Lapatinib Plasma and Tumor Concentrations and Effects on HER Receptor Phosphorylation in Tumor

Neil L Spector, Faith C Robertson, Sarah Bacus, Kimberly Blackwell, Deborah A Smith, Kelli Glenn, Leanne Cartee, Jennifer Harris, Carie L Kimbrough, Mark Gittelman, Eli Avisar, Peter Beitsch, Kevin M Koch, Neil L Spector, Faith C Robertson, Sarah Bacus, Kimberly Blackwell, Deborah A Smith, Kelli Glenn, Leanne Cartee, Jennifer Harris, Carie L Kimbrough, Mark Gittelman, Eli Avisar, Peter Beitsch, Kevin M Koch

Abstract

Purpose: The paradigm shift in cancer treatment from cytotoxic drugs to tumor targeted therapies poses new challenges, including optimization of dose and schedule based on a biologically effective dose, rather than the historical maximum tolerated dose. Optimal dosing is currently determined using concentrations of tyrosine kinase inhibitors in plasma as a surrogate for tumor concentrations. To examine this plasma-tumor relationship, we explored the association between lapatinib levels in tumor and plasma in mice and humans, and those effects on phosphorylation of human epidermal growth factor receptors (HER) in human tumors.

Experimental design: Mice bearing BT474 HER2+ human breast cancer xenografts were dosed once or twice daily (BID) with lapatinib. Drug concentrations were measured in blood, tumor, liver, and kidney. In a randomized phase I clinical trial, 28 treatment-naïve female patients with early stage HER2+ breast cancer received lapatinib 1000 or 1500 mg once daily (QD) or 500 mg BID before evaluating steady-state lapatinib levels in plasma and tumor.

Results: In mice, lapatinib levels were 4-fold higher in tumor than blood with a 4-fold longer half-life. Tumor concentrations exceeded the in vitro IC90 (~ 900 nM or 500 ng/mL) for inhibition of HER2 phosphorylation throughout the 12-hour dosing interval. In patients, tumor levels were 6- and 10-fold higher with QD and BID dosing, respectively, compared to plasma trough levels. The relationship between tumor and plasma concentration was complex, indicating multiple determinants. HER receptor phosphorylation varied depending upon lapatinib tumor concentrations, suggestive of changes in the repertoire of HER homo- and heterodimers.

Conclusion: Plasma lapatinib concentrations underestimated tumor drug levels, suggesting that optimal dosing should be focused on the site of action to avoid to inappropriate dose escalation. Larger clinical trials are required to determine optimal dose and schedule to achieve tumor concentrations that maximally inhibit HER receptors.

Clinical trial registration: NCT00359190.

Conflict of interest statement

Competing Interests: The authors have the following interests: Funding for this study was provided by GSK. Editorial support in the form of development of the first draft, editorial suggestions to draft revisions, assembling tables/figures, collating author comments and referencing was provided by Guissou Dabiri, PhD, and was funded by GSK. Sarah Bacus is employed by Quintiles, Deborah A Smith, Kelli Glenn, Leanne Cartee and Kevin M Koch by GlaxoSmithKline, Jennifer Harris by Pivot Oncology Consulting and Peter Beitsch by Dallas Surgical Group. GlaxoSmithKline markets the drug lapatinib under the propriety names Tykerb (mostly US) and Tyverb (mostly Europe). There are no further patents, products in development or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1. Schema of EGF10027.
Fig 1. Schema of EGF10027.
Twenty-eight patients were randomized to the following treatment groups: lapatinib 500 mg PO BID; lapatinib 1000 mg PO QD; lapatinib 1500 mg PO QD. Patient follow up and data analysis were conducted as indicated.
Fig 2. Lapatinib in human breast cancer…
Fig 2. Lapatinib in human breast cancer xenograft tissue.
Steady-state (Day 3) lapatinib concentrations in blood, tumor, kidney, and liver measured in CB-17 SCID female mice bearing established HER2+ BT474 breast cancer xenografts (n = 45) following treatment with lapatinib (100 mg/kg BID) administered by oral gavage x 5 doses. Results represent the mean +/- standard error.
Fig 3. The relationship between lapatinib concentrations…
Fig 3. The relationship between lapatinib concentrations in tumor and plasma from clinical samples.
Women with early stage HER2+ breast cancer were randomized to oral lapatinib monotherapy at 500 mg twice daily (BID), 1000 mg once daily (QD), or 1500 mg QD. Lapatinib concentrations (μM) in plasma (CP) and tumor (CT) tissue samples obtained simultaneously 9–15 days after initiating therapy (steady state) concentrations (Cmin). (A). Lapatinib concentrations for each patient in the indicated treatment groups. The dotted blue line represents the reported IC90 value for lapatinib in HER2+ breast cancer cell lines. Large circles represent group medians. (B) The complex relationship between lapatinib concentration in tumor and plasma showing a speculative model that suggests determinants: target protein binding (lower Emax curve), and net uptake/efflux (upper Emax curve): grey circle (patient with dysfunctional ABCB1 SNP C3435T); open circle (patient taking lapatinib 500 mg BID also taking grape seed oil), shown with simulation and deviations (dashed lines) illustrating speculation of diminished efflux (left shift) and increased EGFR target binding (right shift). The horizontal dotted line at 4 μM represents the IC50 for the ABCB1 transporter.
Fig 4. Phosphorylation of HER receptors relative…
Fig 4. Phosphorylation of HER receptors relative to tumor and plasma concentrations of lapatinib.
Changes (post-/pre-treatment ratios of optical density (OD values)) in EGFR, HER2, and HER3 phosphorylation in response to lapatinib in clinical tumor samples, and their relationship to tumor (left panel) and plasma (right panel) concentrations of lapatinib.

References

    1. Hurley LH. DNA and its associated processes as targets for cancer therapy. Nature Reviews Cancer 2002;2: 188–200.
    1. Stegmeier F, Warmuth M, Sellers WR, Dorsch M. Targeted cancer therapies in the twenty-first century: lessons from imatinib. Clin Pharmacol Ther. 2010;87: 543–552. 10.1038/clpt.2009.297
    1. Berry DA, Ueno NT, Johnson MM, Lei X, Caputo J, Smith DA, et al. High-dose chemotherapy with autologous hematopoietic stem-cell transplantation in metastatic breast cancer: overview of six randomized trials. J Clin Oncol. 2011;29: 3224–3231. 10.1200/JCO.2010.32.5936
    1. Hanahan D, Weinberg RA: Hallmarks of Cancer: the next generation. Cell 2011;144: 646–674. 10.1016/j.cell.2011.02.013
    1. Hanahan D, Weinberg RA.: Hallmarks of Cancer. Cell. 2000. January 7;100(1):57–70.
    1. Sharma SV, Settleman J. Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes Dev. 2007;21: 3214–3231.
    1. Jain RK, Lee JJ, Hong D, Markman M, Gong J, Naing A, et al.: Phase I oncology studies: evidence that in the era of targeted therapies patients on lower doses do not fare worse. Clin Cancer Res. 2010;16: 1289–1297. 10.1158/1078-0432.CCR-09-2684
    1. Wolf M, Swaisland H, Averbuch S. Development of the novel biologically targeted anticancer agent gefitinib: determining the optimum dose for clinical efficacy. Clin Cancer Res. 2004;10: 4607–4613.
    1. Fabian MA, Biggs WH, Treiber DK, Atteridge CE, Azimioara MD, Benedetti MG, et al. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;2: 329–336.
    1. Rusnak DW, Lackey K, Affleck K, Wood ER, Alligood KJ, Rhodes N, et al. The effects of the novel, reversible epidermal growth factor receptor/ErbB-2 tyrosine kinase inhibitor, GW2016, on the growth of human normal and tumor-derived cell lines in vitro and in vivo. Mol Cancer Ther. 2001;1: 85–94.
    1. Luo FR, Yang Z, Camuso A, Smykla R, McGlinchey K, Fager K, et al. Dasatinib (BMS-354825) pharmacokinetics and pharmacodynamic biomarkers in animal models predict optimal clinical exposure. Clin Cancer Res. 2006; 12: 7180–7186.
    1. Burris H, Dees C, Hurwitz H, Dowlati A, Blackwell K, Marcom K, et al. A phase I safety, pharmacokinetic, and clinical activity study of lapatinib (GW572016), a reversible inhibitor of ErbB1 and ErbB2 tyrosine kinases in heavily pre-treated patients with metastatic carcinomas. J Clin Oncol. 2005; 23: 5305–5313.
    1. Hsieh S, Tobien T, Koch K, Dunn J. Increasing throughput of parallel on-line extraction liquid chromatography/electrospray ionization tandem mass spectrometry system for GLP quantitative bioanalysis in drug development. Rapid Commun Mass Spectrom. 2004;18: 285–292.
    1. Spector NL, Xia W Burris H, Hurwitz H Dees EC, Dowlati A, et al. Study of the Biologic Effects of Lapatinib, a Reversible Inhibitor of ErbB1 and ErbB2 Tyrosine Kinases, on Tumor Growth and Survival Pathways in Patients with Advanced Malignancies. J Clin Oncol. 2005;23: 2502–2512.
    1. Johnston S, Trudeau M, Kaufman B, Boussen M, Blackwell K, LoRusso P, et al. Phase II study of predictive biomarker profiles for response targeting human epidermal growth factor receptor 2 (HER-2) in advanced inflammatory breast cancer with lapatinib monotherapy. J Clin Oncol. 2008;26: 1066–1072. 10.1200/JCO.2007.13.9949
    1. Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 2004;64: 6652–6659.
    1. Real FX, Rettig WJ, Chesa PG, Melamed MR, Old LJ, Mendelsohn J. Expression of Epidermal Growth Factor Receptor in Human Cultured Cells and Tissues: Relationship to Cell Lineages and Stage Differentiation. Cancer Res. 1986;46: 4726–4731.
    1. Siegel-Lakhai WS, Beijnen JH, Vervenne WL, Boot H, Keessen M, Versola M, et al. Phase I pharmacokinetic study of the safety and tolerability of lapatinib (GW572016) in combination with oxaliplatin/fluorouracil/leucovorin (FOLFOX4) in patients with solid tumors. Clin Cancer Res. 2007;13: 4495–4502.
    1. Polli JW, Humphreys JE, Harmon KA, Castellino S, O’Mara MJ, Olson KL, et al. The Role of Efflux and Uptake Transporters in Lapatinib (Tykerb, GW572016) Disposition and Drug Interactions. Drug Metab Dispos. 2008;36: 695–701. 10.1124/dmd.107.018374
    1. Burris HA, Taylor CW, Jones SF, Koch KM, Versola MJ, Arya N, et al. A Phase I and of oral lapatinib administered once or twice daily in patients with solid malignancy. Clin Cancer Res. 2009;15(21): 6702–6708. 10.1158/1078-0432.CCR-09-0369
    1. Kitagawa S, Nabekura T, Nakamura Y, Takahashi T, Kashiwada Y. Inhibition of P-glycoprotein function by tannic acid and pentagalloylglucose. J Pharm Pharmacol. 2007;59: 965–969.
    1. Guy PM, Platko JV, Cantley LC, Cerione RA, Carraway KL. Insect cell-expressed p180ErbB3 possesses an impaired tyrosine kinase activity. Proc Natl Acad Sci USA. 1994;91: 8132–8136.
    1. Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2): 172–187.
    1. Jain RK, Lee JJ, Hong D, Markman M, Gong J, Naing A, et al. Phase I oncology studies: evidence that in the era of targeted therapies patients on lower doses do not fare worse. Clin Cancer Res. 2010;16(4): 1289–1297. 10.1158/1078-0432.CCR-09-2684
    1. Gomez HL, Doval DC, Chavez MA, Ang PC, Aziz Z, Nag S, et al. Efficacy and safety of lapatinib as first-line therapy for ErbB2-amplified locally advanced or metastatic breast cancer. J Clin Oncol. 2008;26(18): 2999–3005. 10.1200/JCO.2007.14.0590
    1. Hendriks BS, Opresko LK, Wiley S, Lauffenburger D. Quantitative Analysis of HER2-mediated Effects on HER2 and Epidermal Growth Factor Receptor Endocytosis. J Biol Chem. 2003;278: 23343–23351.
    1. Shankaran H, Wilery S, Resat H. Modeling the Effects of HER/ErbB1-3 Coexpression on Receptor. Biophys J. 2006;90: 3993–4009.
    1. Fedi P, Pierce J, Di Fiore PP, Kraus MH. Efficient coupling with phosphatidylinositol 3-kinase, but not phospholipase C gamma or GTPase-activating protein, distinguishes ErbB3 signalling from that of other ErbB/EGFR family members. Mol Cell Biol. 1994;14: 492–500.
    1. Holbro T, Beerli RR, Maurer F, Koziczak M, Barbas CF 3rd, Hynes NE. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc Natl Acad Sci USA. 2003;100:8933–8938.
    1. Xia W, Petricoin EF III, Zhao S, Liu L, Osada T, Cheng Q, et al. An heregulin EGFR-HER3 signaling axis can mediate acquired resistance in HER2+ breast cancer models. Breast Cancer Res. 2013;15(5): R85
    1. Xia W, Bacus S, Hegde P, Husain I, Strum J, Liu L, et al. A model of acquired autoresistance to ErbB2 tyrosine kinase inhibitors and a therapeutic strategy to prevent its onset in breast cancer. Proc Natl Acad Sci USA. 2006;103: 7795–7800.
    1. Garrett JT, Olivares MG, Rinehart C, Granja-Ingram ND, Sánchez V, Chakrabarty A, et al. Transcriptional and posttranslational up-regulation of HER3 (ErbB3) compensates for inhibition of the HER2 tyrosine kinase. Proc Natl Acad Sci USA. 2011;108: 5021–5026. 10.1073/pnas.1016140108
    1. Li D, Ambrogio L, Shimamura T, Kubo S, Takahashi M, Chirieac LR, et al. BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models. Oncogene 2008; 27: 4702–4711. 10.1038/onc.2008.109
    1. Amin DM, Sergino N, Ahuja D, McMahon M, Blair JA, Wang D, et al. Resilience and Vulnerability in the HER2-HER3 Tumorigenic Driver. Sci Transl Med. 2010; 2(16): 16ra7 10.1126/scitranslmed.3000389
    1. Chien AJ, Munster PN, Melisko ME, Rugo HS, Park JW, Goga A, et al. Phase I dose-escalation study of 5-day intermittent oral lapatinib therapy in patients with human epidermal growth factor receptor 2-overexpressing breast cancer. J Clin Oncol. 2014;32:1472–1479. 10.1200/JCO.2013.52.1161
    1. Xu N, Wang SQ, Tan D, Gao Y, Lin G, Xi R. EGFR, Wingless and JAK/STAT signaling cooperatively maintain Drosophila intestinal stem cells. Dev Biol. 2011;354(1): 31–43. 10.1016/j.ydbio.2011.03.018
    1. Jiang H, Grenley MO, Bravo M-J, Blumhagen RZ, Edgar BA. EGFR/Ras/MAPK Signaling Mediates Adult Midgut Epithelial Homeostasis and Regeneration in Drosophila. Cell Stem Cell 2011;8: 84–95. 10.1016/j.stem.2010.11.026
    1. Natarajan A, Wagner B, Sibilia M. The EGF receptor is required for efficient liver regeneration. Proc Natl Acad Sci USA. 2007;104: 17081–17086.

Source: PubMed

3
Tilaa