Pharmacokinetics and Exposure-Response of Vosoritide in Children with Achondroplasia

Ming Liang Chan, Yulan Qi, Kevin Larimore, Anu Cherukuri, Lori Seid, Kala Jayaram, George Jeha, Elena Fisheleva, Jonathan Day, Alice Huntsman-Labed, Ravi Savarirayan, Melita Irving, Carlos A Bacino, Julie Hoover-Fong, Keiichi Ozono, Klaus Mohnike, William R Wilcox, William A Horton, Joshua Henshaw, Ming Liang Chan, Yulan Qi, Kevin Larimore, Anu Cherukuri, Lori Seid, Kala Jayaram, George Jeha, Elena Fisheleva, Jonathan Day, Alice Huntsman-Labed, Ravi Savarirayan, Melita Irving, Carlos A Bacino, Julie Hoover-Fong, Keiichi Ozono, Klaus Mohnike, William R Wilcox, William A Horton, Joshua Henshaw

Abstract

Background and objective: Vosoritide, an analog of C-type natriuretic peptide, has been developed for the treatment of children with achondroplasia. The pharmacokinetics of vosoritide and relationships between plasma exposure and efficacy, biomarkers, and safety endpoints were evaluated in a phase II, open-label, dose-escalation study (N = 35 patients aged 5-14 years who received daily subcutaneous injections for 24 months) and a phase III, double-blind, placebo-controlled study (N = 60 patients aged 5-18 years randomized to receive daily subcutaneous injections for 52 weeks).

Methods: Pharmacokinetic parameters for both studies were obtained from non-compartmental analysis. Potential correlations between vosoritide exposure and changes in annualized growth velocity, collagen type X marker (CXM; a biomarker of endochondral ossification), cyclic guanosine monophosphate (cGMP; a biomarker of pharmacological activity), heart rate, and systolic and diastolic blood pressures were then evaluated.

Results: The exposure-response relationships for changes in both annualized growth velocity and the CXM biomarker saturated at 15 μg/kg, while systemic pharmacological activity, as measured by urinary cGMP, was near maximal or saturated at exposures obtained at the highest dose studied (i.e. 30 μg/kg). This suggested that the additional bioactivity was likely in tissues not related to endochondral bone formation. In the phase III study, following subcutaneous administration at the recommended dose of 15 μg/kg to patients with achondroplasia aged 5-18 years, vosoritide was rapidly absorbed with a median time to maximal plasma concentration (Cmax) of 15 minutes, and cleared with a mean half-life of 27.9 minutes after 52 weeks of treatment. Vosoritide exposure (Cmax and area under the concentration-time curve [AUC]) was consistent across visits. No evidence of accumulation with once-daily dosing was observed. Total anti-vosoritide antibody (TAb) responses were detected in the serum of 25 of 60 (42%) treated patients in the phase III study, with no apparent impact of TAb development noted on annualized growth velocity or vosoritide exposure. Across the exposure range obtained with 15 µg/kg in the phase III study, no meaningful correlations between vosoritide plasma exposure and changes in annualized growth velocity or CXM, or changes from predose heart rate, and systolic or diastolic blood pressures were observed.

Conclusions: The results support the recommended dose of vosoritide 15 µg/kg for once-daily subcutaneous administration in patients with achondroplasia aged ≥ 5 years whose epiphyses are not closed.

Clinical trials registration: NCT02055157, NCT03197766, and NCT01603095.

Conflict of interest statement

Ming Liang Chan, Yulan Qi, Kevin Larimore, Anu Cherukuri, Lori Seid, Kala Jayaram, Elena Fisheleva, Jonathan Day, Alice Huntsman Labed and Joshua Henshaw are employees of BioMarin Pharmaceutical Inc. Ravi Savarirayan and Klaus Mohnike have received consulting fees and grants from BioMarin. Melita Irving has received consulting fees from BioMarin. Carlos A. Bacino has received consulting fees, honoraria and grants from BioMarin. Julie Hoover-Fong has received consulting fees from BioMarin, Therachon and Ascendis, and grants from BioMarin. Keiichi Ozono has received consulting fees and honoraria from BioMarin. William R. Wilcox was a consultant to BioMarin and received an honoraria, and was the principal investigator for the clinical trial contract with Emory University. William A. Horton is an inventor on a patent application “Type X collagen assay and methods of use thereof”, submitted by Shriners Hospitals for Children. He has consulted for and/or received speaker honoraria from BioMarin, TherAchon (Pfizer), Ascendis, QED, Relay Therapeutics, Fortress Biotech, OPKO, and Medicell Technologies.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Mean (+SD) concentration-time profiles of vosoritide in plasma observed in the phases II and III studies. a Mean (+SD) concentration-time profiles of vosoritide in plasma observed on day 1 in the phase II study, color-coded by study cohort and dose (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). Each line represents the mean plasma vosoritide concentration across patients receiving 2.5, 7.5, 15.0 or 30.0 µg/kg vosoritide on day 1. Error bars represent the standard deviation of the mean, and only the upper error bars are shown. b Mean (+SD) concentration-time profiles of vosoritide in plasma observed at 15 µg/kg in the phase III study, color-coded by visits. Each line represents the mean plasma vosoritide concentration across patients for each visit. Error bars represent the standard deviation of the mean, and only the upper error bars are shown. The LLOQ for the PK assays used in the phases II and III studies were 391 pg/mL and 137 pg/mL in neat plasma, respectively. SD standard error, LLOQ lower limit of quantitation, PK pharmacokinetic
Fig. 2
Fig. 2
Distribution of individual mean vosoritide AUC0-∞ in plasma by serum TAb status (negative or positive) in the phase III study. The line inside the box represents the median and the box represents the limits of the middle half of the data. The range of the box, from the first quartile (Q1) to the third quartile (Q3), defines the IQR. The standard span of the data is defined within the range from Q1 − 1.5 × IQR to Q3+1.5 × IQR. Whiskers are drawn to the nearest value not beyond the range of the standard span; points beyond are drawn as individual open circles. Distribution of the individual mean vosoritide Cmax is similar and is shown in Fig. S4 of the ESM. AUC0-∞ area under the plasma concentration-time curve from time zero to infinity, TAb total anti-vosoritide antibody, IQR interquartile range, Cmax maximum observed plasma concentration, ESM electronic supplementary material
Fig. 3
Fig. 3
Change in annualized growth velocity from baseline by individual mean AUC in the phase II and III studies. a Points represent the individual change in annualized growth velocity from baseline to day 183 of the phase II study and individual mean vosoritide AUC0-t, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high individual mean AUC0-t (4.28 × 106 pg-min/mL) relative to other patients, but a within-range change in annualized growth velocity (3.56 cm/year). This patient was included in the fit through the data, but was not included in this figure. b Points represent the individual change in annualized growth velocity from baseline to month 24 of the phase II study and individual mean vosoritide AUC0-60min, color-coded by study cohort (C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). c Points represent the individual change in annualized growth velocity from baseline from the phase III study and individual mean vosoritide AUC0-60min. Solid lines represent the fits through the data and the shaded regions represent the 95% confidence intervals. The respective analyses using the individual mean vosoritide Cmax are similar and are shown in Fig. S5 in the ESM. AUC area under the plasma concentration-time curve, AUC0-t AUC from time zero to the time of the last measurable concentration, AUC0-60min area under the plasma concentration-time curve from time zero to 60 min postdose, Cmax maximum observed plasma concentration, ESM electronic supplementary material
Fig. 4
Fig. 4
Change in mean CXM by individual mean AUC in the phase II and III studies. (a) Points represent the individual mean CXM from day 10 to day 183 of the phase II study and individual mean vosoritide AUC0-t, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high individual mean AUC0-t (4.28 × 106 pg-min/mL) relative to other patients, but a within-range mean CXM (12,200 pg/mL). This patient was included in the fit through the data but was not included in this figure. b Points represent the individual mean CXM from day 10 to month 24 of the phase II study and individual mean vosoritide AUC0-60min, color-coded by study cohort (C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). c Points represent the individual mean CXM from week 13 to week 52 in the phase III study and individual mean vosoritide AUC0-60min. Solid lines represent the fits through the data and the shaded regions represent the 95% confidence intervals. The respective analyses using the individual mean vosoritide Cmax are similar and are shown in Fig. S6 in the ESM. AUC0-t area under the plasma concentration-time curve from time zero to the time of the last measurable concentration, AUC0-60min area under the plasma concentration-time curve from time zero to 60 min postdose, CXM collagen type X marker, Cmax maximum observed plasma concentration, ESM electronic supplementary material
Fig. 5
Fig. 5
Distribution of the change from predose to maximum postdose urine cGMP/Cr levels observed in the phase III study. The line inside the box represents the median and the box represents the limits of the middle half of the data. The range of the box, from the first quartile (Q1) to the third quartile (Q3), defines the IQR. The standard span of the data is defined within the range from Q1 − 1.5 × IQR to Q3 + 1.5 × IQR. Whiskers are drawn to the nearest value not beyond the range of the standard span; points beyond are drawn as individual open circles. cGMP cyclic guanosine monophosphate, cGMP/Cr cGMP normalized to creatinine, IQR interquartile range
Fig. 6
Fig. 6
Distribution of the change in CXM from baseline from week 13 to week 52 of the phase III study. The line inside the box represents the median and the box represents the limits of the middle half of the data. The range of the box, from the first quartile (Q1) to the third quartile (Q3), defines the IQR. The standard span of the data is defined within the range from Q1 − 1.5 × IQR to Q3 + 1.5 × IQR. Whiskers are drawn to the nearest value not beyond the range of the standard span; points beyond are drawn as individual open circles. CXM collagen type X marker, IQR interquartile range
Fig. 7
Fig. 7
Visit-matched maximum increase in urine cGMP/Cr by individual AUC in the phase II and III studies. a Points represent the individual patient urine cGMP/Cr data and individual plasma vosoritide AUC0-t at each visit during the initial 6 months of the phase II study, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high AUC0-t (1.63 × 107 pg-min/mL) relative to other patients, but a within-range maximum change from pre-dose cGMP/Cr (8190 pmol/mg Cr). This patient was included in the fit through the data but was not included in this figure. b Points represent the individual patient urine cGMP/Cr data and individual plasma vosoritide AUC0-60min at each visit during the phase III study. Solid lines represent Emax model fits through the data and the shaded regions represent the 95% confidence intervals. The respective analyses using the individual plasma vosoritide Cmax are similar and are shown in Fig. S7 in the ESM. cGMP cyclic guanosine monophosphate, cGMP/Cr cGMP normalized to creatinine, AUC0-t area under the plasma concentration-time curve from time zero to the time of the last measurable concentration, AUC0-60min area under the plasma concentration-time curve from time zero to 60 min postdose, Emax maximum possible effect, ESM electronic supplementary material
Fig. 8
Fig. 8
Visit-matched vosoritide Cmax in plasma and maximum increase in heart rate in the phase II and III studies. a Points represent the individual patient maximum increase in HR and individual plasma vosoritide Cmax at each visit during the initial 6 months of the phase II study, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high Cmax (8.39 × 105 pg/mL) relative to other patients, but a within-range maximum increase in HR (39 bpm). This patient was included in the fit through the data but was not included in this figure. b Points represent the individual patient maximum increase in HR and individual plasma vosoritide Cmax at each visit during the phase III study. Solid lines represent the linear fits through the data and the shaded regions represent the 95% confidence interval. The respective analyses using the individual plasma vosoritide AUC is similar and is shown in Fig. S8 in the ESM. Cmax maximum observed plasma concentration, HR heart rate, bpm beats per min, AUC area under the plasma concentration-time curve, ESM electronic supplementary material
Fig. 9
Fig. 9
Visit-matched vosoritide Cmax in plasma and maximum decrease in systolic and diastolic blood pressures in the phase II and III studies. a Points represent the individual patient maximum decrease in SBP and individual plasma vosoritide Cmax at each visit during the initial 6 months of the phase II study, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high Cmax (8.39 × 105 pg/mL) relative to other patients, but a within-range maximum decrease in SBP (−7 mmHg). This patient was included in the fit through the data but was not included in this figure. b Points represent the individual patient maximum decrease in SBP and individual plasma vosoritide Cmax at each visit during the phase III study. c Points represent the individual patient maximum decrease in DBP and individual plasma vosoritide Cmax at each visit during the initial 6 months of the phase II study, color-coded by study cohort (C1: Cohort 1, 2.5 µg/kg; C2: Cohort 2, 7.5 µg/kg; C3: Cohort 3, 15 µg/kg; C4: Cohort 4, 30 µg/kg). One patient had a high Cmax (8.39 × 105 pg/mL) relative to other patients, but a within-range maximum decrease in DBP (−2 mmHg). This patient was included in the fit through the data but was not included in this figure. d Points represent the individual patient maximum decrease in DBP and individual plasma vosoritide Cmax at each visit during the phase III study. Solid lines represent the linear fits through the data and the shaded regions represent the 95% confidence interval. The respective analyses using the individual patient AUC are similar and are shown in Fig. S9 in the ESM. Cmax maximum observed plasma concentration, SBP systolic blood pressure, DBP diastolic blood pressure, mmHg millimeter of mercury, ESM electronic supplementary material

References

    1. Merker A, Neumeyer L, Hertel NT, Grigelioniene G, Mohnike K, Hagenäs L. Development of body proportions in achondroplasia: Sitting height, leg length, arm span, and foot length. Am J Med Genet A. 2018;176(9):1819–1829.
    1. Vajo Z, Francomano CA, Wilkin DJ. The molecular and genetic basis of fibroblast growth factor receptor 3 disorders: the achondroplasia family of skeletal dysplasias, muenke craniosynostosis, and crouzon syndrome with acanthosis nigricans. Endocr Rev. 2000;21(1):23–39.
    1. Wright MJ, Irving MD. Clinical management of achondroplasia. Arch Dis Child. 2012;97(2):129–134.
    1. Foldynova-Trantirkova S, Wilcox WR, Krejci P. Sixteen years and counting: The current understanding of fibroblast growth factor receptor 3 (fgfr3) signaling in skeletal dysplasias. Hum Mutat. 2011;33(1):29–41.
    1. Horton WA, Hall JG, Hecht JT. Achondroplasia. Lancet. 2007;370(9582):162–172.
    1. Krakow D. Skeletal dysplasias. Clin Perinatol. 2015;42(2):301–19.
    1. Alade Y, Tunkel D, Schulze K, McGready J, Jallo G, Ain M, et al. Cross-sectional assessment of pain and physical function in skeletal dysplasia patients. Clin Genet. 2013;84(3):237–243.
    1. Hunter AG, Bankier A, Rogers JG, Sillence D, Scott CI. Medical complications of achondroplasia: a multicentre patient review. J Med Genet. 1998;35(9):705–712.
    1. Ireland PJ, Johnson S, Donaghey S, Johnston L, McGill J, Zankl A, et al. Developmental milestones in infants and young australasian children with achondroplasia. J Dev Behav Pediatr. 2010;31(1):41–47.
    1. Mahomed NN, Spellmann M, Goldberg MJ. Functional health status of adults with achondroplasia. Am J Med Genet. 1998;78(1):30–35.
    1. Ranke MB, Wit JM. Growth hormone—past, present and future. Nat Rev Endocrinol. 2018;14(5):285–300.
    1. Harada D, Namba N, Hanioka Y, Ueyama K, Sakamoto N, Nakano Y, et al. Final adult height in long-term growth hormone-treated achondroplasia patients. Eur J Pediatr. 2017;176(7):873–879.
    1. Kanaka-Gantenbein C. Present status of the use of growth hormone in short children with bone diseases (diseases of the skeleton) J Pediatr Endocrinol Metab. 2001;14(1):17–26.
    1. Miccoli M, Bertelloni S, Massart F. Height outcome of recombinant human growth hormone treatment in achondroplasia children: a meta-analysis. Horm Res Paediatr. 2016;86(1):27–34.
    1. Donaldson J, Aftab S, Bradish C. Achondroplasia and limb lengthening: results in a UK cohort and review of the literature. J Orthop. 2015;12(1):31–34.
    1. Yasoda A, Kazuwa N. Translational research of c-type natriuretic peptide (CNP) into skeletal dysplasias. Endocr J. 2010;57(8):659–666.
    1. Kozhemyakina E, Lassar AB, Zelzer E. A pathway to bone: Signaling molecules and transcription factors involved in chondrocyte development and maturation. Development. 2015;142(5):817–831.
    1. Chusho H, Tamura N, Ogawa Y, Yasoda A, Suda M, Miyazawa T, et al. Dwarfism and early death in mice lacking c-type natriuretic peptide. Proc Natl Acad Sci USA. 2001;98(7):4016–4021.
    1. Tamura N, Doolittle LK, Hammer RE, Shelton JM, Richardson JA, Garbers DL. Critical roles of the guanylyl cyclase b receptor in endochondral ossification and development of female reproductive organs. Proc Natl Acad Sci USA. 2004;101(49):17300–17305.
    1. Bocciardi R, Giorda R, Buttgereit J, Gimelli S, Divizia MT, Beri S, et al. Overexpression of the c-type natriuretic peptide (CNP) is associated with overgrowth and bone anomalies in an individual with balanced t(2;7) translocation. Hum Mutat. 2007;28(7):724–731.
    1. Kake T, Kitamura H, Adachi Y, Yoshioka T, Watanabe T, Matsushita H, et al. Chronically elevated plasma c-type natriuretic peptide level stimulates skeletal growth in transgenic mice. Am J Physiol Endocrinol Metabol. 2009;297(6):E1339–E1348.
    1. Ko JM, Bae J-S, Choi JS, Miura K, Lee HR, Kim O-H, et al. Skeletal overgrowth syndrome caused by overexpression of c-type natriuretic peptide in a girl with balanced chromosomal translocation, t(1;2)(q41;q37.1). Am J Med Genet A. 2015;167A(5):1033–8.
    1. Moncla A, Missirian C, Cacciagli P, Balzamo E, Legeai-Mallet L, Jouve J-L, et al. A cluster of translocation breakpoints in 2q37 is associated with overexpression of NPPC in patients with a similar overgrowth phenotype. Hum Mutat. 2007;28(12):1183–1188.
    1. Wendt DJ, Dvorak-Ewell M, Bullens S, Lorget F, Bell SM, Peng J, et al. Neutral endopeptidase-resistant c-type natriuretic peptide variant represents a new therapeutic approach for treatment of fibroblast growth factor receptor 3–related dwarfism. J Pharmacol Exp Ther. 2015;353(1):132–149.
    1. Yasoda A, Kitamura H, Fujii T, Kondo E, Murao N, Miura M, et al. Systemic administration of c-type natriuretic peptide as a novel therapeutic strategy for skeletal dysplasias. Endocrinology. 2009;150(7):3138–3144.
    1. Yasoda A, Komatsu Y, Chusho H, Miyazawa T, Ozasa A, Miura M, et al. Overexpression of cnp in chondrocytes rescues achondroplasia through a mapk-dependent pathway. Nat Med. 2004;10(1):80–86.
    1. Krejci P, Masri B, Fontaine V, Mekikian PB, Weis M, Prats H, et al. Interaction of fibroblast growth factor and c-natriuretic peptide signaling in regulation of chondrocyte proliferation and extracellular matrix homeostasis. J Cell Sci. 2005;118(21):5089–5100.
    1. Lorget F, Kaci N, Peng J, Benoist-Lasselin C, Mugniery E, Oppeneer T, et al. Evaluation of the therapeutic potential of a cnp analog in a fgfr3 mouse model recapitulating achondroplasia. Am J Hum Genet. 2012;91(6):1108–1114.
    1. Potter LR, Yoder AR, Flora DR, Antos LK, Dickey DM. Natriuretic peptides: Their structures, receptors, physiologic functions and therapeutic applications. Handb Exp Pharmacol. 2009;191:341–366.
    1. Coghlan RF, Oberdorf JA, Sienko S, Aiona MD, Boston BA, Connelly KJ, et al. A degradation fragment of type x collagen is a real-time marker for bone growth velocity. Sci Transl Med. 2017;9(419):eaan4669.
    1. Shen G. The role of type x collagen in facilitating and regulating endochondral ossification of articular cartilage. Orthod Craniofac Res. 2005;8(1):11–17.
    1. Coghlan RF, Olney RC, Boston BA, Coleman DT, Johnstone B, Horton WA. Norms for clinical use of CXM, a real-time marker of height velocity. J Clin Endocrinol Metab. 2021;106(1):e255–e264.
    1. Savarirayan R, Irving M, Bacino CA, Bostwick B, Charrow J, Cormier-Daire V, et al. C-type natriuretic peptide analogue therapy in children with achondroplasia. New Engl J Med. 2019;381(1):25–35.
    1. Savarirayan RTL, Irving M, Wilcox W, Bacino CA, Hoover-Fong J, Font RU, et al. Once-daily, subcutaneous vosoritide therapy in children with achondroplasia: a randomised, double-blind, phase 3, placebo-controlled, multicentre trial. Lancet. 2020;396(10252):684–692.
    1. Li D. Strategic approaches to optimizing peptide adme properties. AAPS J. 2015;17(1):134–143.
    1. Diao L, Meibohm B. Pharmacokinetics and pharmacokinetic–pharmacodynamic correlations of therapeutic peptides. Clin Pharmacokinet. 2013;52(10):855–868.
    1. Potter LR. Natriuretic peptide metabolism, clearance and degradation. FEBS J. 2011;278(11):1808–1817.
    1. Meibohm B. Pharmacokinetics and pharmacodynamics of peptide and protein therapeutics. In: Crommelin DJA, Sindelar RD, Meibohm B, editors. Pharmaceutical biotechnology. New York: Springer; 2013. pp. 101–132.
    1. Santhekadur PK, Kumar DP, Seneshaw M, Mirshahi F, Sanyal AJ. The multifaceted role of natriuretic peptides in metabolic syndrome. Biomed Pharmacother. 2017;92:826–835.
    1. Chirmule N, Jawa V, Meibohm B. Immunogenicity to therapeutic proteins: Impact on pk/pd and efficacy. AAPS J. 2012;14(2):296–302.
    1. Haymond M, Kappelgaard A-M, Czernichow P, Biller BM, Takano K, Kiess W, et al. Early recognition of growth abnormalities permitting early intervention. Acta Paediatr. 2013;102(8):787–796.
    1. del Pino M, Fano V, Adamo P. Growth in achondroplasia, from birth to adulthood, analysed by the jpa-2 model. J Pediatr Endocrinol Metab. 2020;33(12):1589–1595.
    1. Hoover-Fong JE, Schulze KJ, McGready J, Barnes H, Scott CI. Age-appropriate body mass index in children with achondroplasia: interpretation in relation to indexes of height. Am J Clin Nutr. 2008;88(2):364–371.
    1. Madsen A, Fredwall SO, Maanum G, Henriksen C, Slettahjell HB. Anthropometrics, diet, and resting energy expenditure in norwegian adults with achondroplasia. Am J Med Genet A. 2019;179(9):1745–1755.

Source: PubMed

3
購読する