Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 1/2019

01-01-2019 | Original Article

Nonclinical pharmacokinetics and in vitro metabolism of H3B-6545, a novel selective ERα covalent antagonist (SERCA)

Authors: Nathalie Rioux, Sherri Smith, Manav Korpal, Morgan O’Shea, Sudeep Prajapati, Guo Zhu Zheng, Markus Warmuth, Peter G. Smith

Published in: Cancer Chemotherapy and Pharmacology | Issue 1/2019

Login to get access

Abstract

Purpose

H3B-6545, a novel selective estrogen receptor (ER)α covalent antagonist (SERCA) which inactivates both wild-type and mutant ERα, is in clinical development for the treatment of metastatic breast cancer. Preclinical studies were conducted to characterize the pharmacokinetics and metabolism of H3B-6545 in rat and monkeys.

Methods

The clearance and metabolic profiles of H3B-6545 were studied using rat, monkey and human hepatocytes, and reaction phenotyping was done using recombinant human cytochrome P450 enzymes. Blood stability, protein binding, and permeability were also determined in vitro. Pharmacokinetics of H3B-6545 was assessed after both intravenous and oral dosing. A nonclinical PBPK model was developed to assess in vitro–in vivo correlation of clearance.

Results

H3B-6545 had a terminal elimination half-life of 2.4 h in rats and 4.0 h in monkeys and showed low to moderate bioavailability, in line with the in vitro permeability assessment. Plasma protein binding was similar across species, at 99.5–99.8%. Nine metabolites of H3B-6545 were identified in hepatocyte incubations, none of which were unique to humans. Formation of glutathione-related conjugate of H3B-6545 was minimal in vitro. H3B-6545, a CYP3A substrate, is expected to be mostly cleared via hepatic phase 1 metabolism. Hepatocyte clearance values were used to adequately model the time-concentration profiles in rat and monkey.

Conclusions

We report on the absorption and metabolic fate and disposition of H3B-6545 in rats and dogs and illustrate that in vitro–in vivo correlation of clearance is possible for targeted covalent inhibitors, provided reactivity is not a predominant mechanism of clearance.
Literature
1.
go back to reference Kauhava L, Immonen-Räihä P, Parvinen I, Holli K, Pylkkänen L, Kaljonen A, Helenius H, Kronqvist P, Klemi PJ (2008) Lower recurrence risk through mammographic screening reduces breast cancer treatment costs. Breast 17:550–554CrossRefPubMed Kauhava L, Immonen-Räihä P, Parvinen I, Holli K, Pylkkänen L, Kaljonen A, Helenius H, Kronqvist P, Klemi PJ (2008) Lower recurrence risk through mammographic screening reduces breast cancer treatment costs. Breast 17:550–554CrossRefPubMed
2.
go back to reference Spicer DV, Pike MC (1993) Breast cancer prevention through modulation of endogenous hormones. Breast Cancer Res Treat 28:179–193CrossRefPubMed Spicer DV, Pike MC (1993) Breast cancer prevention through modulation of endogenous hormones. Breast Cancer Res Treat 28:179–193CrossRefPubMed
3.
go back to reference Beslija S, Bonneterre J, Burstein HJ et al (2009) Third consensus on medical treatment of metastatic breast cancer. Ann Oncol 20:1771–1785CrossRefPubMed Beslija S, Bonneterre J, Burstein HJ et al (2009) Third consensus on medical treatment of metastatic breast cancer. Ann Oncol 20:1771–1785CrossRefPubMed
4.
go back to reference Chandarlapaty S, Chen D, He W et al (2016) Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol 2:1310–1315CrossRefPubMedPubMedCentral Chandarlapaty S, Chen D, He W et al (2016) Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol 2:1310–1315CrossRefPubMedPubMedCentral
6.
go back to reference Leung L, Yang X, Strelevitz TJ, Montgomery J, Brown MF, Zientek MA, Banfield C, Gilbert AM, Thorarensen A, Dowty ME (2017) Clearance prediction of targeted covalent inhibitors by in vitro–in vivo extrapolation of hepatic and extrahepatic clearance mechanisms. Drug Metab Dispos 45:1–7CrossRefPubMed Leung L, Yang X, Strelevitz TJ, Montgomery J, Brown MF, Zientek MA, Banfield C, Gilbert AM, Thorarensen A, Dowty ME (2017) Clearance prediction of targeted covalent inhibitors by in vitro–in vivo extrapolation of hepatic and extrahepatic clearance mechanisms. Drug Metab Dispos 45:1–7CrossRefPubMed
8.
go back to reference Smith PG, Puyang X, Furman C et al (2017) Discovery and development of H3B-6545: a novel, oral, selective estrogen receptor covalent antagonist (SERCA) for the treatment of breast cancer. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017, AACR, Washington DC. Cancer Res 77(13 Suppl):Abstract nr DDT01-04 Smith PG, Puyang X, Furman C et al (2017) Discovery and development of H3B-6545: a novel, oral, selective estrogen receptor covalent antagonist (SERCA) for the treatment of breast cancer. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017, AACR, Washington DC. Cancer Res 77(13 Suppl):Abstract nr DDT01-04
9.
go back to reference Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10:1093–1095CrossRef Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10:1093–1095CrossRef
10.
go back to reference Pollack GM, Brouwer KLR, Demby KB, Jones JA (1990) Determination of hepatic blood flow in the rat using sequential infusions of indocyanine green and galactose. Drug Metab Dispos 18:197–202PubMed Pollack GM, Brouwer KLR, Demby KB, Jones JA (1990) Determination of hepatic blood flow in the rat using sequential infusions of indocyanine green and galactose. Drug Metab Dispos 18:197–202PubMed
11.
go back to reference Nakajima M, Nakamura S, Tokudome S, Shimada N, Yamazaki H, Yokoi T (1999) Azelastine N-demethylation by cytochrome P450 (CYP)3A4, CYP2D6, and CYP1A2 in human liver microsomes: evaluation of approach to predict the contribution of multiple CYPs. Drug Metab Dispos 27:1381–1391PubMed Nakajima M, Nakamura S, Tokudome S, Shimada N, Yamazaki H, Yokoi T (1999) Azelastine N-demethylation by cytochrome P450 (CYP)3A4, CYP2D6, and CYP1A2 in human liver microsomes: evaluation of approach to predict the contribution of multiple CYPs. Drug Metab Dispos 27:1381–1391PubMed
13.
go back to reference Takenaka K, Morgan JA, Scheffer GL, Adachi M, Stewart CF, Sun D, Leggas M, Ejendal KF, Hrycyna CA, Schuetz JD (2007) Substrate overlap between Mrp4 and Abcg2/Bcrp affects purine analogue drug cytotoxicity and tissue distribution. Cancer Res 67:6965–6972CrossRefPubMed Takenaka K, Morgan JA, Scheffer GL, Adachi M, Stewart CF, Sun D, Leggas M, Ejendal KF, Hrycyna CA, Schuetz JD (2007) Substrate overlap between Mrp4 and Abcg2/Bcrp affects purine analogue drug cytotoxicity and tissue distribution. Cancer Res 67:6965–6972CrossRefPubMed
15.
go back to reference Rowland M, Benet LZ, Graham GG (1973) Clearance concepts in pharmacokinetics. J Pharmacokinet Biopharm 1:123–136CrossRefPubMed Rowland M, Benet LZ, Graham GG (1973) Clearance concepts in pharmacokinetics. J Pharmacokinet Biopharm 1:123–136CrossRefPubMed
16.
go back to reference Obach RS (1999) Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos 27:1350–1359PubMed Obach RS (1999) Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos 27:1350–1359PubMed
17.
go back to reference Mackenzie PI, Rogers A, Treloar J, Jorgensen BR, Miners JO, Meech R (2008) Identification of UDP glycosyltransferase 3A1 as a UDP N-acetylglucosaminyltransferase. J Biol Chem 283:36205–36210CrossRefPubMedPubMedCentral Mackenzie PI, Rogers A, Treloar J, Jorgensen BR, Miners JO, Meech R (2008) Identification of UDP glycosyltransferase 3A1 as a UDP N-acetylglucosaminyltransferase. J Biol Chem 283:36205–36210CrossRefPubMedPubMedCentral
18.
go back to reference Stopfer P, Marzin K, Narjes H, Gansser D, Shahidi M, Uttereuther-Fischer M, Ebner T (2012) Afatinib pharmacokinetics and metabolism after oral administration to healthy male volunteers. Cancer Chemother Pharmacol 69:1051–1061CrossRefPubMed Stopfer P, Marzin K, Narjes H, Gansser D, Shahidi M, Uttereuther-Fischer M, Ebner T (2012) Afatinib pharmacokinetics and metabolism after oral administration to healthy male volunteers. Cancer Chemother Pharmacol 69:1051–1061CrossRefPubMed
19.
go back to reference Scheers E, Leclercq L, de Jong J, Bode N, Bockx M, Laenen A, Cuyckens F, Skee D, Murphy J, Sukbuntherng J, Mannens G (2015) Absorption, metabolism, and excretion of oral 14C radiolabeled ibrutinib: an open-label, phase I, single-dose study in healthy men. Drug Metab Dispos 43:289–297CrossRefPubMed Scheers E, Leclercq L, de Jong J, Bode N, Bockx M, Laenen A, Cuyckens F, Skee D, Murphy J, Sukbuntherng J, Mannens G (2015) Absorption, metabolism, and excretion of oral 14C radiolabeled ibrutinib: an open-label, phase I, single-dose study in healthy men. Drug Metab Dispos 43:289–297CrossRefPubMed
20.
go back to reference Baillie TA (2016) Targeted covalent inhibitors for drug design. Angew Chem Int Ed 55:13408–13421CrossRef Baillie TA (2016) Targeted covalent inhibitors for drug design. Angew Chem Int Ed 55:13408–13421CrossRef
21.
go back to reference Shibata Y, Chiba M (2015) The role of extrahepatic metabolism in the pharmacokinetics of the targeted covalent inhibitors afatinib, ibrutinib, and neratinib. Drug Metab Dispos 43:375–384CrossRefPubMed Shibata Y, Chiba M (2015) The role of extrahepatic metabolism in the pharmacokinetics of the targeted covalent inhibitors afatinib, ibrutinib, and neratinib. Drug Metab Dispos 43:375–384CrossRefPubMed
22.
go back to reference Commandeur JN, Stijntjes GJ, Vermeulen NP (1995) Enzymes and transport systems involved in the formation and disposition of glutathione S-conjugates. Role of bioactivation and detoxification mechanisms of xenobiotics. Pharmacol Rev 47:271–330PubMed Commandeur JN, Stijntjes GJ, Vermeulen NP (1995) Enzymes and transport systems involved in the formation and disposition of glutathione S-conjugates. Role of bioactivation and detoxification mechanisms of xenobiotics. Pharmacol Rev 47:271–330PubMed
23.
go back to reference Dickinson PA, Cantarini MV, Collier J, Frewer P, Martin S, Pickup K, Ballard P (2016) Metabolic disposition of osimertinib in rats, dogs, and humans: insights into a drug designed to bind covalently to a cysteine residue of epidermal growth factor receptor. Drug Metab Dispos 44:1201–1212CrossRefPubMed Dickinson PA, Cantarini MV, Collier J, Frewer P, Martin S, Pickup K, Ballard P (2016) Metabolic disposition of osimertinib in rats, dogs, and humans: insights into a drug designed to bind covalently to a cysteine residue of epidermal growth factor receptor. Drug Metab Dispos 44:1201–1212CrossRefPubMed
24.
go back to reference Block M (2015) Physiologically based pharmacokinetic and pharmacodynamic modeling in cancer drug development: status, potential and gaps. Expert Opin Drug Metab Toxicol 11:743–756CrossRefPubMed Block M (2015) Physiologically based pharmacokinetic and pharmacodynamic modeling in cancer drug development: status, potential and gaps. Expert Opin Drug Metab Toxicol 11:743–756CrossRefPubMed
Metadata
Title
Nonclinical pharmacokinetics and in vitro metabolism of H3B-6545, a novel selective ERα covalent antagonist (SERCA)
Authors
Nathalie Rioux
Sherri Smith
Manav Korpal
Morgan O’Shea
Sudeep Prajapati
Guo Zhu Zheng
Markus Warmuth
Peter G. Smith
Publication date
01-01-2019
Publisher
Springer Berlin Heidelberg
Published in
Cancer Chemotherapy and Pharmacology / Issue 1/2019
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-018-3716-3

Other articles of this Issue 1/2019

Cancer Chemotherapy and Pharmacology 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine