Paltusotine, a novel oral once-daily nonpeptide SST2 receptor agonist, suppresses GH and IGF-1 in healthy volunteers

Ajay Madan, Stacy Markison, Stephen F Betz, Alan Krasner, Rosa Luo, Theresa Jochelson, Jason Lickliter, R Scott Struthers, Ajay Madan, Stacy Markison, Stephen F Betz, Alan Krasner, Rosa Luo, Theresa Jochelson, Jason Lickliter, R Scott Struthers

Abstract

Purpose: Evaluate the pharmacodynamics, pharmacokinetics, and safety of paltusotine, an orally bioavailable, nonpeptide, somatostatin receptor subtype 2 (SST2) agonist being developed for the treatment of acromegaly and neuroendocrine tumors.

Methods: A randomized, double-blind, placebo-controlled, single center, single and multiple ascending dose phase 1 study was conducted in healthy male volunteers who received (i) single-dose of oral paltusotine 1.25, 2.5, 5, 10, and 20 mg (solution); and 40 and 60 mg (capsules) or (ii) multiple-dose oral paltusotine capsules once daily 5 mg (× 7 days), 10, 20, and 30 mg (× 10 days). Main outcome measures were pharmacodynamics (changes in growth hormone-releasing hormone [GHRH] stimulated growth hormone [GH] and insulin-like growth factor 1 [IGF-1]), pharmacokinetics, safety, and tolerability.

Results: Single-dose cohorts: n = 41 active, n = 14 placebo. Multiple-dose cohorts: n = 24 active, n = 12 placebo. Paltusotine was well tolerated, orally bioavailable, associated with increased plasma concentrations to doses up to 40 mg, and was eliminated with a half-life of approximately 30 h. Single-dose paltusotine 1.25 to 20 mg suppressed GHRH-stimulated GH secretion by 44% to 93% compared to 15% with placebo. Multiple-dose paltusotine 5 to 30 mg administered once daily for 10 days suppressed IGF-1 by 19% to 37% compared to an increase of 2.4% with placebo.

Conclusions: Paltusotine suppresses GH and IGF-1 in a dose-dependent fashion, with a safety profile similar to currently approved SST2 receptor ligands. Paltusotine is a promising once-daily oral nonpeptide SST2 agonist candidate for managing acromegaly and neuroendocrine tumors.

Trial registration: NCT03276858, registered September 8, 2017, retrospectively registered.

Keywords: Acromegaly; IGF-1; Paltusotine; SST2; Somatostatin receptor agonist.

Conflict of interest statement

Ajay Madan, Stacy Markison, Stephen F. Betz, Alan Krasner, Rosa Luo, and R. Scott Struthe are employees of Crinetics Pharmaceuticals, Inc. Theresa Jochelson is a consultant for Crinetics Pharmaceuticals, Inc. Jason Lickliter is an employee of Nucleus Network.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Trial design for single- and multiple-dose cohorts. Dose escalation in a new cohort, soln solution, caps capsule. *n = 8 per single-dose cohort (6 active, 2 placebo), except for cohort S5 in which one participant withdrew prior to dosing and was not replaced (n = 7; 5 active, 2 placebo). †n = 9 per multiple-dose cohort (6 active, 3 placebo). **There were 3 study periods in cohort S4: period A (10 mg solution); period B (10 mg capsules) and period C (10 mg capsules with food). ‡Single-dose pharmacodynamic population (n = 39; paltusotine n = 29; placebo n = 10) included participants in S1, S2, S3, S4A, and S5 cohorts. GHRH challenge was administered on Day 1 and 2 h post-dose on Day 1. §Multiple-dose pharmacodynamic population (n = 31; paltusotine n = 21; placebo n = 10) included participants from the M1, M2, M3, and M4 cohorts who received all doses
Fig. 2
Fig. 2
Growth hormone suppression with single-dose paltusotine. Data shown in panels a and b are mean ± SEM. **P GH growth hormone, GHRH growth hormone-releasing hormone, AUC0-2 area under the serum concentration time curve from 0 to 2 h; EC80 = 80% GH suppression. On Day 1, GHRH was administered 2 h post paltusotine dose. On Day -1, GHRH was administered at approximately the same time of the day as Day 1. a Plasma GH concentrations upon GHRH challenge on Day -1, and Day 1 on paltusotine 10 mg. GH levels were measured at the same time of the day after a GHRH challenge on Day -1 (open circles) and on Day 1 (closed circles). Plasma paltusotine concentrations are shown in black squares. Mean peak GH response on Day 1 and Day 1 were 21 ng/mL and 1.7 ng/mL, respectively. b Mean GHRH-stimulated GH AUC0-2 percent of baseline (Day 1/Day 1) by each dose group. P values were calculated for each dose of paltusotine compared to placebo using ANOVA with Dunnett multiple comparison adjustment. c. Suppression of GH AUC0-2 post GHRH (Day-1–Day 1)/Day-1 as a function of paltusotine concentration (average of 2-, 3-, and 4-h post-dose)
Fig. 3
Fig. 3
Suppression of IGF-1 with repeated administration of paltusotine. Data shown are mean ± SEM. *P IGF-1 insulin-like growth factor-1; GHRH = growth hormone-releasing hormone. a Plasma paltusotine (open squares) and serum IGF-1 (closed circles) concentrations at 10 mg multiple dose from baseline (Day 1) to 10–11 days post-dose (Days 20–21). For this dose group, IGF-1 samples were obtained pre-dose, 6 and 12 h post-dose on Days 1, 1 and 10; pre-dose and 12 h post-dose on Days 2–9; and a single sample at approximately the same time as dosing on Days 11, 14, and 21. Arrows represent days on which paltusotine was administered (Days 1 to 10). Paltusotine concentrations are trough plasma concentrations 24 h post-dose on Days 2 to 11 and measured concentrations on Day 12 to 20 after the last dose on Day 10. Baseline IGF-1 was the average of all pre-first dose values. For each of the subsequent days, during the treatment period, each day’s pre-dose IGF-1 value was averaged with the previous day’s value (e.g., Day 2 pre-dose value was averaged with Day 1 post-dose value). b Dose–response for IGF-1 suppression relative to baseline with multiple-dose paltusotine. Values are IGF-1 percent of baseline 12 h post-dose on Day 7 (5 mg dose group) and on Day 10 (10 mg, 20 mg, 30 mg dose groups). Baseline was the average of Day -1 and Day 1 pre-dose IGF-1 results. P-values were calculated for each dose of paltusotine compared to placebo using ANOVA with Dunnett multiple comparison adjustment

References

    1. Patel YC, Srikant CB. Somatostatin receptors. Trends Endocrinol Metab. 1997;8(10):398–405. doi: 10.1016/s1043-2760(97)00168-9.
    1. Reisine T, Bell GI. Molecular biology of somatostatin receptors. Endocr Rev. 1995;16(4):427–442. doi: 10.1210/edrv-16-4-427.
    1. Reubi JC, Schaer JC, Markwalder R, Waser B, Horisberger U, Laissue J. Distribution of somatostatin receptors in normal and neoplastic human tissues: recent advances and potential relevance. Yale J Biol Med. 1997;70(5–6):471–479.
    1. Strosberg JR, Halfdanarson TR, Bellizzi AM, Chan JA, Dillon JS, Heaney AP, et al. The North American Neuroendocrine Tumor Society consensus guidelines for surveillance and medical management of midgut neuroendocrine tumors. Pancreas. 2017;46(6):707–714. doi: 10.1097/mpa.0000000000000850.
    1. Carmichael JD, Bonert VS, Nuño M, Ly D, Melmed S. Acromegaly clinical trial methodology impact on reported biochemical efficacy rates of somatostatin receptor ligand treatments: a meta-analysis. J Clin Endocrinol Metab. 2014;99(5):1825–1833. doi: 10.1210/jc.2013-3757.
    1. Gadelha MR, Bronstein MD, Brue T, Coculescu M, Fleseriu M, Guitelman M, et al. Pasireotide versus continued treatment with octreotide or lanreotide in patients with inadequately controlled acromegaly (PAOLA): a randomised, phase 3 trial. Lancet Diabetes Endocrinol. 2014;2(11):875–884. doi: 10.1016/s2213-8587(14)70169-x.
    1. Esposito D, Ragnarsson O, Granfeldt D, Marlow T, Johannsson G, Olsson DS. Decreasing mortality and changes in treatment patterns in patients with acromegaly from a nationwide study. Eur J Endocrinol. 2018;178(5):459–469. doi: 10.1530/eje-18-0015.
    1. Strasburger CJ, Karavitaki N, Störmann S, Trainer PJ, Kreitschmann-Andermahr I, Droste M, et al. Patient-reported outcomes of parenteral somatostatin analogue injections in 195 patients with acromegaly. Eur J Endocrinol. 2016;174(3):355–362. doi: 10.1530/eje-15-1042.
    1. Giustina A, Barkan A, Beckers A, Biermasz N, Biller BMK, Boguszewski C, et al. A consensus on the diagnosis and treatment of acromegaly comorbidities: an update. J Clin Endocrinol Metab. 2020 doi: 10.1210/clinem/dgz096.
    1. Katznelson L, Laws ER, Jr, Melmed S, Molitch ME, Murad MH, Utz A, et al. Acromegaly: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. 2014;99(11):3933–3951. doi: 10.1210/jc.2014-2700.
    1. Boyd AE, DeFord LL, Mares JE, Leary CC, Garris JL, Dagohoy CG, et al. Improving the success rate of gluteal intramuscular injections. Pancreas. 2013;42(5):878–882. doi: 10.1097/MPA.0b013e318279d552.
    1. Melmed S, Popovic V, Bidlingmaier M, Mercado M, van der Lely AJ, Biermasz N, et al. Safety and efficacy of oral octreotide in acromegaly: results of a multicenter phase III trial. J Clin Endocrinol Metab. 2015;100(4):1699–1708. doi: 10.1210/jc.2014-4113.
    1. Betz SF, Markison S, Kusnetzow AK, Athanacio JD, Rico-Bautista E, Madan A, et al. Suppression of growth hormone and insulin-like growth factor 1 in rats after oral administration of CRN00808, a small molecule, SST2 selective somatostatin biased agonist. Endocrine Rev. 2018;39(2):i1–i1417.
    1. Madan A, Juo R, Ferrara-Cook C, Struthers RS, van Marle S, Krasner A (2020) Absorption, metabolism, excretion and absolute oral bioavailability [14C]-CRN00808, an orally bioavailable, nonpeptide, selective, somatostatin receptor 2 (sst2) biased agonist for the treatment of acromegaly. OR23–05. J Endo Soc. 4(Suppl):A352-A3.
    1. Hingorani P, Karnad DR, Rohekar P, Kerkar V, Lokhandwala YY, Kothari S. Arrhythmias seen in baseline 24-hour Holter ECG recordings in healthy normal volunteers during phase 1 clinical trials. J Clin Pharmacol. 2016;56(7):885–893. doi: 10.1002/jcph.679.
    1. Min SS, Turner JR, Nada A, DiMino TL, Hynie I, Kleiman R, et al. Evaluation of ventricular arrhythmias in early clinical pharmacology trials and potential consequences for later development. Am Heart J. 2010;159(5):716–729. doi: 10.1016/j.ahj.2010.02.004.
    1. Tiberg F, Roberts J, Cervin C, Johnsson M, Sarp S, Tripathi AP, et al. Octreotide sc depot provides sustained octreotide bioavailability and similar IGF-1 suppression to octreotide LAR in healthy volunteers. Br J Clin Pharmacol. 2015;80(3):460–472. doi: 10.1111/bcp.12698.
    1. SIGNIFOR LAR [prescribing information]. Lebanon, NJ: Recordati Rare Diseases, Inc (2020)
    1. Giusti M, Gussoni G, Cuttica CM, Giordano G. Effectiveness and tolerability of slow release lanreotide treatment in active acromegaly: six-month report on an Italian multicenter study. Italian Multicenter Slow Release Lanreotide Study Group. J Clin Endocrinol Metab. 1996;81(6):2089–2097. doi: 10.1210/jcem.81.6.8964833.
    1. SANDOSTATIN LAR DEPOT [Prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals Corporation (2019)
    1. Mazziotti G, Floriani I, Bonadonna S, Torri V, Chanson P, Giustina A. Effects of somatostatin analogs on glucose homeostasis: a metaanalysis of acromegaly studies. J Clin Endocrinol Metab. 2009;94(5):1500–1508. doi: 10.1210/jc.2008-2332.
    1. Henry RR, Ciaraldi TP, Armstrong D, Burke P, Ligueros-Saylan M, Mudaliar S. Hyperglycemia associated with pasireotide: results from a mechanistic study in healthy volunteers. J Clin Endocrinol Metab. 2013;98(8):3446–3453. doi: 10.1210/jc.2013-1771.
    1. SOMATULINE DEPOT [prescribing informaiton]. Cambridge, MA: Ipsen Biopharmaceuticals, Inc (2019)
    1. MYCAPSSA [prescribing information]. Needham, MA: Chiasma (2020)
    1. Wesolowski CA, Wesolowski MJ, Babyn PS, Wanasundara SN. Time varying apparent volume of distribution and drug half-lives following intravenous bolus injections. PLoS ONE. 2016;11(7):e0158798. doi: 10.1371/journal.pone.0158798.
    1. Madan A, Luo R, Ferrara-Cook C, Struthers RS, van Marle S, Krasner A. Absolute oral bioavailability and absorption, metabolism, excretion of [14C]-Labeled paltusotine (CRN00808), an orally bioavailable, nonpeptide, selective, somatostatin receptor 2 (sst2) biased agonist for the treatment of acromegaly. Endocr Abstr. 2020;70:AEP627. doi: 10.1530/endoabs.70.AEP627.
    1. Lou R, Burke G, Mui C, Shakib S, Ferrara-Cook C, Krasner A et al. Pharmacokinetics and safety of an improved oral formulation of paltusotine, a selective, non-peptide somatostatin receptor 2 (SST2) agonist for the treatment of acromegaly. Endocrine Society 2021. March 20, 2021. 2021. . Accessed 7 Feb 2021
    1. Golor G, Hu K, Ruffin M, Buchelt A, Bouillaud E, Wang Y, et al. A first-in-man study to evaluate the safety, tolerability, and pharmacokinetics of pasireotide (SOM230), a multireceptor-targeted somatostatin analog, in healthy volunteers. Drug Des Devel Ther. 2012;6:71–79. doi: 10.2147/dddt.S29125.
    1. Kuhn JM, Basin C, Mollard M, de Rougé B, Baudoin C, Obach R, et al. Pharmacokinetic study and effects on growth hormone secretion in healthy volunteers of the new somatostatin analogue BIM 23014. Eur J Clin Pharmacol. 1993;45(1):73–77. doi: 10.1007/bf00315353.
    1. Tuvia S, Atsmon J, Teichman SL, Katz S, Salama P, Pelled D, et al. Oral octreotide absorption in human subjects: comparable pharmacokinetics to parenteral octreotide and effective growth hormone suppression. J Clin Endocrinol Metab. 2012;97(7):2362–2369. doi: 10.1210/jc.2012-1179.
    1. Kuhn JM, Legrand A, Ruiz JM, Obach R, De Ronzan J, Thomas F. Pharmacokinetic and pharmacodynamic properties of a long-acting formulation of the new somatostatin analogue, lanreotide, in normal healthy volunteers. Br J Clin Pharmacol. 1994;38(3):213–219. doi: 10.1111/j.1365-2125.1994.tb04344.x.
    1. Trocóniz IF, Cendrós JM, Peraire C, Ramis J, Garrido MJ, Boscani PF, et al. Population pharmacokinetic analysis of lanreotide Autogel in healthy subjects : evidence for injection interval of up to 2 months. Clin Pharmacokinet. 2009;48(1):51–62. doi: 10.2165/0003088-200948010-00004.
    1. Coopmans EC, Korevaar TIM, van Meyel SWF, Daly AF, Chanson P, Brue T, et al. Multivariable prediction mdel for biochemical response to first-generation somatostatin receptor ligands in acromegaly. J Clin Endocrinol Metab. 2020;105(9):2964–2974. doi: 10.1210/clinem/dgaa387.
    1. Schonbrunn A. Selective agonism in somatostatin receptor signaling and regulation. Mol Cell Endocrinol. 2008;286(1–2):35–39. doi: 10.1016/j.mce.2007.09.009.
    1. Oldfield EH, Jane JA, Jr, Thorner MO, Pledger CL, Sheehan JP, Vance ML. Correlation between GH and IGF-1 during treatment for acromegaly. J Neurosurg. 2017;126(6):1959–1966. doi: 10.3171/2016.8.Jns161123.
    1. Lamberti G, Faggiano A, Brighi N, Tafuto S, Ibrahim T, Brizzi MP, et al. Nonconventional doses of somatostatin analogs in patients with progressing well-differentiated neuroendocrine tumor. J Clin Endocrinol Metab. 2020;105(1):194–200. doi: 10.1210/clinem/dgz035.
    1. Strosberg JR. Systemic treatment of gastroenteropancreatic neuroendocrine tumors (GEP-NETS): current approaches and future options. Endocr Pract. 2014;20(2):167–175. doi: 10.4158/ep13262.Ra.
    1. . A study to evaluate the safety and efficacy of paltusotine for the treatment of acromegaly (ACROBAT Evolve). . Accessed 5 Dec 2020.
    1. . An study to evaluate the safety and efficacy of paltusotine for the treatment of acromegaly (ACROBAT Edge). . Accessed 5 Dec 2020
    1. Chen C, Wu D, Guo Z, Xie Q, Reinhart GJ, Madan A, et al. Discovery of sodium R-(+)-4-{2-[5-(2-fluoro-3-methoxyphenyl)-3-(2-fluoro-6-[trifluoromethyl]benzyl)-4 -methyl-2,6-dioxo-3,6-dihydro-2H-pyrimidin-1-yl]-1-phenylethylamino}butyrate (elagolix), a potent and orally available nonpeptide antagonist of the human gonadotropin-releasing hormone receptor. J Med Chem. 2008;51(23):7478–7485. doi: 10.1021/jm8006454.
    1. Duncia JV, Chiu AT, Carini DJ, Gregory GB, Johnson AL, Price WA, et al. The discovery of potent nonpeptide angiotensin II receptor antagonists: a new class of potent antihypertensives. J Med Chem. 1990;33(5):1312–1329. doi: 10.1021/jm00167a007.
    1. Kubo K, Kohara Y, Yoshimura Y, Inada Y, Shibouta Y, Furukawa Y, et al. Nonpeptide angiotensin II receptor antagonists Synthesis and biological activity of potential prodrugs of benzimidazole-7-carboxylic acids. J Med Chem. 1993;36(16):2343–2349. doi: 10.1021/jm00068a011.

Source: PubMed

3
Abonnieren