Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 63(13): 7186-7210, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32453591

ABSTRACT

Acquired resistance to fulvestrant and palbociclib is a new challenge to treatment of estrogen receptor positive (ER+) breast cancer. ER is expressed in most resistance settings; thus, bromodomain and extra-terminal protein inhibitors (BETi) that target BET-amplified ER-mediated transcription have therapeutic potential. Novel pyrrolopyridone BETi leveraged novel interactions with L92/L94 confirmed by a cocrystal structure of 27 with BRD4. Optimization of BETi using growth inhibition in fulvestrant-resistant (MCF-7:CFR) cells was confirmed in endocrine-resistant, palbociclib-resistant, and ESR1 mutant cell lines. 27 was more potent in MCF-7:CFR cells than six BET inhibitors in clinical trials. Transcriptomic analysis differentiated 27 from the benchmark BETi, JQ-1, showing downregulation of oncogenes and upregulation of tumor suppressors and apoptosis. The therapeutic approach was validated by oral administration of 27 in orthotopic xenografts of endocrine-resistant breast cancer in monotherapy and in combination with fulvestrant. Importantly, at an equivalent dose in rats, thrombocytopenia was mitigated.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Fulvestrant/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Humans , MCF-7 Cells , Mice , Models, Molecular , Protein Domains , Pyridones/pharmacokinetics , Receptors, Estrogen/metabolism , Tissue Distribution , Transcription Factors/chemistry , Xenograft Model Antitumor Assays
2.
Breast Cancer Res Treat ; 180(3): 635-646, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32130619

ABSTRACT

PURPOSE: The combination of targeting the CDK4/6 and estrogen receptor (ER) signaling pathways with palbociclib and fulvestrant is a proven therapeutic strategy for the treatment of ER-positive breast cancer. However, the poor physicochemical properties of fulvestrant require monthly intramuscular injections to patients, which limit the pharmacokinetic and pharmacodynamic activity of the compound. Therefore, an orally available compound that more rapidly reaches steady state may lead to a better clinical response in patients. Here, we report the identification of G1T48, a novel orally bioavailable, non-steroidal small molecule antagonist of ER. METHODS: The pharmacological effects and the antineoplastic mechanism of action of G1T48 on tumors was evaluated using human breast cancer cells (in vitro) and xenograft efficacy models (in vivo). RESULTS: G1T48 is a potent and efficacious inhibitor of estrogen-mediated transcription and proliferation in ER-positive breast cancer cells, similar to the pure antiestrogen fulvestrant. In addition, G1T48 can effectively suppress ER activity in multiple models of endocrine therapy resistance including those harboring ER mutations and growth factor activation. In vivo, G1T48 has robust antitumor activity in a model of estrogen-dependent breast cancer (MCF7) and significantly inhibited the growth of tamoxifen-resistant (TamR), long-term estrogen-deprived (LTED) and patient-derived xenograft tumors with an increased response being observed with the combination of G1T48 and the CDK4/6 inhibitor lerociclib. CONCLUSIONS: These data show that G1T48 has the potential to be an efficacious oral antineoplastic agent in ER-positive breast cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , HIV Antibodies/pharmacology , Neoplasms, Hormone-Dependent/drug therapy , Selective Estrogen Receptor Modulators/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Estrogen Antagonists/pharmacology , Female , Humans , Mice , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
J Med Chem ; 62(24): 11301-11323, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31746603

ABSTRACT

The clinical steroidal selective estrogen receptor (ER) degrader (SERD), fulvestrant, is effective in metastatic breast cancer, but limited by poor pharmacokinetics, prompting the development of orally bioavailable, nonsteroidal SERDs, currently in clinical trials. These trials address local breast cancer as well as peripheral metastases, but patients with brain metastases are generally excluded because of the lack of blood-brain barrier penetration. A novel family of benzothiophene SERDs with a basic amino side arm (B-SERDs) was synthesized. Proteasomal degradation of ERα was induced by B-SERDs that achieved the objectives of oral and brain bioavailability, while maintaining high affinity binding to ERα and both potency and efficacy comparable to fulvestrant in cell lines resistant to endocrine therapy or bearing ESR1 mutations. A novel 3-oxyazetidine side chain was designed, leading to 37d, a B-SERD that caused endocrine-resistant ER+ tumors to regress in a mouse orthotopic xenograft model.


Subject(s)
Breast Neoplasms/drug therapy , Drug Design , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor alpha/metabolism , Proteolysis/drug effects , Selective Estrogen Receptor Modulators/chemical synthesis , Selective Estrogen Receptor Modulators/pharmacology , Thiophenes/chemical synthesis , Thiophenes/pharmacology , Animals , Apoptosis , Aromatase Inhibitors/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Estrogen Receptor alpha/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Mutation , Rats , Rats, Sprague-Dawley , Selective Estrogen Receptor Modulators/pharmacokinetics , Thiophenes/chemistry , Thiophenes/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Med Chem ; 60(4): 1325-1342, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28117994

ABSTRACT

Resistance to the selective estrogen receptor modulator tamoxifen and to aromatase inhibitors that lower circulating estradiol occurs in up to 50% of patients, generally leading to an endocrine-independent ER+ phenotype. Selective ER downregulators (SERDs) are able to ablate ER and thus, theoretically, to prevent survival of both endocrine-dependent and -independent ER+ tumors. The clinical SERD fulvestrant is hampered by intramuscular administration and undesirable pharmacokinetics. Novel SERDs were designed using the 6-OH-benzothiophene (BT) scaffold common to arzoxifene and raloxifene. Treatment-resistant (TR) ER+ cell lines (MCF-7:5C and MCF-7:TAM1) were used for optimization, followed by validation in the parent endocrine-dependent cell line (MCF-7:WS8), in 2D and 3D cultures, using ERα in-cell westerns, ERE-luciferase, and cell viability assays, with 2 (GDC-0810/ARN-810) used for comparison. Two BT SERDs with superior in vitro activity to 2 were studied for bioavailability and shown to cause regression of a TR, endocrine-independent ER+ xenograft superior to that with 2.


Subject(s)
Antineoplastic Agents, Hormonal/chemistry , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast/drug effects , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/therapeutic use , Thiophenes/chemistry , Thiophenes/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Mice, Nude , Models, Molecular , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Thiophenes/pharmacology
5.
J Med Chem ; 59(1): 219-237, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26681208

ABSTRACT

Almost 70% of breast cancers are estrogen receptor α (ERα) positive. Tamoxifen, a selective estrogen receptor modulator (SERM), represents the standard of care for many patients; however, 30-50% develop resistance, underlining the need for alternative therapeutics. Paradoxically, agonists at ERα such as estradiol (E2) have demonstrated clinical efficacy in patients with heavily treated breast cancer, although side effects in gynecological tissues are unacceptable. A drug that selectively mimics the actions of E2 in breast cancer therapy but minimizes estrogenic effects in other tissues is a novel, therapeutic alternative. We hypothesized that a selective human estrogen receptor partial agonist (ShERPA) at ERα would provide such an agent. Novel benzothiophene derivatives with nanomolar potency in breast cancer cell cultures were designed. Several showed partial agonist activity, with potency of 0.8-76 nM, mimicking E2 in inhibiting growth of tamoxifen-resistant breast cancer cell lines. Three ShERPAs were tested and validated in xenograft models of endocrine-independent and tamoxifen-resistant breast cancer, and in contrast to E2, ShERPAs did not cause significant uterine growth.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/therapeutic use , Animals , Cell Line, Tumor , Drug Design , Estrogen Receptor alpha/drug effects , Female , Humans , Mice , Models, Molecular , Structure-Activity Relationship , Uterus/drug effects , Uterus/growth & development , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...