Population pharmacokinetic analysis of nanoparticle-bound and free camptothecin after administration of NLG207 in adults with advanced solid tumors

Keith T Schmidt, Alwin D R Huitema, Thomas P C Dorlo, Cody J Peer, Lisa M Cordes, Linda Sciuto, Susan Wroblewski, Yves Pommier, Ravi A Madan, Anish Thomas, William D Figg, Keith T Schmidt, Alwin D R Huitema, Thomas P C Dorlo, Cody J Peer, Lisa M Cordes, Linda Sciuto, Susan Wroblewski, Yves Pommier, Ravi A Madan, Anish Thomas, William D Figg

Abstract

Purpose: NLG207 (formerly CRLX101) is a nanoparticle-drug conjugate (NDC) of the potent topoisomerase I inhibitor, camptothecin (CPT). The present study sought to characterize the complex pharmacokinetics (PK) of NLG207 and better describe CPT release from nanoparticles using a population PK (popPK) model.

Methods: From 27 patients enrolled on two phase II clinical trials (NCT02769962 and NCT03531827), dense sampling was performed up to 48 h post-administration of NLG207 during cycle one and six of treatment; samples were also collected at ~ 360 h post-dose. Conjugated and free CPT concentrations were quantified from each sample, resulting in 477 observations to build a popPK model using non-linear mixed-effects modeling.

Results: The PK of NLG207 was characterized by combining two linear two-compartment models with first-order kinetics each to describe nanoparticle-bound (conjugated) and free CPT. Allometric scaling based on body weight provided the best body-size descriptor for all PK parameters. The typical volumes of distribution of the conjugated CPT central and free CPT central compartments were 3.16 L (BSV CV%; 18.1%) and 21.1 L (CV%; 79.8%), respectively. CPT release from the nanoparticle formulation was characterized via an initial rapid clearance of 5.71 L/h (CV%; 62.6%), which decreased via first-order decay (estimated half-life of 0.307 h) to the steady-state value of 0.0988 L/h (CV%; 33.5%) by ~ 4 h after end of infusion. Renal clearance of free CPT was 0.874 L/h (CV%; 42.2%).

Conclusion: The popPK model confirmed nanoparticle behavior of conjugated CPT and mechanistically characterized CPT release from NLG207. The current analysis provides a strong foundation for future study as a potential predictive tool in ongoing NLG207 clinical trials.

Keywords: Drug release; NLMEM; Nanoparticle; Population pharmacokinetics; Topoisomerase I.

Conflict of interest statement

The author(s) declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Overview of the four-compartment model describing disposition of nanoparticle-bound (conjugated) and free CPT, and the associated calculated parameters. Plasma sample quantitation enabled CMT1 and CMT3 observations for conjugated and free CPT concentrations, respectively. The model can be divided into two portions: conjugated CPT distribution described via estimates V1, V2, and Q1, and free CPT distribution described via estimates V3, V4, and Q3. CL1, or the conversion of conjugated to free CPT, was described using two additive clearance terms: CLB is the “base” rate of conversion, and CLF is a “fast” clearance rate that is modified via a simulated first-order decay term with estimated half-life, t1/2
Fig. 2
Fig. 2
Goodness-of-fit plots for the pharmacokinetic model. Separate plots were generated for the evaluation of conjugated CPT (a–d) and free CPT (e–h). Plots include population prediction against observed data (a, e), individual prediction against observed data (b, f), conditional weighted residuals against population prediction (c, g), and conditional weighted residuals against time after dose (d, h). The solid black lines represent either the line of unity (a, b, e, f) or the zero line (c, d, g, h). Solid blue lines represent the local regression fit of the values
Fig. 3
Fig. 3
Visual predictive checks (VPCs) of conjugated (a) and free CPT (b) for timepoints up to ~ 50 h post-dose of NLG207. “Observations” are reported in units of ng/mL using log scale axis, and “time after dose” is reported in hours. Solid black lines depict the observed median and dashed lines represent the 2.5% and 97.5% percentile concentrations. 95% prediction intervals of the simulated mean and the 2.5 and 97.5% percentiles are represented by dark- and light-gray areas, respectively. Round dots represent observations and asterisks highlight observed percentiles outside of the prediction area

References

    1. Schmidt KT, Peer CJ, Huitema ADR, Williams MD, Wroblewski S, Schellens JHM, Madan RA, Figg WD. Measurement of NLG207 (formerly CRLX101) nanoparticle-bound and released camptothecin in human plasma. J Pharm Biomed Anal. 2020;181:113073. doi: 10.1016/j.jpba.2019.113073.
    1. Wall ME, Wani MC (1996) Camptothecin and taxol: from discovery to clinic. J Ethnopharmacol 51 (1–3):239–253 (discussion 253–234)
    1. Li F, Jiang T, Li Q, Ling X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: did we miss something in CPT analogue molecular targets for treating human disease such as cancer? Am J Cancer Res. 2017;7(12):2350–2394.
    1. Moertel CG, Schutt AJ, Reitemeier RJ, Hahn RG. Phase II study of camptothecin (NSC-100880) in the treatment of advanced gastrointestinal cancer. Cancer Chemother Rep. 1972;56(1):95–101.
    1. Gottlieb JA, Luce JK. Treatment of malignant melanoma with camptothecin (NSC-100880) Cancer Chemother Rep. 1972;56(1):103–105.
    1. Thomas A, Pommier Y. Targeting topoisomerase I in the era of precision medicine. Clin Cancer Res. 2019 doi: 10.1158/1078-0432.CCR-19-1089.
    1. Davis ME. Design and development of IT-101, a cyclodextrin-containing polymer conjugate of camptothecin. Adv Drug Deliv Rev. 2009;61(13):1189–1192. doi: 10.1016/j.addr.2009.05.005.
    1. Clark AJ, Wiley DT, Zuckerman JE, Webster P, Chao J, Lin J, Yen Y, Davis ME. CRLX101 nanoparticles localize in human tumors and not in adjacent, nonneoplastic tissue after intravenous dosing. Proc Natl Acad Sci USA. 2016;113(14):3850–3854. doi: 10.1073/pnas.1603018113.
    1. Chao J, Lin J, Frankel P, Clark AJ, Wiley DT, Garmey E, Fakih M, Lim D, Chung V, Luevanos E, Eliasof S, Davis ME, Yen Y. Pilot trial of CRLX101 in patients with advanced, chemotherapy-refractory gastroesophageal cancer. J Gastrointest Oncol. 2017;8(6):962–969. doi: 10.21037/jgo.2017.08.10.
    1. Keefe SM, Hoffman-Censits J, Cohen RB, Mamtani R, Heitjan D, Eliasof S, Nixon A, Turnbull B, Garmey EG, Gunnarsson O, Waliki M, Ciconte J, Jayaraman L, Senderowicz A, Tellez AB, Hennessy M, Piscitelli A, Vaughn D, Smith A, Haas NB. Efficacy of the nanoparticle-drug conjugate CRLX101 in combination with bevacizumab in metastatic renal cell carcinoma: results of an investigator-initiated phase I-IIa clinical trial. Ann Oncol. 2016;27(8):1579–1585. doi: 10.1093/annonc/mdw188.
    1. Pham E, Birrer MJ, Eliasof S, Garmey EG, Lazarus D, Lee CR, Man S, Matulonis UA, Peters CG, Xu P, Krasner C, Kerbel RS. Translational impact of nanoparticle-drug conjugate CRLX101 with or without bevacizumab in advanced ovarian cancer. Clin Cancer Res. 2015;21(4):808–818. doi: 10.1158/1078-0432.CCR-14-2810.
    1. Sanoff HK, Moon DH, Moore DT, Boles J, Bui C, Blackstock W, O'Neil BH, Subramaniam S, McRee AJ, Carlson C, Lee MS, Tepper JE, Wang AZ. Phase I/II trial of nano-camptothecin CRLX101 with capecitabine and radiotherapy as neoadjuvant treatment for locally advanced rectal cancer. Nanomedicine. 2019;18:189–195. doi: 10.1016/j.nano.2019.02.021.
    1. Voss MH, Hussain A, Vogelzang N, Lee JL, Keam B, Rha SY, Vaishampayan U, Harris WB, Richey S, Randall JM, Shaffer D, Cohn A, Crowell T, Li J, Senderowicz A, Stone E, Figlin R, Motzer RJ, Haas NB, Hutson T. A randomized phase II trial of CRLX101 in combination with bevacizumab versus standard of care in patients with advanced renal cell carcinoma. Ann Oncol. 2017;28(11):2754–2760. doi: 10.1093/annonc/mdx493.
    1. Weiss GJ, Chao J, Neidhart JD, Ramanathan RK, Bassett D, Neidhart JA, Choi CHJ, Chow W, Chung V, Forman SJ, Garmey E, Hwang J, Kalinoski DL, Koczywas M, Longmate J, Melton RJ, Morgan R, Oliver J, Peterkin JJ, Ryan JL, Schluep T, Synold TW, Twardowski P, Davis ME, Yen Y. First-in-human phase 1/2a trial of CRLX101, a cyclodextrin-containing polymer-camptothecin nanopharmaceutical in patients with advanced solid tumor malignancies. Invest New Drugs. 2013;31(4):986–1000. doi: 10.1007/s10637-012-9921-8.
    1. Rapisarda A, Uranchimeg B, Sordet O, Pommier Y, Shoemaker RH, Melillo G. Topoisomerase I-mediated inhibition of hypoxia-inducible factor 1: mechanism and therapeutic implications. Cancer Res. 2004;64(4):1475–1482. doi: 10.1158/0008-5472.can-03-3139.
    1. Duska L, O’Malley DM, Krasner C, Schilder RJ, Matthew C, Moore K, Thaker P, Miller A, Purdy C, Leyco AJ, Smith C, Mercier D, Tennant L, Kennedy E, Vahanian N, Link C (2019) Abstract CT151: a Phase II study of NLG207 (formerly CRLX101) in combination with weekly paclitaxel in patients with recurrent or persistent epithelial ovarian, fallopian tube or primary peritoneal cancer. Cancer Res 79(13):Suppl.
    1. Takahashi N, Surolia I, Thomas A. Targeting DNA repair to drive immune responses: it's time to reconsider the strategy for clinical translation. Clin Cancer Res. 2020 doi: 10.1158/1078-0432.CCR-19-3841.
    1. US Food and Drug Administration (2018) Bioanalytical method validation guidance for industry.
    1. Keizer RJ, Karlsson MO, Hooker A. Modeling and Simulation workbench for NONMEM: Tutorial on Pirana, PsN, and Xpose. CPT Pharm Syst Pharmacol. 2013;2:e50. doi: 10.1038/psp.2013.24.
    1. Cheng J, Khin KT, Jensen GS, Liu A, Davis ME. Synthesis of linear, beta-cyclodextrin-based polymers and their camptothecin conjugates. Bioconjug Chem. 2003;14(5):1007–1017. doi: 10.1021/bc0340924.
    1. Young C, Schluep T, Hwang J, Eliasof S. CRLX101 (formerly IT-101)—a novel nanopharmaceutical of camptothecin in clinical development. Curr Bioact Compd. 2011;7(1):8–14. doi: 10.2174/157340711795163866.
    1. Dosne AG, Bergstrand M, Harling K, Karlsson MO. Improving the estimation of parameter uncertainty distributions in nonlinear mixed effects models using sampling importance resampling. J Pharmacokinet Pharmacodyn. 2016;43(6):583–596. doi: 10.1007/s10928-016-9487-8.
    1. West GB, Brown JH, Enquist BJ. A general model for the origin of allometric scaling laws in biology. Science. 1997;276(5309):122–126. doi: 10.1126/science.276.5309.122.
    1. Eliasof S, Lazarus D, Peters CG, Case RI, Cole RO, Hwang J, Schluep T, Chao J, Lin J, Yen Y, Han H, Wiley DT, Zuckerman JE, Davis ME. Correlating preclinical animal studies and human clinical trials of a multifunctional, polymeric nanoparticle. Proc Natl Acad Sci USA. 2013;110(37):15127–15132. doi: 10.1073/pnas.1309566110.
    1. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet. 2010;49(10):633–659. doi: 10.2165/11535960-000000000-00000.
    1. Schluep T, Hwang J, Hildebrandt IJ, Czernin J, Choi CH, Alabi CA, Mack BC, Davis ME. Pharmacokinetics and tumor dynamics of the nanoparticle IT-101 from PET imaging and tumor histological measurements. Proc Natl Acad Sci USA. 2009;106(27):11394–11399. doi: 10.1073/pnas.0905487106.
    1. Petschauer JS, Madden AJ, Kirschbrown WP, Song G, Zamboni WC. The effects of nanoparticle drug loading on the pharmacokinetics of anticancer agents. Nanomedicine (Lond) 2015;10(3):447–463. doi: 10.2217/nnm.14.179.
    1. Chen YF, Wang YH, Lei CS, Changou CA, Davis ME, Yen Y. Host immune response to anti-cancer camptothecin conjugated cyclodextrin-based polymers. J Biomed Sci. 2019;26(1):85. doi: 10.1186/s12929-019-0583-0.
    1. Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T, Baba Y. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013;13(1):89. doi: 10.1186/1475-2867-13-89.
    1. Park R, Leach WJ, Arieff AI. Determination of liver intracellular pH in vivo and its homeostasis in acute acidosis and alkalosis. Am J Physiol. 1979;236(3):F240–245. doi: 10.1152/ajprenal.1979.236.3.F240.
    1. Skelton LA, Boron WF, Zhou Y. Acid-base transport by the renal proximal tubule. J Nephrol. 2010;23(Suppl 16):S4–18.
    1. Gottlieb JA, Guarino AM, Call JB, Oliverio VT, Block JB. Preliminary pharmacologic and clinical evaluation of camptothecin sodium (NSC-100880) Cancer Chemother Rep. 1970;54(6):461–470.
    1. Mould DR, Holford NH, Schellens JH, Beijnen JH, Hutson PR, Rosing H, ten Bokkel Huinink WW, Rowinsky EK, Schiller JH, Russo M, Ross G. Population pharmacokinetic and adverse event analysis of topotecan in patients with solid tumors. Clin Pharmacol Ther. 2002;71(5):334–348. doi: 10.1067/mcp.2002.123553.
    1. Hospira (2005) 5% Dextrose Injection, USP: Package Insert.
    1. Anderson BJ, Holford NH. Mechanistic basis of using body size and maturation to predict clearance in humans. Drug Metab Pharmacokinet. 2009;24(1):25–36. doi: 10.2133/dmpk.24.25.
    1. Fleury F, Ianoul A, Berjot M, Feofanov A, Alix AJ, Nabiev I. Camptothecin-binding site in human serum albumin and protein transformations induced by drug binding. FEBS Lett. 1997;411(2–3):215–220. doi: 10.1016/s0014-5793(97)00693-5.
    1. Wu H, Infante JR, Keedy VL, Jones SF, Chan E, Bendell JC, Lee W, Zamboni BA, Ikeda S, Kodaira H, Rothenberg ML, Burris HA, 3rd, Zamboni WC. Population pharmacokinetics of PEGylated liposomal CPT-11 (IHL-305) in patients with advanced solid tumors. Eur J Clin Pharmacol. 2013;69(12):2073–2081. doi: 10.1007/s00228-013-1580-y.
    1. Amantea MA, Forrest A, Northfelt DW, Mamelok R. Population pharmacokinetics and pharmacodynamics of pegylated-liposomal doxorubicin in patients with AIDS-related Kaposi's sarcoma. Clin Pharmacol Ther. 1997;61(3):301–311. doi: 10.1016/S0009-9236(97)90162-4.
    1. Hempel G, Reinhardt D, Creutzig U, Boos J. Population pharmacokinetics of liposomal daunorubicin in children. Br J Clin Pharmacol. 2003;56(4):370–377. doi: 10.1046/j.1365-2125.2003.01886.x.

Source: PubMed

3
Sottoscrivi