Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
bioRxiv ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38645034

ABSTRACT

Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)--a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis (TMA) on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated, but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer (SCLC) subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to novel antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.

2.
Sci Adv ; 10(5): eadg7887, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38295166

ABSTRACT

Protein tyrosine phosphatases (PTPs) play major roles in cancer and are emerging as therapeutic targets. Recent reports suggest low-molecular weight PTP (LMPTP)-encoded by the ACP1 gene-is overexpressed in prostate tumors. We found ACP1 up-regulated in human prostate tumors and ACP1 expression inversely correlated with overall survival. Using CRISPR-Cas9-generated LMPTP knockout C4-2B and MyC-CaP cells, we identified LMPTP as a critical promoter of prostate cancer (PCa) growth and bone metastasis. Through metabolomics, we found that LMPTP promotes PCa cell glutathione synthesis by dephosphorylating glutathione synthetase on inhibitory Tyr270. PCa cells lacking LMPTP showed reduced glutathione, enhanced activation of eukaryotic initiation factor 2-mediated stress response, and enhanced reactive oxygen species after exposure to taxane drugs. LMPTP inhibition slowed primary and bone metastatic prostate tumor growth in mice. These findings reveal a role for LMPTP as a critical promoter of PCa growth and metastasis and validate LMPTP inhibition as a therapeutic strategy for treating PCa through sensitization to oxidative stress.


Subject(s)
Prostatic Neoplasms , Male , Humans , Mice , Animals , Molecular Weight , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Tyrosine , Protein Tyrosine Phosphatases/metabolism
3.
Mol Cell ; 82(11): 2021-2031.e5, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35447082

ABSTRACT

The androgen receptor (AR) is a nuclear receptor that governs gene expression programs required for prostate development and male phenotype maintenance. Advanced prostate cancers display AR hyperactivation and transcriptome expansion, in part, through AR amplification and interaction with oncoprotein cofactors. Despite its biological importance, how AR domains and cofactors cooperate to bind DNA has remained elusive. Using single-particle cryo-electron microscopy, we isolated three conformations of AR bound to DNA, showing that AR forms a non-obligate dimer, with the buried dimer interface utilized by ancestral steroid receptors repurposed to facilitate cooperative DNA binding. We identify novel allosteric surfaces which are compromised in androgen insensitivity syndrome and reinforced by AR's oncoprotein cofactor, ERG, and by DNA-binding motifs. Finally, we present evidence that this plastic dimer interface may have been adopted for transactivation at the expense of DNA binding. Our work highlights how fine-tuning AR's cooperative interactions translate to consequences in development and disease.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Cryoelectron Microscopy , DNA/metabolism , Dimerization , Humans , Male , Prostatic Neoplasms/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcriptional Activation
5.
Cancer Cell ; 38(2): 279-296.e9, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32679108

ABSTRACT

Despite the development of second-generation antiandrogens, acquired resistance to hormone therapy remains a major challenge in treating advanced prostate cancer. We find that cancer-associated fibroblasts (CAFs) can promote antiandrogen resistance in mouse models and in prostate organoid cultures. We identify neuregulin 1 (NRG1) in CAF supernatant, which promotes resistance in tumor cells through activation of HER3. Pharmacological blockade of the NRG1/HER3 axis using clinical-grade blocking antibodies re-sensitizes tumors to hormone deprivation in vitro and in vivo. Furthermore, patients with castration-resistant prostate cancer with increased tumor NRG1 activity have an inferior response to second-generation antiandrogen therapy. This work reveals a paracrine mechanism of antiandrogen resistance in prostate cancer amenable to clinical testing using available targeted therapies.


Subject(s)
Androgen Antagonists/pharmacology , Drug Resistance, Neoplasm/genetics , Neuregulin-1/genetics , Prostatic Neoplasms/genetics , Tumor Microenvironment/genetics , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kaplan-Meier Estimate , Male , Mice, SCID , Neuregulin-1/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/prevention & control , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays/methods
6.
Cancer Discov ; 10(7): 1038-1057, 2020 07.
Article in English | MEDLINE | ID: mdl-32376773

ABSTRACT

To study genetic factors influencing the progression and therapeutic responses of advanced prostate cancer, we developed a fast and flexible system that introduces genetic alterations relevant to human disease directly into the prostate glands of mice using tissue electroporation. These electroporation-based genetically engineered mouse models (EPO-GEMM) recapitulate features of traditional germline models and, by modeling genetic factors linked to late-stage human disease, can produce tumors that are metastatic and castration-resistant. A subset of tumors with Trp53 alterations acquired spontaneous WNT pathway alterations, which are also associated with metastatic prostate cancer in humans. Using the EPO-GEMM approach and an orthogonal organoid-based model, we show that WNT pathway activation drives metastatic disease that is sensitive to pharmacologic WNT pathway inhibition. Thus, by leveraging EPO-GEMMs, we reveal a functional role for WNT signaling in driving prostate cancer metastasis and validate the WNT pathway as therapeutic target in metastatic prostate cancer. SIGNIFICANCE: Our understanding of the factors driving metastatic prostate cancer is limited by the paucity of models of late-stage disease. Here, we develop EPO-GEMMs of prostate cancer and use them to identify and validate the WNT pathway as an actionable driver of aggressive metastatic disease.This article is highlighted in the In This Issue feature, p. 890.


Subject(s)
Prostatic Neoplasms/genetics , Tissue Engineering/methods , Wnt Signaling Pathway/genetics , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Neoplasm Metastasis
7.
Nat Commun ; 11(1): 1975, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332851

ABSTRACT

Treatment paradigms for patients with upper tract urothelial carcinoma (UTUC) are typically extrapolated from studies of bladder cancer despite their distinct clinical and molecular characteristics. The advancement of UTUC research is hampered by the lack of disease-specific models. Here, we report the establishment of patient derived xenograft (PDX) and cell line models that reflect the genomic and biological heterogeneity of the human disease. Models demonstrate high genomic concordance with the corresponding patient tumors, with invasive tumors more likely to successfully engraft. Treatment of PDX models with chemotherapy recapitulates responses observed in patients. Analysis of a HER2 S310F-mutant PDX suggests that an antibody drug conjugate targeting HER2 would have superior efficacy versus selective HER2 kinase inhibitors. In sum, the biological and phenotypic concordance between patient and PDXs suggest that these models could facilitate studies of intrinsic and acquired resistance and the development of personalized medicine strategies for UTUC patients.


Subject(s)
Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Aged , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Biopsy , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Female , Gene Expression Profiling , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Immunoconjugates/pharmacology , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , Phenotype , Precision Medicine , Prospective Studies , Quinolines/pharmacology , Retrospective Studies , Sequence Analysis, RNA , Trastuzumab
8.
Nature ; 571(7765): 408-412, 2019 07.
Article in English | MEDLINE | ID: mdl-31243370

ABSTRACT

Mutations in the transcription factor FOXA1 define a unique subset of prostate cancers but the functional consequences of these mutations and whether they confer gain or loss of function is unknown1-9. Here, by annotating the landscape of FOXA1 mutations from 3,086 human prostate cancers, we define two hotspots in the forkhead domain: Wing2 (around 50% of all mutations) and the highly conserved DNA-contact residue R219 (around 5% of all mutations). Wing2 mutations are detected in adenocarcinomas at all stages, whereas R219 mutations are enriched in metastatic tumours with neuroendocrine histology. Interrogation of the biological properties of wild-type FOXA1 and fourteen FOXA1 mutants reveals gain of function in mouse prostate organoid proliferation assays. Twelve of these mutants, as well as wild-type FOXA1, promoted an exaggerated pro-luminal differentiation program, whereas two different R219 mutants blocked luminal differentiation and activated a mesenchymal and neuroendocrine transcriptional program. Assay for transposase-accessible chromatin using sequencing (ATAC-seq) of wild-type FOXA1 and representative Wing2 and R219 mutants revealed marked, mutant-specific changes in open chromatin at thousands of genomic loci and exposed sites of FOXA1 binding and associated increases in gene expression. Of note, ATAC-seq peaks in cells expressing R219 mutants lacked the canonical core FOXA1-binding motifs (GTAAAC/T) but were enriched for a related, non-canonical motif (GTAAAG/A), which was preferentially activated by R219-mutant FOXA1 in reporter assays. Thus, FOXA1 mutations alter its pioneering function and perturb normal luminal epithelial differentiation programs, providing further support for the role of lineage plasticity in cancer progression.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Mutation , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Lineage , Chromatin/genetics , Chromatin/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/chemistry , Humans , Male , Mice , Mice, Inbred NOD , Nucleotide Motifs , Organoids/cytology , Organoids/metabolism
9.
Elife ; 82019 01 15.
Article in English | MEDLINE | ID: mdl-30644358

ABSTRACT

Genomic amplification of the androgen receptor (AR) is an established mechanism of antiandrogen resistance in prostate cancer. Here, we show that the magnitude of AR signaling output, independent of AR genomic alteration or expression level, also contributes to antiandrogen resistance, through upregulation of the coactivator GREB1. We demonstrate 100-fold heterogeneity in AR output within human prostate cancer cell lines and show that cells with high AR output have reduced sensitivity to enzalutamide. Through transcriptomic and shRNA knockdown studies, together with analysis of clinical datasets, we identify GREB1 as a gene responsible for high AR output. We show that GREB1 is an AR target gene that amplifies AR output by enhancing AR DNA binding and promoting EP300 recruitment. GREB1 knockdown in high AR output cells restores enzalutamide sensitivity in vivo. Thus, GREB1 is a candidate driver of enzalutamide resistance through a novel feed forward mechanism.


Subject(s)
Androgen Antagonists/pharmacology , Drug Resistance, Neoplasm , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Benzamides , Cell Line, Tumor , Chromatin/metabolism , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm/drug effects , Humans , Male , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/genetics , Protein Binding/drug effects , Transcription, Genetic/drug effects
10.
Clin Cancer Res ; 25(2): 881-891, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30254080

ABSTRACT

PURPOSE: The impact of androgen receptor (AR) activity in breast cancer biology is unclear. We characterized and tested a novel therapy to an AR-governed target in breast cancer.Experimental Design: We evaluated the expression of prototypical AR gene products human kallikrein 2 (hK2) and PSA in breast cancer models. We screened 13 well-characterized breast cancer cell lines for hK2 and PSA production upon in vitro hormone stimulation by testosterone [dihydrotestosterone (DHT)]. AR-positive lines were further evaluated by exposure to estrogen (17ß-Estradiol) and the synthetic progestin D-Norgestrel. We then evaluated an anti-hK2-targeted radiotherapy platform (hu11B6), labeled with alpha (α)-particle emitting Actinium-225, to specifically treat AR-expressing breast cancer xenografts under hormone stimulation. RESULTS: D-Norgestrel and DHT activated the AR pathway, while 17ß-Estradiol did not. Competitive binding for AR protein showed similar affinity between DHT and D-Norgestrel, indicating direct AR-ligand interaction. In vivo production of hK2 was sufficient to achieve site-specific delivery of therapeutic radionuclide to tumor tissue at >20-fold over background muscle uptake; effecting long-term local tumor control. CONCLUSIONS: [225Ac]hu11B6 targeted radiotherapy was potentiated by DHT and by D-Norgestrel in murine xenograft models of breast cancer. AR activity in breast cancer correlates with kallikrein-related peptidase-2 and can be activated by D-Norgestrel, a common contraceptive, and AR induction can be harnessed for hK2-targeted breast cancer α-emitter radiotherapy.


Subject(s)
Alpha Particles/therapeutic use , Breast Neoplasms/metabolism , Immunoconjugates/administration & dosage , Receptors, Androgen/metabolism , Signal Transduction , Animals , Biomarkers, Tumor , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Hexokinase/antagonists & inhibitors , Humans , Immunoconjugates/pharmacokinetics , Mice , Molecular Targeted Therapy , Radioimmunotherapy , Radiometry , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tissue Distribution , Xenograft Model Antitumor Assays
11.
Nat Commun ; 9(1): 1629, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691406

ABSTRACT

Human kallikrein peptidase 2 (hK2) is a prostate specific enzyme whose expression is governed by the androgen receptor (AR). AR is the central oncogenic driver of prostate cancer (PCa) and is also a key regulator of DNA repair in cancer. We report an innovative therapeutic strategy that exploits the hormone-DNA repair circuit to enable molecularly-specific alpha particle irradiation of PCa. Alpha-particle irradiation of PCa is prompted by molecularly specific-targeting and internalization of the humanized monoclonal antibody hu11B6 targeting hK2 and further accelerated by inherent DNA-repair that up-regulate hK2 (KLK2) expression in vivo. hu11B6 demonstrates exquisite targeting specificity for KLK2. A single administration of actinium-225 labeled hu11B6 eradicates disease and significantly prolongs survival in animal models. DNA damage arising from alpha particle irradiation induces AR and subsequently KLK2, generating a unique feed-forward mechanism, which increases binding of hu11B6. Imaging data in nonhuman primates support the possibility of utilizing hu11B6 in man.


Subject(s)
Alpha Particles/therapeutic use , Prostatic Neoplasms/radiotherapy , Receptors, Androgen/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , DNA Damage/radiation effects , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Tissue Kallikreins/genetics , Tissue Kallikreins/metabolism
12.
Clin Cancer Res ; 24(10): 2408-2416, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29463549

ABSTRACT

Purpose: WST11 vascular targeted photodynamic therapy (VTP) is a local ablation approach relying upon rapid, free radical-mediated destruction of tumor vasculature. A phase III trial showed that VTP significantly reduced disease progression when compared with active surveillance in patients with low-risk prostate cancer. The aim of this study was to identify a druggable pathway that could be combined with VTP to improve its efficacy and applicability to higher risk prostate cancer tumors.Experimental Design: Transcriptome analysis of VTP-treated tumors (LNCaP-AR xenografts) was used to identify a candidate pathway for combination therapy. The efficacy of the combination therapy was assessed in mice bearing LNCaP-AR or VCaP tumors.Results: Gene set enrichment analysis identifies the enrichment of androgen-responsive gene sets within hours after VTP treatment, suggesting that the androgen receptor (AR) may be a viable target in combination with VTP. We tested this hypothesis in mice bearing LNCaP-AR xenograft tumors by using androgen deprivation therapy (ADT), degarelix, in combination with VTP. Compared with either ADT or VTP alone, a single dose of degarelix in concert with VTP significantly inhibited tumor growth. A sharp decline in serum prostate-specific antigen (PSA) confirmed AR inhibition in this group. Tumors treated by VTP and degarelix displayed intense terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining 7 days after treatment, supporting an increased apoptotic frequency underlying the effect on tumor inhibition.Conclusions: Improvement of local tumor control following androgen deprivation combined with VTP provides the rationale and preliminary protocol parameters for clinical trials in patients presented with locally advanced prostate cancer. Clin Cancer Res; 24(10); 2408-16. ©2018 AACR.


Subject(s)
Androgen Antagonists/pharmacology , Neovascularization, Pathologic/metabolism , Photochemotherapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Androgens/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Photochemotherapy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
13.
Cancers (Basel) ; 9(2)2017 Feb 08.
Article in English | MEDLINE | ID: mdl-28208703

ABSTRACT

Salivary gland cancers comprise a small subset of human malignancies, and are classified into multiple subtypes that exhibit diverse histology, molecular biology and clinical presentation. Local disease is potentially curable with surgery, which may be combined with adjuvant radiotherapy. However, metastatic or unresectable tumors rarely respond to chemotherapy and carry a poorer prognosis. Recent molecular studies have shown evidence of androgen receptor signaling in several types of salivary gland cancer, mainly salivary duct carcinoma. Successful treatment with anti-androgen therapy in other androgen receptor-positive malignancies such as prostate and breast cancer has inspired researchers to investigate this treatment in salivary gland cancer as well. In this review, we describe the prevalence, biology, and therapeutic implications of androgen receptor signaling in salivary gland cancer.

14.
Science ; 355(6320): 84-88, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28059768

ABSTRACT

Some cancers evade targeted therapies through a mechanism known as lineage plasticity, whereby tumor cells acquire phenotypic characteristics of a cell lineage whose survival no longer depends on the drug target. We use in vitro and in vivo human prostate cancer models to show that these tumors can develop resistance to the antiandrogen drug enzalutamide by a phenotypic shift from androgen receptor (AR)-dependent luminal epithelial cells to AR-independent basal-like cells. This lineage plasticity is enabled by the loss of TP53 and RB1 function, is mediated by increased expression of the reprogramming transcription factor SOX2, and can be reversed by restoring TP53 and RB1 function or by inhibiting SOX2 expression. Thus, mutations in tumor suppressor genes can create a state of increased cellular plasticity that, when challenged with antiandrogen therapy, promotes resistance through lineage switching.


Subject(s)
Androgen Antagonists/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Retinoblastoma Binding Proteins/genetics , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Benzamides , Cell Line, Tumor , Cell Lineage , Cell Plasticity , Humans , Male , Nitriles , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/genetics , SOXB1 Transcription Factors/genetics
15.
Sci Transl Med ; 8(367): 367ra167, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27903863

ABSTRACT

Targeting the androgen receptor (AR) pathway prolongs survival in patients with prostate cancer, but resistance rapidly develops. Understanding this resistance is confounded by a lack of noninvasive means to assess AR activity in vivo. We report intracellular accumulation of a secreted antigen-targeted antibody (SATA) that can be used to characterize disease, guide therapy, and monitor response. AR-regulated human kallikrein-related peptidase 2 (free hK2) is a prostate tissue-specific antigen produced in prostate cancer and androgen-stimulated breast cancer cells. Fluorescent and radio conjugates of 11B6, an antibody targeting free hK2, are internalized and noninvasively report AR pathway activity in metastatic and genetically engineered models of cancer development and treatment. Uptake is mediated by a mechanism involving the neonatal Fc receptor. Humanized 11B6, which has undergone toxicological tests in nonhuman primates, has the potential to improve patient management in these cancers. Furthermore, cell-specific SATA uptake may have a broader use for molecularly guided diagnosis and therapy in other cancers.


Subject(s)
Antibodies/chemistry , Bone Neoplasms/diagnostic imaging , Histocompatibility Antigens Class I/chemistry , Prostatic Neoplasms/diagnostic imaging , Receptors, Androgen/chemistry , Receptors, Fc/chemistry , Tissue Kallikreins/chemistry , Adenocarcinoma/diagnostic imaging , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Phenotype , Positron-Emission Tomography , Prostatic Neoplasms/pathology , Tomography, X-Ray Computed , Treatment Outcome
16.
Nat Rev Cancer ; 15(12): 701-11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26563462

ABSTRACT

During the past 10 years, preclinical studies implicating sustained androgen receptor (AR) signalling as the primary driver of castration-resistant prostate cancer (CRPC) have led to the development of novel agents targeting the AR pathway that are now in widespread clinical use. These drugs prolong the survival of patients with late-stage prostate cancer but are not curative. In this Review, we highlight emerging mechanisms of acquired resistance to these contemporary therapies, which fall into the three broad categories of restored AR signalling, AR bypass signalling and complete AR independence. This diverse range of resistance mechanisms presents new challenges for long-term disease control, which may be addressable through early use of combination therapies guided by recent insights from genomic landscape studies of CRPC.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Animals , Humans , Male
17.
J Nucl Med ; 55(12): 2045-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25453051

ABSTRACT

UNLABELLED: Antibodies and antibody-drug conjugates targeting the cell surface protein 6 transmembrane epithelial antigen of prostate 1 (STEAP1) are in early clinical development for the treatment of castration-resistant prostate cancer (PCa). In general, antigen expression directly affects the bioactivity of therapeutic antibodies, and the biologic regulation of STEAP1 is unusually complicated in PCa. Paradoxically, STEAP1 can be induced or repressed by the androgen receptor (AR) in different human PCa models, while also expressed in AR-null PCa. Consequently, there is an urgent need to translate diagnostic strategies to establish which regulatory mechanism predominates in patients to situate the appropriate therapy within standard of care therapies inhibiting AR. METHODS: To this end, we prepared and evaluated (89)Zr-labeled MSTP2109A ((89)Zr-2109A), a radiotracer for PET derived from a fully humanized monoclonal antibody to STEAP1 in preclinical PCa models. RESULTS: (89)Zr-2109A specifically localized to the STEAP1-positive human PCa models CWR22Pc, 22Rv1, and PC3. Moreover, (89)Zr-2109A sensitively measured treatment-induced changes (∼66% decline) in STEAP1 expression in CWR22PC in vitro and in vivo, a model we showed to express STEAP1 in an AR-dependent manner. CONCLUSION: These findings highlight the ability of immuno-PET with (89)Zr-2109A to detect acute changes in STEAP1 expression and argue for an expansion of ongoing efforts to image PCa patients with (89)Zr-2109A to maximize the clinical benefit associated with antibodies or antibody-drug conjugates to STEAP1.


Subject(s)
Antigens, Neoplasm/biosynthesis , Immunoconjugates , Oxidoreductases/biosynthesis , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/cerebrospinal fluid , Immunoconjugates/pharmacokinetics , Male , Mice , Oxidoreductases/genetics , Oxidoreductases/physiology , Positron-Emission Tomography , Radioisotopes , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Zirconium
18.
Cancer Discov ; 3(11): 1245-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24027196

ABSTRACT

UNLABELLED: We demonstrate that the androgen receptor (AR) regulates a transcriptional program of DNA repair genes that promotes prostate cancer radioresistance, providing a potential mechanism by which androgen deprivation therapy synergizes with ionizing radiation. Using a model of castration-resistant prostate cancer, we show that second-generation antiandrogen therapy results in downregulation of DNA repair genes. Next, we demonstrate that primary prostate cancers display a significant spectrum of AR transcriptional output, which correlates with expression of a set of DNA repair genes. Using RNA-seq and ChIP-seq, we define which of these DNA repair genes are both induced by androgen and represent direct AR targets. We establish that prostate cancer cells treated with ionizing radiation plus androgen demonstrate enhanced DNA repair and decreased DNA damage and furthermore that antiandrogen treatment causes increased DNA damage and decreased clonogenic survival. Finally, we demonstrate that antiandrogen treatment results in decreased classical nonhomologous end-joining. SIGNIFICANCE: We demonstrate that the AR regulates a network of DNA repair genes, providing a potential mechanism by which androgen deprivation synergizes with radiotherapy for prostate cancer.


Subject(s)
DNA Repair , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Androgen Antagonists/therapeutic use , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Cell Line, Tumor , DNA Damage/radiation effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Male , Metribolone/therapeutic use , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiation, Ionizing , Signal Transduction/genetics , Xenograft Model Antitumor Assays
19.
Elife ; 2: e00499, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23580326

ABSTRACT

The second-generation antiandrogen enzalutamide was recently approved for patients with castration-resistant prostate cancer. Despite its success, the duration of response is often limited. For previous antiandrogens, one mechanism of resistance is mutation of the androgen receptor (AR). To prospectively identify AR mutations that might confer resistance to enzalutamide, we performed a reporter-based mutagenesis screen and identified a novel mutation, F876L, which converted enzalutamide into an AR agonist. Ectopic expression of AR F876L rescued the growth inhibition of enzalutamide treatment. Molecular dynamics simulations performed on antiandrogen-AR complexes suggested a mechanism by which the F876L substitution alleviates antagonism through repositioning of the coactivator recruiting helix 12. This model then provided the rationale for a focused chemical screen which, based on existing antiandrogen scaffolds, identified three novel compounds that effectively antagonized AR F876L (and AR WT) to suppress the growth of prostate cancer cells resistant to enzalutamide. DOI:http://dx.doi.org/10.7554/eLife.00499.001.


Subject(s)
Androgen Antagonists/therapeutic use , Drug Design , Mutation , Androgen Antagonists/chemistry , Cell Line , Drug Resistance, Neoplasm/genetics , Humans , Male , Molecular Dynamics Simulation , Prostatic Neoplasms/drug therapy
20.
Proc Natl Acad Sci U S A ; 107(39): 16759-65, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20823238

ABSTRACT

Androgen receptor (AR) splice variants lacking the ligand binding domain (ARVs), originally isolated from prostate cancer cell lines derived from a single patient, are detected in normal and malignant human prostate tissue, with the highest levels observed in late stage, castration-resistant prostate cancer. The most studied variant (called AR-V7 or AR3) activates AR reporter genes in the absence of ligand and therefore, could play a role in castration resistance. To explore the range of potential ARVs, we screened additional human and murine prostate cancer models using conventional and next generation sequencing technologies and detected several structurally diverse AR isoforms. Some, like AR-V7/AR3, display gain of function, whereas others have dominant interfering activity. We also find that ARV expression increases acutely in response to androgen withdrawal, is suppressed by testosterone, and in some models, is coupled to full-length AR (AR-FL) mRNA production. As expected, constitutively active, ligand-independent ARVs such as AR-V7/AR3 are sufficient to confer anchorage-independent (in vitro) and castration-resistant (in vivo) growth. Surprisingly, this growth is blocked by ligand binding domain-targeted antiandrogens, such as MDV3100, or by selective siRNA silencing of AR-FL, indicating that the growth-promoting effects of ARVs are mediated through AR-FL. These data indicate that the increase in ARV expression in castrate-resistant prostate cancer is an acute response to castration rather than clonal expansion of castration or antiandrogen-resistant cells expressing gain of function ARVs, and furthermore, they provide a strategy to overcome ARV function in the clinic.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Amino Acid Sequence , Androgen Receptor Antagonists , Androgens/pharmacology , Animals , Castration , Cell Line, Tumor , Disease Models, Animal , Exons/genetics , Humans , Male , Mice , Molecular Sequence Data , Protein Structure, Tertiary/genetics , RNA, Messenger/biosynthesis , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...