Molecular imaging of nonsmall cell lung carcinomas expressing active mutant EGFR kinase using PET with [(124)i]-morpholino-IPQA

Skye Hsin-Hsien Yeh, Chien-Feng Lin, Fan-Lin Kong, Hsin-Ell Wang, Ya-Ju Hsieh, Juri G Gelovani, Ren-Shyan Liu, Skye Hsin-Hsien Yeh, Chien-Feng Lin, Fan-Lin Kong, Hsin-Ell Wang, Ya-Ju Hsieh, Juri G Gelovani, Ren-Shyan Liu

Abstract

Mutations in the kinase domain of epidermal growth factor receptor (EGFR) have high levels of basal receptor phosphorylation and are associated with clinical responsiveness to Iressa in patients with nonsmall cell lung cancer (NSCLC). This study aimed to assess the feasibility of morpholino-[(124)I]IPQA derivative as an in vivo PET imaging tool for the expression of different EGFR mutants in NSCLC. In vitro radiotracer accumulation and washout studies demonstrated a rapid accumulation and progressive retention after washout of morpholino-[(131)I]IPQA derivative in high EGFR-expressing H1299 NSCLC derivative cell lines (L858R and E746-A750 del cell lines), but not in EGFR-transfected H1299 cell line and vector-transfected H1299 cell line. Using the morpholino-[(124)I]IPQA derivative, we obtained noninvasive microPET images of EGFR activity in L858R and E746-A750 del subcutaneous tumor xenografts, but not in subcutaneous tumor xenografts grown form control cell line. Different EGFR mutant (activity) tumors have a different morpholino-[ (∗) I]IPQA derivative uptake. However, it still needs to modify the structure of IPQA to increase its water solubility and reduce hepatobiliary clearance. Morpholino-[(124)I]IPQA derivative may be a potential probe for selection of the candidate patients suffering from NSCLC for the small molecule tyrosine kinase inhibitor therapy (e.g., Iressa) in the future.

Figures

Figure 1
Figure 1
The chemical structure of the (E)-But2-enedioic acid [4-(3-[124I]iodoanilino)-quinazolin-6-yl]-amide-(3-morpholin-4-ylpropyl)-amide, termed as morpholino-[124I]IPQA.
Figure 2
Figure 2
Assessment of irreversible binding of morpholino-[131I]IPQA to wild-type and mutant EGFR kinase in four types of human nonsmall cell lung carcinoma cell lines. (a) The autoradiography demonstrates the irreversible and covalent bindings of morpholino-[131I]IPQA to the EGFR kinase domain in different cell lines. (b) The same membrane was stained with anti-EGFR kinase antibody. A single band of radiolabeled protein corresponds to the predominate band of ~170 kDa.
Figure 3
Figure 3
In vitro uptake and washout phases of morpholino-[131I]IPQA in L858R (a), E746-A750 del (b), wild-type EGFR-transfected (c), and EGFR-vector (d). Liner fits of radioactivity accumulation time points show the washout rate at short duration (20–80 minutes, blue dot line) and long duration (60–120 minutes, red dot line) after initial accumulation (from (e) to (g)). Coefficient B (slope) of the exponential washout = A × exp⁡(−BX). Panels (a)–(d) are shown in full scale, and panels (e)–(g) are from the same data in panels (a)–(c) but are shown in smaller scale to visualize differences in washout results.
Figure 4
Figure 4
Reprehensive coronal and axial PET images obtained 24 hours after morpholino-[124I]IPQA administration in mice bearing L858R EGFR (a), E746-A750 del (b), wild-type EGFR-transfected (c), and EGFR-vector ((a)–(c)) subcutaneous tumor xenografts (dot circle). Color coding in the image is set to maximize the visualization of tumor in each projection. The different scale was used in (c). L: L858R EGFR; V: vector; D: E746-A750 del; E: EGFR-vector.
Figure 5
Figure 5
Quantification of accumulation of [124I]IPQA in different tumor xenografts at baseline (blue) and pretreated with Iressa (red), respectively. Error bars represent standard deviation; statically significant difference is indicated by an asterisk (*P < 0.05) or double asterisks (**P < 0.01).

References

    1. ACS. 2012, .
    1. Burris HA., III Shortcomings of current therapies for non-small-cell lung cancer: unmet medical needs. Oncogene. 2009;28(1):S4–S13.
    1. Yarden Y. The EGFR family and its ligands in human cancer: signalling mechanisms and therapeutic opportunities. European Journal of Cancer. 2001;37(4):S3–S8.
    1. Gazdar AF. Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene. 2009;28(1):S24–S31.
    1. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nature Reviews Cancer. 2007;7(3):169–181.
    1. Reade CA, Ganti AK. EGFR targeted therapy in non-small cell lung cancer: potential role of cetuximab. Biologics. 2009;3:215–224.
    1. Fukuoka F. Multi-institutional randomized phase II trial of gefitinib for previously treated patients with advanced non-small-cell lung cancer (The IDEAL 1 Trial) Journal of Clinical Oncology. 2003;21(12):2237–2246.
    1. Kris MG, Natale RB, Herbst RS, et al. Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase, in symptomatic patients with non-small cell lung cancer: a randomized trial. Journal of the American Medical Association. 2003;290(16):2149–2158.
    1. Paez JG, Jänne PA, Lee JC, et al. EGFR mutations in lung, cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304(5676):1497–1500.
    1. Huang S-F, Liu H-P, Li L-H, et al. High frequency of epidermal growth factor receptor mutations with complex patterns in non-small cell lung cancers related to gefitinib responsiveness in Taiwan. Clinical Cancer Research. 2004;10(24):8195–8203.
    1. Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best supportive care in previously treated patients with refractory advanced non-small-cell lung cancer: results from a randomised, placebo-controlled, multicentre study (Iressa Survival Evaluation in Lung Cancer) The Lancet. 2005;366(9496):1527–1537.
    1. Shepherd FA, Tsao M-S. Unraveling the mystery of prognostic and predictive factors in epidermal growth factor receptor therapy. Journal of Clinical Oncology. 2006;24(7):1219–1220.
    1. Riely GJ, Pao W, Pham D, et al. Clinical course of patients with non-small cell lung cancer and epidermal growth factor receptor exon 19 and exon 21 mutations treated with gefitinib or erlotinib. Clinical Cancer Research. 2006;12(3, part 1):839–844.
    1. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. The New England Journal of Medicine. 2004;350(21):2129–2139.
    1. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Medicine. 2005;2(3, article e73)
    1. Jackman DM, Yeap BY, Sequist LV, et al. Exon 19 deletion mutations of epidermal growth factor receptor are associated with prolonged survival in non-small cell lung cancer patients treated with gefitinib or erlotinib. Clinical Cancer Research. 2006;12(13):3908–3914.
    1. Riely GJ, Politi KA, Miller VA, Pao W. Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clinical Cancer Research. 2006;12(24):7232–7241.
    1. Giaccone G. Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer. Journal of Clinical Oncology. 2005;23(14):3235–3242.
    1. Jänne PA. Ongoing first-line studies of epidermal growth factor receptor tyrosine kinase inhibitors in select patient populations. Seminars in Oncology. 2005;32(6, supplement 10):S9–e15.
    1. Gelovani JG. Molecular imaging of epidermal growth factor receptor expression-activity at the kinase level in tumors with positron emission tomography. Cancer and Metastasis Reviews. 2008;27(4):645–653.
    1. Dissoki S, Aviv Y, Laky D, Abourbeh G, Levitzki A, Mishani E. The effect of the [18F]-PEG group on tracer qualification of [4-(phenylamino)-quinazoline-6-YL]-amide moiety-An EGFR putative irreversible inhibitor. Applied Radiation and Isotopes. 2007;65(10):1140–1151.
    1. Mishani E, Abourbeh G, Rozen Y, et al. Novel carbon-11 labeled 4-dimethylamino-but-2-enoic acid [4-(phenylamino)-quinazoline-6-yl]-amides: potential PET bioprobes for molecular imaging of EGFR-positive tumors. Nuclear Medicine and Biology. 2004;31(4):469–476.
    1. Mishani E, Abourbeh G. Cancer molecular imaging: radionuclide-based biomarkers of the epidermal growth factor receptor (EGFR) Current Topics in Medicinal Chemistry. 2007;7(18):1755–1772.
    1. Mishani E, Hagooly A. Strategies for molecular imaging of epidermal growth factor receptor tyrosine kinase in cancer. Journal of Nuclear Medicine. 2009;50(8):1199–1202.
    1. Bonasera TA, Ortu G, Rozen Y, et al. Potential18F-labeled biomarkers for epidermal growth factor receptor tyrosine kinase. Nuclear Medicine and Biology. 2001;28(4):359–374.
    1. Ortu G, Ben-David I, Rozen Y, et al. Labeled EGFr-TK irreversible inhibitor (ML03): in vitro and in vivo properties, potential as pet biomarker for cancer and feasibility as anticancer drug. International Journal of Cancer. 2002;101(4):360–370.
    1. Abourbeh G, Dissoki S, Jacobson O, et al. Evaluation of radiolabeled ML04, a putative irreversible inhibitor of epidermal growth factor receptor, as a bioprobe for PET imaging of EGFR-overexpressing tumors. Nuclear Medicine and Biology. 2007;34(1):55–70.
    1. Pal A, Glekas A, Doubrovin M, et al. Molecular imaging of EGFR kinase activity in tumors with 124I-labeled small molecular tracer and positron emission tomography. Molecular Imaging and Biology. 2006;8(5):262–277.
    1. Su H, Seimbille Y, Ferl GZ, et al. Evaluation of [18F]gefitinib as a molecular imaging probe for the assessment of the epidermal growth factor receptor status in malignant tumors. European Journal of Nuclear Medicine and Molecular Imaging. 2008;35(6):1089–1099.
    1. Wang H, Yu J-M, Yang G-R, et al. Further characterization of the epidermal growth factor receptor ligand 11C-PD153035. Chinese Medical Journal. 2007;120(11):960–964.
    1. Memon AA, Jakobsen S, Dagnaes-Hansen F, Sorensen BS, Keiding S, Nexo E. Positron emission tomography (PET) imaging with [11C]-labeled erlotinib: a micro-PET study on mice with lung tumor xenografts. Cancer Research. 2009;69(3):873–878.
    1. Liu N, Li M, Li X, et al. PET-based biodistribution and radiation dosimetry of epidermal growth factor receptor-selective tracer 11C-PD153035 in humans. Journal of Nuclear Medicine. 2009;50(2):303–308.
    1. Deng W-P, Yang WK, Lai W-F, et al. Non-invasive in vivo imaging with radiolabelled FIAU for monitoring cancer gene therapy using herpes simplex virus type 1 thymidine kinase and ganciclovir. European Journal of Nuclear Medicine and Molecular Imaging. 2004;31(1):99–109.
    1. Fredriksson A, Johnström P, Thorell J-O, et al. In vivo evaluation of the biodistribution of 11C-labeled PD153035 in rats without and with neuroblastoma implants. Life Sciences. 1999;65(2):165–174.
    1. Shaul M, Abourbeh G, Jacobson O, et al. Novel iodine-124 labeled EGFR inhibitors as potential PET agents for molecular imaging in cancer. Bioorganic and Medicinal Chemistry. 2004;12(13):3421–3429.
    1. Mohammed A, Janakiram NB, Li Q, et al. The epidermal growth factor receptor inhibitor gefitinib prevents the progression of pancreatic lesions to carcinoma in a conditional LSL-KrasG12D/+ transgenic mouse model. Cancer Prevention Research. 2010;3(11):1417–1426.
    1. Kelley RK, Ko AH. Erlotinib in the treatment of advanced pancreatic cancer. Biologics. 2008;2(1):83–95.
    1. Ueda S, Ogata S, Tsuda H, et al. The correlation between cytoplasmic overexpression of epidermal growth factor receptor and tumor aggressiveness: poor prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas. 2004;29(1):e1–e8.
    1. Sordella R, Bell DW, Haber DA, Settleman J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science. 2004;305(5687):1163–1167.
    1. Tracy S, Mukohara T, Hansen M, Meyerson M, Johnson BE, Jänne PA. Gefitinib induces apoptosis in the EGFRL858R non-small-cell lung cancer cell line H3255. Cancer Research. 2004;64(20):7241–7244.
    1. Brognard J, Clark AS, Ni Y, Dennis PA. Akt/pbotein kinace B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation. Cancer Research. 2001;61(10):3986–3997.

Source: PubMed

3
订阅