Network


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

Hotspot


Dive into the research topics where Anna Frolov is active.

Publication


Featured researches published by Anna Frolov.


Clinical Cancer Research | 2004

High Levels of Phosphorylated Form of Akt-1 in Prostate Cancer and Non-Neoplastic Prostate Tissues Are Strong Predictors of Biochemical Recurrence

Gustavo Ayala; Timothy C. Thompson; Guang Yang; Anna Frolov; Rile Li; Peter T. Scardino; Makoto Ohori; Thomas M. Wheeler; Wade Harper

Akt is a serine-threonine-kinase that phosphorylates proteins in several pathways regulating aspects of metabolism, apoptosis, and proliferation. Akt signaling promotes proliferation and increased cell survival and is thought to play an important role in prostate cancer progression. Tissue microarrays (640 patients) with triplicate cores of non-neoplastic prostate, BPH, and index tumor were immunostained with antibody to Phospho-Akt (Ser473), digitized, and quantified. The expression index (Intensity*Percentage) was used for statistical analysis. P-Akt-1 staining was found in both the non-neoplastic and cancer tissues, predominantly in cytoplasmic locations. High level P-Akt-1 is expressed almost exclusively in cancer. By Kaplan-Meier actuarial model, high expression of P-Akt-1 in prostate cancer was predictive of a higher probability of recurrence on univariate and multivariate analysis. Akt-1 expression was an independent prognostic indicator of biochemical recurrence-free survival when Gleason 6 and 7 patients were analyzed separately. Surprisingly, a high level of P-Akt-1 expression in non-neoplastic tissues is also an independent predictor of biochemical recurrence. This suggests that some patients might have an inherent predisposition to express a high level of P-Akt-1 and, therefore, to have an adverse prognosis. We conclude that P-Akt-1 is most likely involved in the progression of prostate cancer and is an excellent biomarker for biochemical recurrence.


The American Journal of Surgical Pathology | 2004

High level of androgen receptor is associated with aggressive clinicopathologic features and decreased biochemical recurrence-free survival in prostate: Cancer patients treated with radical prostatectomy

Rile Li; Thomas M. Wheeler; Hong Dai; Anna Frolov; Timothy C. Thompson; Gustavo Ayala

Background:Prostate cancer (PCa) is androgen dependent and is regulated by androgen/androgen receptor (AR) signaling pathway. However, the clinical significance of AR is in question. In this regard, we have correlated levels of AR expression with some well-established clinical and pathologic parameters and assessed the prognostic value of AR expression in PCa patients treated with radical prostatectomy. Design:A total of 640 cases treated with radical prostatectomy were used to build tissue microarrays. Normal prostate tissue, benign prostatic hyperplasia, and index tumor were cored in triplicate (0.6 mm). An array (2 mm) of 177 metastatic PCa was built as well. Slides were immunostained with an antibody to AR and Ki-67 and digitized. Correlations between AR expression and clinicopathologic variables were analyzed by the Spearman test. Biochemical recurrence-free survival analysis was performed using Kaplan-Meier analysis, and Cox proportional hazard regression was used to determine the probability of disease recurrence. Results:AR was found in epithelial nuclei of both benign and cancer tissues. AR index was higher in normal prostate tissues than that in PCa and benign prostatic hyperplasia and decreased in metastases than PCa. High level of AR expression was correlated with clinical stage, lymph node status, extracapsular extension, seminal vesicle invasion, and Gleason score. High levels of AR status also correlated with high Ki-67 index (r2 = 0.211, P = 0.0000). By Kaplan-Meier actuarial model, high expression of AR was predictive of a higher probability of recurrence (P = 0.0046, hazards ratio 2.72 [confidence interval 1.28–4.011]). By multivariate analysis, a high level of AR expression was an independent prognostic indicator of biochemical recurrence-free survival (P = 0.0042; hazards ratio 2.422 [confidence interval 1.32–4.44]). Conclusions:High levels of AR are associated with increased proliferation, markers of aggressive disease and are predictive of decreased biochemical recurrence-free survival independently. This confirms the role of AR in tumor growth and progression in hormonally naive PCa.


Cancer Research | 2005

Role of SRC-1 in the Promotion of Prostate Cancer Cell Growth and Tumor Progression

Irina U. Agoulnik; Ajula Vaid; William E. Bingman; Halime Erdeme; Anna Frolov; Carolyn L. Smith; Gustavo Ayala; Michael Ittmann; Nancy L. Weigel

Prostate cancer is initially androgen dependent and there is evidence that androgen receptor continues to play a role in androgen-independent prostate cancer. Androgen receptor activity depends both on the level of androgens and on the level of coactivators that interact with androgen receptor. Our goal was to evaluate the role of the androgen receptor coactivator SRC-1 in prostate cancer progression. Using tissue arrays to measure SRC-1 protein levels, we found that increased SRC-1 expression in clinically localized, androgen-dependent cancer is associated with clinical and pathologic variables of increased tumor aggressiveness. Interestingly, there was variable expression of SRC-1 in normal prostate tissue which correlated with the staining intensity of the corresponding cancer tissue. To test the contribution of SRC-1, we examined its role in androgen-dependent LNCaP and androgen-independent C4-2 prostate cancer cell lines. Using small interfering RNA to reduce expression of androgen receptor, we found that androgen receptor was required both for cell growth and for basal expression of prostate-specific antigen in the androgen-independent C4-2 cell line. Thus, although the cells can grow in an androgen-depleted medium, they remained androgen receptor dependent. Reduction of SRC-1 expression significantly reduced growth and altered androgen receptor target gene regulation in both LNCaP and C4-2 cell lines whereas it had no effect on the growth of the androgen receptor-negative PC-3 and DU145 prostate cancer cell lines. Although the requirement for androgens and androgen receptor in the development of prostate cancer is well established, our study implicates enhanced androgen receptor activity through elevated expression of SRC-1 in the development of more aggressive disease in men with prostate cancer.


Cancer Research | 2004

Growth and Survival Mechanisms Associated with Perineural Invasion in Prostate Cancer

Gustavo Ayala; Hong Dai; Michael Ittmann; Rile Li; Michael Powell; Anna Frolov; Thomas M. Wheeler; Timothy C. Thompson; David R. Rowley

Perineural invasion (PNI) is the major mechanism of prostate cancer spread outside the prostate. Apoptotic and proliferation indices were determined in PNI cells using the PNI in vitro model and human PNI in tissue microarrays. RNA was extracted from the PNI model and controls and evaluated by cDNA microarray analysis. Differential expression of candidate genes was confirmed by real-time quantitative PCR, fluorescence, and immunohistochemistry using tissue microarrays. Genistein and BAY 11-7085 were added to the supernatant of cocultures and controls in microchamber cultures. The significance of nuclear factor κB (NFκB) nuclear translocation in human PNI was analyzed using Kaplan-Meier analysis. An increase in proliferation and a decrease in apoptosis were observed in human PNI cells and the PNI model as compared with controls. Three of 15 genes up-regulated in the cDNA microarray were involved in the apoptosis signaling pathway (NFκB), and its downstream targets defender against cell death 1 and PIM-2. The increase was corroborated by real-time quantitative PCR and immunofluorescence. NFκB nuclear translocation was seen in the in vitro model and human tissues, where strong nuclear expression was associated with a decrease in recurrence-free survival. Addition of genistein and BAY 11-7085 resulted in a decrease in NFκB, PIM-2 and defender against cell death 1 as well as a reversal of the inhibition of apoptosis. This is the first description of a biological mechanism and functional significance of PNI. Cancer cells in a perineural location acquire a survival and growth advantage using a NFκB survival pathway. Targeting PNI might help detain local spread of the tumor and influence survival.


Cancer Research | 2006

Targeting Aurora kinases for the treatment of prostate cancer.

Edmund Chun Yu Lee; Anna Frolov; Rile Li; Gustavo Ayala; Norman M. Greenberg

Inappropriate expression of the Aurora kinases can induce aberrant mitosis, centrosome irregularities, and chromosomal instability, which lead to anueploidy and cell transformation. Here, we report that Aurora-A and Aurora-B are highly expressed in primary human and mouse prostate cancers and prostate cancer cell lines. In clinical samples, levels of Aurora-A and Aurora-B were significantly elevated in prostatic intraepithelial neoplasia lesions and prostate tumors when compared with the non-neoplastic samples. Interestingly, expression of Aurora-A in non-neoplastic prostates correlated with seminal vesicle invasion (rho = 0.275, P = 0.0169) and in prostate tumor with positive surgical margins (rho = 0.265, P = 0.0161). In addition, nuclear expression of Aurora-B in prostatic intraepithelial neoplasia lesions correlated with clinical staging of the tumor (rho = -0.4, P = 0.0474) whereas cytoplasmic expression in tumors correlated with seminal vesicle invasion (rho = 0.282, P = 0.0098). Cell lines and primary tumors derived from the TRAMP model were also found to express high levels of Aurora-A and Aurora-B. When human PC3, LNCaP, and mouse C1A cells were treated with the potent Aurora kinase inhibitor VX680, which attenuates phosphorylation of histone H3, cancer cell survival was reduced. VX680 could further reduce cell viability >2-fold when used in combination with the chemotherapy drug doxorubicin. Our findings support a functional relationship between Aurora kinase expression and prostate cancer and the application of small-molecule inhibitors in therapeutic modalities.


Cancer Research | 2011

Decreased Expression and Androgen Regulation of the Tumor Suppressor Gene INPP4B in Prostate Cancer

Myles C. Hodgson; Long Jiang Shao; Anna Frolov; Rile Li; Leif E. Peterson; Gustavo Ayala; Michael Ittmann; Nancy L. Weigel; Irina U. Agoulnik

Patients with metastatic prostate cancer who undergo androgen-ablation therapy invariably relapse and develop incurable castration-resistant disease. Activation of the prosurvival Akt pathway accompanies androgen ablation. We discovered that the androgen receptor induces the expression of the tumor suppressor inositol polyphosphate 4-phosphatase type II (INPP4B) but not PTEN in prostate cancer cells. Optimal induction of INPP4B by an androgen receptor required the expression of the transcriptional coactivator NCoR. INPP4B dephosphorylates phosphatidylinositol-3, 4-bisphosphate, which leads to reduced phosphorylation and activity of Akt. In support of a key role for INPP4B in Akt control, INPP4B depletion activated Akt and increased cellular proliferation. The clinical significance of INPP4B in androgen-dependent prostate cancers was determined in normal or primary tumor prostate tissues derived from radical prostatectomy specimens. In primary tumors, the expression of both INPP4B and PTEN was substantially reduced compared with normal tissue. Further, the decreased expression of INPP4B reduced the time to biochemical recurrence. Thus, androgen ablation can activate the Akt pathway via INPP4B downregulation, thereby mitigating the antitumor effects of androgen ablation. Our findings reinforce the concept that patients undergoing androgen ablation may benefit from Akt-targeting therapies.


Nature | 2012

COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis

Jun Qin; San Pin Wu; Chad J. Creighton; Fangyan Dai; Xin Xie; Chiang Min Cheng; Anna Frolov; Gustavo Ayala; Xia Lin; Xin-Hua Feng; Michael Ittmann; Shaw Jenq Tsai; Ming-Jer Tsai; Sophia Y. Tsai

Mutations in phosphatase and tensin homologue (PTEN) or genomic alterations in the phosphatidylinositol-3-OH kinase-signalling pathway are the most common genetic alterations reported in human prostate cancer. However, the precise mechanism underlying how indolent tumours with PTEN alterations acquire metastatic potential remains poorly understood. Recent studies suggest that upregulation of transforming growth factor (TGF)-β signalling triggered by PTEN loss will form a growth barrier as a defence mechanism to constrain prostate cancer progression, underscoring that TGF-β signalling might represent a pre-invasive checkpoint to prevent PTEN-mediated prostate tumorigenesis. Here we show that COUP transcription factor II (COUP-TFII, also known as NR2F2), a member of the nuclear receptor superfamily, serves as a key regulator to inhibit SMAD4-dependent transcription, and consequently overrides the TGF-β-dependent checkpoint for PTEN-null indolent tumours. Overexpression of COUP-TFII in the mouse prostate epithelium cooperates with PTEN deletion to augment malignant progression and produce an aggressive metastasis-prone tumour. The functional counteraction between COUP-TFII and SMAD4 is reinforced by genetically engineered mouse models in which conditional loss of SMAD4 diminishes the inhibitory effects elicited by COUP-TFII ablation. The biological significance of COUP-TFII in prostate carcinogenesis is substantiated by patient sample analysis, in which COUP-TFII expression or activity is tightly correlated with tumour recurrence and disease progression, whereas it is inversely associated with TGF-β signalling. These findings reveal that the destruction of the TGF-β-dependent barrier by COUP-TFII is crucial for the progression of PTEN-mutant prostate cancer into a life-threatening disease, and supports COUP-TFII as a potential drug target for the intervention of metastatic human prostate cancer.


Urology | 2000

PROGNOSTIC VALUE OF p53 NUCLEAR ACCUMULATION AND HISTOPATHOLOGIC FEATURES IN T1 TRANSITIONAL CELL CARCINOMA OF THE URINARY BLADDER

Shahrokh F. Shariat; Alon Z Weizer; Aaran Green; Rodolfo Laucirica; Anna Frolov; Thomas M. Wheeler; Seth P. Lerner

OBJECTIVES To determine whether molecular and histopathologic tumor features can predict disease progression in Stage T1 transitional cell carcinoma of the bladder. METHODS Tumor specimens from 43 patients were analyzed with respect to grade, presence of carcinoma in situ, invasion deep or superficial to the lamina proprias muscularis mucosa, p53 expression using DO-7 and PAb1801 antibodies, age, and sex. Flow cytometry was performed on 30 patients from whom there was adequate paraffin-embedded tissue to assess DNA ploidy. Seven patients underwent immediate cystectomy as primary treatment and 36 patients retained their bladders and were at risk of recurrence and progression. RESULTS The median follow-up was 79 months. Disease recurred in 17 patients (47.2%) and progressed in 6 (16.7%). Only 3 patients (7.0%) died of bladder cancer. None of the parameters investigated was statistically significant in predicting recurrence, progression, or survival. Only carcinoma in situ approached statistical significance (P = 0.0593) as a predictor of progression. Early cystectomy did not have a significant effect on cancer-specific survival (P = 0.3603). The concordance rate between the two p53 antibodies was 88% (P <0.0001). CONCLUSIONS Deep invasion of the lamina propria, p53 positive immunohistochemistry, high grade, and aneuploidy were not significant adverse prognostic factors for either disease progression or survival. Carcinoma in situ associated with Stage T1 transitional cell carcinoma may represent a biologically more aggressive cancer requiring early definitive therapy, but this hypothesis should be evaluated in prospective clinical studies.


Cancer Research | 2006

Stromal Antiapoptotic Paracrine Loop in Perineural Invasion of Prostatic Carcinoma

Gustavo Ayala; Hong Dai; Salahaldin A. Tahir; Rile Li; Terry L. Timme; Michael Ittmann; Anna Frolov; Thomas M. Wheeler; David R. Rowley; Timothy C. Thompson

Caveolin-1 (cav-1) is a major scaffolding component of cell membrane invaginations (caveolae). It is involved in sequestering numerous effectors and signaling molecules and has antiapototic activities in prostate cancer. Perineural invasion (PNI) is associated with decreased apoptosis of cancer cells both in human tissues and the in vitro PNI model. We show here that stromal (perineurium) production of cav-1 is involved in a paracrine antiapoptotic loop in PNI. Transforming growth factor-beta1 is up-regulated in the cancer cells as they approach the nerve and is thought to up-regulate cav-1 in the perineurium of nerves with prostate cancer. Cav-1 is then secreted into the microenvironment and used by prostate cancer cells to inhibit apoptosis. In the in vitro PNI model, this phenomenon is partially reversed by neutralizing cav-1 antibodies or using ganglia from cav-1 knockout mice. Our results show a novel paracrine mechanism used by the prostate cancer in PNI to increase their proliferative activity and decrease apoptosis.


The Journal of Pathology | 2013

Loss of caveolin‐1 in prostate cancer stroma correlates with reduced relapse‐free survival and is functionally relevant to tumour progression

Gustavo Ayala; Matteo Morello; Anna Frolov; Sungyong You; Rile Li; Fabiana Rosati; Gianluca Bartolucci; Giovanna Danza; Rosalyn M. Adam; Timothy C. Thompson; Michael P. Lisanti; Michael R. Freeman; Dolores Di Vizio

Levels of caveolin‐1 (Cav‐1) in tumour epithelial cells increase during prostate cancer progression. Conversely, Cav‐1 expression in the stroma can decline in advanced and metastatic prostate cancer. In a large cohort of 724 prostate cancers, we observed significantly decreased levels of stromal Cav‐1 in concordance with increased Gleason score (p = 0.012). Importantly, reduced expression of Cav‐1 in the stroma correlated with reduced relapse‐free survival (p = 0.009), suggesting a role for stromal Cav‐1 in inhibiting advanced disease. Silencing of Cav‐1 by shRNA in WPMY‐1 prostate fibroblasts resulted in up‐regulation of Akt phosphorylation, and significantly altered expression of genes involved in angiogenesis, invasion, and metastasis, including a > 2.5‐fold increase in TGF‐β1 and γ‐synuclein (SNCG) gene expression. Moreover, silencing of Cav‐1 induced migration of prostate cancer cells when stromal cells were used as attractants. Pharmacological inhibition of Akt caused down‐regulation of TGF‐β1 and SNCG, suggesting that loss of Cav‐1 in the stroma can influence Akt‐mediated signalling in the tumour microenvironment. Cav‐1‐depleted stromal cells exhibited increased levels of intracellular cholesterol, a precursor for androgen biosynthesis, steroidogenic enzymes, and testosterone. These findings suggest that loss of Cav‐1 in the tumour microenvironment contributes to the metastatic behaviour of tumour cells by a mechanism that involves up‐regulation of TGF‐β1 and SNCG through Akt activation. They also suggest that intracrine production of androgens, a process relevant to castration resistance, may occur in the stroma. Copyright

Collaboration


Dive into the Anna Frolov's collaboration.

Top Co-Authors

Avatar

Gustavo Ayala

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Thomas M. Wheeler

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Rile Li

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Timothy C. Thompson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael Ittmann

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Dov Kadmon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David R. Rowley

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hong Dai

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Peter T. Scardino

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Brian J. Miles

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge