Bevacizumab dosing strategy in paediatric cancer patients based on population pharmacokinetic analysis with external validation

Kelong Han, Thomas Peyret, Angelica Quartino, Nathalie H Gosselin, Sridharan Gururangan, Michela Casanova, Johannes H M Merks, Maura Massimino, Jacques Grill, Najat C Daw, Fariba Navid, Jin Jin, David E Allison, Kelong Han, Thomas Peyret, Angelica Quartino, Nathalie H Gosselin, Sridharan Gururangan, Michela Casanova, Johannes H M Merks, Maura Massimino, Jacques Grill, Najat C Daw, Fariba Navid, Jin Jin, David E Allison

Abstract

Aim: The aim of the present study was to evaluate the pharmacokinetics of bevacizumab and various dosing strategies for this agent in paediatric patients.

Methods: Data were collected from 232 paediatric patients (1971 concentrations) in five studies, with a wide range of age (0.5-21 years), body weight (BWT; 5.9-125 kg), and regimens (5-15 mg kg(-1) biweekly or triweekly). Data from 152 patients (1427 concentrations) and 80 patients (544 concentrations) were used for model building and external validation, respectively. Steady-state exposure was simulated under BWT-based, body surface area (BSA)-based, ideal body weight (IBW)-based, and tier-based doses. NONMEM and R were used for analyses.

Results: Typical estimates of clearance, central volume of distribution (V1), and median half-life were 9.04 ml h(-1) , 2851 ml, and 19.6 days, respectively. Clearance decreased with increasing albumin. Clearance and V1 increased with BWT and were higher in male patients. Clearance and V1 were lower in children with primary central nervous system (CNS) tumours than in children with sarcomas, resulting in 49% higher trough (C min) and 29% higher peak (Cmax) concentrations. BWT-adjusted clearance and V1 remained unchanged across ages. Paediatric C min was similar to adult C min under all dosing strategies. Paediatric Cmax exceeded adult Cmax under tier-based doses.

Conclusions: BWT-adjusted pharmacokinetic parameter estimates in paediatric patients were similar to those in adults, and similar across ages. Bevacizumab exposure was higher in children with primary CNS tumours than in children with sarcomas. BSA-based, IBW-based, and tier-based doses offered no substantial advantage over the BWT-based dose currently used in adults for bevacizumab. Given the similarity in pharmacokinetics among many monoclonal antibodies, this may help to develop practical paediatric dosing guidelines for other therapeutic antibodies.

Keywords: bevacizumab; body surface area; central nervous system tumour; dosing strategy; external validation; paediatric.

© 2015 Genentech Inc. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

Figure 1
Figure 1
Weight‐adjusted pharmacokinetic (PK) parameters. (A) Clearance and (B) central volume of distribution obtained from the bevacizumab base model across age. LOESS, locally weighted scatterplot smoothing
Figure 2
Figure 2
(A) Prediction‐corrected visual predictive check for the serum concentration–time profiles of bevacizumab using the final model in paediatric cancer patients. (B) Section of (A) before day 35 after dose administration, where the majority (94%) of the data points lay. CI, confidence interval; Pred, population prediction
Figure 3
Figure 3
Impact of the variation for a single covariate included in the final model on steady‐state bevacizumab exposure in paediatric cancer patients. (A) Trough concentration (Cmin) and (B) peak concentration (Cmax). Red vertical lines represent the ‘base’, defined as the exposure of a typical patient – i.e. a 44‐kg female paediatric patient with an albumin level of 39 g l–1 and sarcomas. The dark blue‐shaded curve at the bottom, with a value at each end, shows the 5th to 95th percentile exposure range across the entire population. Each light blue‐shaded bar represents the influence of a single covariate on the steady‐state exposure after repeated bevacizumab doses of 10 mg kg–1 once every two weeks. The label at left end of the bar represents the covariate being evaluated. The upper and lower values for each covariate capture 90% of the plausible range in the population. The length of each bar describes the potential impact of that particular covariate on bevacizumab steady‐state exposure, with the percentage value in the parentheses at each end representing the percentage change in exposure from the ‘base’. The most influential covariate is at the bottom of the plot for each exposure metric, except for the body weight effect stratified by age group, which is displayed on the top. CI, confidence interval; Cmax, peak concentration; Cmin, trough concentration
Figure 4
Figure 4
External validation for (A) primary CNS tumours and (B) sarcomas. About 95% of the prediction‐corrected observations fall between the 95% prediction interval boundaries, which are very close to the observed 2.5th and 97.5th percentiles. CNS, central nervous system; PI, prediction interval
Figure 5
Figure 5
Simulated steady‐state bevacizumab exposure in paediatric patients under BWT‐, BSA‐, IBW‐, and tier‐based doses. Equivalent doses (once every 2 weeks) were used: 10 mg kg–1 for the BWT‐based dose, 398 mg m–2 for the BSA‐based dose, and 11 mg kg–1 for the IBW‐based dose. The tier‐based dose in each BWT range was determined so that the steady‐state area under the curve (AUC) under these doses matched the adult steady‐state AUC: 180 mg for <15 kg, 360 mg for 15 – 40 kg, 640 mg for >40 kg. Only patients with a BWT below 80 kg are displayed. BSA, body surface area; BWT, total body weight; Cmax, peak concentration; Cmin, trough concentration; IBW, ideal body weight; PI, prediction interval

References

    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003; 9: 669–76.
    1. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350: 2335–42.
    1. Giantonio BJ, Catalano PJ, Meropol NJ, O'Dwyer PJ, Mitchell EP, Alberts SR, Schwartz MA, Benson AB 3rd. Eastern Cooperative Oncology Group Study E3200 . Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol 2007; 25: 1539–44.
    1. Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH. Paclitaxel–carboplatin alone or with bevacizumab for non‐small‐cell lung cancer. N Engl J Med 2006; 355: 2542–50.
    1. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med 2007; 357: 2666–76.
    1. Escudier B, Bellmunt J, Négrier S, Bajetta E, Melichar B, Bracarda S, Ravaud A, Golding S, Jethwa S, Sneller V. Phase III trial of bevacizumab plus interferon alfa‐2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol 2010; 28: 2144–50.
    1. Tewari KS, Sill MW, Long HJ 3rd, Penson RT, Huang H, Ramondetta LM, Landrum LM, Oaknin A, Reid TJ, Leitao MM, Michael HE, Monk BJ. Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med 2014; 370: 734–43.
    1. Pujade‐Lauraine E, Hilpert F, Weber B, Reuss A, Poveda A, Kristensen G, Sorio R, Vergote I, Witteveen P, Bamias A, Pereira D, Wimberger P, Oaknin A, Mirza MR, Follana P, Bollag D, Ray‐Coquard I. Bevacizumab combined with chemotherapy for platinum‐resistant recurrent ovarian cancer: the AURELIA open‐label randomized Phase III trial. J Clin Oncol 2014; 32: 1302–8.
    1. Edlund H, Melin J, Parra‐Guillen ZP, Kloft C. Pharmacokinetics and pharmacokinetic–pharmacodynamic relationships of monoclonal antibodies in children. Clin Pharmacokinet 2015; 54: 35–80.
    1. Crawford JD, Terry ME, Rourke GM. Simplification of drug dosage calculation by application of the surface area principle. Pediatrics 1950; 5: 783–90.
    1. Ross EL, Jorgensen J, DeWitt PE, Okada C, Porter R, Haemer M, Reiter PD. Comparison of 3 body size descriptors in critically ill obese children and adolescents: implications for medication dosing. J Pediatr Pharmacol Ther 2014; 19: 103–10.
    1. Lu JF, Bruno R, Eppler S, Novotny W, Lum B, Gaudreault J. Clinical pharmacokinetics of bevacizumab in patients with solid tumors. Cancer Chemother Pharmacol 2008. Oct; 62: 779–86.
    1. Glade Bender JL, Adamson PC, Reid JM, Xu L, Baruchel S, Shaked Y, Kerbel RS, Cooney‐Qualter EM, Stempak D, Chen HX, Nelson MD, Krailo MD, Ingle AM, Blaney SM, Kandel JJ, Yamashiro DJ, Study C's OG. Phase I trial and pharmacokinetic study of bevacizumab in pediatric patients with refractory solid tumors: a Children's Oncology Group Study. J Clin Oncol 2008; 26: 399–405.
    1. Turner DC, Navid F, Daw NC, Mao S, Wu J, Santana VM, Neel M, Rao B, Willert JR, Loeb DM, Harstead KE, Throm SL, Freeman BB 3rd, Stewart CF. Population pharmacokinetics of bevacizumab in children with osteosarcoma: implications for dosing. Clin Cancer Res 2014; 20: 2783–92.
    1. Gururangan S, Fangusaro J, Poussaint TY, McLendon RE, Onar‐Thomas A, Wu S, Packer RJ, Banerjee A, Gilbertson RJ, Fahey F, Vajapeyam S, Jakacki R, Gajjar A, Goldman S, Pollack IF, Friedman HS, Boyett JM, Fouladi M, Kun LE. Efficacy of bevacizumab plus irinotecan in children with recurrent low‐grade gliomas – a Pediatric Brain Tumor Consortium study. Neuro Oncol 2014; 16: 310–7.
    1. Beal S, Sheiner LB, Boeckmann A, Bauer RJ. NONMEM User's Guides (1989–2009). Ellicott City, MD, USA: Icon Development Solutions, 2009.
    1. Lindbom L, Ribbing J, Jonsson EN. Perl‐speaks‐NONMEM (PsN) – a Perl module for NONMEM related programming. Comput Methods Programs Biomed 2004; 75: 85–94.
    1. R Core Team (2015). R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. [online] Available at: (last accessed Aug 15, 2015).
    1. Dong JQ, Salinger DH, Endres CJ, Gibbs JP, Hsu CP, Stouch BJ, Hurh E, Gibbs MA. Quantitative prediction of human pharmacokinetics for monoclonal antibodies: retrospective analysis of monkey as a single species for first‐in‐human prediction. Clin Pharmacokinet 2011; 50: 131–42.
    1. Karlsson MO, Savic RM. Diagnosing model diagnostics. Clin Pharmacol Ther 2007; 82: 17–20.
    1. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER) . Guidance for industry population pharmacokinetics [online]. Available at (last accessed Aug 15, 2015).
    1. Committee for Medicinal Products for Human Use (CHMP) . Guideline on reporting the results of population pharmacokinetic analyses [online]. Doc. ref. CHMP/EWP/185990/06, London. Available at (last accessed Aug 15, 2015).
    1. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction‐corrected visual predictive checks for diagnosing nonlinear mixed‐effects models. AAPS J 2011; 13: 143–51.
    1. Ette EI. Stability and performance of a population pharmacokinetic model. J Clin Pharmacol 1997; 37: 486–95.
    1. Savic RM, Karlsson MO. Importance of shrinkage in empirical Bayes estimates for diagnostics: problems and solutions. AAPS J 2009; 11: 558–69.
    1. Ogden CL, Kuczmarski RJ, Flegal KM, Mei Z, Guo S, Wei R, Grummer‐Strawn LM, Curtin LR, Roche AF, Johnson CL. Centers for Disease Control and Prevention 2000 growth charts for the United States: improvements to the 1977 National Center for Health Statistics version. Pediatrics 2002; 109: 45–60.
    1. Midgley R, Kerr D. Bevacizumab – current status and future directions. Ann Oncol 2005; 16: 999–1004.
    1. US. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research (CBER) . Guidance for industry. The content and format for pediatric use supplements [online]. Available at (last accessed Aug 15, 2015).
    1. Dostalek M, Gardner I, Gurbaxani BM, Rose RH, Chetty M. Pharmacokinetics, pharmacodynamics and physiologically‐based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet 2013; 52: 83–124.
    1. Weisman MH, Moreland LW, Furst DE, Weinblatt ME, Keystone EC, Paulus HE, Teoh LS, Velagapudi RB, Noertersheuser PA, Granneman GR, Fischkoff SA, Chartash EK. Efficacy, pharmacokinetic, and safety assessment of adalimumab, a fully human anti‐tumor necrosis factor‐alpha monoclonal antibody, in adults with rheumatoid arthritis receiving concomitant methotrexate: a pilot study. Clin Ther 2003; 25: 1700–21.
    1. Höcker B, Kovarik JM, Daniel V, Opelz G, Fehrenbach H, Holder M, Hoppe B, Hoyer P, Jungraithmayr TC, Köpf‐Shakib S, Laube GF, Müller‐Wiefel DE, Offner G, Plank C, Schröder M, Weber LT, Zimmerhackl LB, Tönshoff B. Pharmacokinetics and immunodynamics of basiliximab in pediatric renal transplant recipients on mycophenolate mofetil comedication. Transplantation 2008; 86: 1234–40.
    1. Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, Macfarlane JD, Antoni C, Leeb B, Elliott MJ, Woody JN, Schaible TF, Feldmann M. Therapeutic efficacy of multiple intravenous infusions of anti‐tumor necrosis factor alpha monoclonal antibody combined with low‐dose weekly methotrexate in rheumatoid arthritis. Arthritis Rheum 1998; 41: 1552–63.
    1. Ordás I, Mould DR, Feagan BG, Sandborn WJ. Anti‐TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics‐based dosing paradigms. Clin Pharmacol Ther 2012; 91: 635–46.
    1. Bien E, Rapala M, Krawczyk M, Balcerska A. The serum levels of soluble interleukin‐2 receptor alpha and lactate dehydrogenase but not of B2‐microglobulin correlate with selected clinico‐pathological prognostic factors and response to therapy in childhood soft tissue sarcomas. J Cancer Res Clin Oncol 2010; 136: 293–305.
    1. Bien E, Krawczyk M, Izycka‐Swieszewska E, Trzonkowski P, Kazanowska B, Adamkiewicz‐Drozynska E, Balcerska A. Serum IL‐10 and IL‐12 levels reflect the response to chemotherapy but are influenced by G‐CSF therapy and sepsis in children with soft tissue sarcomas. Postepy Hig Med Dosw (Online) 2013; 67: 517–28.
    1. Yang J, Zhao H, Garnett C, Rahman A, Gobburu JV, Pierce W, Schechter G, Summers J, Keegan P, Booth B, Wang Y. The combination of exposure‐response and case–control analyses in regulatory decision making. J Clin Pharmacol 2013; 53: 160–6.
    1. Keizer RJ, Huitema AD, Schellens JH, Beijnen JH. Clinical pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet 2010; 49: 493–507.
    1. Jastaniah W, Aseeri M. A cross‐sectional study comparing variation in body surface area and chemotherapy dosing in pediatric oncology using two different methods. J Oncol Pharm Pract 2010; 16: 189–93.
    1. Mosteller RD. Simplified calculation of body‐surface area. N Engl J Med 1987; 317: 1098.
    1. Haycock GB, Schwartz GJ, Wisotsky DH. Geometric method for measuring body surface area: a height–weight formula validated in infants, children, and adults. J Pediatr 1978; 93: 62–6.
    1. Du Bois D, Du Bois EF. A formula to estimate the approximate surface area if height and weight be known. Arch Intern Med 1916; 17: 863–71.
    1. Shi R, Derendorf H. Pediatric dosing and body size in biotherapeutics. Pharmaceutics 2010; 2: 389–418.

Source: PubMed

3
Tilaa