Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39113611

ABSTRACT

Phenotypic plasticity is a recognized mechanism driving therapeutic resistance in prostate cancer (PCa) patients. While underlying molecular causations driving phenotypic plasticity have been identified, therapeutic success is yet to be achieved. To identify putative master regulator transcription factors (MR-TF) driving phenotypic plasticity in PCa, this work utilized a multiomic approach using genetically engineered mouse models of prostate cancer combined with patient data to identify MYBL2 as a significantly enriched transcription factor in PCa exhibiting phenotypic plasticity. Genetic inhibition of Mybl2 using independent murine PCa cell lines representing phenotypic plasticity demonstrated Mybl2 loss significantly decreased in vivo growth as well as cell fitness and repressed gene expression signatures involved in pluripotency and stemness. Because MYBL2 is currently not druggable, a MYBL2 gene signature was employed to identify cyclin-dependent kinase-2 (CDK2) as a potential therapeutic target. CDK2 inhibition phenocopied genetic loss of Mybl2 and significantly decreased in vivo tumor growth associated with enrichment of DNA damage. Together, this work demonstrates MYBL2 as an important MR-TF driving phenotypic plasticity in PCa. Further, high MYBL2 activity identifies PCa that would be responsive to CDK2 inhibition.

2.
J Med Chem ; 67(15): 13147-13173, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39078366

ABSTRACT

Fungi have historically been the source of numerous important medicinal compounds, but full exploitation of their genetic potential for drug development has been hampered in traditional discovery paradigms. Here we describe a radically different approach, top-down drug discovery (TD3), starting with a massive digital search through a database of over 100,000 fully genomicized fungi to identify loci encoding molecules with a predetermined human target. We exemplify TD3 by the selection of cyclin-dependent kinases (CDKs) as targets and the discovery of two molecules, 1 and 2, which inhibit therapeutically important human CDKs. 1 and 2 exhibit a remarkable mechanism, forming a site-selective covalent bond to the CDK active site Lys. We explored the structure-activity relationship via semi- and total synthesis, generating an analog, 43, with improved kinase selectivity, bioavailability, and efficacy. This work highlights the power of TD3 to identify mechanistically and structurally novel molecules for the development of new medicines.


Subject(s)
Cyclin-Dependent Kinases , Drug Discovery , Protein Kinase Inhibitors , Humans , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Animals , Genomics/methods , Models, Molecular
3.
Nat Cancer ; 2(4): 444-456, 2021 04.
Article in English | MEDLINE | ID: mdl-33899001

ABSTRACT

Prostate cancers are considered to be immunologically 'cold' tumors given the very few patients who respond to checkpoint inhibitor (CPI) therapy. Recently, enrichment of interferon-stimulated genes (ISGs) predicted a favorable response to CPI across various disease sites. The enhancer of zeste homolog-2 (EZH2) is overexpressed in prostate cancer and known to negatively regulate ISGs. In the present study, we demonstrate that EZH2 inhibition in prostate cancer models activates a double-stranded RNA-STING-ISG stress response upregulating genes involved in antigen presentation, Th1 chemokine signaling and interferon response, including programmed cell death protein 1 (PD-L1) that is dependent on STING activation. EZH2 inhibition substantially increased intratumoral trafficking of activated CD8+ T cells and increased M1 tumor-associated macrophages, overall reversing resistance to PD-1 CPI. Our study identifies EZH2 as a potent inhibitor of antitumor immunity and responsiveness to CPI. These data suggest EZH2 inhibition as a therapeutic direction to enhance prostate cancer response to PD-1 CPI.


Subject(s)
Programmed Cell Death 1 Receptor , Prostatic Neoplasms , CD8-Positive T-Lymphocytes , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Interferons/pharmacology , Male , Prostatic Neoplasms/drug therapy , RNA, Double-Stranded
4.
Mol Cancer Res ; 18(8): 1176-1188, 2020 08.
Article in English | MEDLINE | ID: mdl-32461304

ABSTRACT

The increased treatment of metastatic castration-resistant prostate cancer (mCRPC) with second-generation antiandrogen therapies (ADT) has coincided with a greater incidence of lethal, aggressive variant prostate cancer (AVPC) tumors that have lost dependence on androgen receptor (AR) signaling. These AR-independent tumors may also transdifferentiate to express neuroendocrine lineage markers and are termed neuroendocrine prostate cancer (NEPC). Recent evidence suggests kinase signaling may be an important driver of NEPC. To identify targetable kinases in NEPC, we performed global phosphoproteomics comparing several AR-independent to AR-dependent prostate cancer cell lines and identified multiple altered signaling pathways, including enrichment of RET kinase activity in the AR-independent cell lines. Clinical NEPC patient samples and NEPC patient-derived xenografts displayed upregulated RET transcript and RET pathway activity. Genetic knockdown or pharmacologic inhibition of RET kinase in multiple mouse and human models of NEPC dramatically reduced tumor growth and decreased cell viability. Our results suggest that targeting RET in NEPC tumors with high RET expression could be an effective treatment option. Currently, there are limited treatment options for patients with aggressive neuroendocrine prostate cancer and none are curative. IMPLICATIONS: Identification of aberrantly expressed RET kinase as a driver of tumor growth in multiple models of NEPC provides a significant rationale for testing the clinical application of RET inhibitors in patients with AVPC.


Subject(s)
Carcinoma, Neuroendocrine/drug therapy , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Prostatic Neoplasms/drug therapy , Proteomics/methods , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Male , Mice , PC-3 Cells , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
5.
Nat Commun ; 10(1): 278, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655535

ABSTRACT

Neuroendocrine prostate cancer (NEPC), a lethal form of the disease, is characterized by loss of androgen receptor (AR) signaling during neuroendocrine transdifferentiation, which results in resistance to AR-targeted therapy. Clinically, genomically and epigenetically, NEPC resembles other types of poorly differentiated neuroendocrine tumors (NETs). Through pan-NET analyses, we identified ONECUT2 as a candidate master transcriptional regulator of poorly differentiated NETs. ONECUT2 ectopic expression in prostate adenocarcinoma synergizes with hypoxia to suppress androgen signaling and induce neuroendocrine plasticity. ONEUCT2 drives tumor aggressiveness in NEPC, partially through regulating hypoxia signaling and tumor hypoxia. Specifically, ONECUT2 activates SMAD3, which regulates hypoxia signaling through modulating HIF1α chromatin-binding, leading NEPC to exhibit higher degrees of hypoxia compared to prostate adenocarcinomas. Treatment with hypoxia-activated prodrug TH-302 potently reduces NEPC tumor growth. Collectively, these results highlight the synergy between ONECUT2 and hypoxia in driving NEPC, and emphasize the potential of hypoxia-directed therapy for NEPC patients.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Smad3 Protein/genetics , Transcription Factors/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Datasets as Topic , Disease Progression , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neuroendocrine Tumors/pathology , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Smad3 Protein/metabolism , Transcription Factors/genetics , Up-Regulation , Xenograft Model Antitumor Assays
6.
Adv Exp Med Biol ; 1210: 301-318, 2019.
Article in English | MEDLINE | ID: mdl-31900914

ABSTRACT

The RB tumor suppressor is one of the most commonly deleted/mutated genes in human cancers. In prostate cancer specifically, mutation of RB is most frequently observed in aggressive, metastatic disease. As one of the earliest tumor suppressors to be identified, the molecular functions of RB that are lost in tumor development have been studied for decades. Earlier work focused on the canonical RB pathway connecting mitogenic signaling to the cell cycle via Cyclin/CDK inactivation of RB, thereby releasing the E2F transcription factors. More in-depth analysis revealed that RB-E2F complexes regulate cellular processes beyond proliferation. Most recently, "non-canonical" roles for RB function have been expanded beyond its E2F interactions, which may play a particular role in advanced prostate cancer. For example, in mouse models of prostate cancer, loss of RB has been shown to induce lineage plasticity, which enables resistance to androgen deprivation therapy. This increased understanding of the potential downstream functions of RB in prostate cancer may lead the way to identifying therapeutic vulnerabilities in cells following RB loss.


Subject(s)
Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Retinoblastoma Protein/metabolism , Animals , Disease Progression , Humans , Male
7.
Oncotarget ; 9(30): 21613-21627, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29765563

ABSTRACT

Pancreatic cancer has a dismal prognosis particularly in patients presenting with unresectable tumors. We performed a bibliometric analysis of clinical trials for pancreatic cancer conducted between 2014-2016 focusing on patients that presented with unresectable (locally advanced or metastatic) tumors. We discuss a range of studies that employed FOLFIRINOX, the gemcitabine + nab-paclitaxel combination and studies that used molecularly-targeted therapy. Major areas of focus have been dual targeting of EGFR and VEGFR, immunotherapy or a multimodal approach - combining chemotherapy with radiotherapy. We also point out the need for molecular selection for low prevalence subtypes. Key insights sourced from these pivotal trials should improve clinical outcomes for this devastating cancer.

8.
Urol Oncol ; 36(8): 375-379, 2018 08.
Article in English | MEDLINE | ID: mdl-29395951

ABSTRACT

Prostate cancer initiation, development and progression is driven by androgen receptor (AR) signaling. Androgen deprivation therapy is the primary treatment for patients that present with locally advanced or metastatic disease. However, androgen deprivation therapy is not curative, and patients will progress to castrate-resistant disease (CRPC). Although most patient's progress to CRPC via restoration of AR signaling (CRPC-Ad), approximately a quarter of patients will progress via mechanisms independent of AR signaling. This highly lethal phenotype is termed aggressive variant prostate cancer (AVPC). Data from clinical and preclinical studies demonstrate that AVPC involves combinatorial loss-of-function mutations in key tumor suppressor genes, low to absent AR levels, and re-expression of reprogramming, stem, and neuroendocrine related gene signatures. Further, AVPC is shown to evolve from a CRPC-Ad phenotype. Overall, lineage plasticity underlying progression to AVPC is thought to be provoked by genome-wide chromatin remodeling. Here, we will discuss an emerging focus on key drivers of chromatin remodeling in AVPC, and how their identification could provide noninvasive biomarkers to predict or detect AVPC emergence, and therapeutic targets to prevent or reverse progression to AVPC.


Subject(s)
Drug Resistance, Neoplasm/genetics , Epigenomics/methods , Prostatic Neoplasms, Castration-Resistant/genetics , Disease Progression , Humans , Male , Prostatic Neoplasms, Castration-Resistant/pathology
9.
J Mol Med (Berl) ; 94(7): 799-808, 2016 07.
Article in English | MEDLINE | ID: mdl-27277823

ABSTRACT

UNLABELLED: Aberrant protein folding represents the molecular basis of many important human diseases. Although the discovery of new anti-misfolding drugs is a major priority in molecular therapeutics, there is currently no generalizable protein folding assay for use in cell-based high throughput screening (HTS) of chemical libraries, or for in vivo imaging. We molecularly engineered a bioluminescence-based biosensor composed of rationally split Firefly luciferase reporter fragments flanking a test protein, and used this in a protein-fragment complementation assay to quantitatively measure folding of the test protein. We comprehensively validated this biosensor in vitro, in cells, and by optically imaging protein folding and misfolding in living mice using several test proteins including enhanced green fluorescent protein, Renilla luciferase, Gaussia luciferase, and SIRT1. Applications of this novel biosensor are potentially far-reaching in both cell-based HTS approaches to discover new anti-misfolding drugs, and when using the same biosensor in validation studies of drug candidates in small animal models. KEY MESSAGES: Novel anti-misfolding drugs are needed as molecular therapeutics for many diseases. We developed first in vivo imaging protein folding biosensor to aid drug discovery. Biosensor created by flanking a test protein with rationally split Firefly luciferase. Biosensor validated by detecting folding of test proteins EGFP, Rluc, Gluc, and SIRT1. Generalizable molecular biosensor for translational applications in drug screening.


Subject(s)
Biosensing Techniques/methods , High-Throughput Screening Assays , Molecular Imaging/methods , Protein Engineering/methods , Protein Folding , A549 Cells , Animals , Drug Discovery , Female , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Luciferases, Renilla/genetics , Luciferases, Renilla/metabolism , Mice , Mice, Nude , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transfection
10.
Curr Cancer Drug Targets ; 13(4): 400-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23517597

ABSTRACT

Pancreatic cancer has a poor prognosis, with only 10% survival one year following diagnosis. Despite significant advances in conventional therapies (chemotherapy and radiotherapy), little improvement in patient survival has occurred in the last decade. Therefore, there is a critical need for novel and effective therapeutic approaches for this cancer. This article reviews current concepts in the pathogenesis and treatment of pancreatic cancer, the latter including tumor resection approaches and the current standard of care. We further describe recent advances in new and combination therapies, which result only in modest increases in survival, and discuss challenges in drug delivery and limiting toxicity.


Subject(s)
Pancreatic Neoplasms/therapy , Combined Modality Therapy , Humans
11.
Am J Pathol ; 180(6): 2590-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22507839

ABSTRACT

The acute response to vascular cell injury, which underpins vasculo-occlusive pathologies such as atherogenesis and restenosis after percutaneous coronary intervention, involves a complex series of molecular events that alter patterns of gene expression and favor a synthetic phenotype. One transcription factor that has been implicated in this process is the evolutionarily conserved mammalian stress response pathway regulator activating transcription factor 4 (ATF-4). Here, we show for the first time that both mRNA and protein levels of ATF-4 are induced in smooth muscle cells (SMCs) by the potent migratory factor PDGF-BB through PDGFR-ß. PDGF-BB also stimulates the expression of tenascin-C (TN-C), an extracellular matrix glycoprotein that regulates the activity of focal adhesion complexes, facilitating the SMC migration that underlies negative vascular remodeling in response to injury. Overexpression of ATF-4 increased transcript levels of the four TN-C isoforms in rat vascular SMCs, and ATF-4 knockdown inhibited PDGF-BB-inducible TN-C expression in vitro and injury-inducible TN-C protein expression in the balloon-injured rat artery wall. Furthermore, we show that ATF-4 is required for PDGF-BB-inducible SMC migration in response to injury. PDGF-BB-induced migration was also compromised in ATF-4 null mEFs, and this effect was rescued by the addition of TN-C. Our findings thus demonstrate the role of ATF-4 in both injury- and PDGF-BB-inducible TN-C expression and cell migration.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Proto-Oncogene Proteins c-sis/physiology , Tenascin/biosynthesis , Activating Transcription Factor 4/deficiency , Activating Transcription Factor 4/genetics , Animals , Becaplermin , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/physiopathology , Carotid Artery, Common/metabolism , Dose-Response Relationship, Drug , Fibroblasts/physiology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Gene Silencing , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Tenascin/genetics , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL