Network


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

Hotspot


Dive into the research topics where Behrouz Zand is active.

Publication


Featured researches published by Behrouz Zand.


Nature Communications | 2013

Tumour angiogenesis regulation by the miR-200 family

Chad V. Pecot; Rajesha Rupaimoole; Da Yang; Rehan Akbani; Cristina Ivan; Chunhua Lu; Sherry Y. Wu; Hee Dong Han; Maitri Y. Shah; Cristian Rodriguez-Aguayo; Justin Bottsford-Miller; Yuexin Liu; Sang Bae Kim; Anna K. Unruh; Vianey Gonzalez-Villasana; Li Huang; Behrouz Zand; Myrthala Moreno-Smith; Lingegowda S. Mangala; Morgan Taylor; Heather J. Dalton; Vasudha Sehgal; Yunfei Wen; Yu Kang; Keith A. Baggerly; Ju Seog Lee; Prahlad T. Ram; Murali Ravoori; Vikas Kundra; Xinna Zhang

The miR-200 family is well known to inhibit the epithelial-mesenchymal transition, suggesting it may therapeutically inhibit metastatic biology. However, conflicting reports regarding the role of miR-200 in suppressing or promoting metastasis in different cancer types have left unanswered questions. Here we demonstrate a difference in clinical outcome based on miR-200s role in blocking tumour angiogenesis. We demonstrate that miR-200 inhibits angiogenesis through direct and indirect mechanisms by targeting interleukin-8 and CXCL1 secreted by the tumour endothelial and cancer cells. Using several experimental models, we demonstrate the therapeutic potential of miR-200 delivery in ovarian, lung, renal and basal-like breast cancers by inhibiting angiogenesis. Delivery of miR-200 members into the tumour endothelium resulted in marked reductions in metastasis and angiogenesis, and induced vascular normalization. The role of miR-200 in blocking cancer angiogenesis in a cancer-dependent context defines its utility as a potential therapeutic agent.


Blood | 2012

Platelets increase the proliferation of ovarian cancer cells

Min Soon Cho; Justin Bottsford-Miller; Hernan Vasquez; Rebecca L. Stone; Behrouz Zand; Michael H. Kroll; Anil K. Sood; Vahid Afshar-Kharghan

Platelets promote metastasis and angiogenesis, but their effect on tumor cell growth is uncertain. Here we report a direct proliferative effect of platelets on cancer cells both in vitro and in vivo. Incubation of platelets with ovarian cancer cells from murine (ID8 and 2C6) or human (SKOV3 and OVCAR5) origin increased cell proliferation. The proliferative effect of platelets was not dependent on direct contact with cancer cells, and preincubation of platelets with blocking antibodies against platelet adhesion molecules did not alter their effect on cancer cells. The proliferative effect of platelets was reduced by fixing platelets with paraformaldehyde, preincubating platelets with a TGF-β1-blocking antibody, or reducing expression of TGF-βR1 receptor on cancer cells with siRNA. Infusing platelets into mice with orthotopic ovarian tumors significantly increased the proliferation indices in these tumors. Ovarian cancer patients with thrombocytosis had higher tumor proliferation indices compared with patients with normal platelet counts.


Cancer Cell | 2014

Hematogenous Metastasis of Ovarian Cancer: Rethinking Mode of Spread

Sunila Pradeep; Seung W. Kim; Sherry Y. Wu; Masato Nishimura; Pradeep Chaluvally-Raghavan; Takahito Miyake; Chad V. Pecot; Sun Jin Kim; Hyun Jin Choi; Farideh Z. Bischoff; Julie Ann Mayer; Li Huang; Alpa M. Nick; Carolyn S. Hall; Cristian Rodriguez-Aguayo; Behrouz Zand; Heather J. Dalton; Thiruvengadam Arumugam; Ho Jeong Lee; Hee Dong Han; Min Soon Cho; Rajesha Rupaimoole; Lingegowda S. Mangala; Vasudha Sehgal; Sang Cheul Oh; Jinsong Liu; Ju Seog Lee; Robert L. Coleman; Prahlad T. Ram; Gabriel Lopez-Berestein

Ovarian cancer has a clear predilection for metastasis to the omentum, but the underlying mechanisms involved in ovarian cancer spread are not well understood. Here, we used a parabiosis model that demonstrates preferential hematogenous metastasis of ovarian cancer to the omentum. Our studies revealed that the ErbB3-neuregulin 1 (NRG1) axis is a dominant pathway responsible for hematogenous omental metastasis. Elevated levels of ErbB3 in ovarian cancer cells and NRG1 in the omentum allowed for tumor cell localization and growth in the omentum. Depletion of ErbB3 in ovarian cancer impaired omental metastasis. Our results highlight hematogenous metastasis as an important mode of ovarian cancer metastasis. These findings have implications for designing alternative strategies aimed at preventing and treating ovarian cancer metastasis.


Nature Communications | 2014

Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression

Rajesha Rupaimoole; Sherry Y. Wu; Sunila Pradeep; Cristina Ivan; Chad V. Pecot; Kshipra M. Gharpure; Archana S. Nagaraja; Guillermo N. Armaiz-Pena; Michael McGuire; Behrouz Zand; Heather J. Dalton; Justyna Filant; Justin Bottsford Miller; Chunhua Lu; Nouara C. Sadaoui; Lingegowda S. Mangala; Morgan Taylor; Twan van den Beucken; Elizabeth Koch; Cristian Rodriguez-Aguayo; Li Huang; Menashe Bar-Eli; Bradly G. Wouters; Milan Radovich; Mircea Ivan; George A. Calin; Wei Zhang; Gabriel Lopez-Berestein; Anil K. Sood

Cancer-related deregulation of miRNA biogenesis has been suggested, but the underlying mechanisms remain elusive. Here we report a previously unrecognized effect of hypoxia in the downregulation of Drosha and Dicer in cancer cells that leads to dysregulation of miRNA biogenesis and increased tumour progression. We show that hypoxia-mediated downregulation of Drosha is dependent on ETS1/ELK1 transcription factors. Moreover, mature miRNA array and deep sequencing studies reveal altered miRNA maturation in cells under hypoxic conditions. At a functional level, this phenomenon results in increased cancer progression in vitro and in vivo, and data from patient samples are suggestive of miRNA biogenesis downregulation in hypoxic tumours. Rescue of Drosha by siRNAs targeting ETS1/ELK1 in vivo results in significant tumour regression. These findings provide a new link in the mechanistic understanding of global miRNA downregulation in the tumour microenvironment. MicroRNAs play important roles in the maintenance of cellular homeostasis through the post-transcriptional regulation of gene expression. Here, the authors implicate loss of the miRNA biogenesis factor Drosha and altered miRNA maturation in tumour progression under hypoxic conditions.


Journal of the National Cancer Institute | 2013

Role of Focal Adhesion Kinase in Regulating YB–1–Mediated Paclitaxel Resistance in Ovarian Cancer

Yu Kang; Wei Hu; Cristina Ivan; Heather J. Dalton; Takahito Miyake; Chad V. Pecot; Behrouz Zand; Tao Liu; Jie Huang; Nicholas B. Jennings; Rajesha Rupaimoole; Morgan Taylor; Sunila Pradeep; Sherry Y. Wu; Chunhua Lu; Yunfei Wen; Jianfei Huang; Jinsong Liu; Anil K. Sood

BACKGROUND We previously found focal adhesion kinase (FAK) inhibition sensitizes ovarian cancer to taxanes; however, the mechanisms are not well understood. METHODS We characterized the biologic response of taxane-resistant and taxane-sensitive ovarian cancer models to a novel FAK inhibitor (VS-6063). We used reverse-phase protein arrays (RPPA) to identify novel downstream targets in taxane-resistant cell lines. Furthermore, we correlated clinical and pathological data with nuclear and cytoplasmic expression of FAK and YB-1 in 105 ovarian cancer samples. Statistical tests were two-sided, and P values were calculated with Student t test or Fisher exact test. RESULTS We found that VS-6063 inhibited FAK phosphorylation at the Tyr397 site in a time- and dose-dependent manner. The combination of VS-6063 and paclitaxel markedly decreased proliferation and increased apoptosis, which resulted in 92.7% to 97.9% reductions in tumor weight. RPPA data showed that VS-6063 reduced levels of AKT and YB-1 in taxane-resistant cell lines. FAK inhibition enhanced chemosensitivity in taxane-resistant cells by decreasing YB-1 phosphorylation and subsequently CD44 in an AKT-dependent manner. In human ovarian cancer samples, nuclear FAK expression was associated with increased nuclear YB-1 expression (χ²) = 37.7; P < .001). Coexpression of nuclear FAK and YB-1 was associated with statistically significantly worse median overall survival (24.9 vs 67.3 months; hazard ratio = 2.64; 95% confidence interval = 1.38 to 5.05; P = .006). CONCLUSIONS We have identified a novel pathway whereby FAK inhibition with VS-6063 overcomes YB-1-mediated paclitaxel resistance by an AKT-dependent pathway. These findings have implications for clinical trials aimed at targeting FAK.


Cell Reports | 2014

Autocrine Effects of Tumor-Derived Complement

Min Soon Cho; Hernan Vasquez; Rajesha Rupaimoole; Sunila Pradeep; Sherry Y. Wu; Behrouz Zand; Hee Dong Han; Cristian Rodriguez-Aguayo; Justin Bottsford-Miller; Jie Huang; Takahito Miyake; Hyun Jin Choi; Heather J. Dalton; Cristina Ivan; Keith A. Baggerly; Gabriel Lopez-Berestein; Anil K. Sood; Vahid Afshar-Kharghan

SUMMARY We describe a role for the complement system in enhancing cancer growth. Cancer cells secrete complement proteins that stimulate tumor growth upon activation. Complement promotes tumor growth via a direct autocrine effect that is partially independent of tumor-infiltrating cytotoxic T cells. Activated C5aR and C3aR signal through the PI3K/AKT pathway in cancer cells, and silencing the PI3K or AKT gene in cancer cells eliminates the progrowth effects of C5aR and C3aR stimulation. In patients with ovarian or lung cancer, higher tumoral C3 or C5aR mRNA levels were associated with decreased overall survival. These data identify a role for tumor-derived complement proteins in promoting tumor growth, and they therefore have substantial clinical and therapeutic implications.


Journal of Clinical Investigation | 2013

ATP11B mediates platinum resistance in ovarian cancer

Myrthala Moreno-Smith; Jyotsnabaran Halder; Paul S. Meltzer; Tamas A. Gonda; Lingegowda S. Mangala; Rajesha Rupaimoole; Chunhua Lu; Archana S. Nagaraja; Kshipra M. Gharpure; Yu Kang; Cristian Rodriguez-Aguayo; Pablo Vivas-Mejia; Behrouz Zand; Rosemarie Schmandt; Hua Wang; Robert R. Langley; Nicholas B. Jennings; Cristina Ivan; Jeremy E. Coffin; Guillermo N. Armaiz; Justin Bottsford-Miller; Sang Bae Kim; Margaret S. Halleck; Mary J.C. Hendrix; William Bornman; Menashe Bar-Eli; Ju Seog Lee; Zahid H. Siddik; Gabriel Lopez-Berestein; Anil K. Sood

Platinum compounds display clinical activity against a wide variety of solid tumors; however, resistance to these agents is a major limitation in cancer therapy. Reduced platinum uptake and increased platinum export are examples of resistance mechanisms that limit the extent of DNA damage. Here, we report the discovery and characterization of the role of ATP11B, a P-type ATPase membrane protein, in cisplatin resistance. We found that ATP11B expression was correlated with higher tumor grade in human ovarian cancer samples and with cisplatin resistance in human ovarian cancer cell lines. ATP11B gene silencing restored the sensitivity of ovarian cancer cell lines to cisplatin in vitro. Combined therapy of cisplatin and ATP11B-targeted siRNA significantly decreased cancer growth in mice bearing ovarian tumors derived from cisplatin-sensitive and -resistant cells. In vitro mechanistic studies on cellular platinum content and cisplatin efflux kinetics indicated that ATP11B enhances the export of cisplatin from cells. The colocalization of ATP11B with fluorescent cisplatin and with vesicular trafficking proteins, such as syntaxin-6 (STX6) and vesicular-associated membrane protein 4 (VAMP4), strongly suggests that ATP11B contributes to secretory vesicular transport of cisplatin from Golgi to plasma membrane. In conclusion, inhibition of ATP11B expression could serve as a therapeutic strategy to overcome cisplatin resistance.


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.


Clinical Cancer Research | 2012

Metronomic Activity of CD44-Targeted Hyaluronic Acid-Paclitaxel in Ovarian Carcinoma

Sun Joo Lee; Sukhen C. Ghosh; Hee Dong Han; Rebecca L. Stone; Justin Bottsford-Miller; De Yue Shen; Edmond Auzenne; Alejandro Lopez-Araujo; Chunhua Lu; Masato Nishimura; Chad V. Pecot; Behrouz Zand; Duangmani Thanapprapasr; Nicholas B. Jennings; Yu Kang; Jie Huang; Wei Hu; Jim Klostergaard; Anil K. Sood

Purpose: Most primary human ovarian tumors and peritoneal implants, as well as tumor vascular endothelial cells, express the CD44 family of cell surface proteoglycans, the natural ligand for which is hyaluronic acid. Metronomic dosing, the frequent administration of chemotherapeutics at substantially lower than maximum tolerated doses (MTD), has been shown to result in reduced normal tissue toxicity and to minimize “off-treatment” exposure resulting in an improved therapeutic ratio. Experimental Design: We tested the hypothesis that hyaluronic acid (HA) conjugates of paclitaxel (TXL; HA-TXL) would exert strong antitumor effects with metronomic (MET) dosing and induce antiangiogenic effects superior to those achieved with MTD administration or with free TXL. Female nude mice bearing SKOV3ip1 or HeyA8 ovarian cancer cells were treated intraperitoneally (i.p.) with MET HA-TXL regimens (or MTD administration) to determine therapeutic and biologic effects. Results: All MET HA-TXL–treated mice and the MTD group revealed significantly reduced tumor weights and nodules compared with controls (all P values < 0.05) in the chemotherapy-sensitive models. However, the MTD HA-TXL–treated mice showed significant weight loss compared with control mice, whereas body weights were not affected in the metronomic groups in HeyA8-MDR model, reflecting reduced toxicity. In the taxane-resistant HeyA8-MDR model, significant reduction in tumor weight and nodule counts was noted in the metronomic groups whereas the response of the MTD group did not achieve significance. While both MTD and metronomic regimens reduced proliferation (Ki-67) and increased apoptosis (TUNEL, terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling), only metronomic treatment resulted in significant reductions in angiogenesis (CD31, microvessel density). Moreover, metronomic treatment resulted in substantial increases in thrombospondin-1 (Tsp-1), an inhibitor of angiogenesis. Conclusions: This study showed that MET HA-TXL regimens have substantial antitumor activity in ovarian carcinoma, likely via a predominant antiangiogenic mechanism. Clin Cancer Res; 18(15); 4114–21. ©2012 AACR.


Molecular Cancer Therapeutics | 2014

Therapeutic Silencing of KRAS Using Systemically Delivered siRNAs

Chad V. Pecot; Sherry Y. Wu; Seth Bellister; Justyna Filant; Rajesha Rupaimoole; Takeshi Hisamatsu; Rajat Bhattacharya; Anshumaan Maharaj; Salma H. Azam; Cristian Rodriguez-Aguayo; Archana S. Nagaraja; Maria Pia Morelli; Kshipra M. Gharpure; Trent A. Waugh; Vianey Gonzalez-Villasana; Behrouz Zand; Heather J. Dalton; Scott Kopetz; Gabriel Lopez-Berestein; Lee M. Ellis; Anil K. Sood

Despite being among the most common oncogenes in human cancer, to date, there are no effective clinical options for inhibiting KRAS activity. We investigated whether systemically delivered KRAS siRNAs have therapeutic potential in KRAS-mutated cancer models. We identified KRAS siRNA sequences with notable potency in knocking down KRAS expression. Using lung and colon adenocarcinoma cell lines, we assessed antiproliferative effects of KRAS silencing in vitro. For in vivo experiments, we used a nanoliposomal delivery platform, DOPC, for systemic delivery of siRNAs. Various lung and colon cancer models were used to determine efficacy of systemic KRAS siRNA based on tumor growth, development of metastasis, and downstream signaling. KRAS siRNA sequences induced >90% knockdown of KRAS expression, significantly reducing viability in mutant cell lines. In the lung cancer model, KRAS siRNA treatment demonstrated significant reductions in primary tumor growth and distant metastatic disease, while the addition of CDDP was not additive. Significant reductions in Ki-67 indices were seen in all treatment groups, whereas significant increases in caspase-3 activity were only seen in the CDDP treatment groups. In the colon cancer model, KRAS siRNA reduced tumor KRAS and pERK expression. KRAS siRNAs significantly reduced HCP1 subcutaneous tumor growth, as well as outgrowth of liver metastases. Our studies demonstrate a proof-of-concept approach to therapeutic KRAS targeting using nanoparticle delivery of siRNA. This study highlights the potential translational impact of therapeutic RNA interference, which may have broad applications in oncology, especially for traditional “undruggable” targets. Mol Cancer Ther; 13(12); 2876–85. ©2014 AACR.

Collaboration


Dive into the Behrouz Zand'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

Justin Bottsford-Miller

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

Wei Hu

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

Chad V. Pecot

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Rebecca A. Previs

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alpa M. Nick

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