patients. This study indicated that tion in HER2-positive breast carcinoma metastases is high, which 89Zr-DFO-trastuzumab PET has superior advantages over single- warrants further assessment in a phase II trial (94). site biopsy, because this noninvasive imaging technology can assess variation in levels of HER2 and target engagement in both the primary and metastatic tumor lesions simultaneously. Nota- PET Imaging of EGFR (HER1) bly, bifunctional chelators have certain impact on the in vitro EGFR and its relatives (i.e., ErbB2/HER2, ErbB3/HER3, and stability and in vivo performance of 89Zr-labeled trastuzumab and ErbB4/HER4) are well-known oncogenic drivers in several types antibody–drug conjugates (80). of cancers such as lung cancer, breast cancer, and glioblastoma. Pertuzumab is another HER2-targeting mAb approved by EGFR was among the first RTKs for which the ligand binding the FDA after a survival benefit was achieved for patients with mechanism was studied (95). Inhibitors for this specific RTK, HER2-positive metastatic breast cancer. Importantly, pertuzumab including antibody therapeutics (e.g., cetuximab and panitumu- binds to a HER2 binding site distinct from that of trastuzumab, mab) and small-molecule TKIs (e.g., erlotinib, gefitinib, , augmenting the binding and treatment efficacy of each other (81). and ), have been the most successful examples of Thus, it is of great importance to noninvasively detect the biodis- molecularly targeted therapies in cancers (95, 96). tribution of pertuzumab-specific binding sites when synergistic Cetuximab is a chimeric EGFR-specific IgG1 mAb and functions regimens containing trastuzumab and pertuzumab are given for by preventing ligand activation and receptor dimerization (97). breast cancer patients. In this setting, Marquez and colleagues It was shown in a phase I first-in-human study that 89Zr-Df- conducted an in vivo study in breast cancer models using 89Zr- cetuximab was safe and well tolerated (20). PET imaging using DFO-pertuzumab and observed significant tracer uptake in MDA- 89Zr-labeled cetuximab may also select NSCLC patients, head and MB-231 xenografts (82). More importantly, pretargeting strate- neck cancer patients, and colorectal cancer patients who will gies using unlabeled trastuzumab could further enhance 89Zr- potentially benefit from cetuximab treatment (18, 19, 98). In DFO-pertuzumab accumulation in tumors. Combination treat- recent years, theranostic approaches using mAbs have represented ments with trastuzumab and pertuzumab have also shown a rapidly expanding component of cancer treatments (12). Song enhanced antitumor effects in xenograft models of human and colleagues prepared 64Cu/177Lu-labeled versions of cetuxi- ovarian cancer (83), and the addition of pertuzumab to che- mab and assessed the theranostic efficacy in an esophageal squa- motherapy in patients with platinum-resistant ovarian carci- mous cell carcinoma model (99). The authors elaborately found noma demonstrated favorable trends in progression-free sur- that 64Cu-PCTA-cetuximab immunoPET evaluated EGFR expres- vival (84). Based these preclinical and clinical data, our group sion levels in tumors, and 177Lu-cetuximab radioimmunotherapy synthesized 64Cu-NOTA-pertuzumab and broadened the appli- effectively inhibited tumor growth much more thoroughly than cation of the probe to detect both subcutaneous and orthotopic that of cetuximab alone (Fig. 3C and D). Thus, the pair of ovarian cancer models (85). These solid data demonstrated that 64Cu/177Lu-cetuximab may provide a tailored treatment strategy

1630 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of RTKs

for EGFR-positive patients by immunoPET imaging of EGFR administration of 64Cu-DOTA- is safe, the diagnostic expression and by selective therapeutic radiation delivery. efficacy was limited in solid tumors (116). Comparatively, 89Zr- is a fully humanized mAb that binds to EGFR Df-lumretuzumab PET could visualize intra- and inter patient and is FDA approved for use in receptor-expressing colorectal tumor heterogeneity and target accessibility, and the most opti- cancers without KRAS mutations (100). Wei and colleagues mal PET conditions were found to be 4 and 7 days after admin- synthesized 89Zr-DFO-panitumumab and this probe accumulat- istration of 89Zr-Df-lumretuzumab with 100 mg cold antibody ed in various subcutaneous athymic nude female xenograft mod- (Fig. 3E; ref. 115). In the absence of side effects of lumretuzumab els (101). Lindenberg and colleagues then calculated the maxi- in treating HER3-expressing solid tumors (122), 89Zr-Df-lumre- mum dosing for effective imaging with 89Zr-DFO-panitumumab tuzumab may have potential value in guiding lumretuzumab in 3 patients, and results from this study indicated that injection of treatment of patients with HER3-positive solid tumors. More approximately 1 mCi (37 MBq) of 89Zr-DFO-panitumumab recently, Warnders and colleagues constructed a HER3 signal- intravenously was safe and detected lung metastases from colo- ing–specific probe, 89Zr-MSB0010853, and reported that 89Zr- rectal cancer, but imaging of the included 3 patients failed to MSB0010853 PET could detect HER3-overexpressing H441 demonstrate significant radiotracer accumulation in tumors for NSCLC cancer models in vivo (123). Although development of all time points and failed to detect tumors in the liver due to therapeutic HER3 antibodies is still in an early phase (122, 124), physiologic hepatic excretion of the probe (102). Future studies these preliminary results shed light on the feasibility of nonin- may add unlabeled panitumumab to optimize the radiotracer's vasive HER3-specific PET imaging for mapping HER3 expression uptake in tumors (103). More recently, (GA201), and distribution of HER3 antibodies. a novel humanized anti-EGFR IgG1 isotype mAb, has been foundtoefficiently inhibit EGFR pathway activation in head PET Imaging of Other RTKs and neck cancer patients and may serve as a potential probe for Apart from the above RTKs, there are 17 more RTK subfamilies visualizing EGFR-expressing tumors after radiolabeling with and nearly half of these RTK families are poorly understood (2). isotopes (104, 105). One example is the platelet-derived growth factor receptor-alpha Repebody, a novel nonantibody protein scaffold (106), has (PDGFR-a), which has been reported to be involved in tumor also been engineered to monitor EGFR expression (107, 108). angiogenesis and maintenance of several cancer types including One advantage of repebody-based PET imaging probes is their thyroid cancer (125–127). Importantly, PDGFR-a is associated small molecular weight (30 kDa), which may enable early with lymph node metastases in papillary thyroid carcinoma (PTC; visualization of various EGFR-expressing solid tumors as early ref. 127). Michael and colleagues described a PDGFRa-specific as 1 hour after injection of the probes. Furthermore, due to immunoPET probe—64Cu-NOTA-D13C6—for PTC and demon- superior circulation clearance and tissue penetration, those strated that PET imaging using this agent could delineate PDGFRa probes could be labeled with positron emitters with shorter expression in PTC models in vivo and could therefore potentially half-lives, such as 64Cu (t , 12.7 hours) and 18F(t ,110 1/2 1/2 serve as a novel and promising radiotracer for imaging PDGFRa- minutes; ref. 109). positive metastasis from PTC (128). Another example is the As we previously reviewed (110), PET imaging using bispecific -like growth factor-1 receptor (IGF-1R), which has a peptide- and antibody-based heterodimers may demonstrate well-established role in malignant tumors and has become a higher targeting efficacy and specificity than their monospecific promising target for cancer therapy and imaging (129, 130). As peers. We generated a bispecific immunoconjugate (denoted as R1507 is a mAb directed against IGF-1R, Heskamp and colleagues Bs-F(ab) ) by linking a cetuximab Fab and a TRC105 Fab (target- 2 developed 111In-R1507 and 89Zr-Df-R1507 and reported that ing CD105), and confirmed that 64Cu-NOTA-Bs-F(ab) had a 2 both of the tracers could be applied to noninvasively determine synergistic improvement on both affinity and specificity in visu- IGF-1R expression in vivo in breast cancer xenografts (131). Our alizing small U87MG tumor nodules (<5 mm in diameter; group also developed an IGF-1R–specific PET probe (denoted as ref. 111). Similar results were reported by Kwon and colleagues 89Zr-DFO-1A2G11) and demonstrated highly specific uptake of who developed an immunoconjugate targeting HER2 and EGFR the tracer in IGF-1R–positive pancreatic tumor models (132), and verified its diagnostic efficacy in breast cancer models (112), implying the potential value of this probe in identifying patients and by Mahmood and colleagues who prepared PET probes that may benefit from anti–IGF-1R therapy. specific to EGFR and HER3 and monitored resistance to PI3K In addition, RTKs are important drug targets, and several and protein kinase B (PKB, also known as Akt) inhibitors using small-molecule inhibitors have been developed and approved breast cancer models as well (66). by the FDA in addition to mAbs. Actually, development of radiolabeled TKIs and their analogues is under preclinical and PET Imaging of HER3 clinical translational research (133), and various radiolabeled TKIs have been developed to image tumors in both preclinical Unlike other members of the family, HER3 has relatively weak and clinical studies for several RTKs, such as EGFR (134, 135), kinase activity but still can form highly activated heterodimers HER2 (136), VEGFR (137), tropomyosin receptor kinase (Trk; with EGFR and HER2. Activating mutations in HER3 have been ref. 138), stem cell growth factor receptor (SCFR; ref. 139), and identified in multiple cancer types and therefore HER3 is now IGF-1R (129, 140, 141). being examined as a direct therapeutic target (113). Preclinical and clinical PET imaging has also been developed using anti- HER3 antibodies, such as lumretuzumab (114, 115), patritumab Conclusion and Future Perspectives (116), and mAb3481 (117), or using peptides (118), and affi- Currently, the role of spatial deregulation of RTKs in tumori- bodies (119–121). In a pilot clinical trial that included 11 genesis may be vastly underappreciated, and the development of participants, Lockhart and colleagues reported that although high-resolution immunoPET techniques for detecting RTK

www.aacrjournals.org Mol Cancer Ther; 17(8) August 2018 1631

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

activity in normal and tumor tissues will be essential for studying demonstrated the success of 68Ga-HER2-nanobody in a clinical the role of spatial RTK patterning in the heterogeneous tumor setting (94), and it is easier for immunoPET probes derived from environment. In this state-of-the-art review, we presented typical sdAbs to traverse the blood–brain barrier (90), future studies may examples of deregulated RTKs in cancers and summarized strat- further design noninvasive means to detect RTKs through the use egies to prepare noninvasive PET imaging probes specific for these of sdAbs (150). targets. The current review demonstrates that noninvasive immu- In the development of antibody-based diagnostic, therapeu- noPET could accurately delineate RTK status within a tumor or tic, or theranostic probes, we should pay attention to the fact across multiple tumors within a patient. Visualization of RTK that the immunodeficient strains of animals we use in preclin- phenotype using immunoPET could facilitate selection of ical studies may impact the in vivo fate and performance of the patients for targeted therapies and response assessment/predic- investigated immunoPET probes. For example, a recent study tion. In addition, a greater understanding of the spatial expression reported that immunoPET radiotracers had inefficient tumor of RTKs with the help of immunoPET could affect ongoing efforts targeting and high off-target binding to the spleen in highly to develop therapeutic drugs targeting RTKs. immunodeficient mouse models (151). This was consistent In 2017, the development of antibody therapeutics pro- with a previous study which reported that antibody–drug ceeded at a fast pace, and this is expected to continue in the conjugates had limited antitumor activity in NSG mice coming years. Several RTK-targeting therapeutic antibodies (152). In this setting, pretargeting strategies may also be har- [such as , (vic-)trastuzumab duocarmazine, and nessed for optimizing both the imaging quality and therapy DS-8201 for HER2, and for EGFR] effects in the coming future (151, 153, 154). are undergoing evaluation in late-stage clinical studies of Besides assessing RTKs and selecting proper patients for patients with cancer (142). For mAb-based immunoPET subsequent molecularly targeted therapy using either small probes, 89Zr is utilized in clinical trials much more extensively molecules or antibodies, noninvasive imaging of RTKs could than any other radiometals. DFO and DFO-based bifunctional facilitate image-guided radionuclide therapy. Actually, therapy chelators are the most commonly used chelators for 89Zr- of B-cell lymphomas with radiolabeled mAbs has produced radiolabeling (143), and there are two simple protocols that impressive clinical results because two radiolabeled anti-CD20 canbefollowedtodo89Zr radiolabeling (144, 145). However, antibodies, 131I- and 90Y-, DFO is not an optimal chelator for 89Zr coordination because were approved by FDA more than a decade ago (155, 156). of 89Zr transchelation, which will cause high bone uptake of RTKs are ideal candidates for investigating and performing 89Zr. This was exemplified by a recent study by Vugts and radioimmunotherapy (157, 158). Therefore, radiolabeled anti- colleagues, in which the authors reported that, when compared bodies or antibody fragments targeting markers included in the with DFO, the bifunctional isothiocyanate variant of desfer- current review and other potential neoantigens may provide rioxamine (denoted as octadentate DFO) was advantageous another therapeutic option for patients with cancer in the era of for 89Zr labeling because 89Zr-DFO-trastuzumab had molecularly targeted therapy and precision medicine. enhanced stability in in vitro studies and superior performance We firmly believe that immunoPET will allow for better cancer in in vivo studies over 89Zr-DFO-trastuzumab (80). Therefore, patient management in the clinical setting with the fast develop- there is still room for future studies to optimize radiolabeling ment of this field. Considering most data with immunoPET strategies when developing 89Zr-immunoconjugates (143). probes have been generated in preclinical stages or in small groups With the advent and maturation of click chemistry (146), future of patients, further studies are still needed to push clinical trans- studies may further harness this powerful method to synthesize lation of some of these promising probes and to confirm the value and assess antibody-based PET imaging probes with improved of clinically reported probes. in vivo performance. In addition to singly targeted imaging probes, antibody-based heterodimers or dual-targeting probes Disclosure of Potential Conflicts of Interest may have higher targeting efficacy and superior specificity than No potential conflicts of interest were disclosed. their corresponding monospecific peers (110, 147). Generally, adequate tissue penetration ability, which is inverse- Acknowledgments ly proportional to the size of an imaging probe, and high signal- This work was partially sponsored by the Ph.D. Innovation Fund of Shanghai to-noise ratio, which is closely related to the circulation of a probe, Jiao Tong University School of Medicine (No. BXJ201736) and the China are two important properties of an immunoPET probe. sdAbs, Scholarship Council (No. 201706230067) to W. Wei, the Shanghai Key Dis- also known as nanobodies or heavy chain–only antibodies bear- cipline of Medical Imaging (No. 2017ZZ02005) to Q.Y. Luo, the American Cancer Society (125246-RSG-13-099-01-CCE), and the NIH (P30CA014520) ing a variable region (VHH), hit the sweet spot and have emerged to W. Cai. E.B. Ehlerding was partially sponsored by the NIH (T32CA009206 as substitutes for their full-size counterparts in diagnostic or and T32GM008505). therapeutic applications (87). Enzymatic methods such as sortase have been used to append metal chelators to enable labeling Received February 5, 2018; revised April 19, 2018; accepted June 4, 2018; sdAbs with 64Cu or 89Zr (148, 149). Because a first-in-human trial published first August 1, 2018.

References 1. Casaletto JB, McClatchey AI. Spatial regulation of receptor tyrosine 3. Cai W, Chen K, Mohamedali KA, Cao Q, Gambhir SS, Rosenblum MG, kinases in development and cancer. Nat Rev Cancer 2012;12: et al. PET of vascular endothelial growth factor receptor expression. J Nucl 387–400. Med 2006;47:2048–56. 2. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. 4. Cai W, Niu G, Chen X. Multimodality imaging of the HER-kinase axis in Cell 2010;141:1117–34. cancer. Eur J Nucl Med Mol Imaging 2008;35:186–208.

1632 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of RTKs

5. Peruzzi B, Bottaro DP. Targeting the c-Met signaling pathway in cancer. everolimus efficacy in patients with metastatic renal cell carcinoma. J Nucl Clin Cancer Res 2006;12:3657–60. Med 2017;58:905–10. 6. Regad T. Targeting RTK signaling pathways in cancer. Cancers (Basel) 27. Zhang Y, Hong H, Cai W. PET tracers based on Zirconium-89. Curr 2015;7:1758–84. Radiopharm 2011;4:131–9. 7. McCabe KE, Wu AM. Positive progress in immunoPET–not just a coin- 28. Hernandez R, Sun H, England CG, Valdovinos HF, Ehlerding EB, Barnhart cidence. Cancer Biother Radiopharm 2010;25:253–61. TE, et al. CD146-targeted immunoPET and NIRF imaging of hepatocel- 8. Aluicio-Sarduy E, Ellison PA, Barnhart TE, Cai W, Nickles RJ, Engle JW. PET lular carcinoma with a dual-labeled . Theranostics radiometals for antibody labeling. J Labelled Comp Radiopharm 2018; 2016;6:1918–33. [Epub ahead of print]. 29. England CG, Jiang D, Ehlerding EB, Rekoske BT, Ellison PA, Hernandez R, 9. Cai W, Chen X. Multimodality molecular imaging of tumor angiogenesis. et al. (89)Zr-labeled for imaging of T-cell infiltration in a J Nucl Med 2008;49:113S–28S. humanized murine model of lung cancer. Eur J Nucl Med Mol Imaging 10. Perik PJ, Lub-De Hooge MN, Gietema JA, van der Graaf WT, de Korte MA, 2018;45:110–20. Jonkman S, et al. Indium-111-labeled trastuzumab scintigraphy in 30. Sierra JR, Tsao MS. c-MET as a potential therapeutic target and biomarker patients with human epidermal growth factor receptor 2-positive meta- in cancer. Ther Adv Med Oncol 2011;3:S21–35. static breast cancer. J Clin Oncol 2006;24:2276–82. 31. Salgia R. MET in lung cancer: biomarker selection based on scientific 11. James ML, Gambhir SS. A molecular imaging primer: modalities, imaging rationale. Mol Cancer Ther 2017;16:555–65. agents, and applications. Physiol Rev 2012;92:897–965. 32. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. 12. Moek KL, Giesen D, Kok IC, de Groot DJA, Jalving M, Fehrmann RSN, et al. MET amplification leads to gefitinib resistance in lung cancer by activating Theranostics using antibodies and antibody-related therapeutics. J Nucl ERBB3 signaling. Science 2007;316:1039–43. Med 2017;58:83S–90S. 33. Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, et al. 13. Dijkers EC, Kosterink JG, Rademaker AP, Perk LR, van Dongen GA, Bart J, Preexistence and clonal selection of MET amplification in EGFR mutant et al. Development and characterization of clinical-grade 89Zr-trastuzu- NSCLC. Cancer Cell 2010;17:77–88. mab for HER2/neu immunoPET imaging. J Nucl Med 2009;50:974–81. 34. Johnson ML, Yu HA, Hart EM, Weitner BB, Rademaker AW, Patel JD, et al. 14. Gebhart G, Lamberts LE, Wimana Z, Garcia C, Emonts P, Ameye L, et al. Phase I/II Study of HSP90 Inhibitor AUY922 and Erlotinib for EGFR- Molecular imaging as a tool to investigate heterogeneity of advanced mutant lung cancer with acquired resistance to epidermal growth factor HER2-positive breast cancer and to predict patient outcome under tras- inhibitors. J Clin Oncol 2015;33:1666–73. tuzumab emtansine (T-DM1): the ZEPHIR trial. Ann Oncol 2016;27: 35. Merchant M, Ma X, Maun HR, Zheng Z, Peng J, Romero M, et al. 619–24. Monovalent antibody design and mechanism of action of onartuzumab, 15. Ulaner GA, Hyman DM, Lyashchenko SK, Lewis JS, Carrasquillo JA. 89Zr- a MET antagonist with anti-tumor activity as a therapeutic agent. Proc Natl Trastuzumab PET/CT for detection of human epidermal growth factor Acad Sci U S A 2013;110:E2987–96. receptor 2-positive metastases in patients with human epidermal growth 36. Rolfo C, Van Der Steen N, Pauwels P, Cappuzzo F. Onartuzumab in lung factor receptor 2-negative primary breast cancer. Clin Nucl Med 2017; cancer: the fall of Icarus? Expert Rev Anticancer Ther 2015;15:487–9. 42:912–7. 37. Spigel DR, Ervin TJ, Ramlau RA, Daniel DB, Goldschmidt JH, Jr. 16. Mortimer JE, Bading JR, Park JM, Frankel PH, Carroll MI, Tran TT, et al. Blumenschein GR, Jr. et al. Randomized phase II trial of Onartuzumab Tumor Uptake of (64)Cu-DOTA-trastuzumab in patients with metastatic in combination with erlotinib in patients with advanced non-small-cell breast cancer. J Nucl Med 2018;59:38–43. lung cancer. J Clin Oncol 2013;31:4105–14. 17. O'Donoghue JA, Lewis JS, Pandit-Taskar N, Fleming SE, Schoder H, Larson 38. Spigel DR, Edelman MJ, O'Byrne K, Paz-Ares L, Mocci S, Phan S, et al. SM, et al. Pharmacokinetics, biodistribution, and radiation dosimetry for Results from the phase III randomized trial of onartuzumab plus erlotinib (89)Zr-Trastuzumab in patients with esophagogastric cancer. J Nucl Med versus erlotinib in previously treated stage IIIB or IV non-small-cell lung 2018;59:161–6. cancer: METLung. J Clin Oncol 2017;35:412–20. 18. Menke-van der Houven van Oordt CW, Gootjes EC, Huisman MC, Vugts 39. Zhang Y, Xia M, Jin K, Wang S, Wei H, Fan C, et al. Function of the c-Met DJ, Roth C, Luik AM, et al. 89Zr-cetuximab PET imaging in patients with receptor tyrosine kinase in carcinogenesis and associated therapeutic advanced colorectal cancer. Oncotarget 2015;6:30384–93. opportunities. Mol Cancer 2018;17:45. 19. Even AJ, Hamming-Vrieze O, van Elmpt W, Winnepenninckx VJ, 40. Hay RV, Cao B, Skinner RS, Su Y, Zhao P, Gustafson MF, et al. Nuclear Heukelom J, Tesselaar ME, et al. Quantitative assessment of Zirconi- imaging of Met-expressing human and canine cancer xenografts with um-89 labeled cetuximab using PET/CT imaging in patients with radiolabeled monoclonal antibodies (MetSeek). Clin Cancer Res advanced head and neck cancer: a theragnostic approach. Oncotarget 2005;11:7064s–9s. 2017;8:3870–80. 41. Perk LR, Stigter-van Walsum M, Visser GW, Kloet RW, Vosjan MJ, Leemans 20. van Loon J, Even AJG, Aerts H, Ollers M, Hoebers F, van Elmpt W, et al. PET CR, et al. Quantitative PET imaging of Met-expressing human cancer imaging of zirconium-89 labelled cetuximab: a phase I trial in patients xenografts with 89Zr-labelled monoclonal antibody DN30. Eur J Nucl with head and neck and lung cancer. Radiother Oncol 2017;122:267–73. Med Mol Imaging 2008;35:1857–67. 21. Bahce I, Huisman MC, Verwer EE, Ooijevaar R, Boutkourt F, Vugts DJ, et al. 42. Jagoda EM, Lang L, Bhadrasetty V, Histed S, Williams M, Kramer-Marek G, Pilot study of (89)Zr-bevacizumab positron emission tomography et al. Immuno-PET of the receptor Met using the in patients with advanced non-small cell lung cancer. EJNMMI Res 1-armed antibody onartuzumab. J Nucl Med 2012;53:1592–600. 2014;4:35. 43. Li K, Tavare R, Zettlitz KA, Mumenthaler SM, Mallick P, Zhou Y, et al. Anti- 22. Gaykema SB, Brouwers AH, Lub-de Hooge MN, Pleijhuis RG, Timmer- MET immunoPET for non-small cell lung cancer using novel fully human Bosscha H, Pot L, et al. 89Zr-bevacizumab PET imaging in primary breast antibody fragments. Mol Cancer Ther 2014;13:2607–17. cancer. J Nucl Med 2013;54:1014–8. 44. Pool M, Terwisscha van Scheltinga AGT, Kol A, Giesen D, de Vries EGE, 23. Jansen MH, Lagerweij T, Sewing AC, Vugts DJ, van Vuurden DG, Molthoff Lub-de Hooge MN. (89)Zr-Onartuzumab PET imaging of c-MET receptor CF, et al. Bevacizumab targeting diffuse intrinsic pontine glioma: results dynamics. Eur J Nucl Med Mol Imaging 2017;44:1328–36. of 89Zr-Bevacizumab PET imaging in brain tumor models. Mol Cancer 45. Eccles SA, Massey A, Raynaud FI, Sharp SY, Box G, Valenti M, et al. NVP- Ther 2016;15:2166–74. AUY922: a novel heat shock protein 90 inhibitor active against xenograft 24. Oosting SF, Brouwers AH, van Es SC, Nagengast WB, Oude Munnink TH, tumor growth, angiogenesis, and metastasis. Cancer Res 2008;68: Lub-de Hooge MN, et al. 89Zr-bevacizumab PET visualizes heterogeneous 2850–60. tracer accumulation in tumor lesions of renal cell carcinoma patients and 46. Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson differential effects of antiangiogenic treatment. J Nucl Med 2015;56:63–9. DH, et al. Rilotumumab plus epirubicin, cisplatin, and capecitabine as 25. Oosting SF, van Asselt SJ, Brouwers AH, Bongaerts AH, Steinberg JD, de first-line therapy in advanced MET-positive gastric or gastro-oesophageal Jong JR, et al. 89Zr-Bevacizumab PET visualizes disease manifestations in junction cancer (RILOMET-1): a randomised, double-blind, placebo- patients with von hippel-lindau disease. J Nucl Med 2016;57:1244–50. controlled, phase 3 trial. Lancet Oncol 2017;18:1467–82. 26. van Es SC, Brouwers AH, Mahesh SVK, Leliveld-Kors AM, de Jong IJ, Lub- 47. Price EW, Carnazza KE, Carlin SD, Cho A, Edwards KJ, Sevak KK, et al. (89) de Hooge MN, et al. (89)Zr-Bevacizumab PET: potential early indicator of Zr-DFO-AMG102 Immuno-PET to determine local hepatocyte growth

www.aacrjournals.org Mol Cancer Ther; 17(8) August 2018 1633

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

factor protein levels in tumors for enhanced patient selection. J Nucl Med 69. Pereira PMR, Abma L, Henry KE, Lewis JS. Imaging of human epidermal 2017;58:1386–94. growth factor receptors for patient selection and response monitoring – 48. Luo H, Hong H, Slater MR, Graves SA, Shi S, Yang Y, et al. PET of c-met in From PET imaging and beyond. Cancer Lett 2018;419:139–51. cancer with (6)(4)Cu-labeled hepatocyte growth factor. J Nucl Med 70. J NT, Pandya DN, Pailloux SL, Ogasawara A, Vanderbilt AN, Gill HS, et al. 2015;56:758–63. Evaluation of a 3-hydroxypyridin-2-one (2,3-HOPO) based macrocyclic 49. Burggraaf J, Kamerling IM, Gordon PB, Schrier L, de Kam ML, Kales AJ, chelator for (89)Zr(4þ) and its use for immunopet imaging of HER2 et al. Detection of colorectal polyps in humans using an intravenously positive model of ovarian carcinoma in mice. Theranostics 2016;6: administered fluorescent peptide targeted against c-Met. Nat Med 511–21. 2015;21:955–61. 71. Ma T, Sun X, Cui L, Gao L, Wu Y, Liu H, et al. Molecular imaging reveals 50. Esfahani SA, Heidari P, Kim SA, Ogino S, Mahmood U. Optical imaging of trastuzumab-induced epidermal growth factor receptor downregulation mesenchymal epithelial transition factor (MET) for enhanced detection in vivo. J Nucl Med 2014;55:1002–7. and characterization of primary and metastatic hepatic tumors. Thera- 72. Janjigian YY, Viola-Villegas N, Holland JP, Divilov V, Carlin SD, Gomes- nostics 2016;6:2028–38. DaGama EM, et al. Monitoring afatinib treatment in HER2-positive 51. Arulappu A, Battle M, Eisenblaetter M, McRobbie G, Khan I, Monypenny J, gastric cancer with 18F-FDG and 89Zr-trastuzumab PET. J Nucl Med et al. c-Met PET imaging detects early-stage locoregional recurrence of 2013;54:936–43. basal-like breast cancer. J Nucl Med 2016;57:765–70. 73. Dijkers EC, Oude Munnink TH, Kosterink JG, Brouwers AH, Jager PL, de 52. Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of Jong JR, et al. Biodistribution of 89Zr-trastuzumab and PET imaging of angiogenesis. Cell 2011;146:873–87. HER2-positive lesions in patients with metastatic breast cancer. Clin 53. Simon T, Gagliano T, Giamas G. Direct effects of anti-angiogenic therapies Pharmacol Ther 2010;87:586–92. on tumor cells: VEGF signaling. Trends Mol Med 2017;23:282–92. 74. McGranahan N, Swanton C. Biological and therapeutic impact of intra- 54. Bahce I, Yaqub M, Smit EF, Lammertsma AA, van Dongen GA, Hendrikse tumor heterogeneity in cancer evolution. Cancer Cell 2015;27:15–26. NH. Personalizing NSCLC therapy by characterizing tumors using TKI- 75. Ulaner GA, Hyman DM, Ross DS, Corben A, Chandarlapaty S, Goldfarb S, PET and immuno-PET. Lung Cancer 2017;107:1–13. et al. Detection of HER2-positive metastases in patients with HER2- 55. van Asselt SJ, Oosting SF, Brouwers AH, Bongaerts AH, de Jong JR, Lub- negative primary breast cancer using 89Zr-trastuzumab PET/CT. J Nucl de Hooge MN, et al. Everolimus Reduces (89)Zr-Bevacizumab tumor Med 2016;57:1523–8. uptake in patients with neuroendocrine tumors. J Nucl Med 2014;55: 76. Paik S, Kim C, Wolmark N. HER2 status and benefit from adjuvant 1087–92. trastuzumab in breast cancer. N Engl J Med 2008;358:1409–11. 56. Nagengast WB, de Vries EG, Hospers GA, Mulder NH, de Jong JR, Hollema 77. Gaykema SB, Schroder CP, Vitfell-Rasmussen J, Chua S, Oude Munnink H, et al. In vivo VEGF imaging with radiolabeled bevacizumab in a human TH, Brouwers AH, et al. 89Zr-trastuzumab and 89Zr-bevacizumab PET to ovarian tumor xenograft. J Nucl Med 2007;48:1313–9. evaluate the effect of the HSP90 inhibitor NVP-AUY922 in metastatic 57. Cai W, Rao J, Gambhir SS, Chen X. How molecular imaging is speeding up breast cancer patients. Clin Cancer Res 2014;20:3945–54. antiangiogenic drug development. Mol Cancer Ther 2006;5:2624–33. 78. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. 58. Jansen MH, Veldhuijzen van Zanten SEM, van Vuurden DG, Huisman Trastuzumab in combination with versus chemotherapy MC, Vugts DJ, Hoekstra OS, et al. Molecular drug imaging: (89)Zr- alone for treatment of HER2-positive advanced gastric or gastro-oeso- bevacizumab PET in children with diffuse intrinsic pontine glioma. phageal junction cancer (ToGA): a phase 3, open-label, randomised J Nucl Med 2017;58:711–6. controlled trial. Lancet 2010;376:687–97. 59. Veldhuijzen van Zanten SEM, Sewing ACP, van Lingen A, Hoekstra OS, 79. Ock CY, Lee KW, Kim JW, Kim JS, Kim TY, Lee KH, et al. Optimal patient Wesseling P, Meel MH, et al. Multiregional tumor drug-uptake imaging by selection for trastuzumab treatment in HER2-positive advanced gastric PET and microvascular morphology in end-stage diffuse intrinsic pontine cancer. Clin Cancer Res 2015;21:2520–9. glioma. J Nucl Med 2018;59:612–5. 80. Vugts DJ, Klaver C, Sewing C, Poot AJ, Adamzek K, Huegli S, et al. 60. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Staehler M, et al. Comparison of the octadentate bifunctional chelator DFO-pPhe-NCS Sorafenib for treatment of renal cell carcinoma: final efficacy and safety and the clinically used hexadentate bifunctional chelator DFO-pPhe-NCS results of the phase III treatment approaches in renal cancer global for (89)Zr-immuno-PET. Eur J Nucl Med Mol Imaging 2017;44:286–95. evaluation trial. J Clin Oncol 2009;27:3312–8. 81. Lua WH, Gan SK, Lane DP, Verma CS. A search for synergy in the binding 61. Bueno MJ, Mouron S, Quintela-Fandino M. Personalising and targeting kinetics of trastuzumab and pertuzumab whole and F(ab) to Her2. NPJ antiangiogenic resistance: a complex and multifactorial approach. Br J Breast Cancer 2015;1:15012. Cancer 2017;116:1119–25. 82. Marquez BV, Ikotun OF, Zheleznyak A, Wright B, Hari-Raj A, Pierce RA, 62. Dedania VS, Bakri SJ. Current perspectives on ranibizumab. Clin Ophthal- et al. Evaluation of (89)Zr-pertuzumab in breast cancer xenografts. Mol mol 2015;9:533–42. Pharm 2014;11:3988–95. 63. Nagengast WB, Lub-de Hooge MN, Oosting SF, den Dunnen WF, Warn- 83. Faratian D, Zweemer AJ, Nagumo Y, Sims AH, Muir M, Dodds M, et al. ders FJ, Brouwers AH, et al. VEGF-PET imaging is a noninvasive biomarker Trastuzumab and pertuzumab produce changes in morphology and showing differential changes in the tumor during sunitinib treatment. estrogen receptor signaling in ovarian cancer xenografts revealing new Cancer Res 2011;71:143–53. treatment strategies. Clin Cancer Res 2011;17:4451–61. 64. Meyer JP, Edwards KJ, Kozlowski P, Backer MV, Backer JM, Lewis JS. 84. Kurzeder C, Bover I, Marme F, Rau J, Pautier P, Colombo N, et al. Double- Selective imaging of VEGFR-1 and VEGFR-2 using 89Zr-labeled single- blind, placebo-controlled, randomized phase III trial evaluating pertu- chain VEGF mutants. J Nucl Med 2016;57:1811–6. zumab combined with chemotherapy for low tumor human epidermal 65. Luo H, England CG, Graves SA, Sun H, Liu G, Nickles RJ, et al. PET Imaging growth factor receptor 3 mRNA-expressing platinum-resistant ovarian of VEGFR-2 expression in lung cancer with 64Cu-labeled ramucirumab. cancer (PENELOPE). J Clin Oncol 2016;34:2516–25. J Nucl Med 2016;57:285–90. 85. Jiang D, Im HJ, Sun H, Valdovinos HF, England CG, Ehlerding EB, et al. 66. Laffon E, Marthan R. A three-time-point method for assessing kinetic Radiolabeled pertuzumab for imaging of human epidermal growth factor parameters of (64)Cu-labeled ramucirumab trapping in VEGFR-2 posi- receptor 2 expression in ovarian cancer. Eur J Nucl Med Mol Imaging tive lung tumors. Phys Med 2017;43:1–5. 2017;44:1296–305. 67. Wimana Z, Gebhart G, Guiot T, Vanderlinden B, Larsimont D, Doumont 86. Ulaner GA, Lyashchenko SK, Riedl C, Ruan S, Zanzonico PB, Lake D, et al. G, et al. N-Acetylcysteine breaks resistance to trastuzumab caused by First-in-human HER2-targeted imaging using (89)Zr-pertuzumab PET/ MUC4 overexpression in human HER2 positive BC-bearing nude mice CT: dosimetry and clinical application in patients with breast cancer. monitored by (89)Zr-Trastuzumab and (18)F-FDG PET imaging. Onco- J Nucl Med 2017;59:900–6. target 2017;8:56185–98. 87. Chakravarty R, Goel S, Cai W. Nanobody: the "magic bullet" for molecular 68. Gebhart G, Flamen P, De Vries EG, Jhaveri K, Wimana Z. Imaging imaging? Theranostics 2014;4:386–98. diagnostic and therapeutic targets: human epidermal growth factor recep- 88. Wu AM. Engineered antibodies for molecular imaging of cancer. Methods tor 2. J Nucl Med 2016;57:81S–8S. 2014;65:139–47.

1634 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of RTKs

89. Vaidyanathan G, McDougald D, Choi J, Koumarianou E, Weitzel D, mal growth factor receptor over-expressing tumors. Bioconjug Chem Osada T, et al. Preclinical evaluation of 18F-labeled anti-HER2 nanobody 2017;28:2361–71. conjugates for imaging HER2 receptor expression by immuno-PET. J Nucl 110. Ehlerding EB, Sun L, Lan X, Zeng D, Cai W. Dual-targeted molecular Med 2016;57:967–73. imaging of cancer. J Nucl Med 2018;59:390–5. 90. Zhou Z, Vaidyanathan G, McDougald D, Kang CM, Balyasnikova I, 111. Luo H, Hernandez R, Hong H, Graves SA, Yang Y, England CG, et al. Devoogdt N, et al. Fluorine-18 labeling of the HER2-targeting single- Noninvasive brain cancer imaging with a bispecific antibody fragment, domain antibody 2Rs15d using a residualizing label and preclinical generated via click chemistry. Proc Natl Acad Sci U S A 2015;112: evaluation. Mol Imaging Biol 2017;19:867–77. 12806–11. 91. Lam K, Chan C, Reilly RM. Development and preclinical studies of (64) 112. Kwon LY, Scollard DA, Reilly RM. (64)Cu-Labeled trastuzumab Fab- Cu-NOTA-pertuzumab F(ab')2 for imaging changes in tumor HER2 PEG24-EGF radioimmunoconjugates bispecific for HER2 and EGFR: expression associated with response to trastuzumab by PET/CT. MAbs pharmacokinetics, biodistribution, and tumor imaging by PET in com- 2017;9:154–64. parison to monospecific agents. Mol Pharm 2017;14:492–501. 92. Cleeren F, Lecina J, Ahamed M, Raes G, Devoogdt N, Caveliers V, et al. Al 113. Gala K, Chandarlapaty S. Molecular pathways: HER3 targeted therapy. (18)F-labeling of heat-sensitive biomolecules for positron emission Clin Cancer Res 2014;20:1410–6. tomography imaging. Theranostics 2017;7:2924–39. 114. Terwisscha van Scheltinga AG, Lub-de Hooge MN, Abiraj K, Schroder CP, 93. Xavier C, Blykers A, Vaneycken I, D'Huyvetter M, Heemskerk J, Lahoutte T, Pot L, Bossenmaier B, et al. ImmunoPET and biodistribution with human et al. (18)F-nanobody for PET imaging of HER2 overexpressing tumors. epidermal growth factor receptor 3 targeting antibody (8)(9)Zr-RG7116. Nucl Med Biol 2016;43:247–52. MAbs 2014;6:1051–8. 94. Keyaerts M, Xavier C, Heemskerk J, Devoogdt N, Everaert H, Ackaert C, 115. Bensch F, Lamberts LE, Smeenk MM, Jorritsma-Smit A, Lub-de Hooge et al. Phase I study of 68Ga-HER2-nanobody for PET/CT assessment of MN, Terwisscha van Scheltinga AGT, et al. (89)Zr-Lumretuzumab PET HER2 expression in breast carcinoma. J Nucl Med 2016;57:27–33. imaging before and during HER3 antibody lumretuzumab treatment in 95. Lemmon MA, Schlessinger J, Ferguson KM. The EGFR family: not so patients with solid tumors. Clin Cancer Res 2017;23:6128–37. prototypical receptor tyrosine kinases. Cold Spring Harb Perspect Biol 116. Lockhart AC, Liu Y, Dehdashti F, Laforest R, Picus J, Frye J, et al. Phase 1 2014;6:a020768. evaluation of [(64)Cu]DOTA-patritumab to assess dosimetry, apparent 96. Tomas A, Futter CE, Eden ER. EGF receptor trafficking: consequences for receptor occupancy, and safety in subjects with advanced solid tumors. signaling and cancer. Trends Cell Biol 2014;24:26–34. Mol Imaging Biol 2016;18:446–53. 97. Weiner LM, Surana R, Wang S. Monoclonal antibodies: versatile platforms 117. Pool M, Kol A, de Jong S, de Vries EGE, Lub-de Hooge MN, Terwisscha van for cancer immunotherapy. Nat Rev Immunol 2010;10:317–27. Scheltinga AGT. (89)Zr-mAb3481 PET for HER3 tumor status assessment 98. van Dijk LK, Yim CB, Franssen GM, Kaanders JH, Rajander J, Solin O, et al. during lapatinib treatment. MAbs 2017;9:1370–8. PET of EGFR with (64) Cu-cetuximab-F(ab')2 in mice with head and neck 118. Larimer BM, Phelan N, Wehrenberg-Klee E, Mahmood U. Phage display squamous cell carcinoma xenografts. Contrast Media Mol Imaging selection, in vitro characterization, and correlative PET imaging of a novel 2016;11:65–70. HER3 peptide. Mol Imaging Biol 2018;20:300–8. 99. Song IH, Lee TS, Park YS, Lee JS, Lee BC, Moon BS, et al. Immuno-PET 119. Da Pieve C, Allott L, Martins CD, Vardon A, Ciobota DM, Kramer-Marek imaging and radioimmunotherapy of 64Cu-/177Lu-labeled anti-EGFR G, et al. Efficient [(18)F]AlF radiolabeling of ZHER3:8698 affibody antibody in esophageal squamous cell carcinoma model. J Nucl Med molecule for imaging of HER3 positive tumors. Bioconjug Chem 2016;57:1105–11. 2016;27:1839–49. 100. Ciombor KK, Bekaii-Saab T. A comprehensive review of sequencing and 120. Rosestedt M, Andersson KG, Mitran B, Tolmachev V, Lofblom J, Orlova A, combination strategies of targeted agents in metastatic colorectal cancer. et al. Affibody-mediated PET imaging of HER3 expression in malignant Oncologist 2018;23:25–34. tumours. Sci Rep 2015;5:15226. 101. Wei L, Shi J, Afari G, Bhattacharyya S. Preparation of clinical-grade (89) Zr- 121. Orlova A, Malm M, Rosestedt M, Varasteh Z, Andersson K, Selvaraju RK, panitumumab as a positron emission tomography biomarker for eval- et al. Imaging of HER3-expressing xenografts in mice using a (99m)Tc uating epidermal growth factor receptor-targeted therapy. J Labelled (CO) 3-HEHEHE-Z HER3:08699 affibody molecule. Eur J Nucl Med Mol Comp Radiopharm 2014;57:25–35. Imaging 2014;41:1450–9. 102. Lindenberg L, Adler S, Turkbey IB, Mertan F, Ton A, Do K, et al. Dosimetry 122. Meulendijks D, Jacob W, Martinez-Garcia M, Taus A, Lolkema MP, Voest and first human experience with (89)Zr-panitumumab. Am J Nucl Med EE, et al. First-in-Human Phase I study of lumretuzumab, a glycoengi- Mol Imaging 2017;7:195–203. neered humanized Anti-HER3 monoclonal antibody, in patients with 103. Nayak TK, Garmestani K, Milenic DE, Brechbiel MW. PET and MRI of metastatic or advanced HER3-positive solid tumors. Clin Cancer Res metastatic peritoneal and pulmonary colorectal cancer in mice with 2016;22:877–85. human epidermal growth factor receptor 1-targeted 89Zr-labeled pani- 123. Warnders FJ, Terwisscha van Scheltinga AGT, Knuehl C, van Roy M, de tumumab. J Nucl Med 2012;53:113–20. Vries EFJ, Kosterink JGW, et al. Human epidermal growth factor receptor 104. Temam S, Spicer J, Farzaneh F, Soria JC, Oppenheim D, McGurk M, et al. 3-specific tumor uptake and biodistribution of (89)Zr-MSB0010853 An exploratory, open-label, randomized, multicenter study to investigate visualized by real-time and noninvasive PET imaging. J Nucl Med the pharmacodynamics of a glycoengineered antibody (imgatuzumab) 2017;58:1210–5. and cetuximab in patients with operable head and neck squamous cell 124. Juric D, Dienstmann R, Cervantes A, Hidalgo M, Messersmith W, Blu- carcinoma. Ann Oncol 2017;28:2827–35. menschein GR Jr, et al. Safety and pharmacokinetics/pharmacodynamics 105. Pool M, Kol A, Lub-de Hooge MN, Gerdes CA, de Jong S, de Vries EG, et al. of the first-in-class dual action HER3/EGFR antibody MEHD7945A in Extracellular domain shedding influences specific tumor uptake and locally advanced or metastatic epithelial tumors. Clin Cancer Res organ distribution of the EGFR PET tracer 89Zr-imgatuzumab. Oncotar- 2015;21:2462–70. get 2016;7:68111–21. 125. Oseini AM, Roberts LR. PDGFRalpha: a new therapeutic target in the 106. Lee SC, Park K, Han J, Lee JJ, Kim HJ, Hong S, et al. Design of a binding treatment of hepatocellular carcinoma? Expert Opin Ther Targets scaffold based on variable lymphocyte receptors of jawless vertebrates by 2009;13:443–54. module engineering. Proc Natl Acad Sci U S A 2012;109:3299–304. 126. Lopez-Campistrous A, Adewuyi EE, Benesch MGK, Ko YM, Lai R, Thiesen 107. Pyo A, Yun M, Kim HS, Kim TY, Lee JJ, Kim JY, et al. (64)Cu-Labeled A, et al. PDGFRalpha regulates follicular cell differentiation driving repebody molecules for imaging of epidermal growth factor receptor- treatment resistance and disease recurrence in papillary thyroid cancer. expressing tumors. J Nucl Med 2018;59:340–6. EBioMedicine 2016;12:86–97. 108. Yun M, Kim DY, Lee JJ, Kim HS, Kim HS, Pyo A, et al. A High-affinity 127. Zhang J, Wang P, Dykstra M, Gelebart P, Williams D, Ingham R, et al. repebody for molecular imaging of EGFR-expressing malignant tumors. Platelet-derived growth factor receptor-alpha promotes lymphatic metas- Theranostics 2017;7:2620–33. tases in papillary thyroid cancer. J Pathol 2012;228:241–50. 109. Goux M, Becker G, Gorre H, Dammicco S, Desselle A, Egrise D, et al. 128. Wagner M, Wuest M, Hamann I, Lopez-Campistrous A, McMullen TPW, Nanofitin as a new molecular-imaging agent for the diagnosis of epider- Wuest F. Molecular imaging of platelet-derived growth factor receptor-

www.aacrjournals.org Mol Cancer Ther; 17(8) August 2018 1635

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. Wei et al.

alpha (PDGFRalpha) in papillary thyroid cancer using immuno-PET. 143. Heskamp S, Raave R, Boerman O, Rijpkema M, Goncalves V, Denat F. (89) Nucl Med Biol 2018;58:51–8. Zr-immuno-positron emission tomography in oncology: state-of-the-art 129. Sun Y, Sun X, Shen B. Molecular Imaging of IGF-1R in Cancer. Mol (89)Zr radiochemistry. Bioconjug Chem 2017;28:2211–23. Imaging 2017;16:1536012117736648. 144. Verel I, Visser GW, Boellaard R, Stigter-van Walsum M, Snow GB, van 130. Orlova A, Hofstrom C, Strand J, Varasteh Z, Sandstrom M, Andersson K, Dongen GA. 89Zr immuno-PET: comprehensive procedures for the et al. [99mTc(CO)3]þ-(HE)3-ZIGF1R:4551, a new Affibody conjugate production of 89Zr-labeled monoclonal antibodies. J Nucl Med 2003; for visualization of insulin-like growth factor-1 receptor expression in 44:1271–81. malignant tumours. Eur J Nucl Med Mol Imaging 2013;40:439–49. 145. Perk LR, Vosjan MJ, Visser GW, Budde M, Jurek P, Kiefer GE, et al. p- 131. Heskamp S, van Laarhoven HW, Molkenboer-Kuenen JD, Franssen GM, Isothiocyanatobenzyl-desferrioxamine: a new bifunctional chelate Versleijen-Jonkers YM, Oyen WJ, et al. ImmunoSPECT and immunoPET for facile radiolabeling of monoclonal antibodies with zirconium- of IGF-1R expression with the radiolabeled antibody R1507 in a triple- 89 for immuno-PET imaging. Eur J Nucl Med Mol Imaging 2010;37: negative breast cancer model. J Nucl Med 2010;51:1565–72. 250–9. 132. England CG, Kamkaew A, Im HJ, Valdovinos HF, Sun H, Hernandez R, 146. Meyer JP, Adumeau P, Lewis JS, Zeglis BM. Click chemistry and radio- et al. ImmunoPET imaging of insulin-like growth factor 1 receptor in a chemistry: the first 10 years. Bioconjug Chem 2016;27:2791–807. subcutaneous mouse model of pancreatic cancer. Mol Pharm 2016;13: 147. Luo H, Hong H, Yang SP, Cai W. Design and applications of bispecific 1958–66. heterodimers: molecular imaging and beyond. Mol Pharm 2014;11: 133. Tolmachev V, Stone-Elander S, Orlova A. Radiolabelled receptor-tyrosine- 1750–61. kinase targeting drugs for patient stratification and monitoring of therapy 148. Popp MW, Antos JM, Grotenbreg GM, Spooner E, Ploegh HL. Sortagging: response: prospects and pitfalls. Lancet Oncol 2010;11:992–1000. a versatile method for protein labeling. Nat Chem Biol 2007;3:707–8. 134. Bahce I, Smit EF, Lubberink M, van der Veldt AA, Yaqub M, Windhorst AD, 149. Massa S, Vikani N, Betti C, Ballet S, Vanderhaegen S, Steyaert J, et al. et al. Development of [(11)C]erlotinib positron emission tomography for Sortase A-mediated site-specific labeling of camelid single-domain anti- in vivo evaluation of EGF receptor mutational status. Clin Cancer Res body-fragments: a versatile strategy for multiple molecular imaging 2013;19:183–93. modalities. Contrast Media Mol Imaging 2016;11:328–39. 135. Yaqub M, Bahce I, Voorhoeve C, Schuit RC, Windhorst AD, Hoekstra OS, 150. Ingram JR, Schmidt FI, Ploegh HL. Exploiting nanobodies' singular traits. et al. Quantitative and simplified analysis of 11C-erlotinib studies. J Nucl Annu Rev Immunol 2018;36:695–715. Med 2016;57:861–6. 151. Sharma SK, Chow A, Monette S, Vivier D, Pourat J, Edwards KJ, et al. Fc- 136. Henry KE, Ulaner GA, Lewis JS. Human epidermal growth factor receptor mediated anomalous biodistribution of therapeutic antibodies in immu- 2-targeted PET/single-photon emission computed tomography imaging nodeficient mouse models. Cancer Res 2018;78:1820–32. of breast cancer: noninvasive measurement of a biomarker integral to 152. Stefan N, Gebleux R, Waldmeier L, Hell T, Escher M, Wolter FI, et al. tumor treatment and prognosis. PET Clin 2017;12:269–88. Highly potent, anthracycline-based antibody-drug conjugates generated 137. Slobbe P, Poot AJ, Haumann R, Schuit RC, Windhorst AD, van Dongen by enzymatic, site-specific conjugation. Mol Cancer Ther 2017;16: GA. Two anti-angiogenic TKI-PET tracers, [(11)C] and [(11) 879–92. C]: radiosynthesis, in vivo metabolism and initial biodistribu- 153. Kraeber-Bodere F, Rousseau C, Bodet-Milin C, Frampas E, Faivre-Chauvet tion studies in rodents. Nucl Med Biol 2016;43:612–24. A, Rauscher A, et al. A pretargeting system for tumor PET imaging and 138. Bernard-Gauthier V, Mahringer A, Vesnaver M, Fricker G, Schirrmacher R. radioimmunotherapy. Front Pharmacol 2015;6:54. Design and synthesis of a fluorinated quinazoline-based type-II Trk 154. Keinanen O, Fung K, Pourat J, Jallinoja V, Vivier D, Pillarsetty NK, et al. inhibitor as a scaffold for PET radiotracer development. Bioorg Med Pretargeting of internalizing trastuzumab and cetuximab with a (18)F- Chem Lett 2017;27:2771–5. tetrazine tracer in xenograft models. EJNMMI Res 2017;7:95. 139. Peng Z, Maxwell DS, Sun D, Bhanu Prasad BA, Pal A, Wang S, et al. 155. England CG, Rui L, Cai W. Lymphoma: current status of clinical and analogs as potential agents for PET imaging of Bcr-Abl and c-KIT preclinical imaging with radiolabeled antibodies. Eur J Nucl Med Mol expression at a kinase level. Bioorg Med Chem 2014;22:623–32. Imaging 2017;44:517–32. 140. Su X, Cheng K, Liu Y, Hu X, Meng S, Cheng Z. PET imaging of insulin-like 156. Green DJ, Press OW. Whither radioimmunotherapy: to be or not to be? growth factor type 1 receptor expression with a 64Cu-labeled affibody Cancer Res 2017;77:2191–6. molecule. Amino Acids 2015;47:1409–19. 157. Levy A, Nigro G, Sansonetti PJ, Deutsch E. Candidate immune biomarkers 141. Majo VJ, Arango V, Simpson NR, Prabhakaran J, Kassir SA, Underwood MD, for radioimmunotherapy. Biochim Biophys Acta 2017;1868:58–68. et al. Synthesis and in vitro evaluation of [18F]BMS-754807: a potential PET 158. Tagawa ST, Milowsky MI, Morris M, Vallabhajosula S, Christos P, Akhtar ligand for IGF-1R. Bioorg Med Chem Lett 2013;23:4191–4. NH, et al. Phase II study of lutetium-177-labeled anti-prostate-specific 142. Kaplon H, Reichert JM. Antibodies to watch in 2018. MAbs 2018;10: membrane antigen monoclonal antibody J591 for metastatic castration- 183–203. resistant prostate cancer. Clin Cancer Res 2013;19:5182–91.

1636 Mol Cancer Ther; 17(8) August 2018 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research. PET Imaging of Receptor Tyrosine Kinases in Cancer

Weijun Wei, Dalong Ni, Emily B. Ehlerding, et al.

Mol Cancer Ther 2018;17:1625-1636.

Updated version Access the most recent version of this article at: http://mct.aacrjournals.org/content/17/8/1625

Cited articles This article cites 157 articles, 65 of which you can access for free at: http://mct.aacrjournals.org/content/17/8/1625.full#ref-list-1

Citing articles This article has been cited by 1 HighWire-hosted articles. Access the articles at: http://mct.aacrjournals.org/content/17/8/1625.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://mct.aacrjournals.org/content/17/8/1625. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mct.aacrjournals.org on October 1, 2021. © 2018 American Association for Cancer Research.