Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Breast Cancer Res Treat ; 179(1): 67-77, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31562570

ABSTRACT

PURPOSE: Fulvestrant is a selective estrogen receptor downregulator (SERD) that is approved for first- or second-line use as a single agent or in combination with cyclin dependent kinase or phosphatidylinositol 3-kinase inhibitors for the treatment of metastatic breast cancer. Fulvestrant exhibits exceptionally effective antitumor activity in preclinical models of breast cancer, a success that has been attributed to its robust SERD activity despite modest receptor downregulation in patient tumors. By modeling human exposures in animal models we probe the absolute need for SERD activity. METHODS: Three xenograft models of endocrine therapy-resistant breast cancer were used to evaluate the efficacy of fulvestrant administered in doses historically used in preclinical studies in the field or by using a dose regimen intended to model clinical exposure levels. Pharmacokinetic and pharmacodynamic analyses were conducted to evaluate plasma exposure and intratumoral ER downregulation. RESULTS: A clinically relevant 25 mg/kg dose of fulvestrant exhibited antitumor efficacy comparable to the historically used 200 mg/kg dose, but at this lower dose it did not result in robust ER downregulation. Further, the antitumor efficacy of the lower dose of fulvestrant was comparable to that observed for other oral SERDs currently in development. CONCLUSION: The use of clinically unachievable exposure levels of fulvestrant as a benchmark in preclinical development of SERDs may negatively impact the selection of those molecules that are advanced for clinical development. Further, these studies suggest that antagonist efficacy, as opposed to SERD activity, is likely to be the primary driver of clinical response.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacokinetics , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/pharmacokinetics , Fulvestrant/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Hormonal/administration & dosage , Dose-Response Relationship, Drug , Estrogen Receptor Antagonists/administration & dosage , Estrogen Receptor alpha/antagonists & inhibitors , Female , Fulvestrant/administration & dosage , Mice , Xenograft Model Antitumor Assays
2.
J Med Chem ; 64(16): 11841-11856, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34251202

ABSTRACT

Breast cancer remains a leading cause of cancer death in women, representing a significant unmet medical need. Here, we disclose our discovery efforts culminating in a clinical candidate, 35 (GDC-9545 or giredestrant). 35 is an efficient and potent selective estrogen receptor degrader (SERD) and a full antagonist, which translates into better antiproliferation activity than known SERDs (1, 6, 7, and 9) across multiple cell lines. Fine-tuning the physiochemical properties enabled once daily oral dosing of 35 in preclinical species and humans. 35 exhibits low drug-drug interaction liability and demonstrates excellent in vitro and in vivo safety profiles. At low doses, 35 induces tumor regressions either as a single agent or in combination with a CDK4/6 inhibitor in an ESR1Y537S mutant PDX or a wild-type ERα tumor model. Currently, 35 is being evaluated in Phase III clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carbolines/therapeutic use , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Carbolines/chemistry , Carbolines/pharmacokinetics , Dogs , Estrogen Receptor Antagonists/chemistry , Estrogen Receptor Antagonists/pharmacokinetics , Female , Humans , MCF-7 Cells , Macaca fascicularis , Mice , Molecular Structure , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
3.
J Med Chem ; 62(3): 1593-1608, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30640465

ABSTRACT

Herein, we report the identification and synthesis of a series of tricyclic indazoles as a novel class of selective estrogen receptor degrader antagonists. Replacement of a phenol, present in our previously reported tetrahydroisoquinoline scaffold, with an indazole group led to the removal of a reactive metabolite signal in an in vitro glutathione trapping assay. Further optimization, guided by X-ray crystal structures and NMR conformational work, varied the alkyl side chain and pendant aryl group and resulted in compounds with low turnover in human hepatocytes and enhanced chemical stability. Compound 9 was profiled as a representative of the series in terms of pharmacology and demonstrated the desired estrogen receptor α degrader-antagonist profile and demonstrated activity in a xenograft model of breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/therapeutic use , Heterocyclic Compounds, 3-Ring/therapeutic use , Indazoles/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Dogs , Drug Screening Assays, Antitumor , Estrogen Receptor Antagonists/chemical synthesis , Estrogen Receptor Antagonists/pharmacokinetics , Estrogen Receptor alpha/metabolism , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Indazoles/chemical synthesis , Indazoles/pharmacokinetics , MCF-7 Cells , Male , Mice, SCID , Microsomes, Liver/metabolism , Molecular Structure , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
4.
Cancer Chemother Pharmacol ; 83(1): 151-160, 2019 01.
Article in English | MEDLINE | ID: mdl-30386887

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.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/pharmacokinetics , Estrogen Receptor alpha/antagonists & inhibitors , Hepatocytes/metabolism , Indazoles/pharmacology , Indazoles/pharmacokinetics , Microsomes, Liver/metabolism , Pyridines/pharmacology , Pyridines/pharmacokinetics , Animals , Biological Availability , Cells, Cultured , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Dogs , Drug Evaluation, Preclinical , Female , Hepatocytes/drug effects , Humans , In Vitro Techniques , Macaca fascicularis , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Protein Binding , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
5.
Expert Opin Drug Metab Toxicol ; 13(5): 537-544, 2017 May.
Article in English | MEDLINE | ID: mdl-27696906

ABSTRACT

INTRODUCTION: Endometrial cancer is the most common malignancy of the female genital tract in industrialized countries. The traditional treatment of endometrial cancer is based on a surgical approach. In recent years, systemic endocrine therapy has demonstrated good efficacy in recurrent or metastatic setting, delaying progression, ameliorating quality of life and palliating symptoms. Areas covered: Phase I and II studies on selective estrogen receptor down-regulators used for the treatment of endometrial cancer treatment have been reviewed. The pharmacokinetic and pharmacodynamic features of selective receptor down-regulators have been also investigated. Expert opinion: Selective estrogen receptor down-regulators may exhibit clinical efficacy in the treatment of gynecological malignancies due to their pure estrogen receptor antagonist properties. However, up to now data are still limited and some unsolved questions remain. Fulvestrant has poor oral bioavailability and low pharmacodynamic characteristics. Further trials are required to examine new selective estrogen receptor down-regulator agents with better pharmacodynamic and pharmacokinetic profiles.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Endometrial Neoplasms/drug therapy , Estradiol/analogs & derivatives , Animals , Antineoplastic Agents, Hormonal/pharmacokinetics , Antineoplastic Agents, Hormonal/pharmacology , Biological Availability , Disease Progression , Endometrial Neoplasms/pathology , Estradiol/pharmacokinetics , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogen Receptor Antagonists/pharmacokinetics , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant , Humans , Quality of Life
SELECTION OF CITATIONS
SEARCH DETAIL