First-in-human uPAR PET: Imaging of Cancer Aggressiveness

Morten Persson, Dorthe Skovgaard, Malene Brandt-Larsen, Camilla Christensen, Jacob Madsen, Carsten H Nielsen, Tine Thurison, Thomas Levin Klausen, Søren Holm, Annika Loft, Anne Kiil Berthelsen, Michael Ploug, Helle Pappot, Klaus Brasso, Niels Kroman, Liselotte Højgaard, Andreas Kjaer, Morten Persson, Dorthe Skovgaard, Malene Brandt-Larsen, Camilla Christensen, Jacob Madsen, Carsten H Nielsen, Tine Thurison, Thomas Levin Klausen, Søren Holm, Annika Loft, Anne Kiil Berthelsen, Michael Ploug, Helle Pappot, Klaus Brasso, Niels Kroman, Liselotte Højgaard, Andreas Kjaer

Abstract

A first-in-human clinical trial with Positron Emission Tomography (PET) imaging of the urokinase-type plasminogen activator receptor (uPAR) in patients with breast, prostate and bladder cancer, is described. uPAR is expressed in many types of human cancers and the expression is predictive of invasion, metastasis and indicates poor prognosis. uPAR PET imaging therefore holds promise to be a new and innovative method for improved cancer diagnosis, staging and individual risk stratification. The uPAR specific peptide AE105 was conjugated to the macrocyclic chelator DOTA and labeled with (64)Cu for targeted molecular imaging with PET. The safety, pharmacokinetic, biodistribution profile and radiation dosimetry after a single intravenous dose of (64)Cu-DOTA-AE105 were assessed by serial PET and computed tomography (CT) in 4 prostate, 3 breast and 3 bladder cancer patients. Safety assessment with laboratory blood screening tests was performed before and after PET ligand injection. In a subgroup of the patients, the in vivo stability of our targeted PET ligand was determined in collected blood and urine. No adverse or clinically detectable side effects in any of the 10 patients were found. The ligand exhibited good in vivo stability and fast clearance from plasma and tissue compartments by renal excretion. In addition, high uptake in both primary tumor lesions and lymph node metastases was seen and paralleled high uPAR expression in excised tumor tissue. Overall, this first-in-human study therefore provides promising evidence for safe use of (64)Cu-DOTA-AE105 for uPAR PET imaging in cancer patients.

Keywords: PET; clinical trial; uPAR.

Conflict of interest statement

Competing interests: MPE, JM & AK are inventor of a patent on the composition of matter of uPAR PET. MPE, JM, CHN & AK are co-founders of a start-up company (Curasight) that has licensed the uPAR PET patent and is currently raising funds to commercialize uPAR PET technology. Pending patent for MPE, JM & AK: Positron Emitting Radionuclides Labeled Peptides for Human uPAR PET Imaging (WO 2014/086364 A1). The authors declare that their spouses, partners, or children have no financial relationships relevant to the submitted work. Inquiries should be directed to the corresponding author.

Figures

Fig 1
Fig 1
uPAR PET imaging and overview of first-in-human uPAR PET study design. (A) Schematic of the uPAR PET ligand 64Cu-DOTA-AE105 showing the chemical structure, a chromatogram of the final product, a transverse PET/CT image from a prostate cancer patient with tumor uptake of 64Cu-DOTA-AE105 and a Pymol visualization of uPAR (surface representation) in complex with the targeting peptide shown as a cartoon representation. (B) Clinical trial events after single dose injection of 64Cu-DOTA-AE105. Timeline denotes injection, acquisition of serial PET/CT imaging, and collection of blood and tissue specimens. (C) Patient characteristics.
Fig 2
Fig 2
Whole-body distribution and PK of 64Cu-DOTA-AE105. (A) Maximum intensity projection PET images at 1, 3 and 24 hours following injection of 64Cu-DOTA-AE105 (patient 10). The highest accumulation of activity was in the liver, bowel and bladder. (B) Decay corrected SUV values in blood and major organs plotted individually for n = 10 patients. For each patient ROIs were drawn on selected organ/tissue of interest at all three consecutive PET scans.
Fig 3
Fig 3
Metabolic analysis of 64Cu-DOTA-AE105 in plasma and urine. Data are shown for patient 10. (A) Relative time-dependent activity concentrations in plasma. Plasma half life was estimated to 8.5 min. (B) Chromatograms of 64Cu-DOTA-AE105 in plasma. Data are shown for standard (Rt=11.8 min) and for the time points: 1, 10, 30 and 120 minutes. A single metabolite was formed (Rt=11.1 min) Vertical lines discriminate peaks corresponding to the uPAR PET ligand 64Cu-DOTA-AE105. (C) Urine collection data. Time-dependent excretion of activity (top) and accumulated activity (bottom) are displayed. (D) Chromatograms of 64Cu-DOTA-AE105 in urine. Data are shown for standard (Rt=11.8 min) and for the time points 20, 30 and 50 minutes. A single metabolite was found (Rt=11.1 min), corresponding to the single metabolite formed in plasma.
Fig 4
Fig 4
Dosimetry and whole-body PET/CT imaging of 64Cu-DOTA-AE105. (A) Mean absorbed dose per unit administrated (mGy/MBq) of major organs and tissues (table S4) were derived from 6 patients (patient 3,4,5,6,8 and 10) from serial whole-body PET scans acquired over 24 hours time interval following injection of 64Cu-DOTA-AE105 using VOI-based time activity data (table S5). (B). Coronal whole-body PET/CT images (patient 5) show time dependent biodistribution at 1,3 and 24 hours and demonstrates accumulation of activity mainly in the liver and bowel.
Fig 5
Fig 5
uPAR PET imaging in bladder cancer. (A) Representative transverse CT, PET and co-registered PET/CT images of a primary tumor lesion (blue circle), top images, with intense uptake of 64Cu-DOTA-AE105 (patient 7). (B) Bottom images show a uPAR positive inguinal lymph node metastasis (blue arrow) with high uptake (patient 1). (C) Tumor-to-liver, tumor-to-blood, tumor-to-kidney and tumor-to-muscle ratios as a function of time post injection. Data are averages ± SD (n = 1 ROI per time point, n = 3 patients).
Fig 6
Fig 6
uPAR PET imaging in breast cancer. (A) Representative transverse CT, PET and co-registered PET/CT images of a primary tumor lesion (blue arrow), top images, with intense uptake of 64Cu-DOTA-AE105 (patient 8). (B) Bottom images show an uPAR positive axillary lymph node metastasis (blue arrow) with high uptake in the same patient. (C) Tumor-to-liver, tumor-to-blood, tumor-to-kidney and tumor-to-muscle ratios as a function of time post injection. Data are averages ± SD (n = 1 ROI per time point, n = 3 patients).
Fig 7
Fig 7
uPAR PET imaging in prostate cancer. (A) Representative transverse CT, PET and co-registered PET/CT images of a primary tumor lesion (blue arrow), top images, with high uptake of 64Cu-DOTA-AE105 (patient 4). (B) Bottom images show an uPAR positive regional lymph node metastasis (blue arrow) with high uptake (patient 9). (C) Tumor-to-liver, tumor-to-blood, tumor-to-kidney and tumor-to-muscle ratios as a function of time post injection. Data are averages ± SD (n = 1 ROI per time point, n = 3 patients).

References

    1. Gambhir SS. Molecular imaging of cancer with positron emission tomography. Nat Rev Cancer. 2002;2(9):683–93.
    1. Kayani I, Groves AM. 18F-fluorodeoxyglucose PET/CT in cancer imaging. Clinical medicine. 2006;6(3):240–4.
    1. Rohren EM, Turkington TG, Coleman RE. Clinical applications of PET in oncology. Radiology. 2004;231(2):305–32.
    1. Weber WA, Czernin J, Phelps ME, Herschman HR. Technology Insight: novel imaging of molecular targets is an emerging area crucial to the development of targeted drugs. Nature clinical practice Oncology. 2008;5(1):44–54.
    1. Persson M KA. Urokinase-type plasminogen activator receptor (uPAR) as a promising new imaging target: potential clinical applications. Clinical Physiology and Functional Imaging; 2013.
    1. Jacobsen B, Ploug M. The urokinase receptor and its structural homologue C4.4A in human cancer: expression, prognosis and pharmacological inhibition. Curr Med Chem. 2008;15(25):2559–73.
    1. Dass K, Ahmad A, Azmi AS, Sarkar SH, Sarkar FH. Evolving role of uPA/uPAR system in human cancers. Cancer Treat Rev. 2008;34(2):122–36.
    1. Dano K, Behrendt N, Hoyer-Hansen G, Johnsen M, Lund LR, Ploug M, Romer J. Plasminogen activation and cancer. Thromb Haemost. 2005;93(4):676–81.
    1. Ganesh S, Sier CF, Heerding MM, Griffioen G, Lamers CB, Verspaget HW. Urokinase receptor and colorectal cancer survival. Lancet. 1994;344(8919):401–2.
    1. Foekens JA, Peters HA, Look MP, Portengen H, Schmitt M, Kramer MD, Brunner N, Janicke F, Meijer-van Gelder ME, Henzen-Logmans SC. et al. The urokinase system of plasminogen activation and prognosis in 2780 breast cancer patients. Cancer Res. 2000;60(3):636–43.
    1. Riisbro R, Christensen IJ, Piironen T, Greenall M, Larsen B, Stephens RW, Han C, Hoyer-Hansen G, Smith K, Brunner N. et al. Prognostic significance of soluble urokinase plasminogen activator receptor in serum and cytosol of tumor tissue from patients with primary breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2002;8(5):1132–41.
    1. Pappot H, Hoyer-Hansen G, Ronne E, Hansen HH, Brunner N, Dano K, Grondahl-Hansen J. Elevated plasma levels of urokinase plasminogen activator receptor in non-small cell lung cancer patients. Eur J Cancer. 1997;33(6):867–72.
    1. Pappot H, Pfeiffer P, Grondahlhansen J, Skov B. Presence of urokinase plasminogen activator, its inhibitor and receptor in small cell lung cancer and non-small cell lung cancer. Int J Oncol. 1997;10(1):177–82.
    1. Pyke C, Ralfkiaer E, Ronne E, Hoyer-Hansen G, Kirkeby L, Dano K. Immunohistochemical detection of the receptor for urokinase plasminogen activator in human colon cancer. Histopathology. 1994;24(2):131–8.
    1. Almasi CE, Brasso K, Iversen P, Pappot H, Hoyer-Hansen G, Dano K, Christensen IJ. Prognostic and predictive value of intact and cleaved forms of the urokinase plasminogen activator receptor in metastatic prostate cancer. Prostate. 2011;71(8):899–907.
    1. Shariat SF, Roehrborn CG, McConnell JD, Park S, Alam N, Wheeler TM, Slawin KM. Association of the circulating levels of the urokinase system of plasminogen activation with the presence of prostate cancer and invasion, progression, and metastasis. J Clin Oncol. 2007;25(4):349–55.
    1. Miyake H, Hara I, Yamanaka K, Gohji K, Arakawa S, Kamidono S. Elevation of serum levels of urokinase-type plasminogen activator and its receptor is associated with disease progression and prognosis in patients with prostate cancer. Prostate. 1999;39(2):123–9.
    1. Dohn LH, Illemann M, Hoyer-Hansen G, Christensen IJ, Hostmark J, Litlekalsoy J, von der Maase H, Pappot H, Laerum OD. Urokinase-type plasminogen activator receptor (uPAR) expression is associated with T-stage and survival in urothelial carcinoma of the bladder. Urologic oncology; 2015.
    1. Kriegbaum MC, Persson M, Haldager L, Alpizar-Alpizar W, Jacobsen B, Gardsvoll H, Kjaer A, Ploug M. Rational targeting of the urokinase receptor (uPAR): development of antagonists and non-invasive imaging probes. Curr Drug Targets. 2011;12(12):1711–28.
    1. Li D, Liu S, Shan H, Conti P, Li Z. Urokinase plasminogen activator receptor (uPAR) targeted nuclear imaging and radionuclide therapy. Theranostics. 2013;3(7):507–15.
    1. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70.
    1. Ploug M, Ostergaard S, Gardsvoll H, Kovalski K, Holst-Hansen C, Holm A, Ossowski L, Dano K. Peptide-derived antagonists of the urokinase receptor. affinity maturation by combinatorial chemistry, identification of functional epitopes, and inhibitory effect on cancer cell intravasation. Biochemistry. 2001;40(40):12157–68.
    1. Li ZB, Niu G, Wang H, He L, Yang L, Ploug M, Chen X. Imaging of urokinase-type plasminogen activator receptor expression using a 64Cu-labeled linear peptide antagonist by microPET. Clinical cancer research: an official journal of the American Association for Cancer Research. 2008;14(15):4758–66.
    1. Persson M, Madsen J, Ostergaard S, Jensen MM, Jorgensen JT, Juhl K, Lehmann C, Ploug M, Kjaer A. Quantitative PET of human urokinase-type plasminogen activator receptor with 64Cu-DOTA-AE105: implications for visualizing cancer invasion. J Nucl Med. 2012;53(1):138–45.
    1. Persson M HM, Madsen J, Jørgensen TJD, Jensen KJ, Kjaer A, Ploug M. Improved PET Imaging of uPAR Expression Using new 64Cu-labeled Cross-Bridged Peptide Ligands: Comparative in vitro and in vivo Studies. Theranostics. 2013;3(9):618–32.
    1. Persson M, El Ali HH, Binderup T, Pfeifer A, Madsen J, Rasmussen P, Kjaer A. Dosimetry of 64Cu-DOTA-AE105, a PET tracer for uPAR imaging. Nucl Med Biol. 2014;41(3):290–5.
    1. Agency EM. Guideline EMA/CPMP/ICH/286/1995. EMA; 2009.
    1. Dano K, Andreasen PA, Grondahl-Hansen J, Kristensen P, Nielsen LS, Skriver L. Plasminogen activators, tissue degradation, and cancer. Advances in cancer research. 1985;44:139–266.
    1. Yang Y, Adelstein SJ, Kassis AI. General approach to identifying potential targets for cancer imaging by integrated bioinformatics analysis of publicly available genomic profiles. Mol Imaging. 2011;10(2):123–34.
    1. Sah BR, Burger IA, Schibli R, Friebe M, Dinkelborg L, Graham K, Borkowski S, Bacher-Stier C, Valencia R, Srinivasan A. et al. Dosimetry and First Clinical Evaluation of the New 18F-Radiolabeled Bombesin Analogue BAY 864367 in Patients with Prostate Cancer. J Nucl Med. 2015;56(3):372–8.
    1. Deloar HM, Fujiwara T, Shidahara M, Nakamura T, Watabe H, Narita Y, Itoh M, Miyake M, Watanuki S. Estimation of absorbed dose for 2-[F-18]fluoro-2-deoxy-D-glucose using whole-body positron emission tomography and magnetic resonance imaging. European journal of nuclear medicine. 1998;25(6):565–74.
    1. Deloar HM, Fujiwara T, Shidahara M, Nakamura T, Yamadera A, Itoh M. Internal absorbed dose estimation by a TLD method for 18F-FDG and comparison with the dose estimates from whole body PET. Physics in medicine and biology. 1999;44(2):595–606.
    1. Anderson CJ, Dehdashti F, Cutler PD, Schwarz SW, Laforest R, Bass LA, Lewis JS, McCarthy DW. 64Cu-TETA-octreotide as a PET imaging agent for patients with neuroendocrine tumors. J Nucl Med. 2001;42(2):213–21.
    1. Lewis JS, Laforest R, Dehdashti F, Grigsby PW, Welch MJ, Siegel BA. An imaging comparison of 64Cu-ATSM and 60Cu-ATSM in cancer of the uterine cervix. J Nucl Med. 2008;49(7):1177–82.
    1. Pfeifer A, Knigge U, Mortensen J, Oturai P, Berthelsen AK, Loft A, Binderup T, Rasmussen P, Elema D, Klausen TL. et al. Clinical PET of Neuroendocrine Tumors Using 64Cu-DOTATATE: First-in-Humans Study. J Nucl Med. 2012;53(8):1207–15.
    1. Tamura K, Kurihara H, Yonemori K, Tsuda H, Suzuki J, Kono Y, Honda N, Kodaira M, Yamamoto H, Yunokawa M. et al. 64Cu-DOTA-trastuzumab PET imaging in patients with HER2-positive breast cancer. J Nucl Med. 2013;54(11):1869–75.
    1. Wieser G, Mansi R, Grosu AL, Schultze-Seemann W, Dumont-Walter RA, Meyer PT, Maecke HR, Reubi JC, Weber WA. Positron emission tomography (PET) imaging of prostate cancer with a gastrin releasing peptide receptor antagonist--from mice to men. Theranostics. 2014;4(4):412–9.
    1. Boswell CA, Sun X, Niu W, Weisman GR, Wong EH, Rheingold AL, Anderson CJ. Comparative in vivo stability of copper-64-labeled cross-bridged and conventional tetraazamacrocyclic complexes. J Med Chem. 2004;47(6):1465–74.
    1. Jones-Wilson TM, Deal KA, Anderson CJ, McCarthy DW, Kovacs Z, Motekaitis RJ, Sherry AD, Martell AE, Welch MJ. The in vivo behavior of copper-64-labeled azamacrocyclic complexes. Nucl Med Biol. 1998;25(6):523–30.
    1. Ferdani R, Stigers DJ, Fiamengo AL, Wei L, Li BT, Golen JA, Rheingold AL, Weisman GR, Wong EH, Anderson CJ. Synthesis, Cu(II) complexation, 64Cu-labeling and biological evaluation of cross-bridged cyclam chelators with phosphonate pendant arms. Dalton Trans. 2012;41(7):1938–50.
    1. Stigers DJ, Ferdani R, Weisman GR, Wong EH, Anderson CJ, Golen JA, Moore C, Rheingold AL. A new phosphonate pendant-armed cross-bridged tetraamine chelator accelerates copper(ii) binding for radiopharmaceutical applications. Dalton Trans. 2010;39(7):1699–701.
    1. Anderson CJ, Wadas TJ, Wong EH, Weisman GR. Cross-bridged macrocyclic chelators for stable complexation of copper radionuclides for PET imaging. Q J Nucl Med Mol Imaging. 2008;52(2):185–92.
    1. Sprague JE, Peng Y, Fiamengo AL, Woodin KS, Southwick EA, Weisman GR, Wong EH, Golen JA, Rheingold AL, Anderson CJ. Synthesis, characterization and in vivo studies of Cu(II)-64-labeled cross-bridged tetraazamacrocycle-amide complexes as models of peptide conjugate imaging agents. J Med Chem. 2007;50(10):2527–35.
    1. Wong EH, Weisman GR, Hill DC, Reed DP, Rogers ME, Condon JS, Fagan MA, Calabrese JC, Lam KC, Guzei IA. et al. Synthesis and characterization of cross-bridged cyclams and pendant-armed derivatives and structural studies of their copper(II) complexes. J Am Chem Soc. 2000;122(43):10561–72.
    1. Guo Y, Ferdani R, Anderson CJ. Preparation and biological evaluation of (64)cu labeled tyr(3)-octreotate using a phosphonic Acid-based cross-bridged macrocyclic chelator. Bioconjug Chem. 2012;23(7):1470–7.
    1. Wei L, Ye Y, Wadas TJ, Lewis JS, Welch MJ, Achilefu S, Anderson CJ. (64)Cu-labeled CB-TE2A and diamsar-conjugated RGD peptide analogs for targeting angiogenesis: comparison of their biological activity. Nucl Med Biol. 2009;36(3):277–85.
    1. Wadas TJ, Eiblmaier M, Zheleznyak A, Sherman CD, Ferdani R, Liang K, Achilefu S, Anderson CJ. Preparation and biological evaluation of 64Cu-CB-TE2A-sst2-ANT, a somatostatin antagonist for PET imaging of somatostatin receptor-positive tumors. J Nucl Med. 2008;49(11):1819–27.
    1. Szekanecz Z, Haines GK, Koch AE. Differential expression of the urokinase receptor (CD87) in arthritic and normal synovial tissues. Journal of clinical pathology. 1997;50(4):314–9.
    1. Ronday HK, Smits HH, Van Muijen GN, Pruszczynski MS, Dolhain RJ, Van Langelaan EJ, Breedveld FC, Verheijen JH. Difference in expression of the plasminogen activation system in synovial tissue of patients with rheumatoid arthritis and osteoarthritis. British journal of rheumatology. 1996;35(5):416–23.
    1. Fuhrman B. The urokinase system in the pathogenesis of atherosclerosis. Atherosclerosis. 2012;222(1):8–14.
    1. Svensson PA, Olson FJ, Hagg DA, Ryndel M, Wiklund O, Karlstrom L, Hulthe J, Carlsson LM, Fagerberg B. Urokinase-type plasminogen activator receptor is associated with macrophages and plaque rupture in symptomatic carotid atherosclerosis. International journal of molecular medicine. 2008;22(4):459–64.
    1. Cermik TF, Mavi A, Basu S, Alavi A. Impact of FDG PET on the preoperative staging of newly diagnosed breast cancer. Eur J Nucl Med Mol Imaging. 2008;35(3):475–83.
    1. Siggelkow W, Rath W, Buell U, Zimny M. FDG PET and tumour markers in the diagnosis of recurrent and metastatic breast cancer. Eur J Nucl Med Mol Imaging. 2004;31(Suppl 1):S118–24.
    1. Loeb S, Bjurlin MA, Nicholson J, Tammela TL, Penson DF, Carter HB, Carroll P, Etzioni R. Overdiagnosis and overtreatment of prostate cancer. Eur Urol. 2014;65(6):1046–55.
    1. Hansen HD, Ettrup A, Herth MM, Dyssegaard A, Ratner C, Gillings N, Knudsen GM. Direct comparison of [(18) F] and [(18) F] altanserin for 5-HT2A receptor imaging with PET. Synapse. 2013;67(6):328–37.
    1. Gillings N. A restricted access material for rapid analysis of [(11)C]-labeled radiopharmaceuticals and their metabolites in plasma. Nucl Med Biol. 2009;36(8):961–5.
    1. Thurison T, Christensen IJ, Lund IK, Nielsen HJ, Hoyer-Hansen G. Circulating intact and cleaved forms of the urokinase-type plasminogen activator receptor: biological variation, reference intervals and clinical useful cut-points. Clin Chim Acta. 2015;439:84–90.

Source: PubMed

3
Suscribir