Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353221

ABSTRACT

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , HEK293 Cells , Heterografts , Humans , Indazoles/pharmacology , Ligands , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Polymorphism, Single Nucleotide , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
2.
iScience ; 13: 488-497, 2019 Mar 29.
Article in English | MEDLINE | ID: mdl-30898619

ABSTRACT

Werner syndrome protein (WRN) is a RecQ enzyme involved in the maintenance of genome integrity. Germline loss-of-function mutations in WRN led to premature aging and predisposition to cancer. We evaluated synthetic lethal (SL) interactions between WRN and another human RecQ helicase, BLM, with DNA damage response genes in cancer cell lines. We found that WRN was SL with a DNA mismatch repair protein MutL homolog 1, loss of which is associated with high microsatellite instability (MSI-H). MSI-H cells exhibited increased double-stranded DNA breaks, altered cell cycles, and decreased viability in response to WRN knockdown, in contrast to microsatellite stable (MSS) lines, which tolerated depletion of WRN. Although WRN is the only human RecQ enzyme with a distinct exonuclease domain, only loss of helicase activity drives the MSI SL interaction. This SL interaction in MSI cancer cells positions WRN as a relevant therapeutic target in patients with MSI-H tumors.

3.
ACS Med Chem Lett ; 10(1): 50-55, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30655946

ABSTRACT

The further optimization of ER-α degradation efficacy of a series of ER modulators by refining side-chain substitution led to efficacious selective estrogen receptor degraders (SERDs). A fluoromethyl azetidine group was found to be preferred and resulted in the identification of bis-phenol chromene 17ha. In a tamoxifen-resistant breast cancer xenograft model, 17ha (ER-α degradation efficacy = 97%) demonstrated tumor regression, together with robust reduction of intratumoral ER-α levels. However, despite superior oral exposure, 5a (ER-α degradation efficacy = 91%) had inferior activity. This result suggests that optimizing ER-α degradation efficacy leads to compounds with robust effects in a model of tamoxifen-resistant breast cancer. Compound 17ha (GDC-0927) was evaluated in clinical trials in women with metastatic estrogen receptor-positive breast cancer.

5.
Bioorg Med Chem Lett ; 29(3): 367-372, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30587451
6.
J Med Chem ; 61(17): 7917-7928, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30086626

ABSTRACT

About 75% of breast cancers are estrogen receptor alpha (ER-α) positive, and women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, but resistance often emerges. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and shows some activity in patients who have progressed on antihormonal agents. However, fulvestrant must be administered by intramuscular injections that limit its efficacy. We describe the optimization of ER-α degradation efficacy of a chromene series of ER modulators resulting in highly potent and efficacious SERDs such as 14n. When examined in a xenograft model of tamoxifen-resistant breast cancer, 14n (ER-α degradation efficacy = 91%) demonstrated robust activity, while, despite superior oral exposure, 15g (ER-α degradation efficacy = 82%) was essentially inactive. This result suggests that optimizing ER-α degradation efficacy in the MCF-7 cell line leads to compounds with robust effects in models of tamoxifen-resistant breast cancer derived from an MCF-7 background.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzopyrans/chemistry , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Estrogen Receptor alpha/metabolism , Selective Estrogen Receptor Modulators/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice , Rats , Selective Estrogen Receptor Modulators/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Elife ; 52016 07 13.
Article in English | MEDLINE | ID: mdl-27410477

ABSTRACT

ER-targeted therapeutics provide valuable treatment options for patients with ER+ breast cancer, however, current relapse and mortality rates emphasize the need for improved therapeutic strategies. The recent discovery of prevalent ESR1 mutations in relapsed tumors underscores a sustained reliance of advanced tumors on ERα signaling, and provides a strong rationale for continued targeting of ERα. Here we describe GDC-0810, a novel, non-steroidal, orally bioavailable selective ER downregulator (SERD), which was identified by prospectively optimizing ERα degradation, antagonism and pharmacokinetic properties. GDC-0810 induces a distinct ERα conformation, relative to that induced by currently approved therapeutics, suggesting a unique mechanism of action. GDC-0810 has robust in vitro and in vivo activity against a variety of human breast cancer cell lines and patient derived xenografts, including a tamoxifen-resistant model and those that harbor ERα mutations. GDC-0810 is currently being evaluated in Phase II clinical studies in women with ER+ breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Cinnamates/administration & dosage , Indazoles/administration & dosage , Receptors, Estrogen/administration & dosage , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Mice , Prospective Studies , Rats , Treatment Outcome
8.
Bioorg Med Chem Lett ; 25(22): 5163-7, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26463130

ABSTRACT

Selective estrogen receptor degraders (SERDs) have shown promise for the treatment of ER+ breast cancer. Disclosed herein is the continued optimization of our indazole series of SERDs. Exploration of ER degradation and antagonism in vitro followed by in vivo antagonism and oral exposure culminated in the discovery of indazoles 47 and 56, which induce tumor regression in a tamoxifen-resistant breast cancer xenograft.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor Antagonists/therapeutic use , Indazoles/therapeutic use , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cinnamates/therapeutic use , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/metabolism , Female , Indazoles/chemistry , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
9.
J Med Chem ; 58(12): 4888-904, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-25879485

ABSTRACT

Approximately 80% of breast cancers are estrogen receptor alpha (ER-α) positive, and although women typically initially respond well to antihormonal therapies such as tamoxifen and aromatase inhibitors, resistance often emerges. Although a variety of resistance mechanism may be at play in this state, there is evidence that in many cases the ER still plays a central role, including mutations in the ER leading to constitutively active receptor. Fulvestrant is a steroid-based, selective estrogen receptor degrader (SERD) that both antagonizes and degrades ER-α and is active in patients who have progressed on antihormonal agents. However, fulvestrant suffers from poor pharmaceutical properties and must be administered by intramuscular injections that limit the total amount of drug that can be administered and hence lead to the potential for incomplete receptor blockade. We describe the identification and characterization of a series of small-molecule, orally bioavailable SERDs which are potent antagonists and degraders of ER-α and in which the ER-α degrading properties were prospectively optimized. The lead compound 11l (GDC-0810 or ARN-810) demonstrates robust activity in models of tamoxifen-sensitive and tamoxifen-resistant breast cancer, and is currently in clinical trials in women with locally advanced or metastatic estrogen receptor-positive breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/metabolism , Proteolysis/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Small Molecule Libraries/therapeutic use , Tamoxifen/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Dogs , Drug Discovery , Drug Resistance, Neoplasm/drug effects , Female , Heterografts , Humans , Mice , Rats , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics
10.
J Clin Oncol ; 31(28): 3525-30, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24002508

ABSTRACT

PURPOSE: ARN-509 is a novel androgen receptor (AR) antagonist for the treatment of castration-resistant prostate cancer (CRPC). ARN-509 inhibits AR nuclear translocation and AR binding to androgen response elements and, unlike bicalutamide, does not exhibit agonist properties in the context of AR overexpression. This first-in-human phase I study assessed safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of ARN-509 in men with metastatic CRPC. PATIENTS AND METHODS: Thirty patients with progressive CRPC received continuous daily oral ARN-509 at doses between 30 and 480 mg, preceded by administration of a single dose followed by a 1-week observation period with pharmacokinetic sampling. Positron emission tomography/computed tomography imaging was conducted to monitor [(18)F]fluoro-α-dihydrotestosterone (FDHT) binding to AR in tumors before and during treatment. Primary objective was to determine pharmacokinetics, safety, and recommended phase II dose. RESULTS: Pharmacokinetics were linear and dose proportional. Prostate-specific antigen declines at 12 weeks (≥ 50% reduction from baseline) were observed in 46.7% of patients. Reduction in FDHT uptake was observed at all doses, with a plateau in response at ≥ 120-mg dose, consistent with saturation of AR binding. The most frequently reported adverse event was grade 1/2 fatigue (47%). One dose-limiting toxicity event (grade 3 abdominal pain) occurred at the 300-mg dose. Dose escalation to 480 mg did not identify a maximum-tolerated dose. CONCLUSION: ARN-509 was safe and well tolerated, displayed dose-proportional pharmacokinetics, and demonstrated pharmacodynamic and antitumor activity across all dose levels tested. A maximum efficacious dose of 240 mg daily was selected for phase II exploration based on integration of preclinical and clinical data.


Subject(s)
Androgen Antagonists/therapeutic use , Androgen Receptor Antagonists/therapeutic use , Bone Neoplasms/drug therapy , Castration , Prostatic Neoplasms/drug therapy , Thiohydantoins/therapeutic use , Aged , Aged, 80 and over , Androgen Antagonists/pharmacokinetics , Androgen Receptor Antagonists/pharmacokinetics , Bone Neoplasms/secondary , Diagnostic Imaging , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Grading , Neoplastic Cells, Circulating/pathology , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Thiohydantoins/pharmacokinetics , Tissue Distribution
11.
Cancer Discov ; 3(9): 1020-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23779130

ABSTRACT

UNLABELLED: Despite the impressive clinical activity of the second-generation antiandrogens enzalutamide and ARN-509 in patients with prostate cancer, acquired resistance invariably emerges. To identify the molecular mechanisms underlying acquired resistance, we developed and characterized cell lines resistant to ARN-509 and enzalutamide. In a subset of cell lines, ARN-509 and enzalutamide exhibit agonist activity due to a missense mutation (F876L) in the ligand-binding domain of the androgen receptor (AR). AR F876L is sufficient to confer resistance to ARN-509 and enzalutamide in in vitro and in vivo models of castration-resistant prostate cancer (CRPC). Importantly, the AR F876L mutant is detected in plasma DNA from ARN-509-treated patients with progressive CRPC. Thus, selective outgrowth of AR F876L is a clinically relevant mechanism of second-generation antiandrogen resistance that can potentially be targeted with next-generation antiandrogens. SIGNIFICANCE: A missense mutation in the ligand-binding domain of the androgen receptor F876L confers resistance to the second-generation antiandrogens enzalutamide and ARN-509 in preclinical models of AR function and prostate cancer and is detected in plasma DNA from ARN-509-treated patients with progressive disease. These results chart a new path for the discovery and development of next-generation antiandrogens that could be coupled with a blood-based companion diagnostic to guide treatment decisions.


Subject(s)
Androgen Antagonists/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Thiohydantoins/therapeutic use , Benzamides , Cell Line, Tumor , DNA/blood , Drug Resistance, Neoplasm/genetics , Humans , Male , Molecular Targeted Therapy , Mutation, Missense/genetics , Nitriles , Phenylthiohydantoin/therapeutic use , Receptors, Androgen/genetics
12.
Cancer Res ; 72(6): 1494-503, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22266222

ABSTRACT

Continued reliance on the androgen receptor (AR) is now understood as a core mechanism in castration-resistant prostate cancer (CRPC), the most advanced form of this disease. While established and novel AR pathway-targeting agents display clinical efficacy in metastatic CRPC, dose-limiting side effects remain problematic for all current agents. In this study, we report the discovery and development of ARN-509, a competitive AR inhibitor that is fully antagonistic to AR overexpression, a common and important feature of CRPC. ARN-509 was optimized for inhibition of AR transcriptional activity and prostate cancer cell proliferation, pharmacokinetics, and in vivo efficacy. In contrast to bicalutamide, ARN-509 lacked significant agonist activity in preclinical models of CRPC. Moreover, ARN-509 lacked inducing activity for AR nuclear localization or DNA binding. In a clinically valid murine xenograft model of human CRPC, ARN-509 showed greater efficacy than MDV3100. Maximal therapeutic response in this model was achieved at 30 mg/kg/d of ARN-509, whereas the same response required 100 mg/kg/d of MDV3100 and higher steady-state plasma concentrations. Thus, ARN-509 exhibits characteristics predicting a higher therapeutic index with a greater potential to reach maximally efficacious doses in man than current AR antagonists. Our findings offer preclinical proof of principle for ARN-509 as a promising therapeutic in both castration-sensitive and castration-resistant forms of prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Prostatic Neoplasms/drug therapy , Thiohydantoins/therapeutic use , Androgen Antagonists/pharmacokinetics , Anilides/pharmacokinetics , Anilides/therapeutic use , Animals , Antineoplastic Agents, Hormonal/blood , Antineoplastic Agents, Hormonal/pharmacokinetics , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Nitriles/pharmacokinetics , Nitriles/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/blood , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/therapeutic use , Rats , Receptors, Androgen/drug effects , Thiohydantoins/blood , Thiohydantoins/chemical synthesis , Thiohydantoins/pharmacokinetics , Tosyl Compounds/pharmacokinetics , Tosyl Compounds/therapeutic use , Xenograft Model Antitumor Assays
13.
BMC Cancer ; 10: 11, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20067634

ABSTRACT

BACKGROUND: The X-linked Inhibitor of Apoptosis (XIAP) has attracted much attention as a cancer drug target. It is the only member of the IAP family that can directly inhibit caspase activity in vitro, and it can regulate apoptosis and other biological processes through its C-terminal E3 ubiquitin ligase RING domain. However, there is controversy regarding XIAP's role in regulating tumor cell proliferation and survival under normal growth conditions in vitro. METHODS: We utilized siRNA to systematically knock down XIAP in ten human tumor cell lines and then monitored both XIAP protein levels and cell viability over time. To examine the role of XIAP in the intrinsic versus extrinsic cell death pathways, we compared the viability of XIAP depleted cells treated either with a variety of mechanistically distinct, intrinsic pathway inducing agents, or the canonical inducer of the extrinsic pathway, TNF-related apoptosis-inducing ligand (TRAIL). RESULTS: XIAP knockdown had no effect on the viability of six cell lines, whereas the effect in the other four was modest and transient. XIAP knockdown only sensitized tumor cells to TRAIL and not the mitochondrial pathway inducing agents. CONCLUSIONS: These data indicate that XIAP has a more central role in regulating death receptor mediated apoptosis than it does the intrinsic pathway mediated cell death.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , X-Linked Inhibitor of Apoptosis Protein/metabolism , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Mitochondria/metabolism , Models, Biological , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/chemistry
14.
PLoS One ; 4(2): e4455, 2009.
Article in English | MEDLINE | ID: mdl-19209227

ABSTRACT

Tumor cell death is modulated by an intrinsic cell death pathway controlled by the pro- and anti-apoptotic members of the Bcl-2 family. Up-regulation of anti-apoptotic Bcl-2 family members has been shown to suppress cell death in pre-clinical models of human cancer and is implicated in human tumor progression. Previous gain-of-function studies in the RIP1-Tag2 model of pancreatic islet carcinogenesis, involving uniform or focal/temporal over-expression of Bcl-x(L), demonstrated accelerated tumor formation and growth. To specifically assess the role of endogenous Bcl-x in regulating apoptosis and tumor progression in this model, we engineered a pancreatic beta-cell-specific knockout of both alleles of Bcl-x using the Cre-LoxP system of homologous recombination. Surprisingly, there was no appreciable effect on tumor cell apoptosis rates or on tumor growth in the Bcl-x knockout mice. Other anti-apoptotic Bcl-2 family members were expressed but not substantively altered at the mRNA level in the Bcl-x-null tumors, suggestive of redundancy without compensatory transcriptional up-regulation. Interestingly, the incidence of invasive carcinomas was reduced, and tumor cells lacking Bcl-x were impaired in invasion in a two-chamber trans-well assay under conditions mimicking hypoxia. Thus, while the function of Bcl-x in suppressing apoptosis and thereby promoting tumor growth is evidently redundant, genetic ablation implicates Bcl-x in selectively facilitating invasion, consistent with a recent report documenting a pro-invasive capability of Bcl-x(L) upon exogenous over-expression.


Subject(s)
Apoptosis , Gene Deletion , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , bcl-X Protein/deficiency , Animals , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , GTPase-Activating Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness , Neuroendocrine Tumors/genetics , Organ Specificity , Pancreatic Neoplasms/genetics , Transcription, Genetic , Up-Regulation/genetics , bcl-X Protein/metabolism
15.
Bioorg Med Chem Lett ; 18(23): 6093-6, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18954983

ABSTRACT

We report the identification of KD5170, a potent mercaptoketone-based Class I and II-histone deacetylase inhibitor that demonstrates broad spectrum cytotoxic activity against a range of human tumor-derived cell lines. KD5170 exhibits robust and sustained histone H3 hyperacetylation in HCT-116 xenograft tumors following single oral or i.v. dose and inhibition of tumor growth following chronic dosing.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Prodrugs/pharmacology , Pyridines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Molecular Structure , Prodrugs/chemistry , Pyridines/chemistry , Structure-Activity Relationship , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
16.
Bioorg Med Chem Lett ; 18(24): 6482-5, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18954984

ABSTRACT

In an effort to discover novel non-hydroxamic acid histone deacetylase (HDAC) inhibitors, a novel alpha-mercaptoketone was identified in a high-throughput screen. Lead optimization of the screening hit, led to a number of potent HDAC inhibitors. In particular, alpha-mercaptoketone 19y (KD5150) exhibited nanomolar in vitro activity and inhibition of tumor growth in vivo.


Subject(s)
Drug Screening Assays, Antitumor , Histone Deacetylase Inhibitors , Ketones/chemistry , Antineoplastic Agents/therapeutic use , Chelating Agents/pharmacology , Chemistry, Pharmaceutical , Drug Design , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Models, Chemical , Neoplasms/drug therapy , Prodrugs/chemistry , Structure-Activity Relationship , Zinc/chemistry
17.
Mol Cancer Ther ; 7(6): 1494-505, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18566220

ABSTRACT

Histone deacetylase inhibitors have emerged as promising anticancer drugs. Using an unbiased ultrahigh throughput screening system, a novel mercaptoketone-based histone deacetylase inhibitor series was identified that was optimized to the lead compound, KD5170. KD5170 inhibited the proliferation of myeloma cell lines and the viability of CD138(+) primary myeloma cells by induction of apoptosis, accompanied by an increase of acetylation of histones and activation of caspase-3, caspase-8, and caspase-9. Treatment with KD5170 caused a loss of mitochondrial membrane potential resulting in release of apoptogenic factors such as cytochrome c, Smac, and apoptosis-inducing factor. Furthermore, KD5170 induced oxidative stress and oxidative DNA damage in myeloma cells as evidenced by the up-regulation of heme oxygenase-1 and H2A.X phosphorylation. Combination of KD5170 with proteasome inhibitor bortezomib or tumor necrosis factor-related apoptosis-inducing ligand synergistically enhanced the antimyeloma activity. We further found that resistance of myeloma cells to KD5170 was associated with activation of the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway under treatment with KD5170. Pretreatment with the mitogen-activated protein kinase inhibitor U0126 restored sensitivity to KD5170, suggesting that the combination of KD5170 with U0126 could overcome drug resistance. Growth of myeloma tumor xenografts in KD5170-treated nude mice was significantly inhibited and survival was prolonged. Histone acetylation was increased in spleen and tumor tissues of animals treated with KD5170. Our data indicate that KD5170 has potent antimyeloma activity in vitro and in vivo, which is mediated by DNA damage and mitochondrial signaling and subsequent induction of apoptosis.


Subject(s)
DNA Damage , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Ketones/pharmacology , Mitochondria/metabolism , Multiple Myeloma/enzymology , Pyridines/pharmacology , Signal Transduction/drug effects , Sulfonamides/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Boronic Acids/pharmacology , Bortezomib , Caspases/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Histones/metabolism , Humans , Ketones/chemistry , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects , Protein Transport/drug effects , Pyrazines/pharmacology , Pyridines/chemistry , Sulfonamides/chemistry , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism
18.
Mol Cancer Ther ; 7(5): 1054-65, 2008 May.
Article in English | MEDLINE | ID: mdl-18483295

ABSTRACT

Histone deacetylase (HDAC) inhibitors have garnered significant attention as cancer drugs. These therapeutic agents have recently been clinically validated with the market approval of vorinostat (SAHA, Zolinza) for treatment of cutaneous T-cell lymphoma. Like vorinostat, most of the small-molecule HDAC inhibitors in clinical development are hydroxamic acids, whose inhibitory activity stems from their ability to coordinate the catalytic Zn2+ in the active site of HDACs. We sought to identify novel, nonhydroxamate-based HDAC inhibitors with potentially distinct pharmaceutical properties via an ultra-high throughput small molecule biochemical screen against the HDAC activity in a HeLa cell nuclear extract. An alpha-mercaptoketone series was identified and chemically optimized. The lead compound, KD5170, exhibits HDAC inhibitory activity with an IC50 of 0.045 micromol/L in the screening biochemical assay and an EC50 of 0.025 micromol/L in HeLa cell-based assays that monitor histone H3 acetylation. KD5170 also exhibits broad spectrum classes I and II HDAC inhibition in assays using purified recombinant human isoforms. KD5170 shows significant antiproliferative activity against a variety of human tumor cell lines, including the NCI-60 panel. Significant tumor growth inhibition was observed after p.o. dosing in human HCT-116 (colorectal cancer), NCI-H460 (non-small cell lung carcinoma), and PC-3 (prostate cancer) s.c. xenografts in nude mice. In addition, a significant increase in antitumor activity and time to end-point occurred when KD5170 was combined with docetaxel in xenografts of the PC-3 prostate cancer cell line. The biological and pharmaceutical profile of KD5170 supports its continued preclinical and clinical development as a broad spectrum anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pyridines/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays
19.
Oncogene ; 24(44): 6597-604, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16007165

ABSTRACT

Here, we describe the surprising residual capability of the Rb pathway to negatively regulate proliferation and tumorigenesis in a SV40 large T antigen (Tag)-driven mouse model of pancreatic islet carcinogenesis. Heterogeneous Tag expression during all progression stages suggested that a threshold level of the T antigen oncoprotein might be deterministic for beta-cell hyperproliferation and led us to hypothesize that Tag might not be fully inhibiting the tumor suppressor activity of Rb. Moreover, genomic profiling of these tumors by array CGH pointed to regions of loss on chromosomes 6 and 14, where the Rb pathway inhibitor p27 and Rb itself, respectively, reside. Indeed, genetic ablation of the p27(Kip1) or Rb genes accentuated Tag-induced tumorigenesis, with loss of Rb in particular broadly enhancing multiple parameters of tumorigenesis including the frequency and growth rates of premalignant lesions, of nascent solid tumors, and of invasive carcinomas. The data indicate that attenuation rather than complete inactivation of Rb tumor suppressor gene function, in the context of p53 inhibition, is sufficient to initiate tumorigenesis in this model of islet cell cancer, with the demonstrable possibility that subsequent losses of Rb or its regulators can enhance malignant progression. The results may be relevant to human papillomavirus (HPV)-induced cervical neoplasias where E7 oncogene expression levels or activity (in the case of intermediate/low-risk HPV subtypes) incompletely inhibits Rb tumor suppressor functions, as well as to other neoplasias where initiating oncogenic or tumor suppressor events reduce but do not abrogate Rb function.


Subject(s)
Genes, Retinoblastoma , Islets of Langerhans/pathology , Pancreatic Neoplasms/genetics , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27 , Humans , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Pancreatic Neoplasms/pathology , Tumor Suppressor Proteins/genetics
20.
Cancer Cell ; 5(5): 443-53, 2004 May.
Article in English | MEDLINE | ID: mdl-15144952

ABSTRACT

Tumors develop through successive stages characterized by changes in gene expression and protein function. Gene expression profiling of pancreatic islet tumors in a mouse model of cancer revealed upregulation of cathepsin cysteine proteases. Cathepsin activity was assessed using chemical probes allowing biochemical and in vivo imaging, revealing increased activity associated with the angiogenic vasculature and invasive fronts of carcinomas, and differential expression in immune, endothelial, and cancer cells. A broad-spectrum cysteine cathepsin inhibitor was used to pharmacologically knock out cathepsin function at different stages of tumorigenesis, impairing angiogenic switching in progenitor lesions, as well as tumor growth, vascularity, and invasiveness. Cysteine cathepsins are also upregulated during HPV16-induced cervical carcinogenesis, further encouraging consideration of this protease family as a therapeutic target in human cancers.


Subject(s)
Cathepsins/metabolism , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Islet Cell/blood supply , Carcinoma, Islet Cell/pathology , Cathepsins/antagonists & inhibitors , Cell Transformation, Neoplastic , Cysteine Proteinase Inhibitors/pharmacology , Female , Gene Expression Profiling , Humans , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Staging , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/physiology , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Viral/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...