Network


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

Hotspot


Dive into the research topics where Khandan Keyomarsi is active.

Publication


Featured researches published by Khandan Keyomarsi.


Nature Communications | 2014

2′-OMe-phosphorodithioate-modified siRNAs show increased loading into the RISC complex and enhanced anti-tumour activity

Sherry Y. Wu; Xianbin Yang; Kshipra M. Gharpure; Hiroto Hatakeyama; Martin Egli; Michael McGuire; Archana S. Nagaraja; Takahito Miyake; Rajesha Rupaimoole; Chad V. Pecot; Morgan Taylor; Sunila Pradeep; Malgorzata Sierant; Cristian Rodriguez-Aguayo; Hyun J. Choi; Rebecca A. Previs; Guillermo N. Armaiz-Pena; Li Huang; Carlos Martinez; Tom Hassell; Cristina Ivan; Vasudha Sehgal; Richa Singhania; Hee Dong Han; Chang Su; Ji Hoon Kim; Heather J. Dalton; Chandra Kovvali; Khandan Keyomarsi; Nigel A.J. McMillan

Improving small interfering RNA (siRNA) efficacy in target cell populations remains a challenge to its clinical implementation. Here, we report a chemical modification, consisting of phosphorodithioate (PS2) and 2-O-Methyl (2-OMe) MePS2 on one nucleotide that significantly enhances potency and resistance to degradation for various siRNAs. We find enhanced potency stems from an unforeseen increase in siRNA loading to the RNA-induced silencing complex, likely due to the unique interaction mediated by 2-OMe and PS2. We demonstrate the therapeutic utility of MePS2 siRNAs in chemoresistant ovarian cancer mouse models via targeting GRAM domain containing 1B (GRAMD1B), a protein involved in chemoresistance. GRAMD1B silencing is achieved in tumours following MePS2-modified siRNA treatment, leading to a synergistic anti-tumour effect in combination with paclitaxel. Given the previously limited success in enhancing siRNA potency with chemically modified siRNAs, our findings represent an important advance in siRNA design with the potential for application in numerous cancer types.


Breast Cancer Research and Treatment | 2012

Targeting low molecular weight cyclin E (LMW-E) in breast cancer

Angela Nanos-Webb; Natalie A. Jabbour; Asha S. Multani; Hannah Wingate; Nassima Oumata; Hervé Galons; Benoît Joseph; Laurent Meijer; Kelly K. Hunt; Khandan Keyomarsi

Low molecular weight cyclin E (LMW-E) plays an important oncogenic role in breast cancer. LMW-E, which is not found in normal tissue, can promote the formation of aggressive tumors and can lead to increased genomic instability and tumorigenesis. Additionally, breast cancer patients whose tumors express LMW-E have a very poor prognosis. Therefore, we investigated LMW-E as a potential specific target for treatment either alone or in combination therapy. We hypothesized that because LMW-E binds to CDK2 more efficiently than full length cyclin E, resulting in increased activity, CDK inhibitors could be used to target tumors with LMW-E bound to CDK2. To test the hypothesis, an inducible full length and LMW-E MCF7-Tet-On system was established. Cyclin E (full length (EL) or LMW-E) is only expressed upon induction of the transgene. The doubling times of cells were unchanged when the transgenes were induced. However, upon induction, the kinase activity associated with LMW-E was much higher than that in the EL induced cells or any of the uninduced cells. Additionally only the LMW-E induced cells underwent chromosome aberrations and increased polyploidy. By examining changes in proliferation and survival in cells with induced full length and LMW-E, CDK inhibitors alone were determined to be insufficient to specifically inhibit LMW-E expressing cells. However, in combination with Doxorubicin, the CDK inhibitor, Roscovitine (Seliciclib, CYC202), synergistically led to increased cell death in LMW-E expressing cells. Clinically, the combination of CDK inhibitors and chemotherapy such as Doxorubicin provides a viable personalized treatment strategy for those breast cancer patients whose tumors express the LMW-E.


Breast Cancer Research | 2004

Low-molecular-weight cyclin E: the missing link between biology and clinical outcome

Said Akli; Khandan Keyomarsi

Cyclin E, a key mediator of transition during the G1/S cellular division phase, is deregulated in a wide variety of human cancers. Our group recently reported that overexpression and generation of low-molecular-weight (LMW) isoforms of cyclin E were associated with poor clinical outcome among breast cancer patients. However, the link between LMW cyclin E biology in mediating a tumorigenic phenotype and clinical outcome is unknown. To address this gap in knowledge, we assessed the role of LMW isoforms in breast cancer cells; we found that these forms of cyclin E induced genomic instability and resistance to p21, p27, and antiestrogens in breast cancer. These findings suggest that high levels of LMW isoforms of cyclin E not only can predict failure to endocrine therapy but also are true prognostic indicators because of their influence on cell proliferation and genetic instability.


Investigational New Drugs | 2015

A phase 1 study with dose expansion of the CDK inhibitor dinaciclib (SCH 727965) in combination with epirubicin in patients with metastatic triple negative breast cancer

Zahi Mitri; Cansu Karakas; Caimiao Wei; Brian Briones; Holly Simmons; Nuhad K. Ibrahim; Ricardo H. Alvarez; James L. Murray; Khandan Keyomarsi; Stacy L. Moulder

SummaryPurpose, Low molecular weight cyclin E (LMW-E) isoforms, overexpressed in a majority (~70xa0%) of triple-negative breast cancers (TNBC), were found in preclinical models to mediate tumorigenesis through binding and activation of CDK2. CDK1/CDK2 inhibitors, such as dinaciclib, combined with anthracyclines, were synergistic in decreasing viability of TNBC cell lines. Based on this data, a phase 1 study was conducted to determine the maximum tolerated dose of dinaciclib in combination with epirubicin in patients with metastatic TNBC. Methods, Cohorts of at least 2 patients were treated with escalating doses of dinaciclib given on day 1 followed by standard dose of epirubicin given on day 2 of a 21xa0dayxa0cycle. No intra-patient dose escalation was allowed. An adaptive accrual design based upon toxicity during cycle 1 determined entry into therapy cohorts. The target acceptable dose limiting toxicity (DLT) to advance to the next treatment level was 30xa0%. Results, Between 9/18/2012 and 7/18/2013, 9 patients were enrolled and treated at MD Anderson Cancer Center. DLTs included febrile neutropenia (grade 3, nu2009=u20092), syncope (grade 3, nu2009=u20092) and vomiting (grade 3, nu2009=u20091). Dose escalation did not proceed past the second cohort due to toxicity. After further accrual, the first dose level was also found to be too toxic. No treatment responses were noted, median time to progression was 5.5xa0weeks (range 3–12xa0weeks). Thus, accrual was stopped rather than explore the −1 dose level. Conclusion, The combination of dinaciclib and epirubicin is associated with substantial toxicities and does not appear to be an effective treatment option for TNBC.


Breast Cancer Research and Treatment | 2009

Cyclin E deregulation is an early event in the development of breast cancer

Alexandra Shaye; Aysegul A. Sahin; Qiang Hao; Kelly K. Hunt; Khandan Keyomarsi; Isabelle Bedrosian

Cyclin E has been shown to be overexpressed in some human breast cancers, however, data to support deregulation of cyclin E as an early event in human mammary tumor development is lacking. We analyzed surgical specimens from 183 patients with breast carcinomas and evaluated cyclin E expression in areas of invasive carcinoma, adjacent carcinoma in situ (CIS), and non-neoplastic breast parenchyma. Overexpression of cyclin E was seen in one-third of invasive carcinoma samples, one-third of the CIS component and nearly half of the non-neoplastic breast epithelial cells adjacent to carcinoma (44% vs. 33%, Pxa0≤xa00.05). Nuclear labeling for cyclin E was highly concordant between areas of in invasive carcinoma, CIS and non-neoplastic breast epithelial cells from the same patient (Pxa0<xa00.0001). Localization of cyclin E to the cytoplasm was seen in a small proportion of tumor samples. Our findings suggest that cyclin E deregulation is an early event in the progression from histologically benign mammary epithelial cells to invasive carcinoma and occurs through both overexpression and altered cellular localization.


Molecular Oncology | 2013

EVI1 splice variants modulate functional responses in ovarian cancer cells

Punashi Dutta; Tuyen Bui; Kyle A. Bauckman; Khandan Keyomarsi; Gordon B. Mills; Meera Nanjundan

Amplification of 3q26.2, found in many cancer lineages, is a frequent and early event in ovarian cancer. We previously defined the most frequent region of copy number increase at 3q26.2 to EVI1 (ecotropic viral integration site‐1) and MDS1 (myelodysplastic syndrome 1) (aka MECOM), an observation recently confirmed by the cancer genome atlas (TCGA). MECOM is increased at the DNA, RNA, and protein level and likely contributes to patient outcome. Herein, we report that EVI1 is aberrantly spliced, generating multiple variants including a Del190–515 variant (equivalent to previously reported) expressed in >90% of advanced stage serous epithelial ovarian cancers. Although EVI1Del190–515 lacks ∼70% of exon 7, it binds CtBP1 as well as SMAD3, important mediators of TGFβ signaling, similar to wild type EVI1. This contrasts with EVI1 1–268 which failed to interact with CtBP1. Interestingly, the EVI1Del190–515 splice variant preferentially localizes to PML nuclear bodies compared to wild type and EVI1Del427–515. While wild type EVI1 efficiently repressed TGFβ‐mediated AP‐1 (activator protein‐1) and plasminogen activator inhibitor‐1 (PAI‐1) promoters, EVI1Del190–515 elicited a slight increase in both promoter activities. Expression of EVI1 and EVI1Del427–515 (but not EVI1Del190–515) in OVCAR8 ovarian cancer cells increased cyclin E1 LMW expression and cell cycle progression. Furthermore, knockdown of specific EVI1 splice variants (both MDS1/EVI1 and EVI1Del190–515) markedly increased claudin‐1 mRNA and protein expression in HEY ovarian and MDA‐MB‐231 breast cancer cells. Changes in claudin‐1 were associated with alterations in specific epithelial–mesenchymal transition markers concurrent with reduced migratory potential. Collectively, EVI1 is frequently aberrantly spliced in ovarian cancer with specific forms eliciting altered functions which could potentially contribute to ovarian cancer pathophysiology.


Nature Communications | 2016

PAF-Wnt signaling-induced cell plasticity is required for maintenance of breast cancer cell stemness

Xin Wang; Youn-Sang Jung; Sohee Jun; Sun-Hye Lee; Wenqi Wang; Andrea Schneider; Young Sun Oh; Steven Hsesheng Lin; Bum Joon Park; Junjie Chen; Khandan Keyomarsi; Jae-Il Park

Cancer stem cells (CSCs) contribute to tumour heterogeneity, therapy resistance and metastasis. However, the regulatory mechanisms of cancer cell stemness remain elusive. Here we identify PCNA-associated factor (PAF) as a key molecule that controls cancer cell stemness. PAF is highly expressed in breast cancer cells but not in mammary epithelial cells (MECs). In MECs, ectopic expression of PAF induces anchorage-independent cell growth and breast CSC marker expression. In mouse models, conditional PAF expression induces mammary ductal hyperplasia. Moreover, PAF expression endows MECs with a self-renewing capacity and cell heterogeneity generation via Wnt signalling. Conversely, ablation of endogenous PAF induces the loss of breast cancer cell stemness. Further cancer drug repurposing approaches reveal that NVP-AUY922 downregulates PAF and decreases breast cancer cell stemness. Our results unveil an unsuspected role of the PAF-Wnt signalling axis in modulating cell plasticity, which is required for the maintenance of breast cancer cell stemness.


American Journal of Pathology | 2016

Cytoplasmic Cyclin E and Phospho-Cyclin-Dependent Kinase 2 Are Biomarkers of Aggressive Breast Cancer.

Cansu Karakas; Anna Biernacka; Tuyen Bui; Aysegul A. Sahin; Min Yi; Said Akli; Jolie R. Schafer; Angela Alexander; Opoku Adjapong; Kelly K. Hunt; Khandan Keyomarsi

Cyclin E and its co-activator, phospho-cyclin-dependent kinase 2 (p-CDK2), regulate G1 to S phase transition and their deregulation induces oncogenesis. Immunohistochemical assessments of these proteins in cancer have been reported but were based only on their nuclear expression. However, the oncogenic forms of cyclin E (low molecular weight cyclin E or LMW-E) in complex with CDK2 are preferentially mislocalized to the cytoplasm. Here, we used separate nuclear and cytoplasmic scoring systems for both cyclin E and p-CDK2 expression to demonstrate altered cellular accumulation of these proteins using immunohistochemical analysis. We examined the specificity of different cyclin E antibodies and evaluated their concordance between immunohistochemical and Western blot analyses in a panel of 14 breast cell lines. Nuclear versus cytoplasmic staining of cyclin E readily differentiated full-length from LMW-E, respectively. We also evaluated the expression of cyclin E and p-CDK2 in 1676 breast carcinoma patients by immunohistochemistry. Cytoplasmic cyclin E correlated strongly with cytoplasmic p-CDK2 (Pxa0<xa00.0001), high tumor grade, negative estrogen/progesterone receptor status, and human epidermal growth factor receptor 2 positivity (all Pxa0<xa00.0001). In multivariable analysis, cytoplasmic cyclin E plus phosphorylated CDK2 (as one variable) predicted breast cancer recurrence-free and overall survival. These results suggest that cytoplasmic cyclin E and p-CDK2 can be readily detected with immunohistochemistry andxa0used as clinical biomarkers for aggressive breast cancer.


Cancer Research | 2016

Cyclin E Associates with the Lipogenic Enzyme ATP-Citrate Lyase to Enable Malignant Growth of Breast Cancer Cells

Kimberly S. Lucenay; Iman Doostan; Cansu Karakas; Tuyen Bui; Zhiyong Ding; Gordon B. Mills; Kelly K. Hunt; Khandan Keyomarsi

Cyclin E is altered in nearly a third of invasive breast cancers where it is a powerful independent predictor of survival in women with stage I-III disease. Full-length cyclin E is posttranslationally cleaved into low molecular weight (LMW-E) isoforms, which are tumor-specific and accumulate in the cytoplasm because they lack a nuclear localization sequence. We hypothesized that aberrant localization of cytosolic LMW-E isoforms alters target binding and activation ultimately contributing to LMW-E-induced tumorigenicity. To address this hypothesis, we used a retrovirus-based protein complementation assay to find LMW-E binding proteins in breast cancer, identifying ATP-citrate lyase (ACLY), an enzyme in the de novo lipogenesis pathway, as a novel LMW-E-interacting protein in the cytoplasm. LMW-E upregulated ACLY enzymatic activity, subsequently increasing lipid droplet formation, thereby providing cells with essential building blocks to support growth. ACLY was also required for LMW-E-mediated transformation, migration, and invasion of breast cancer cells in vitro along with tumor growth in vivo In clinical specimens of breast cancer, the absence of LMW-E and low expression of adipophilin (PLIN2), a marker of lipid droplet formation, associated with favorable prognosis, whereas overexpression of both proteins correlated with a markedly worse prognosis. Taken together, our findings establish a novel relationship between LMW-E isoforms of cyclin E and aberrant lipid metabolism pathways in breast cancer tumorigenesis, warranting further investigation in additional malignancies exhibiting their expression. Cancer Res; 76(8); 2406-18. ©2016 AACR.


Advances in Experimental Medicine and Biology | 2007

Cell cycle deregulation in breast cancer : Insurmountable chemoresistance or achilles' heel?

Laura A. Lambert; Khandan Keyomarsi

Deregulation of the G1 cyclin, cyclin E, has been shown to be both the most powerful predictor of prognosis in early stage breast cancer as well as a significant determinant of tumor aggressiveness. It may also contribute to treatment failure due to chemoresistance. Because some form of cell cycle deregulation is present in all malignant cells,3 increasing understanding of these processes is starting to provide new opportunities to overcome the cells resistance mechanisms. One particular form of cyclin E deregulation, the generation of hyperactive low molecular weight isoforms, is especially intriguing. Because only tumor cells contain the machinery necessary to generate these isoforms, they not only provide a mechanism of targeting critical cell cycle events, but their presence may also provide both a means of increased specificity for targeting malignant cells, as well as an objective measure of response. This review describes the mechanisms of resistance to commonly used systemic therapies for the treatment of breast cancer, with particular respect to the role of the cell cycle. The mechanisms and effects of the deregulation of cyclin E in breast cancer are reviewed and novel approaches to circumventing chemoresistance through abrogation of the malignant cell cycle are proposed.

Collaboration


Dive into the Khandan Keyomarsi's collaboration.

Top Co-Authors

Avatar

Kelly K. Hunt

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cansu Karakas

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Tuyen Bui

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jason P.W. Carey

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Said Akli

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Smruthi Vijayaraghavan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Angela Alexander

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Aysegul A. Sahin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hannah Wingate

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Iman Doostan

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge