Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Daksh Thaper is active.

Publication


Featured researches published by Daksh Thaper.


Molecular Cancer Therapeutics | 2013

Synergistic Targeting of PI3K/AKT Pathway and Androgen Receptor Axis Significantly Delays Castration-Resistant Prostate Cancer Progression In Vivo

Christian Thomas; Francois Lamoureux; Claire Crafter; Barry R. Davies; Eliana Beraldi; Ladan Fazli; Soo Jin Kim; Daksh Thaper; Martin Gleave; Amina Zoubeidi

The progression to castration-resistant prostate cancer (CRPC) correlates with gain-of-function of the androgen receptor (AR) and activation of AKT. However, as single agents, AR or AKT inhibitors result in a reciprocal feedback loop. Therefore, we hypothesized that combination of an AKT inhibitor with an antiandrogen might result in a more profound, long-lasting remission of CRPC. Here, we report that the AKT inhibitor AZD5363 potently inhibits proliferation and induces apoptosis in prostate cancer cell lines expressing the AR and has anticancer activity in vivo in androgen-sensitive and castration-resistant phases of the LNCaP xenograft model. However, we found that the effect of castration-resistant tumor growth inhibition and prostate-specific antigen (PSA) stabilization is transient and resistance occurs with increasing PSA after approximately 30 days of treatment. Mechanistically, we found that single agent AZD5363 induces increase of AR binding to androgen response element, AR transcriptional activity, and AR-dependent genes such as PSA and NKX3.1 expression. These effects were overcome by the combination of AZD5363 with the antiandrogen bicalutamide, resulting in synergistic inhibition of cell proliferation and induction of apoptosis in vitro, and prolongation of tumor growth inhibition and PSA stabilization in CRPC in vivo. This study provides a preclinical proof-of-concept that combination of an AKT inhibitor with antiandrogen results in prolonged disease stabilization in a model of CRPC. Mol Cancer Ther; 12(11); 2342–55. ©2013 AACR.


Cancers | 2014

The Multifaceted Roles of STAT3 Signaling in the Progression of Prostate Cancer.

Jennifer L. Bishop; Daksh Thaper; Amina Zoubeidi

The signal transducer and activator of transcription (STAT)3 governs essential functions of epithelial and hematopoietic cells that are often dysregulated in cancer. While the role for STAT3 in promoting the progression of many solid and hematopoietic malignancies is well established, this review will focus on the importance of STAT3 in prostate cancer progression to the incurable metastatic castration-resistant prostate cancer (mCRPC). Indeed, STAT3 integrates different signaling pathways involved in the reactivation of androgen receptor pathway, stem like cells and the epithelial to mesenchymal transition that drive progression to mCRPC. As equally important, STAT3 regulates interactions between tumor cells and the microenvironment as well as immune cell activation. This makes it a major factor in facilitating prostate cancer escape from detection of the immune response, promoting an immunosuppressive environment that allows growth and metastasis. Based on the multifaceted nature of STAT3 signaling in the progression to mCRPC, the promise of STAT3 as a therapeutic target to prevent prostate cancer progression and the variety of STAT3 inhibitors used in cancer therapies is discussed.


Cancer Discovery | 2017

The Master Neural Transcription Factor BRN2 is an Androgen Receptor Suppressed Driver of Neuroendocrine Differentiation in Prostate Cancer

Jennifer L. Bishop; Daksh Thaper; Sepideh Vahid; Alastair H. Davies; Kirsi Ketola; Hidetoshi Kuruma; Randy Jama; Ka Mun Nip; Arkhjamil Angeles; Fraser Johnson; Alexander W. Wyatt; Ladan Fazli; Martin Gleave; Dong Lin; Mark A. Rubin; Colin Collins; Yuzhuo Wang; Himisha Beltran; Amina Zoubeidi

Mechanisms controlling the emergence of lethal neuroendocrine prostate cancer (NEPC), especially those that are consequences of treatment-induced suppression of the androgen receptor (AR), remain elusive. Using a unique model of AR pathway inhibitor-resistant prostate cancer, we identified AR-dependent control of the neural transcription factor BRN2 (encoded by POU3F2) as a major driver of NEPC and aggressive tumor growth, both in vitro and in vivo Mechanistic studies showed that AR directly suppresses BRN2 transcription, which is required for NEPC, and BRN2-dependent regulation of the NEPC marker SOX2. Underscoring its inverse correlation with classic AR activity in clinical samples, BRN2 expression was highest in NEPC tumors and was significantly increased in castration-resistant prostate cancer compared with adenocarcinoma, especially in patients with low serum PSA. These data reveal a novel mechanism of AR-dependent control of NEPC and suggest that targeting BRN2 is a strategy to treat or prevent neuroendocrine differentiation in prostate tumors. SIGNIFICANCE Understanding the contribution of the AR to the emergence of highly lethal, drug-resistant NEPC is critical for better implementation of current standard-of-care therapies and novel drug design. Our first-in-field data underscore the consequences of potent AR inhibition in prostate tumors, revealing a novel mechanism of AR-dependent control of neuroendocrine differentiation, and uncover BRN2 as a potential therapeutic target to prevent emergence of NEPC. Cancer Discov; 7(1); 54-71. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1.


International Journal of Cancer | 2015

Hsp27 regulates EGF/β-catenin mediated epithelial to mesenchymal transition in prostate cancer.

Thomas Cordonnier; Jennifer L. Bishop; Masaki Shiota; Ka Mun Nip; Daksh Thaper; Sepideh Vahid; Devon Heroux; Martin Gleave; Amina Zoubeidi

Increased expression of the molecular chaperone Hsp27 is associated with the progression of prostate cancer (PCa) to castration‐resistant disease, which is lethal due to metastatic spread of the prostate tumor. Metastasis requires epithelial to mesenchymal transition (EMT), which endows cancer cells with the ability to disseminate from the primary tumor and colonize new tissue sites. A wide variety of secreted factors promote EMT, and while overexpression and constitutive activation of epidermal growth factor (EGF) signaling is associated with poor prognosis of PCa, a precise role of EGF in PCa progression to metastasis remains unclear. Here, we show that Hsp27 is required for EGF‐induced cell migration, invasion and MMPs activity as well as the expression of EMT markers including Fibronectin, Vimentin and Slug with concomitant decrease of E‐cadherin. Mechanistically, we found that Hsp27 is required for EGF‐induced AKT and GSK3β phosphorylation and β‐catenin nuclear translocation. Moreover, silencing Hsp27 decreases EGF dependent phosphorylation of β‐catenin on tyrosine 142 and 654, enhances β‐catenin ubiquitination and degradation, prevents β‐catenin nuclear translocation and binding to the Slug promoter. These data suggest that Hsp27 is required for EGF‐mediated EMT via modulation of the β‐catenin/Slug signaling pathway. Together, our findings underscore the importance of Hsp27 in EGF induced EMT in PCa and highlight the use of Hsp27 knockdown as a useful strategy for patients with advanced disease.


Scientific Reports | 2016

Molecular chaperone Hsp27 regulates the Hippo tumor suppressor pathway in cancer

Sepideh Vahid; Daksh Thaper; Kate Gibson; Jennifer L. Bishop; Amina Zoubeidi

Heat shock protein 27 (Hsp27) is a molecular chaperone highly expressed in aggressive cancers, where it is involved in numerous pro-tumorigenic signaling pathways. Using functional genomics we identified for the first time that Hsp27 regulates the gene signature of transcriptional co-activators YAP and TAZ, which are negatively regulated by the Hippo Tumor Suppressor pathway. The Hippo pathway inactivates YAP by phosphorylating and increasing its cytoplasmic retention with the 14.3.3 proteins. Gain and loss of function experiments in prostate, breast and lung cancer cells showed that Hsp27 knockdown induced YAP phosphorylation and cytoplasmic localization while overexpression of Hsp27 displayed opposite results. Mechanistically, Hsp27 regulates the Hippo pathway by accelerating the proteasomal degradation of ubiquitinated MST1, the core Hippo kinase, resulting in reduced phosphorylation/activity of LATS1 and MOB1, its downstream effectors. Importantly, our in vitro results were supported by data from human tumors; clinically, high expression of Hsp27 in prostate tumors is correlated with increased expression of YAP gene signature and reduced phosphorylation of YAP in lung and invasive breast cancer clinical samples. This study reveals for the first time a link between Hsp27 and the Hippo cascade, providing a novel mechanism of deregulation of this tumor suppressor pathway across multiple cancers.


Molecular and Cellular Endocrinology | 2014

Insulin-like growth factor-I induces CLU expression through Twist1 to promote prostate cancer growth

Ario Takeuchi; Masaki Shiota; Eliana Beraldi; Daksh Thaper; Kiyoshi Takahara; Naokazu Ibuki; Michael Pollak; Michael E. Cox; Seiji Naito; Martin Gleave; Amina Zoubeidi

Clusterin (CLU) is cytoprotective molecular chaperone that is highly expressed in castrate-resistant prostate cancer (CRPC). CRPC is also characterized by increased insulin-like growth factor (IGF)-I responsiveness which induces prostate cancer survival and CLU expression. However, how IGF-I induces CLU expression and whether CLU is required for IGF-mediated growth signaling remain unknown. Here we show that IGF-I induced CLU via STAT3-Twist1 signaling pathway. In response to IGF-I, STAT3 was phosphorylated, translocated to the nucleus and bound to the Twist1 promoter to activate Twist1 transcription. In turn, Twist1 bound to E-boxes on the CLU promoter and activated CLU transcription. Inversely, we demonstrated that knocking down Twist1 abrogated IGF-I induced CLU expression, indicating that Twist1 mediated IGF-I-induced CLU expression. When PTEN knockout mice were crossed with lit/lit mice, the resultant IGF-I deficiency suppressed Twist1 as well as CLU gene expression in mouse prostate glands. Moreover, both Twist1 and CLU knockdown suppressed prostate cancer growth accelerated by IGF-I, suggesting the relevance of this signaling not only in an in vitro, but also in an in vivo. Collectively, this study indicates that IGF-I induces CLU expression through sequential activation of STAT3 and Twist1, and suggests that this signaling cascade plays a critical role in prostate cancer pathogenesis.


Recent Patents on Anti-cancer Drug Discovery | 2017

Chaperoning the Cancer: The Proteostatic Functions of the Heat Shock Proteins in Cancer

Sepideh Vahid; Daksh Thaper; Amina Zoubeidi

BACKGROUND Protein homeostasis (proteostasis) is vital for the survival of cells in physiological and pathological conditions. Particularly, cancer cells are in constant state of cellular stress due to rapid proliferation and decreased quality control in proteosynthesis and therefore, are exceedingly dependent on the homeostasis pathways. Among the complex biological mechanisms regulating proteostasis are the highly conserved molecular chaperones, heat shock proteins (HSPs). HSPs assist cell survival by catalysing the proper folding of proteins, modulation of the apoptotic machinery and finally regulating the protein degradation machinery, providing either the stability or the degradation of selected proteins under stress conditions. Inevitably, HSPs are upregulated in malignancies and participate in different hallmarks of cancer, with indispensable roles in the onset and progression of the disease. Moreover, high levels of HSPs contribute to poor prognosis and treatment resistance in various cancers. Therefore these molecular chaperones present as attractive targets for anti-cancer therapy. OBJECTIVE This review describes how HSPs regulate different hallmarks of cancer and provides an overview on the most relevant patents which have recently appeared in the literature. METHODS The patents were extracted from Google Patents (2012-2016) while the clinical trial results were mined from www.clinicaltrial.gov. RESULTS AND CONCLUSION Review of literature shows that the proteostatic functions of HSPs can modify different hallmarks of cancer. Moreover, targeting HSPs (notably HSP27, HSP70 and HSP90) exhibited positive results in clinical trials so far. However, more studies should be designed to optimize the efficacy of mono or combination therapy in various malignancies.


Embo Molecular Medicine | 2018

SEMA3C drives cancer growth by transactivating multiple receptor tyrosine kinases via Plexin B1

James W. Peacock; Ario Takeuchi; Norihiro Hayashi; Liangliang Liu; Kevin Tam; Nader Al Nakouzi; Nastaran Khazamipour; Tabitha Tombe; Takashi Dejima; Kevin C.K. Lee; Masaki Shiota; Daksh Thaper; Wilson C.W. Lee; Daniel H.F. Hui; Hidetoshi Kuruma; Larissa Ivanova; Parvin Yenki; Ivy Z.F. Jiao; Shahram Khosravi; Alice L.-F. Mui; Ladan Fazli; Amina Zoubeidi; Mads Daugaard; Martin Gleave; Christopher J. Ong

Growth factor receptor tyrosine kinase (RTK) pathway activation is a key mechanism for mediating cancer growth, survival, and treatment resistance. Cognate ligands play crucial roles in autocrine or paracrine stimulation of these RTK pathways. Here, we show SEMA3C drives activation of multiple RTKs including EGFR, ErbB2, and MET in a cognate ligand‐independent manner via Plexin B1. SEMA3C expression levels increase in castration‐resistant prostate cancer (CRPC), where it functions to promote cancer cell growth and resistance to androgen receptor pathway inhibition. SEMA3C inhibition delays CRPC and enzalutamide‐resistant progression. Plexin B1 sema domain‐containing:Fc fusion proteins suppress RTK signaling and cell growth and inhibit CRPC progression of LNCaP xenografts post‐castration in vivo. SEMA3C inhibition represents a novel therapeutic strategy for treatment of advanced prostate cancer.


Cancer Research | 2017

Abstract 3189: Neuronal transcription factor BRN2 is an androgen suppressed driver of neuroendocrine differentiation in prostate cancer

Jennifer L. Bishop; Daksh Thaper; Sepideh Vahid; Ravi Shashi Nayana Munuganti; Paul Ahn; Alastair H. Davies; Kirsi Ketola; Ka Mun Nip; Dong Lin; Yuzhuo Wang; Himisha Beltran; Amina Zoubeidi

Neuroendocrine prostate cancer (NEPC) is an incurable, rapidly progressing and lethal disease. NEPC is increasingly recognized as a highly therapy resistant tumor variant that evolves from castration-resistant prostate cancer (CRPC) in a subset of patients treated with potent androgen receptor (AR) pathway inhibitors like enzalutamide (ENZ). Importantly, mechanisms by which the AR directly controls the emergence of NEPC from CRPC under the selective pressure of ENZ remain elusive. As the AR is the cornerstone therapeutic target in men with CRPC, understanding its contribution to the development of NEPC is critical to better implement current standard-of-care therapies such as ENZ, and to identify novel therapeutic targets for this incurable disease. Hallmarks of NEPC are resistance to ENZ and the loss or reduced activity of the AR. Thus, we hypothesized that a consequence of ENZ treatment and resistance in CRPC is relief of AR-mediated suppression of factors that drive NEPC. To investigate this, we developed a model of prostate cancer that mimics clinical progression to CRPC and ENZ resistance (ENZR CPRC). Mirroring what is observed in some patients who progress on ENZ, 25% of our ENZR CRPC tumors and derived cell lines showed strong reduction in classic activity of the AR and a NEPC phenotype. By interrogating NEPC-like ENZR CRPC and ENZ-treated CRPC tumors and cell lines with RNA-Seq and mechanistic in vitro and in vivo studies, as well as human tumors with RNA-Seq and IHC, we identified the master neural transcription factor BRN2 as a central and clinically relevant driver of NEPC differentiation. Specifically, we found AR binding in the BRN2 enhancer directly represses BRN2 transcription and this release drives NEPC marker expression and aggressive growth of ENZR CRPC. Our data also reveal a striking overlap of AR and SOX binding motifs in the enhancer region of BRN2 creating a competitive binding scenario between AR and SOX2, another cell-fate determining transcription factor associated with NEPC. We discovered that upregulation of BRN2 further enhances SOX2 expression and that a BRN2-SOX2 interaction contributes to NEPC differentiation. Importantly, we found BRN2 is highly expressed in human NEPC and metastatic CRPC, especially in patients with low AR activity, highlighting its clinical relevance to disease that is difficult to treat with mainstay therapies. These data underscore the consequences of potent AR inhibition in CRPC, revealing a novel mechanism of AR-dependent control of NEPC via direct suppression of BRN2. This work uncovers BRN2 as a clinically relevant potential therapeutic target to prevent emergence of NEPC from ENZR CRPC. Citation Format: Jennifer L. Bishop, Daksh Thaper, Sepideh Vahid, Ravi S. Munuganti, Paul Ahn, Alastair Davies, Kirsi Ketola, Ka Mun Nip, Dong Lin, Yuzhuo Wang, Himisha Beltran, Amina Zoubeidi. Neuronal transcription factor BRN2 is an androgen suppressed driver of neuroendocrine differentiation in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3189. doi:10.1158/1538-7445.AM2017-3189


Cancer Research | 2015

Abstract 4956: Hsp27 negatively affects Hippo tumor suppressor pathway to regulate cell survival in cancer

Sepideh Vahid; Daksh Thaper; Amina Zoubeidi

Introduction: Heat shock protein 27 (Hsp27) is a molecular chaperone highly and uniformly expressed in treatment resistant cancers like castrate resistant prostate cancer (CRPC). Hsp27 regulates activity of several oncogenic pathways and its levels correlate with aggressive tumor behaviour, drug resistance and tumor growth. Similarly, dysregulation of the Hippo tumor suppressor pathway, which restricts organ size and cell proliferation, occurs in many types of cancers. In healthy cells, activation of the Hippo pathway results in phosphorylation and cytoplasmic retention of two transcriptional co-activators YAP and TAZ, whereas in cancer YAP/TAZ are free to translocate to the nucleus and increase cell proliferation by promoting the activities of certain transcriptional factors including TEAD1. Inactivation of the Hippo pathway correlates with poor patient outcome and progression of tumors as well as an increase in migration, invasion and metastatic potential of cancer cells. Therefore it is of great importance to establish the correlation between Hsp27 and the Hippo pathway to further discover suitable targets in the treatment of metastatic malignancies. Methods: Hsp27 gain and loss of function experiments were done on 3 different cancer cell lines and the functional effects on every step of the pathway were monitored via Western blots and Immunofluorescence. Activity of YAP/TAZ after Hsp27 gain and loss of function was monitored by conducting qRT-PCR on TEAD target genes. Transcriptional activity of TEAD1 was also examined using a TEAD-dependent Luciferase reporter construct. Co-immunoprecipitation assay was conducted to analyze protein interactions in the absence/presence of Hsp27. Pathway activity in prostate cancer will be assessed by immunohistochemistry staining of core components of the pathway in patients’ tissue samples. Results: Our preliminary findings indicate that Hsp27 negatively affects the Hippo pathway. We found that targeting Hsp27 using siRNA in the PC3 (prostate cancer), A549 (lung cancer) and MDA-MB-453 (triple negative breast cancer) leads to increased cytoplasmic retention of p-YAP compared to control siRNA treated cells. Moreover, using immunofluorescence, we observed reduced nuclear translocation of the YAP/TAZ as well as sequestration of these components with cytoplasmic 14-3-3 proteins in siRNA treated PC3 cells. Furthermore inhibition of Hsp27 in prostate and lung tumour cells resulted in suppression of TEAD transcriptional activity analyzed by qRT-PCR and luciferase assay. Hsp27 over-expression experiments yielded opposite results compared to knockdown. Conclusion: Hsp27 overexpression contributes to inactivation of Hippo pathway. Targeting Hsp27 leads to inactivation of YAP and TAZ onco-proteins affecting cancer cell survival. Impact: Our data further supports the significance of targeting Hsp27 as a treatment option in cancers, especially metastatic malignancies like CRPC. Citation Format: Sepideh Vahid, Daksh Thaper, Amina Zoubeidi. Hsp27 negatively affects Hippo tumor suppressor pathway to regulate cell survival in cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4956. doi:10.1158/1538-7445.AM2015-4956

Collaboration


Dive into the Daksh Thaper's collaboration.

Top Co-Authors

Avatar

Amina Zoubeidi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sepideh Vahid

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Martin Gleave

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ladan Fazli

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Eliana Beraldi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ka Mun Nip

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alastair H. Davies

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Dong Lin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge