Network


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

Hotspot


Dive into the research topics where Rebecca A. Previs is active.

Publication


Featured researches published by Rebecca A. Previs.


Clinical Cancer Research | 2015

Molecular Pathways: Translational and Therapeutic Implications of the Notch Signaling Pathway in Cancer

Rebecca A. Previs; Robert L. Coleman; Adrian L. Harris; Anil K. Sood

Over 100 years have passed since the first observation of the notched wing phenotype in Drosophila melanogaster, and significant progress has been made to characterize the role of the Notch receptor, its ligands, downstream targets, and cross-talk with other signaling pathways. The canonical Notch pathway with four Notch receptors (Notch1-4) and five ligands (DLL1, 3-4, Jagged 1-2) is an evolutionarily conserved cell signaling pathway that plays critical roles in cell-fate determination, differentiation, development, tissue patterning, cell proliferation, and death. In cancer, these roles have a critical impact on tumor behavior and response to therapy. Because the role of Notch remains tissue and context dependent, alterations within this pathway may lead to tumor suppressive or oncogenic phenotypes. Although no FDA-approved therapies currently exist for the Notch pathway, multiple therapeutics (e.g., demcizumab, tarextumab, GSI MK-0752, R04929097, and PF63084014) have been developed to target different aspects of this pathway for both hematologic and solid malignancies. Understanding the context-specific effects of the Notch pathway will be important for individualized therapies targeting this pathway. Clin Cancer Res; 21(5); 955–61. ©2014 AACR.


Cell Reports | 2015

Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis

Rajesha Rupaimoole; Jaehyuk Lee; Monika Haemmerle; Hui Ling; Rebecca A. Previs; Sunila Pradeep; Sherry Y. Wu; Cristina Ivan; Manuela Ferracin; Jennifer B. Dennison; Niki Zacharias Millward; Archana S. Nagaraja; Kshipra M. Gharpure; Michael McGuire; Nidhin Sam; Guillermo N. Armaiz-Pena; Nouara C. Sadaoui; Cristian Rodriguez-Aguayo; George A. Calin; Ronny Drapkin; Jeffery Kovacs; Gordon B. Mills; Wei Zhang; Gabriel Lopez-Berestein; Pratip Bhattacharya; Anil K. Sood

Long noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer.


Clinical Cancer Research | 2015

Differential platelet levels affect response to taxane-based therapy in ovarian cancer

Justin Bottsford-Miller; Hyun Jin Choi; Heather J. Dalton; Rebecca L. Stone; Min Soon Cho; Monika Haemmerle; Alpa M. Nick; Sunila Pradeep; Behrouz Zand; Rebecca A. Previs; Chad V. Pecot; Erin K. Crane; Wei Hu; Susan K. Lutgendorf; Vahid Afshar-Kharghan; Anil K. Sood

Purpose: We hypothesized that platelet levels during therapy could serve as a biomarker for response to therapy and that manipulation of platelet levels could impact responsiveness to chemotherapy. Experimental Design: The medical records of patients with recurrent or progressive ovarian cancer were retrospectively queried for changes in platelet and CA-125 levels during primary therapy. In vitro coculture experiments and in vivo orthotopic models of human ovarian cancer in mice were used to test the effect of modulating platelet levels on tumor growth and responsiveness to docetaxel. Results: Thrombocytosis at the diagnosis of ovarian cancer was correlated with decreased interval to progression (P = 0.05) and median overall survival (P = 0.007). Mean platelet levels corrected during primary therapy and rose at recurrence. Contrary to treatment-responsive patients, in a cohort of patients refractory to primary therapy, platelet levels did not normalize during therapy. In A2780, HeyA8, and SKOV3-ip1 ovarian cancer cell lines, platelet coculture protected against apoptosis (P < 0.05). In orthotopic models of human ovarian cancer, platelet depletion resulted in 70% reduced mean tumor weight (P < 0.05). Compared with mice treated with docetaxel, mice treated with both docetaxel and platelet-depleting antibody had a 62% decrease in mean tumor weight (P = 0.04). Platelet transfusion increased mean aggregate tumor weight 2.4-fold (P < 0.05), blocked the effect of docetaxel on tumor growth (P = 0.55) and decreased tumor cell apoptosis. Pretransfusion aspirinization of the platelets blocked the growth-promoting effects of transfusion. Conclusions: Platelet-driven effects of chemotherapy response may explain clinical observations. Clin Cancer Res; 21(3); 602–10. ©2014 AACR.


Oncogene | 2016

Hypoxia-upregulated microRNA-630 targets Dicer, leading to increased tumor progression.

Rajesha Rupaimoole; Cristina Ivan; Da Yang; Kshipra M. Gharpure; Sherry Y. Wu; Chad V. Pecot; Rebecca A. Previs; Archana S. Nagaraja; Guillermo N. Armaiz-Pena; Michael McGuire; Sunila Pradeep; Lingegowda S. Mangala; Cristian Rodriguez-Aguayo; Li Huang; Menashe Bar-Eli; Wei Zhang; Gabriel Lopez-Berestein; George A. Calin; Anil K. Sood

MicroRNAs (miRNAs) are small RNA molecules that affect cellular processes by controlling gene expression. Recent studies have shown that hypoxia downregulates Drosha and Dicer, key enzymes in miRNA biogenesis, causing a decreased pool of miRNAs in cancer and resulting in increased tumor growth and metastasis. Here we demonstrate a previously unrecognized mechanism by which hypoxia downregulates Dicer. We found that miR-630, which is upregulated under hypoxic conditions, targets and downregulates Dicer expression. In an orthotopic mouse model of ovarian cancer, delivery of miR-630 using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) nanoliposomes resulted in increased tumor growth and metastasis, and decreased Dicer expression. Treatment with the combination of anti-miR-630 and anti-vascular endothelial growth factor antibody in mice resulted in rescue of Dicer expression and significantly decreased tumor growth and metastasis. These results indicate that targeting miR-630 is a promising approach to overcome Dicer deregulation in cancer. As demonstrated in the study, use of DOPC nanoliposomes for anti-miR delivery serves as a better alternative approach to cell line-based overexpression of sense or antisense miRNAs, while avoiding potential in vitro selection effects. Findings from this study provide a new understanding of miRNA biogenesis downregulation observed under hypoxia and suggest therapeutic avenues to target this dysregulation in cancer.


Clinical Cancer Research | 2015

Immunotherapy Targeting Folate Receptor Induces Cell Death Associated with Autophagy in Ovarian Cancer

Yunfei Wen; Whitney S. Graybill; Rebecca A. Previs; Wei Hu; Cristina Ivan; Lingegowda S. Mangala; Behrouz Zand; Alpa M. Nick; Nicholas B. Jennings; Heather J. Dalton; Vasudha Sehgal; Prahlad T. Ram; J. Lee; Pablo Vivas-Mejia; Robert E. Coleman; Anil K. Sood

Purpose: Cancer cells are highly dependent on folate metabolism, making them susceptible to drugs that inhibit folate receptor activities. Targeting overexpressed folate receptor alpha (FRα) in cancer cells offers a therapeutic opportunity. We investigated the functional mechanisms of MORAB-003 (farletuzumab), a humanized mAb against FRα, in ovarian cancer models. Experimental Design: We first examined FRα expression in an array of human ovarian cancer cell lines and then assessed the in vivo effect of MORAB-003 on tumor growth and progression in several orthotopic mouse models of ovarian cancer derived from these cell lines. Molecular mechanisms of tumor cell death induced by MORAB-003 were investigated by cDNA and protein expression profiling analysis. Mechanistic studies were performed to determine the role of autophagy in MORAB-003–induced cell death. Results: MORAB-003 significantly decreased tumor growth in the high-FRα IGROV1 and SKOV3ip1 models but not in the low-FRα A2780 model. MORAB-003 reduced proliferation, but had no significant effect on apoptosis. Protein expression and cDNA microarray analyses showed that MORAB-003 regulated an array of autophagy-related genes. It also significantly increased expression of LC3 isoform II and enriched autophagic vacuolization. Blocking autophagy with hydroxychloroquine or bafilomycin A1 reversed the growth inhibition induced by MORAB-003. In addition, alteration of FOLR1 gene copy number significantly correlated with shorter disease-free survival in patients with ovarian serous cancer. Conclusions: MORAB-003 displays prominent antitumor activity in ovarian cancer models expressing FRα at high levels. Blockade of folate receptor by MORAB-003 induced sustained autophagy and suppressed cell proliferation. Clin Cancer Res; 21(2); 448–59. ©2014 AACR.


Oncogene | 2016

Sustained adrenergic signaling leads to increased metastasis in ovarian cancer via increased PGE2 synthesis

Archana S. Nagaraja; Piotr L. Dorniak; Nouara C. Sadaoui; Yu Kang; Tan Lin; Guillermo N. Armaiz-Pena; Sherry Y. Wu; Rajesha Rupaimoole; Julie K. Allen; Kshipra M. Gharpure; Sunila Pradeep; Behrouz Zand; Rebecca A. Previs; Jean M. Hansen; Cristina Ivan; Cristian Rodriguez-Aguayo; Peiying Yang; Gabriel Lopez-Berestein; Susan K. Lutgendorf; Steve W. Cole; Anil K. Sood

Adrenergic stimulation adversely affects tumor growth and metastasis, but the underlying mechanisms are not well understood. Here, we uncovered a novel mechanism by which catecholamines induce inflammation by increasing prostaglandin E2 (PGE2) levels in ovarian cancer cells. Metabolic changes in tumors isolated from patients with depression and mice subjected to restraint stress showed elevated PGE2 levels. Increased metabolites, PTGS2 and PTGES protein levels were found in Skov3-ip1 and HeyA8 cells treated with norepinephrine (NE), and these changes were shown to be mediated by ADRB2 receptor signaling. Silencing PTGS2 resulted in significantly decreased migration and invasion in ovarian cancer cells in the presence of NE and decreased tumor burden and metastasis in restraint stress orthotopic models. In human ovarian cancer samples, concurrent increased ADRB2, PTGS2 and PTGES expression was associated with reduced overall and progression-free patient survival. In conclusion, increased adrenergic stimulation results in increased PGE2 synthesis via ADRB2–Nf-kB–PTGS2 axis, which drives tumor growth and metastasis.


Clinical Cancer Research | 2015

Rac1/Pak1/p38/MMP-2 Axis Regulates Angiogenesis in Ovarian Cancer

Vianey Gonzalez-Villasana; Enrique Fuentes-Mattei; Cristina Ivan; Heather J. Dalton; Cristian Rodriguez-Aguayo; Ricardo J. Fernandez-de Thomas; Paloma Monroig; Guermarie Velazquez-Torres; Rebecca A. Previs; Sunila Pradeep; Nermin Kahraman; Huamin Wang; Pinar Kanlikilicer; Bulent Ozpolat; George A. Calin; Anil K. Sood; Gabriel Lopez-Berestein

Purpose: Zoledronic acid is being increasingly recognized for its antitumor properties, but the underlying functions are not well understood. In this study, we hypothesized that zoledronic acid inhibits ovarian cancer angiogenesis preventing Rac1 activation. Experimental Design: The biologic effects of zoledronic acid were examined using a series of in vitro [cell invasion, cytokine production, Rac1 activation, reverse-phase protein array, and in vivo (orthotopic mouse models)] experiments. Results: There was significant inhibition of ovarian cancer (HeyA8-MDR and OVCAR-5) cell invasion as well as reduced production of proangiogenic cytokines in response to zoledronic acid treatment. Furthermore, zoledronic acid inactivated Rac1 and decreased the levels of Pak1/p38/matrix metalloproteinase-2 in ovarian cancer cells. In vivo, zoledronic acid reduced tumor growth, angiogenesis, and cell proliferation and inactivated Rac1 in both HeyA8-MDR and OVCAR-5 models. These in vivo antitumor effects were enhanced in both models when zoledronic acid was combined with nab-paclitaxel. Conclusions: Zoledronic acid has robust antitumor and antiangiogenic activity and merits further clinical development as ovarian cancer treatment. Clin Cancer Res; 21(9); 2127–37. ©2015 AACR.


Cancer Cell | 2015

Erythropoietin Stimulates Tumor Growth via EphB4

Sunila Pradeep; Jie Huang; Edna Mora; Alpa M. Nick; Min Soon Cho; Sherry Y. Wu; Kyunghee Noh; Chad V. Pecot; Rajesha Rupaimoole; Martin Stein; Stephan Brock; Yunfei Wen; Chiyi Xiong; Kshipra M. Gharpure; Jean M. Hansen; Archana S. Nagaraja; Rebecca A. Previs; Pablo Vivas-Mejia; Hee Dong Han; Wei Hu; Lingegowda S. Mangala; Behrouz Zand; Loren J. Stagg; John E. Ladbury; Bulent Ozpolat; S. Neslihan Alpay; Masato Nishimura; Rebecca L. Stone; Koji Matsuo; Guillermo N. Armaiz-Pena

While recombinant human erythropoietin (rhEpo) has been widely used to treat anemia in cancer patients, concerns about its adverse effects on patient survival have emerged. A lack of correlation between expression of the canonical EpoR and rhEpos effects on cancer cells prompted us to consider the existence of an alternative Epo receptor. Here, we identified EphB4 as an Epo receptor that triggers downstream signaling via STAT3 and promotes rhEpo-induced tumor growth and progression. In human ovarian and breast cancer samples, expression of EphB4 rather than the canonical EpoR correlated with decreased disease-specific survival in rhEpo-treated patients. These results identify EphB4 as a critical mediator of erythropoietin-induced tumor progression and further provide clinically significant dimension to the biology of erythropoietin.


Cancer Research | 2014

Notch3 Pathway Alterations in Ovarian Cancer

Wei Hu; Tao Liu; Cristina Ivan; Yunjie Sun; Jie Huang; Lingegowda S. Mangala; Takahito Miyake; Heather J. Dalton; Sunila Pradeep; R. Rupaimoole; Rebecca A. Previs; Hee Dong Han; Justin Bottsford-Miller; Behrouz Zand; Yu Kang; Chad V. Pecot; Alpa M. Nick; Sherry Y. Wu; Ju Seog Lee; Vasudha Sehgal; Prahlad T. Ram; Jinsong Liu; Susan L. Tucker; Gabriel Lopez-Berestein; Keith A. Baggerly; Robert L. Coleman; Anil K. Sood

The Notch pathway plays an important role in the growth of high-grade serous ovarian (HGS-OvCa) and other cancers, but its clinical and biologic mechanisms are not well understood. Here, we found that the Notch pathway alterations are prevalent and significantly related to poor clinical outcome in patients with ovarian cancer. Particularly, Notch3 alterations, including amplification and upregulation, were highly associated with poor patient survival. Targeting Notch3 inhibited ovarian cancer growth and induced apoptosis. Importantly, we found that dynamin-mediated endocytosis was required for selectively activating Jagged-1-mediated Notch3 signaling. Cleaved Notch3 expression was the critical determinant of response to Notch-targeted therapy. Collectively, these data identify previously unknown mechanisms underlying Notch3 signaling and identify new, biomarker-driven approaches for therapy.


Journal of the National Cancer Institute | 2016

Role of Increased n-acetylaspartate Levels in Cancer

Behrouz Zand; Rebecca A. Previs; Niki Zacharias; Rajesha Rupaimoole; Takashi Mitamura; Archana S. Nagaraja; Michele Guindani; Heather J. Dalton; Lifeng Yang; Joelle Baddour; Abhinav Achreja; Wei Hu; Chad V. Pecot; Cristina Ivan; Sherry Y. Wu; Christopher R. McCullough; Kshipra M. Gharpure; Einav Shoshan; Sunila Pradeep; Lingegowda S. Mangala; Cristian Rodriguez-Aguayo; Ying Wang; Alpa M. Nick; Michael A. Davies; Guillermo N. Armaiz-Pena; Jinsong Liu; Susan K. Lutgendorf; Keith A. Baggerly; Menashe Bar Eli; Gabriel Lopez-Berestein

BACKGROUND The clinical and biological effects of metabolic alterations in cancer are not fully understood. METHODS In high-grade serous ovarian cancer (HGSOC) samples (n = 101), over 170 metabolites were profiled and compared with normal ovarian tissues (n = 15). To determine NAT8L gene expression across different cancer types, we analyzed the RNA expression of cancer types using RNASeqV2 data available from the open access The Cancer Genome Atlas (TCGA) website (http://www.cbioportal.org/public-portal/). Using NAT8L siRNA, molecular techniques and histological analysis, we determined cancer cell viability, proliferation, apoptosis, and tumor growth in in vitro and in vivo (n = 6-10 mice/group) settings. Data were analyzed with the Students t test and Kaplan-Meier analysis. Statistical tests were two-sided. RESULTS Patients with high levels of tumoral NAA and its biosynthetic enzyme, aspartate N-acetyltransferase (NAT8L), had worse overall survival than patients with low levels of NAA and NAT8L. The overall survival duration of patients with higher-than-median NAA levels (3.6 years) was lower than that of patients with lower-than-median NAA levels (5.1 years, P = .03). High NAT8L gene expression in other cancers (melanoma, renal cell, breast, colon, and uterine cancers) was associated with worse overall survival. NAT8L silencing reduced cancer cell viability (HEYA8: control siRNA 90.61% ± 2.53, NAT8L siRNA 39.43% ± 3.00, P < .001; A2780: control siRNA 90.59% ± 2.53, NAT8L siRNA 7.44% ± 1.71, P < .001) and proliferation (HEYA8: control siRNA 74.83% ± 0.92, NAT8L siRNA 55.70% ± 1.54, P < .001; A2780: control siRNA 50.17% ± 4.13, NAT8L siRNA 26.52% ± 3.70, P < .001), which was rescued by addition of NAA. In orthotopic mouse models (ovarian cancer and melanoma), NAT8L silencing reduced tumor growth statistically significantly (A2780: control siRNA 0.52 g ± 0.15, NAT8L siRNA 0.08 g ± 0.17, P < .001; HEYA8: control siRNA 0.79 g ± 0.42, NAT8L siRNA 0.24 g ± 0.18, P = .008, A375-SM: control siRNA 0.55 g ± 0.22, NAT8L siRNA 0.21 g ± 0.17 g, P = .001). NAT8L silencing downregulated the anti-apoptotic pathway, which was mediated through FOXM1. CONCLUSION These findings indicate that the NAA pathway has a prominent role in promoting tumor growth and represents a valuable target for anticancer therapy.Altered energy metabolism is a hallmark of cancer (1). Proliferating cancer cells have much greater metabolic requirements than nonproliferating differentiated cells (2,3). Moreover, altered cancer metabolism elevates unique metabolic intermediates, which can promote cancer survival and progression (4,5). Furthermore, emerging evidence suggests that proliferating cancer cells exploit alternative metabolic pathways to meet their high demand for energy and to accumulate biomass (6-8).

Collaboration


Dive into the Rebecca A. Previs's collaboration.

Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Heather J. Dalton

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Robert L. Coleman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jean M. Hansen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sunila Pradeep

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wei Hu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Behrouz Zand

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Guillermo N. Armaiz-Pena

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cristina Ivan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rajesha Rupaimoole

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge