Network


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

Hotspot


Dive into the research topics where Erkan Yuca is active.

Publication


Featured researches published by Erkan Yuca.


Molecular therapy. Nucleic acids | 2013

Therapeutic Silencing of Bcl-2 by Systemically Administered siRNA Nanotherapeutics Inhibits Tumor Growth by Autophagy and Apoptosis and Enhances the Efficacy of Chemotherapy in Orthotopic Xenograft Models of ER (−) and ER (+) Breast Cancer

Ibrahim Tekedereli; S. Neslihan Alpay; Ugur Akar; Erkan Yuca; Cristian Ayugo-Rodriguez; He Dong Han; Anil K. Sood; Gabriel Lopez-Berestein; Bulent Ozpolat

Bcl-2 is overexpressed in about a half of human cancers and 50–70% of breast cancer patients, thereby conferring resistance to conventional therapies and making it an excellent therapeutic target. Small interfering RNA (siRNA) offers novel and powerful tools for specific gene silencing and molecularly targeted therapy. Here, we show that therapeutic silencing of Bcl-2 by systemically administered nanoliposomal (NL)-Bcl-2 siRNA (0.15 mg siRNA/kg, intravenous) twice a week leads to significant antitumor activity and suppression of growth in both estrogen receptor-negative (ER(−)) MDA-MB-231 and ER-positive (+) MCF7 breast tumors in orthotopic xenograft models (P < 0.05). A single intravenous injection of NL-Bcl-2-siRNA provided robust and persistent silencing of the target gene expression in xenograft tumors. NL-Bcl-2-siRNA treatment significantly increased the efficacy of chemotherapy when combined with doxorubicin in both MDA-MB-231 and MCF-7 animal models (P < 0.05). NL-Bcl-2-siRNA treatment-induced apoptosis and autophagic cell death, and inhibited cyclin D1, HIF1α and Src/Fak signaling in tumors. In conclusion, our data provide the first evidence that in vivo therapeutic targeting Bcl-2 by systemically administered nanoliposomal-siRNA significantly inhibits growth of both ER(−) and ER(+) breast tumors and enhances the efficacy of chemotherapy, suggesting that therapeutic silencing of Bcl-2 by siRNA is a viable approach in breast cancers.


Clinical Cancer Research | 2012

Highly specific targeting of the TMPRSS2/ERG fusion gene using liposomal nanovectors

Longjiang Shao; Ibrahim Tekedereli; Jianghua Wang; Erkan Yuca; Susan Tsang; Anil K. Sood; Gabriel Lopez-Berestein; Bulent Ozpolat; Michael Ittmann

Purpose: The TMPRSS2/ERG (T/E) fusion gene is present in half of all prostate cancer tumors. Fusion of the oncogenic ERG gene with the androgen-regulated TMPRSS2 gene promoter results in expression of fusion mRNAs in prostate cancer cells. The junction of theTMPRSS2- and ERG-derived portions of the fusion mRNA constitutes a cancer-specific target in cells containing the T/E fusion gene. Targeting the most common alternatively spliced fusion gene mRNA junctional isoforms in vivo using siRNAs in liposomal nanovectors may potentially be a novel, low-toxicity treatment for prostate cancer. Experimental Design: We designed and optimized siRNAs targeting the two most common T/E fusion gene mRNA junctional isoforms (type III or type VI). Specificity of siRNAs was assessed by transient co-transfection in vitro. To test their ability to inhibit growth of prostate cancer cells expressing these fusion gene isoforms in vivo, specific siRNAs in liposomal nanovectors were used to treat mice bearing orthotopic or subcutaneous xenograft tumors expressing the targeted fusion isoforms. Results: The targeting siRNAs were both potent and highly specific in vitro. In vivo they significantly inhibited tumor growth. The degree of growth inhibition was variable and was correlated with the extent of fusion gene knockdown. The growth inhibition was associated with marked inhibition of angiogenesis and, to a lesser degree, proliferation and a marked increase in apoptosis of tumor cells. No toxicity was observed. Conclusions: Targeting the T/E fusion junction in vivo with specific siRNAs delivered via liposomal nanovectors is a promising therapy for men with prostate cancer. Clin Cancer Res; 18(24); 6648–57. ©2012 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Molecular circuit involving KLK4 integrates androgen and mTOR signaling in prostate cancer

Yang Jin; Su Qu; Martina Tesikova; Ling Wang; Alexandr Kristian; Gunhild M. Mælandsmo; Haiying Kong; Tianzhou Zhang; Carmen Jerónimo; Manuel R. Teixeira; Erkan Yuca; Ibrahim Tekedereli; Kivanc Gorgulu; Neslihan Alpay; Anil K. Sood; Gabriel Lopez-Berestein; Håvard E. Danielsen; Bulent Ozpolat; Fahri Saatcioglu

Significance All cancer lesions sustain alterations in signaling pathways, which are the drivers of disease initiation and progression. Study of altered signaling in cancer is thus important to develop more effective therapeutic regimens as well as better prognostic markers. In this study, we show that two of the most frequently altered signaling pathways in prostate cancer, the androgen receptor and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin pathways, are dependent on kallikrein related peptidase 4 (KLK4), whose expression is highly prostate enriched. Our results suggest that KLK4 has a central role in prostate cancer survival and that KLK4 silencing may have significant therapeutic efficacy. The androgen receptor (AR) and the phosphoinositide 3-kinase (PI3K)/protein kinase B/mammalian target of rapamycin (mTOR) signaling are two of the major proliferative pathways in a number of tissues and are the main therapeutic targets in various disorders, including prostate cancer (PCa). Previous work has shown that there is reciprocal feedback regulation of PI3K and AR signaling in PCa, suggesting that cotargeting both pathways may enhance therapeutic efficacy. Here we show that proteins encoded by two androgen-regulated genes, kallikrein related peptidase 4 (KLK4) and promyelocytic leukemia zinc finger (PLZF), integrate optimal functioning of AR and mTOR signaling in PCa cells. KLK4 interacts with PLZF and decreases its stability. PLZF in turn interacts with AR and inhibits its function as a transcription factor. PLZF also activates expression of regulated in development and DNA damage responses 1, an inhibitor of mTORC1. Thus, a unique molecular switch is generated that regulates both AR and PI3K signaling. Consistently, KLK4 knockdown results in a significant decline in PCa cell proliferation in vitro and in vivo, decreases anchorage-independent growth, induces apoptosis, and dramatically sensitizes PCa cells to apoptosis-inducing agents. Furthermore, in vivo nanoliposomal KLK4 siRNA delivery in mice bearing PCa tumors results in profound remission. These results demonstrate that the activities of AR and mTOR pathways are maintained by KLK4, which may thus be a viable target for therapy.


Cell Reports | 2014

Antagonism of Tumoral Prolactin Receptor Promotes Autophagy-Related Cell Death

Yunfei Wen; Behrouz Zand; Bulent Ozpolat; Miroslaw J. Szczepanski; Chunhua Lu; Erkan Yuca; Amy R. Carroll; Neslihan Alpay; Chandra Bartholomeusz; Ibrahim Tekedereli; Yu Kang; Rajesha Rupaimoole; Chad V. Pecot; Heather J. Dalton; Anadulce Hernandez; Anna Lokshin; Susan K. Lutgendorf; Jinsong Liu; Walter N. Hittelman; Wen Y. Chen; Gabriel Lopez-Berestein; Marta Szajnik; Naoto T. Ueno; Robert L. Coleman; Anil K. Sood

Therapeutic upregulation of macroautophagy in cancer cells provides an alternative mechanism for cell death. Prolactin (PRL) and its receptor (PRLR) are considered attractive therapeutic targets because of their roles as growth factors in tumor growth and progression. We utilized G129R, an antagonist peptide of PRL, to block activity of the tumoral PRL/PRLR axis, which resulted in inhibition of tumor growth in orthotopic models of human ovarian cancer. Prolonged treatment with G129R induced the accumulation of redundant autolysosomes in 3D cancer spheroids, leading to a type II programmed cell death. This inducible autophagy was a noncanonical beclin-1-independent pathway and was sustained by an astrocytic phosphoprotein (PEA-15) and protein kinase C zeta interactome. Lower levels of tumoral PRL/PRLR in clinical samples were associated with longer patient survival. Our findings provide an understanding of the mechanisms of tumor growth inhibition through targeting PRL/PRLR and may have clinical implications.


Nature Communications | 2015

The ZNF304-integrin axis protects against anoikis in cancer

Paloma Monroig; Ming Chuan Hsu; Guillermo Armaiz Pena; Cristian Rodriguez-Aguayo; Vianey Gonzalez-Villasana; Rajesha Rupaimoole; Archana S. Nagaraja; Selanere Mangala; Hee Dong Han; Erkan Yuca; Sherry Y. Wu; Cristina Ivan; Tyler Moss; Prahlad T. Ram; Huamin Wang; Alexandra Gol-Chambers; Ozgur Ozkayar; Pinar Kanlikilicer; Enrique Fuentes-Mattei; Nermin Kahraman; Sunila Pradeep; Bulent Ozpolat; Susan L. Tucker; Mien Chie Hung; Keith A. Baggerly; Geoffrey Bartholomeusz; George A. Calin; Anil K. Sood; Gabriel Lopez-Berestein

Ovarian cancer (OC) is a highly metastatic disease, but no effective strategies to target this process are currently available. Here, an integrative computational analysis of the Cancer Genome Atlas OC data set and experimental validation identifies a zinc finger transcription factor ZNF304 associated with OC metastasis. High tumoral ZNF304 expression is associated with poor overall survival in OC patients. Through reverse phase protein array analysis, we demonstrate that ZNF304 promotes multiple proto-oncogenic pathways important for cell survival, migration and invasion. ZNF304 transcriptionally regulates β1 integrin, which subsequently regulates Src/focal adhesion kinase and paxillin and prevents anoikis. In vivo delivery of ZNF304 siRNA by a dual assembly nanoparticle leads to sustained gene silencing for 14 days, increased anoikis and reduced tumour growth in orthotopic mouse models of OC. Taken together, ZNF304 is a transcriptional regulator of β1 integrin, promotes cancer cell survival and protects against anoikis in OC.


Embo Molecular Medicine | 2015

STAMP2 increases oxidative stress and is critical for prostate cancer

Yang Jin; Ling Wang; Su Qu; Xia Sheng; Alexandr Kristian; Gunhild M. Mælandsmo; Nora Pällmann; Erkan Yuca; Ibrahim Tekedereli; Kivanc Gorgulu; Neslihan Alpay; Anil K. Sood; Gabriel Lopez-Berestein; Ladan Fazli; Paul S. Rennie; Björn Risberg; Håkon Wæhre; Håvard E. Danielsen; Bulent Ozpolat; Fahri Saatcioglu

The six transmembrane protein of prostate 2 (STAMP2) is an androgen‐regulated gene whose mRNA expression is increased in prostate cancer (PCa). Here, we show that STAMP2 protein expression is increased in human PCa compared with benign prostate that is also correlated with tumor grade and treatment response. We also show that STAMP2 significantly increased reactive oxygen species (ROS) in PCa cells through its iron reductase activity which also depleted NADPH levels. Knockdown of STAMP2 expression in PCa cells inhibited proliferation, colony formation, and anchorage‐independent growth, and significantly increased apoptosis. Furthermore, STAMP2 effects were, at least in part, mediated by activating transcription factor 4 (ATF4), whose expression is regulated by ROS. Consistent with in vitro findings, silencing STAMP2 significantly inhibited PCa xenograft growth in mice. Finally, therapeutic silencing of STAMP2 by systemically administered nanoliposomal siRNA profoundly inhibited tumor growth in two established preclinical PCa models in mice. These data suggest that STAMP2 is required for PCa progression and thus may serve as a novel therapeutic target.


Clinical Cancer Research | 2017

A population of heterogeneous breast cancer patient-derived xenografts demonstrate broad activity of PARP inhibitor in BRCA1/2 wild-type tumors

Kurt W. Evans; Erkan Yuca; Argun Akcakanat; Stephen Scott; Natalia Paez Arango; Xiaofeng Zheng; Ken Chen; Coya Tapia; Emily Tarco; Agda Karina Eterovic; Dalliah Mashon Black; Jennifer K. Litton; Timothy A. Yap; Debu Tripathy; Gordon B. Mills; Funda Meric-Bernstam

Background: Breast cancer patients who do not respond to neoadjuvant therapy have a poor prognosis. There is a pressing need for novel targets and models for preclinical testing. Here we report characterization of breast cancer patient-derived xenografts (PDX) largely generated from residual tumors following neoadjuvant chemotherapy. Experimental Design: PDXs were derived from surgical samples of primary or locally recurrent tumors. Normal and tumor DNA sequencing, RNASeq, and reverse phase protein arrays (RPPA) were performed. Phenotypic profiling was performed by determining efficacy of a panel of standard and investigational agents. Results: Twenty-six PDXs were developed from 25 patients. Twenty-two were generated from residual disease following neoadjuvant chemotherapy, and 24 were from triple-negative breast cancer (TNBC). These PDXs harbored a heterogeneous set of genomic alterations and represented all TNBC molecular subtypes. On RPPA, PDXs varied in extent of PI3K and MAPK activation. PDXs also varied in their sensitivity to chemotherapeutic agents. PI3K, mTOR, and MEK inhibitors repressed growth but did not cause tumor regression. The PARP inhibitor talazoparib caused dramatic regression in five of 12 PDXs. Notably, four of five talazoparib-sensitive models did not harbor germline BRCA1/2 mutations, but several had somatic alterations in homologous repair pathways, including ATM deletion and BRCA2 alterations. Conclusions: PDXs capture the molecular and phenotypic heterogeneity of TNBC. Here we show that PARP inhibition can have activity beyond germline BRCA1/2 altered tumors, causing regression in a variety of molecular subtypes. These models represent an opportunity for the discovery of rational combinations with targeted therapies and predictive biomarkers. Clin Cancer Res; 23(21); 6468–77. ©2017 AACR.


Breast Cancer Research | 2017

Selinexor (KPT-330) demonstrates anti-tumor efficacy in preclinical models of triple-negative breast cancer

Natalia Paez Arango; Erkan Yuca; Ming Zhao; Kurt W. Evans; Stephen Scott; Charissa Kim; Ana M. Gonzalez-Angulo; Filip Janku; Naoto Ueno; Debu Tripathy; Argun Akcakanat; Aung Naing; Funda Meric-Bernstam

BackgroundSelinexor (KPT-330) is an oral agent that has been shown to inhibit the nuclear exporter XPO1. Given the pressing need for novel therapies for triple-negative breast cancer (TNBC), we sought to determine the antitumor effects of selinexor in vitro and in vivo.MethodsTwenty-six breast cancer cell lines of different breast cancer subtypes were treated with selinexor in vitro. Cell proliferation assays were used to measure the half-maximal inhibitory concentration (IC50) and to test the effects in combination with chemotherapy. In vivo efficacy was tested both as a single agent and in combination therapy in TNBC patient-derived xenografts (PDXs).ResultsSelinexor demonstrated growth inhibition in all 14 TNBC cell lines tested; TNBC cell lines were more sensitive to selinexor (median IC50 44 nM, range 11 to 550 nM) than were estrogen receptor (ER)-positive breast cancer cell lines (median IC50 > 1000 nM, range 40 to >1000 nM; P = 0.017). In multiple TNBC cell lines, selinexor was synergistic with paclitaxel, carboplatin, eribulin, and doxorubicin in vitro. Selinexor as a single agent reduced tumor growth in vivo in four of five different TNBC PDX models, with a median tumor growth inhibition ratio (T/C: treatment/control) of 42% (range 31 to 73%) and demonstrated greater antitumor efficacy in combination with paclitaxel or eribulin (average T/C ratios of 27% and 12%, respectively).ConclusionsCollectively, these findings strongly suggest that selinexor is a promising therapeutic agent for TNBC as a single agent and in combination with standard chemotherapy.


Patient Derived Tumor Xenograft Models#R##N#Promise, Potential and Practice | 2017

Modeling breast cancer heterogeneity with patient-derived xenografts

Kurt W. Evans; N. Paez-Arango; A. Akcakanat; Erkan Yuca; F. Meric-Bernstam

Abstract To better understand the biology underlying breast cancer progression and improve the development and application of therapies, it is essential to model the heterogeneity inherent to breast cancer. The ability to graft patient tumor fragments into immune-deficient mice is enabling development of relatively large collections of unique breast cancer models. Several different techniques, including orthotopic and heterotopic implantations, are being used to generate breast cancer patient-derived xenograft (PDX), and the consensus across techniques is that molecular and phenotypic traits exhibited by the patient tumors are for the most part maintained in the xenografts, although some genetic drift has been reported. At present, pools of breast cancer PDXs are being used in preclinical testing, and the coming years will illuminate whether the use of PDXs enables broader preclinical testing and increased therapeutic success.


Cancer Research | 2017

Abstract P3-07-01: Selinexor, a selective inhibitor of nuclear export, demonstrates efficacy in preclinical models of triple negative breast cancer

N Paez Arango; Kurt W. Evans; Ming Zhao; Erkan Yuca; Stephen Scott; Filip Janku; Naoto Ueno; Debu Tripathy; Charissa Kim; Aung Naing; M-B Funda

Background: Approximately 15% of all breast cancers are categorized as triple negative (TNBC) for which the only chemotherapy is known to be effective, yet often fails to achieve remission. Nuclear exporter XPO1 (Exportin1 or CRM1) is a promising target for cancer therapy that mediates the transport of multiple tumor suppressors and cell cycle regulators that have been known to be relevant predictors in the mechanism and severity of TNBC. Given the pressing need for novel therapies for this disease, we sought to determine the antitumor effects of selinexor, a novel inhibitor of nuclear export, on triple negative breast cancers in vitro and in vivo as well as to address its mechanism of action. Methods: 26 breast cancer cell lines of different breast cancer subtypes were treated with selinexor in vitro. Using cell proliferation assays the half maximal inhibitory concentration (IC50) was calculated using isobologram curves after 3 days of treatment; sensitivity was defined as IC50 Results: Selinexor demonstrated growth inhibition in all fourteen TNBC cell lines tested; TNBC cell lines were more sensitive to selinexor (median IC50 44nM, range 11 - 550nM), compared to ER+ cells lines (median IC50 of 13000 nM, range of 40nM - > 1000 nM; P=0.017). Treatment with selinexor decreased expression levels of XPO1, as well as survivin and XIAP, and induced apoptosis. In multiple TNBC cell lines selinexor was synergistic with paclitaxel, carboplatin, eribulin and doxorubicin in vitro (median combination index 0.6, range 0.5-0.8). Selinexor as a single agent reduced tumor growth in vivo in 4 of 5 different TNBC PDX models with a median tumor growth inhibition ratio score (T/C) of 48% (range 34-59%) and demonstrated greater antitumor efficacy in combination with paclitaxel or eribulin with an average T/C score of 27% and 12% respectively. Conclusions: Selinexor is a promising therapeutic agent for triple negative breast cancer and it has potential as a combination agent with standard chemotherapy. Citation Format: Paez Arango N, Evans KW, Zhao M, Yuca E, Scott SM, Janku F, Ueno NT, Tripathy D, Kim C, Naing A, Funda M-B. Selinexor, a selective inhibitor of nuclear export, demonstrates efficacy in preclinical models of triple negative breast cancer [abstract]. In: Proceedings of the 2016 San Antonio Breast Cancer Symposium; 2016 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2017;77(4 Suppl):Abstract nr P3-07-01.

Collaboration


Dive into the Erkan Yuca's collaboration.

Top Co-Authors

Avatar

Kurt W. Evans

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Stephen Scott

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Funda Meric-Bernstam

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bulent Ozpolat

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gabriel Lopez-Berestein

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Debu Tripathy

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ibrahim Tekedereli

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Argun Akcakanat

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ming Zhao

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge