Network


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

Hotspot


Dive into the research topics where Roxan Ara is active.

Publication


Featured researches published by Roxan Ara.


Cancer Letters | 2015

Bone marrow derived myeloid cells orchestrate antiangiogenic resistance in glioblastoma through coordinated molecular networks

Bhagelu R. Achyut; Adarsh Shankar; Asm Iskander; Roxan Ara; Kartik Angara; Peng Zeng; Robert A. Knight; A. G. Scicli; Ali S. Arbab

Glioblastoma (GBM) is a hypervascular and malignant form of brain tumors. Anti-angiogenic therapies (AAT) were used as an adjuvant against VEGF-VEGFR pathway to normalize blood vessels in clinical and preclinical studies, which resulted into marked hypoxia and recruited bone marrow derived cells (BMDCs) to the tumor microenvironment (TME). In vivo animal models to track BMDCs and investigate molecular mechanisms in AAT resistance are rare. We exploited recently established chimeric mouse to develop orthotopic U251 tumor, which uses as low as 5 × 10(6) GFP+ BM cells in athymic nude mice and engrafted >70% GFP+ cells within 14 days. Our unpublished data and published studies have indicated the involvement of immunosuppressive myeloid cells in therapeutic resistance in glioma. Similarly, in the present study, vatalanib significantly increased CD68+ myeloid cells, and CD133+, CD34+ and Tie2+ endothelial cell signatures. Therefore, we tested inhibition of CSF1R+ myeloid cells using GW2580 that reduced tumor growth by decreasing myeloid (Gr1+ CD11b+ and F4/80+) and angiogenic (CD202b+ and VEGFR2+) cell signatures in TME. CSF1R blockade significantly decreased inflammatory, proangiogenic and immunosuppressive molecular signatures compared to vehicle, vatalanib or combination. TCK1 or CXCL7, a potent chemoattractant and activator of neutrophils, was observed as most significantly decreased cytokine in CSF1R blockade. ERK MAPK pathway was involved in cytokine network regulation. In conclusion, present study confirmed the contribution of myeloid cells in GBM development and therapeutic resistance using chimeric mouse model. We identified novel molecular networks including CXCL7 chemokine as a promising target for future studies. Nonetheless, survival studies are required to assess the beneficial effect of CSF1R blockade.


Cancer Biology & Therapy | 2016

Chimeric Mouse model to track the migration of bone marrow derived cells in glioblastoma following anti-angiogenic treatments

Bhagelu R. Achyut; Adarsh Shankar; Asm Iskander; Roxan Ara; Robert A. Knight; A. G. Scicli; Ali S. Arbab

ABSTRACT Bone marrow derived cells (BMDCs) have been shown to contribute in the tumor development. In vivo animal models to investigate the role of BMDCs in tumor development are poorly explored. We established a novel chimeric mouse model using as low as 5 × 106 GFP+ BM cells in athymic nude mice, which resulted in >70% engraftment within 14 d. In addition, chimera was established in NOD-SCID mice, which displayed >70% with in 28 d. Since anti-angiogenic therapies (AAT) were used as an adjuvant against VEGF-VEGFR pathway to normalize blood vessels in glioblastoma (GBM), which resulted into marked hypoxia and recruited BMDCs to the tumor microenvironment (TME). We exploited chimeric mice in athymic nude background to develop orthotopic U251 tumor and tested receptor tyrosine kinase inhibitors and CXCR4 antagonist against GBM. We were able to track GFP+ BMDCs in the tumor brain using highly sensitive multispectral optical imaging instrument. Increased tumor growth associated with the infiltration of GFP+ BMDCs acquiring suppressive myeloid and endothelial phenotypes was seen in TME following treatments. Immunofluorescence study showed GFP+ cells accumulated at the site of VEGF, SDF1 and PDGF expression, and at the periphery of the tumors following treatments. In conclusion, we developed a preclinical chimeric model of GBM and phenotypes of tumor infiltrated BMDCs were investigated in context of AATs. Chimeric mouse model could be used to study detailed cellular and molecular mechanisms of interaction of BMDCs and TME in cancer.


Histology and Histopathology | 2017

Vascular mimicry in glioblastoma following anti-angiogenic and anti-20-HETE therapies

Kartik Angara; Mohammad H. Rashid; Adarsh Shankar; Roxan Ara; Asm Iskander; Thaiz F. Borin; Meenu Jain; Bhagelu R. Achyut; Ali S. Arbab

Glioblastoma (GBM) is one hypervascular and hypoxic tumor known among solid tumors. Antiangiogenic therapeutics (AATs) have been tested as an adjuvant to normalize blood vessels and control abnormal vasculature. Evidence of relapse exemplified in the progressive tumor growth following AAT reflects development of resistance to AATs. Here, we identified that GBM following AAT (Vatalanib) acquired an alternate mechanism to support tumor growth, called vascular mimicry (VM). We observed that Vatalanib induced VM vessels are positive for periodic acid-Schiff (PAS) matrix but devoid of any endothelium on the inner side and lined by tumor cells on the outer-side. The PAS+ matrix is positive for basal laminae (laminin) indicating vascular structures. Vatalanib treated GBM displayed various stages of VM such as initiation (mosaic), sustenance, and full-blown VM. Mature VM structures contain red blood cells (RBC) and bear semblance to the functional blood vessel-like structures, which provide all growth factors to favor tumor growth. Vatalanib treatment significantly increased VM especially in the core of the tumor, where HIF-1α was highly expressed in tumor cells. VM vessels correlate with hypoxia and are characterized by co-localized MHC-1+ tumor and HIF-1α expression. Interestingly, 20-HETE synthesis inhibitor HET0016 significantly decreased GBM tumors through decreasing VM structures both at the core and at periphery of the tumors. In summary, AAT induced resistance characterized by VM is an alternative mechanism adopted by tumors to make functional vessels by transdifferentiation of tumor cells into endothelial-like cells to supply nutrients in the event of hypoxia. AAT induced VM is a potential therapeutic target of the novel formulation of HET0016. Our present study suggests that HET0016 has a potential to target therapeutic resistance and can be combined with other antitumor agents in preclinical and clinical trials.


Scientific Reports | 2017

Intravenous Formulation of HET0016 Decreased Human Glioblastoma Growth and Implicated Survival Benefit in Rat Xenograft Models

Meenu Jain; Nipuni Dhanesha H Gamage; Meshal Alsulami; Adarsh Shankar; Bhagelu R. Achyut; Kartik Angara; Mohammad H. Rashid; Asm Iskander; Thaiz F. Borin; Zhi Wenbo; Roxan Ara; Meser M. Ali; Iryna Lebedyeva; Wilson B. Chwang; Austin M. Guo; Hassan Bagher-Ebadian; Ali S. Arbab

Glioblastoma (GBM) is a hypervascular primary brain tumor with poor prognosis. HET0016 is a selective CYP450 inhibitor, which has been shown to inhibit angiogenesis and tumor growth. Therefore, to explore novel treatments, we have generated an improved intravenous (IV) formulation of HET0016 with HPßCD and tested in animal models of human and syngeneic GBM. Administration of a single IV dose resulted in 7-fold higher levels of HET0016 in plasma and 3.6-fold higher levels in tumor at 60 min than that in IP route. IV treatment with HPßCD-HET0016 decreased tumor growth, and altered vascular kinetics in early and late treatment groups (p < 0.05). Similar growth inhibition was observed in syngeneic GL261 GBM (p < 0.05). Survival studies using patient derived xenografts of GBM811, showed prolonged survival to 26 weeks in animals treated with focal radiation, in combination with HET0016 and TMZ (p < 0.05). We observed reduced expression of markers of cell proliferation (Ki-67), decreased neovascularization (laminin and αSMA), in addition to inflammation and angiogenesis markers in the treatment group (p < 0.05). Our results indicate that HPßCD-HET0016 is effective in inhibiting tumor growth through decreasing proliferation, and neovascularization. Furthermore, HPßCD-HET0016 significantly prolonged survival in PDX GBM811 model.


Scientific Reports | 2017

Canonical NFκB signaling in myeloid cells is required for the glioblastoma growth

Bhagelu R. Achyut; Kartik Angara; Meenu Jain; Thaiz F. Borin; Mohammad H. Rashid; Asm Iskander; Roxan Ara; Ravindra Kolhe; Shelby Howard; Natasha Venugopal; Paulo C. Rodriguez; Jennifer W. Bradford; Ali S. Arbab

Tumor development and therapeutic resistance are linked with tumor-associated macrophage (TAM) and myeloid-derived suppressor cell (MDSC) infiltration in tumors via chemokine axis. Chemokine expression, which determines the pro or anti-inflammatory status of myeloid cells, are partly regulated by the nuclear factor-kappa B (NF-κB) pathway. Here, we identified that conditional deletion of canonical NF-κB signaling (p65) in myeloid cells inhibited syngeneic glioblastoma (GBM) through decreased CD45 infiltration in tumors, as characterized by decreased TAMs (CD206+) and MDSCs (Gr1+ CD11b+), increased dendritic cells (CD86+) and cytotoxic T cells (CD8+) in the p65 knockout (KO) mice. Proinflammatory cytokines (IFNγ, MCP1, MIP1α, and TNFα) and myeloid differentiation factor (Endoglin) were increased in myeloid cells from p65 KO tumor, which demonstrated an influence on CD8+T cell proliferation. In contrast, p65KO athymic chimeric mice with human GBM, failed to inhibit tumor growth, confirming the contribution of T cells in an immune competent model. The analysis of human datasets and GBM tumors revealed higher expression of p65 in GBM-associated CD68+ macrophages compared to neighboring stroma. Thus, canonical NF-κB signaling has an anti-inflammatory role and is required for macrophage polarization, immune suppression, and GBM growth. Combining an NF-κB inhibitor with standard therapy could improve antitumor immunity in GBM.


PLOS ONE | 2017

HET0016 decreases lung metastasis from breast cancer in immune-competent mouse model

Thaiz F. Borin; Adarsh Shankar; Kartik Angara; Mohammad H. Rashid; Meenu Jain; Asm Iskander; Roxan Ara; Iryna Lebedyeva; Hasan Korkaya; Bhagelu R. Achyut; Ali S. Arbab

Distant metastasis is the primary cause of death in the majority of the cancer types. Recently, much importance has been given to tumor microenvironment (TME) in the development of invasive malignant tumors, as well as the metastasis potential. The ability of tumor cells to modulate TME and to escape immune-mediated attack by releasing immunosuppressive cytokines has become a hallmark of breast cancer. Our study shows the effect of IV formulation of HET0016 (HPßCD-HET0016) a selective inhibitor of 20-HETE synthesis, administered intravenously in immune-competent in vivo mouse model of murine breast cancer. 4T1 luciferase positive cells were implanted to the mammary fat pad in Balb/c mice. Treatment started on day 15, and was administered for 5 days a week for 3 weeks. The development of metastasis was detected via optical imaging. Blood, spleen, lungs, bone marrow and tumor were collected for flow cytometry, to investigate changes in myeloid-derived suppressive cells (MDSCs) populations and endothelial phenotype. Tumor and lungs were collected for protein analysis. Our results show that HPßCD-HET0016: (1) decreased tumor volume and lung metastasis compared to the vehicle group; (2) reduced migration and invasion of tumor cells and levels of metalloproteinases in the lungs of animals treated with HPßCD-HET0016 via PI3K/AKT pathway; and (3) decreased expression of pro-inflammatory cytokines, growth factors and granulocytic MDSCs population in the lung microenvironment in treated animals. Thus, HPßCD-HET0016 showed potential in treating lung metastasis in a preclinical mouse model and needs further investigations on TME.


Cancer Research | 2017

Abstract 1043: CSF-1R inhibitor prevented pre-metastatic lung niches in metastatic mammary tumor

Thaiz F. Borin; Kartik Angara; Mohammad H. Rashid; Adarsh Shankar; Asm Iskander; Roxan Ara; Meenu Jain; Bhagelu R. Achyut; Ali S. Arbab

Exosomes are small vesicle cellular products originating from the endocytic pathway abundantly secreted by tumor cells. These exosomes have the ability to alter their immediate microenvironment (ME) through cell-cell interaction by fusion with plasma membrane and subsequent endocytosis or release of their cargo. Exosomes are critical modulators of pre-metastatic niche creation by increasing the recruitment of inflammatory cells. In breast cancer, tumor-derived exosomes recruit myeloid derived suppressor cells in the creation of an immunosuppressive pro-tumorigenic lung niches. Colony stimulating factor 1-receptor (CSF1R), which is a key regulator of myeloid cell proliferation, survival, and differentiation can be blocked by a selective inhibitor GW2580. Overexpression of CSF-1 has been implicated in the creation of increased number of metastatic niches in numerous cancers. In our study, we have demonstrated that the exosomes secreted under hypoxic conditions can initiate early pre-metastatic niche creation in lungs in a metastatic breast cancer model compared to normoxia-secreted exosomes. Exosomes were injected intravenously into Balb/c female mice three days after the implantation of 4T1 breast tumor and continued for a week with injections on alternated days. The animals were pre-treated with GW2580 the day before tumor implantation and continued for a week concomitantly with exosomes on alternate days. Lungs, bone marrow, spleen and brain tissues were collected and analyzed by flow cytometer to detect myeloid and angiogenic cells populations. GW2580 was able to prevent myeloid cell infiltration in lungs and bone marrow. Further, we observed significant increase in anti-tumorigenic M1-macrophage population in the lungs of exosome treated animals pre-treated with GW2580. However, these findings were not observed in the bone marrows of the same group. Vasculogenic leukocyte and angiogenic myeloid cell populations were significantly decreased in the lung of exosome treated animals pre-treated with GW2580. A similar decrease in these populations of cells was not seen in the lungs of animals pre-treated with GW2580 only. These surprising results have led us to hypothesize that GW2580 treatment can prevent the effects of exosomes, which causes infiltration of myeloid cells in the lung to create metastatic niche. These observations indicate a role of CSF1R inhibitor in preventing the distant metastatic niche formation. Citation Format: Thaiz F. Borin, Kartik Angara, Mohammad Rashid, Adarsh Shankar, Asm Iskander, Roxan Ara, Meenu Jain, Bhagelu R. Achyut, Ali S. Arbab. CSF-1R inhibitor prevented pre-metastatic lung niches in metastatic mammary tumor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1043. doi:10.1158/1538-7445.AM2017-1043


Neoplasia | 2018

CXCR2-Expressing Tumor Cells Drive Vascular Mimicry in Antiangiogenic Therapy–Resistant Glioblastoma

Kartik Angara; Thaiz F. Borin; Mohammad H. Rashid; Iryna Lebedyeva; Roxan Ara; Ping-Chang Lin; Asm Iskander; Roni J. Bollag; Bhagelu R. Achyut; Ali S. Arbab

BACKGROUND: Glioblastoma (GBM) was shown to relapse faster and displayed therapeutic resistance to antiangiogenic therapies (AATs) through an alternative tumor cell-driven mechanism of neovascularization called vascular mimicry (VM). We identified highly upregulated interleukin 8 (IL-8)-CXCR2 axis in tumor cells in high-grade human glioma and AAT-treated orthotopic GBM tumors. METHODS: Human GBM tissue sections and tissue array were used to ascertain the clinical relevance of CXCR2-positive tumor cells in the formation of VM. We utilized U251 and U87 human tumor cells to understand VM in an orthotopic GBM model and AAT-mediated enhancement in VM was modeled using vatalanib (anti-VEGFR2) and avastin (anti-VEGF). Later, VM was inhibited by SB225002 (CXCR2 inhibitor) in a preclinical study. RESULTS: Overexpression of IL8 and CXCR2 in human datasets and histological analysis was identified as a bonafide candidate to validate VM through in vitro and animal model studies. AAT-treated tumors displayed a higher number of CXCR2-positive GBM-stem cells with endothelial-like phenotypes. Stable knockdown of CXCR2 expression in tumor cells led to decreased tumor growth as well as incomplete VM structures in the animal models. Similar data were obtained following SB225002 treatment. CONCLUSIONS: The present study suggests that tumor cell autonomous IL-8-CXCR2 pathway is instrumental in AAT-mediated resistance and VM formation in GBM. Therefore, CXCR2 can be targeted through SB225002 and can be combined with standard therapies to improve the therapeutic outcomes in clinical trials.


Cancer Growth and Metastasis | 2017

Suppression of Breast Cancer Metastasis Using Stapled Peptides Targeting the WASF Regulatory Complex

John K. Cowell; Yong Teng; N. George Bendzunas; Roxan Ara; Ali S. Arbab; Eileen J. Kennedy

The WASF3 gene facilitates the metastatic phenotype, and its inactivation leads to suppression of invasion and metastasis regardless of the genetic background of the cancer cell. This reliance on WASF3 to facilitate metastasis suggests that targeting its function could serve as an effective strategy to suppress metastasis. WASF3 stability and function are regulated by the WASF Regulatory Complex (WRC) of proteins, particularly CYFIP1 and NCKAP1. Knockdown of these proteins in vitro leads to disruption of the WRC and suppression of invasion. We have used mouse xenograft models of breast cancer metastasis to assess whether targeting the WRC complex suppresses metastasis in vivo. Stapled peptides targeting the WASF3-CYFIP1 interface (WAHM1) and the CYFIP1-NCKAP1 interface (WANT3) suppress the development of lung and liver metastases. Targeting these critical protein-protein interactions, therefore, could potentially be developed into a therapeutic strategy to control cancer cell invasion and metastasis.


Journal of Cancer Science & Therapy | 2016

Anti-VEGFR2 Driven Nuclear Translocation of VEGFR2 and AcquiredMalignant Hallmarks are Mutation Dependent in Glioblastoma

Adarsh Shankar; Meenu Jain; Mei Jing Lim; Kartik Angara; Syed A Arbab; Isk; er Asm; Roxan Ara; Ali S. Arbab; Bhagelu R. Achyut

OBJECTIVE Anti-angiogenic therapies (AATs), targeting VEGF-VEGFR pathways, are being used as an adjuvant to normalize glioblastoma (GBM) vasculature. Unexpectedly, clinical trials have witnessed transient therapeutic effect followed by aggressive tumor recurrence. In pre-clinical studies, targeting VEGFR2 with vatalanib, increased GBM growth under hypoxic microenvironment. There is limited understanding of these unanticipated results. Here, we investigated tumor cell associated phenotypes in response to VEGFR2 blockade. METHODS Human U251 cells were orthotopically implanted in mice (day 0) and were treated with vehicle or vatalanib on day 8. Tumor specimens were collected for immunohistochemistry and protein array. Nuclear translocation of VEGFR2 was analyzed through IHC and western blot. In vitro studies were performed in U251 (p53 and EGFR mutated) and U87 (p53 and EGFR wildtype) cells following vehicle or vatalanib treatments under normoxia (21% O2) and hypoxia (1% O2). Proliferation, cell cycle and apoptosis assays were done to analyze tumor cell phenotypes after treatments. RESULTS Vatalanib treated animals displayed distinct patterns of VEGFR2 translocation into nuclear compartment of U251 tumor cells. In vitro studies suggest that vatalanib significantly induced nuclear translocation of VEGFR2, characterized in chromatin bound fraction, especially in U251 tumor cells grown under normoxia and hypoxia. Anti-VEGFR2 driven nuclear translocation of VEGFR2 was associated with increased cell cycle and proliferation, decreased apoptosis, and displayed increased invasiveness in U251 compared to U87 cells. CONCLUSIONS Study suggests that AAT- induced molecular and phenotypic alterations in tumor cells are associated with mutation status and are responsible for aggressive tumor growth. Therefore, mutation status of the tumor in GBM patients should be taken in to consideration before applying targeted therapy to overcome unwanted effects.

Collaboration


Dive into the Roxan Ara's collaboration.

Top Co-Authors

Avatar

Ali S. Arbab

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Bhagelu R. Achyut

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Kartik Angara

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Asm Iskander

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Meenu Jain

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Thaiz F. Borin

Faculdade de Medicina de São José do Rio Preto

View shared research outputs
Top Co-Authors

Avatar

Adarsh Shankar

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. G. Scicli

Henry Ford Health System

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge