1,362
Views
6
CrossRef citations to date
0
Altmetric
REVIEW

Epidermal growth factor receptor imaging in human head and neck cancer xenografts

, , &
Pages 1263-1267 | Received 15 May 2015, Accepted 11 Jun 2015, Published online: 06 Aug 2015

ABSTRACT

Molecular imaging of specific biomarkers can have prognostic, predictive or monitoring value in head and neck squamous cell carcinoma (HNSCC). The epidermal growth factor receptor (EGFR) is involved in various radiation resistance mechanisms as it steers the pathways related to DNA damage repair, proliferation, hypoxia and apoptosis. Radiolabeled labeled F(ab’)2 fragments of the EGFR antibody cetuximab can be applied for non-invasive imaging of this receptor. Preclinical studies have shown that radioresistant tumors had a higher tracer uptake after irradiation, probably due to upregulation of membranous EGFR, thereby increasing target availability possibly as a compensation mechanism. Tumors with increased EGFR availability were also more responsive to the EGFR inhibitor cetuximab. Potentially, radionuclide imaging of the EGFR can be applied for monitoring treatment regimens in clinical practice.

Improvements have been implemented in cancer treatment over the last decennia. These include the introduction of hypoxia modification and targeting the epithelial growth factor receptor (EGFR) in combination with radiotherapy [Citation1–5]. By adaptations based on the characteristics of the tumor of the patient, treatment outcome can be improved and overall treatment-related side effects can be reduced. The heterogeneity of the patient population drives the search for tumor-specific biomarkers. This heterogeneity, as well as the observation that specific characteristics of the microenvironment are maintained when human tumors are transplanted in immune compromised mice, makes these relevant models for testing new therapies [Citation6].

In this review we will discuss recent developments of the application of radiolabeled EGFR antibodies for quantifying EGFR expression in human head and neck cancer xenografts.

Imaging biomarkers for head and neck squamous cell carcinoma treatment

Molecular imaging of specific biomarkers can have prognostic, predictive or monitoring value in head and neck squamous cell carcinoma (HNSCC) [Citation7]. A promising approach to evaluate therapy outcome is the development of imaging tracers for early response monitoring. As tumor glucose consumption, proliferation and hypoxia are often enhanced as compared to normal (adjacent) tissues, tracers targeting these features were tested as imaging biomarkers. By conducting multiple positron emission tomography (PET) scans, prior to and early after onset of treatment, molecular imaging can reveal patient-specific information on (therapy-induced) changes in the tumor microenvironment [Citation7]. A biomarker of interest is the EGFR as it steers the pathways related to DNA damage repair, proliferation, hypoxia and apoptosis in HNSCC [Citation8–11]. The chimeric antibody cetuximab binds with high affinity to the EGFR and has been used in treating HNSCC, though it remains unclear which patients will benefit from this therapy [Citation5]. Imaging with tracers produced by direct labeling of the target drugs themselves provides information on the pharmacodynamic and pharmacokinetic characteristics of the drugs [Citation12,Citation13]. Cetuximab has been labeled with several radionuclides and has been investigated preclinically in several HNSCC models [Citation14–16]. We have labeled cetuximab with 111In for SPECT-imaging, showing good tumor uptake and optimal tumor-to-background images at 3–7 days after injection in mice with subcutaneous human HNSCC FaDu tumors [Citation17]. We developed 111In-labeled F(ab’)2 fragments of cetuximab, which in preclinical models exhibit rapid clearance from the blood stream and better tumor penetration relative to the whole IgG [Citation18–20]. After intravenous injection 111In-cetuximab-F(ab’)2 specifically targets systemically accessible EGFR with an optimum tumor-to-background ratio at 24 hours after tracer injection. Furthermore, the tracer could distinguish between several HNSCC xenografts with different EGFR expression profiles and in three of four models this correlated with sensitivity to EGFR-inhibition with cetuximab [Citation9,Citation21]. Tracers based on antibody fragments have considerable renal uptake which can be reduced by co-injection of basic amino acids, (i.e. arginine and lysine) or the gelatine-based plasma expander gelofusine [Citation22–24]. In addition, when using anti-EGFR based tracers, the liver can act as a basin, which could limit tracer uptake in the tumor. Administering higher protein doses could counteract this effect, thus careful dosing of the tracer is crucial [Citation25].

Cetuximab sensitivity is not solely determined by the amount of EGFR available at the tumor cell surface. For example, the EGFR can be bypassed via signaling through ErbB family members, activating the same downstream pathways. Many ErbB family members are also frequently overexpressed in HNSCC, albeit usually to a lesser degree than EGFR [Citation26,Citation27]. Other transmembrane receptors like insuline-like growth factor receptor (IGF1R) and hepatocyte growth factor receptor (c-MET) are also coupled to the same pathways, and are two well documented sources of treatment resistance in HNSCC [Citation9]. A preclinical study reported an additive effect of blocking ERK phosphorylation (with the anti-IGF-1R antibody cixutumumab) together with cetuximab in HNSCC xenografts [Citation28]. Combination of inhibitors could potentially lead to more effective treatment of HNSCC. In addition to other membrane receptor proteins negating EGFR inhibition, the downstream pathways P13-K/AKT, RAS/ERK and STAT can be independently constitutively activated [Citation29–32]. This would omit the need for EGFR activation and result in continued tumor cell survival. For example, constitutive activation of STAT3 enables tumor growth without the requirement of growth factors [Citation33]. In vitro studies have shown that blocking of STAT3 can induce apoptosis, inhibit cell proliferation and suppress angiogenesis [Citation34]. However, STAT3 inhibition will also affect normal tissues and so far few translational studies have been reported.

For the clinic, PET scanning is preferred over SPECT scanning because of the better resolution [Citation35]. Cetuximab has been labeled with Zirconium-89 (89Zr). The relatively long half-life of the radionuclide (78.4 hour) matches with the pharmacokinetics of the IgG, and 89Zr-cetuximab is considered a potential useful tracer for the clinic [Citation15,Citation16,Citation36]. Cetuximab- F(ab’)2 labeled with the PET radionuclide copper-64 (64Cu β+ 656 keV, half-life = 12.7 hour), is in good concordance with the half-life of the radionuclide and the kinetics of the F(ab’)2 fragments in vivo. In HNSCC models UT-SCC-8 and UT-SCC-45, 64Cu-cetuximab-F(ab’)2 visualized the heterogeneously expressed EGFR and showed tumor-specific uptake, potentially making it a more effective tracer for monitoring EGFR-inhibition therapy than 89Zr-cetuximab. It should be noted that anti-EGFR antibodies like cetuximab do not cross-react with murine EGFR. Therefore, the interaction of anti-EGFR antibody-based tracers with EGFR expressed in normal tissues like the liver and skin cannot be studied in HNSCC xenograft models [Citation37].

Monitoring of HNSCC treatment

The effect of radiotherapy on EGFR expression in two HNSCC xenograft models was visualized by 111In-cetuximab-F(ab’)2 SPECT. Radioresistant tumors had a higher tracer uptake after irradiation, probably due to upregulation of membranous EGFR, thereby increasing target availability possibly as a compensation mechanism [Citation38]. Tumors with increased EGFR availability were also more responsive to the EGFR inhibitor cetuximab. Recently, similar findings were reported as a single dose of irradiation significantly increased cell-surface expression of EGFR in HNSCC [Citation39]. The hypothesis that an increase in membranous EGFR would result in an increase in cetuximab targeting, and thus inhibit downstream cell survival signaling, should be further investigated clinically. We investigated whether tumor perfusion and vessel density could have affected the radiation-induced (increase in) tumor uptake of 111In-cetuximab-F(ab’)2 [Citation38]. Tumor perfusion and vessel density were evaluated and differed between the HNSCC models. In one of the models it appeared that there was an increase in vessel density and tumor perfusion after irradiation. This could result in an increase of tracer uptake in the tumor, also mediated by the enhanced permeability and retention (EPR) effect. However, autoradiography correlated strongly with EGFR distribution as determined immunohistochemically indicating that the tracer reaches the EGFR irrespective of the (change in) tumor perfusion.

Further investigation of EGFR-inhibition treatment monitoring in addition to radiotherapy is clinically relevant. We investigated 111In-cetuximab-F(ab’)2 tumor uptake in HNSCC xenografts treated with radiotherapy and/or cetuximab. This enabled us to visualize changes in EGFR-expression in response to EGFR inhibition with cetuximab and EGFR inhibition in combination with radiotherapy [Citation40]. Surprisingly, in both HNSCC models, an increase of tracer uptake in the tumor correlated to non-response [Citation40]. However, the mechanism is yet unclear as higher 111In-cetuximab-F(ab’)2 uptake was not in all cases accompanied by an increase in membranous EGFR expression as determined after immunohistochemical staining. However, findings from immunohistochemical staining have some limits, as it is a static evaluation, inconsiderate of cellular dynamics and tumor heterogeneity. This restricts the value of EGFR staining from single biopsies and supports longitudinal EGFR monitoring with EGFR-targeting tracers.

Future prospects

A phase II clinical trial aimed to investigate the predictive value of 89Zr-cetuximab in determining treatment outcome of chemoradiation with cetuximab in HNSCC patients, but has been discontinued due to logistic complications [Citation41]. After the positive results of combining cetuximab with radiotherapy in HNSCC the combination of radiotherapy with panitumumab, a human monoclonal antibody that targets EGFR, showed no benefit [Citation5,Citation42]. More studies are needed to elucidate the role of anti-EGFR-based tracers in the management of HNSCC. Other EGFR-targeting antibodies could also have a role in predicting or monitoring HNSCC treatment. For example, panitumumab, a human anti-EGFR monoclonal antibody that does not evoke an immunological response, is approved for the treatment of patients with EGFR-expressing metastatic colorectal cancer with disease progression after chemotherapy [Citation43,Citation44]. To assess which receptors might be dominant in survival signaling, it would be interesting to compare tracers targeting different ErbB receptors in relation to treatment response monitoring. For example, as cetuximab only targets ErbB1, it could be combined with a radiolabeled tracer targeting the HER2, as like the EGFR, HER2 is known to lead to activation of the P13-K/AKT pathway [Citation45].

Conclusion

Methods to facilitate personalized therapy are widely investigated and shape the future of cancer treatment as new biomarkers are identified on genetic, molecular and metabolic levels. Translational research forms the bridge between preclinical development and clinical application, although this is not always straightforward. Many imaging biomarkers have been characterized, but only a few have reached clinical implementation, urging for a greater continuity between concept development and applicability. Future studies should exploit the benefits of radionuclide imaging in finding predictive and monitoring agents for drug efficacy and treatment regimens in order to progress to improved personalized healthcare.

Declaration of interest: Financial support: Dutch Cancer Society, grant no. NKB-KUN 2010-4688. The authors have no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Rojas A, Vojnovic B, Johns H, Joiner MC, Martindale C, Fowler JF, et al. Radiosensitisation in normal tissues with oxygen, carbogen or nicotinamide: Therapeutic gain comparisons for fractionated x-ray schedules. Radiother Oncol 1996;39:53–64.
  • Overgaard J, Hansen HS, Overgaard M, Bastholt L, Berthelsen A, Specht L, et al. A randomized double-blind phase III study of nimorazole as a hypoxic radiosensitizer of primary radiotherapy in supraglottic larynx and pharynx carcinoma. Results of the Danish Head and Neck Cancer Study DAHANCA) Protocol 5-85. Radiother Oncol 1998;46: 135–46.
  • Janssens GO, Rademakers SE, Terhaard CH, Doornaert PA, Bijl HP, van den Ende P, et al. Accelerated radiotherapy with carbogen and nicotinamide for laryngeal cancer: Results of a phase III randomized trial. J Clin Oncol 2012;30: 1777–83.
  • Milas L, Mason K, Hunter N, Petersen S, Yamakawa M, Ang K, et al. In vivo enhancement of tumor radioresponse by C225 antiepidermal growth factor receptor antibody. Clin Cancer Res 2000;6:701–8.
  • Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, et al. Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006;354: 567–78.
  • Stegeman H1, Rademakers SE, Span PN, Takes RP, van der Kogel AJ, Kaanders JH, et al. Hypoxia, metabolism, and growth factor signaling in head and neck squamous cell carcinoma: Correlation between primary and xenograft tumors. Head Neck 2014;36:1288–95.
  • Bussink J, van Herpen CM, Kaanders JH, Oyen WJ. PET-CT for response assessment and treatment adaptation in head and neck cancer. Lancet Oncol 2010;11:661–9.
  • Yewale C, Baradia D, Vhora I, Patil S, Misra A. Epidermal growth factor receptor targeting in cancer: A review of trends and strategies. Biomaterials 2013;34:8690–707.
  • Stegeman H, Kaanders JH, van der Kogel AJ, Iida M, Wheeler DL, Span PN, et al. Predictive value of hypoxia, proliferation and tyrosine kinase receptors for EGFR-inhibition and radiotherapy sensitivity in head and neck cancer models. Radiother Oncol 2013;106:383–9.
  • Bussink J, van der Kogel AJ, Kaanders JH. Activation of the PI3-K/AKT pathway and implications for radioresistance mechanisms in head and neck cancer. Lancet Oncol 2008;9:288–96.
  • van Dijk LK, Boerman OC, Kaanders JH, Bussink J. PET imaging in head and neck cancer patients to monitor treatment response: A future role for EGFR-targeted imaging. Clin Cancer Res Epub 2015 Apr 30. pii: clincanres.0348.2015.
  • Thurber GM, Schmidt MM, Wittrup KD. Factors determining antibody distribution in tumors. Trends Pharmaco Sci 2008;29:57–61.
  • Jain RK. Transport of molecules, particles, and cells in solid tumors. Ann Rev Biomed Eng 1999;1:241–63.
  • Sihver W, Pietzsch J, Krause M, Baumann M, Steinbach J, Pietzsch HJ. Radiolabeled cetuximab conjugates for EGFR targeted cancer diagnostics and therapy. Pharmaceuticals Basel) 2014;7:311–38.
  • Perk LR, Visser GW, Vosjan MJ, Stigter-van Walsum M, Tijink BM, Leemans CR, et al.89)Zr as a PET surrogate radioisotope for scouting biodistribution of the therapeutic radiometals 90)Y and 177)Lu in tumor-bearing nude mice after coupling to the internalizing antibody cetuximab. J Nucl Med 2005;46:1898–906.
  • Cai W, Chen K, He L, Cao Q, Koong A, Chen X. Quantitative PET of EGFR expression in xenograft-bearing mice using 64Cu-labeled cetuximab, a chimeric anti-EGFR monoclonal antibody. Eur J Nucl Med Mol Imaging 2007;34: 850–8.
  • Hoeben BA, Molkenboer-Kuenen JD, Oyen WJ, Peeters WJ, Kaanders JH, Bussink J, et al. Radiolabeled cetuximab: dose optimization for epidermal growth factor receptor imaging in a head-and-neck squamous cell carcinoma model. Int J Cancer 2011;129:870–8.
  • van Dijk LK, Hoeben BA, Kaanders JH, Franssen GM, Boerman OC, Bussink J. Imaging of epidermal growth factor receptor expression in head and neck cancer with SPECT/CT and 111In-labeled cetuximab-F (ab’)2. J Nucl Med 2013;54:2118–24.
  • Lofblom J, Feldwisch J, Tolmachev V, Carlsson J, Stahl S, Frejd FY. Affibody molecules: Engineered proteins for therapeutic, diagnostic and biotechnological applications. FEBS Letters 2010;584:2670–80.
  • Wu AM. Engineered antibodies for molecular imaging of cancer. Methods 2014;65:139–47.
  • van Dijk LK, Hoeben BA, Stegeman H, Kaanders JH, Franssen GM, Boerman OC, et al. 111In-cetuximab-F (ab’)2 SPECT imaging for quantification of accessible epidermal growth factor receptors EGFR) in HNSCC xenografts. Radiother Oncol 2013;108:484–8.
  • Gainkam LO, Caveliers V, Devoogdt N, Vanhove C, Xavier C, Boerman O, et al. Localization, mechanism and reduction of renal retention of technetium-99m labeled epidermal growth factor receptor-specific nanobody in mice. Contrast Media Mol Imaging 2011;6:85–92.
  • Garkavij M, Samarzija M, Ewers SB, Jakopovic M, Tezak S, Tennvall J. Concurrent radiotherapy and tumor targeting with 111In-HMFG1-F (ab’)2 in patients with MUC1- positive non-small cell lung cancer. Anticancer Res 2005;25: 4663–71.
  • Kobayashi H, Yoo TM, Kim IS, Kim MK, Le N, Webber KO, et al. L-lysine effectively blocks renal uptake of 125I- or 99mTc-labeled anti-Tac disulfide-stabilized Fv fragment. Cancer Res 1996;56:3788–95.
  • Kareem H, Sandstrom K, Elia R, Gedda L, Anniko M, Lundqvist H, et al. Blocking EGFR in the liver improves the tumor-to-liver uptake ratio of radiolabeled EGF. Tumour Biol 2010;31:79–87.
  • Bei R, Budillon A, Masuelli L, Cereda V, Vitolo D, Di Gennaro E, et al. Frequent overexpression of multiple ErbB receptors by head and neck squamous cell carcinoma contrasts with rare antibody immunity in patients. J Pathol 2004;204:317–25.
  • Bei R, Pompa G, Vitolo D, Moriconi E, Ciocci L, Quaranta M, et al. Co-localization of multiple ErbB receptors in stratified epithelium of oral squamous cell carcinoma. J Pathol 2001;195:343–8.
  • Mehra R, Serebriiskii IG, Dunbrack RL, Jr., Robinson MK, Burtness B, Golemis EA. Protein-intrinsic and signaling network-based sources of resistance to EGFR- and ErbB family-targeted therapies in head and neck cancer. Drug Resist Updat 2011;14:260–79.
  • Riesterer O, Yang Q, Raju U, Torres M, Molkentine D, Patel N, et al. Combination of anti-IGF-1R antibody A12 and ionizing radiation in upper respiratory tract cancers. Int J Radiat Oncol Biol Phys 2011;79:1179–87.
  • Yamatodani T, Ekblad L, Kjellen E, Johnsson A, Mineta H, Wennerberg J. Epidermal growth factor receptor status and persistent activation of Akt and p44/42 MAPK pathways correlate with the effect of cetuximab in head and neck and colon cancer cell lines. J Cancer Research Clin Oncol 2009; 135:395–402.
  • Janmaat ML, Kruyt FA, Rodriguez JA, Giaccone G. Response to epidermal growth factor receptor inhibitors in non-small cell lung cancer cells: Limited antiproliferative effects and absence of apoptosis associated with persistent activity of extracellular signal-regulated kinase or Akt kinase pathways. Clin Cancer Res 2003;9:2316–26.
  • Yoshida T, Okamoto I, Okabe T, Iwasa T, Satoh T, Nishio K, et al. Matuzumab and cetuximab activate the epidermal growth factor receptor but fail to trigger downstream signaling by Akt or Erk. Int J Cancer 2008;122:1530–8.
  • Kijima T, Niwa H, Steinman RA, Drenning SD, Gooding WE, Wentzel AL, et al. STAT3 activation abrogates growth factor dependence and contributes to head and neck squamous cell carcinoma tumor growth in vivo. Differentiation 2002;13:355–62.
  • Siveen KS, Sikka S, Surana R, Dai X, Zhang J, Kumar AP, et al. Targeting the STAT3 signaling pathway in cancer: Role of synthetic and natural inhibitors. Biochim Biophys Acta 2014;1845:136–54.
  • Histed SN, Lindenberg ML, Mena E, Turkbey B, Choyke PL, Kurdziel KA. Review of functional/anatomical imaging in oncology. Nuc Med Commun 2012;33:349–61.
  • Zhang Y, Hong H, Cai W. PET tracers based on Zirconium-89. Curr Radiopharm 2011;4:131–9.
  • Van Dijk LK, Yim C-B, Boerman OC, Franssen GM, Kaanders JHAM, Rajander J, Solin O, Gronroos TJ, Bussink J .PET of EGFR with 64Cu-cetuximab-F(ab’)2 in mice with head and neck squamous cell carcinoma xenografts In: Contrast Media and Molecular Imaging. DOI: 10.1002/cmmi.1659..
  • van Dijk LK, Boerman OC, Franssen GM, Lok J, Kaanders JH, Bussink J. Early response monitoring with 18F-FDG PET and cetuximab-F (ab’)2-SPECT after radiotherapy of human head and neck squamous cell carcinomas in a mouse model. J Nucl Med 2014;55: 1665–70.
  • Wattenberg MM, Kwilas AR, Gameiro SR, Dicker AP, Hodge JW. Expanding the use of monoclonal antibody therapy of cancer by using ionising radiation to upregulate antibody targets. Br J Cancer 2014;110:1472–80.
  • van Dijk LK, Boerman OC, Franssen GM, Kaanders JH, Bussink J. 111In-cetuximab-F (ab’)2 SPECT and 18F-FDG PET for prediction and response monitoring of combined-modality treatment of human head and neck carcinomas in a mouse model. J Nucl Med 2015;56: 287–92.
  • Heukelom J, Hamming O, Bartelink H, Hoebers F, Giralt J, Herlestam T, et al. Adaptive and innovative Radiation Treatment FOR improving Cancer treatment outcomE ARTFORCE); a randomized controlled phase II trial for individualized treatment of head and neck cancer. BMC Cancer 2013;13:84.
  • Mesía R, Henke M, Fortin A, Minn H, Yunes Ancona AC, Cmelak A, et al. Chemoradiotherapy with or without panitumumab in patients with unresected, locally advanced squamous-cell carcinoma of the head and neck CONCERT-1): A randomised, controlled, open-label phase 2 trial. Lancet Oncol 2015;16:208–20.
  • Poulin-Costello M, Azoulay L, Van Cutsem E, Peeters M, Siena S, Wolf M. An analysis of the treatment effect of panitumumab on overall survival from a phase 3, randomized, controlled, multicenter trial 20020408) in patients with chemotherapy refractory metastatic colorectal cancer. Targeted Oncol 2013;8:127–36.
  • Voigt M, Braig F, Gothel M, Schulte A, Lamszus K, Bokemeyer C, et al. Functional dissection of the epidermal growth factor receptor epitopes targeted by panitumumab and cetuximab. Neoplasia 2012;14:1023–151.
  • Baselga J, Swain SM. Novel anticancer targets: Revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 2009;9:463–75.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.