Network


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

Hotspot


Dive into the research topics where Justin Bottsford-Miller is active.

Publication


Featured researches published by Justin Bottsford-Miller.


The New England Journal of Medicine | 2012

Paraneoplastic Thrombocytosis in Ovarian Cancer

Rebecca L. Stone; Alpa M. Nick; Iain A. McNeish; Frances R. Balkwill; Hee Dong Han; Justin Bottsford-Miller; Rajesha Rupaimoole; Guillermo N. Armaiz-Pena; Chad V. Pecot; Jermaine Coward; Michael T. Deavers; Hernan Vasquez; Diana L. Urbauer; Charles N. Landen; Wei Hu; Hannah Gershenson; Koji Matsuo; Mian M.K. Shahzad; Erin R. King; Ibrahim Tekedereli; Bulent Ozpolat; Edward H. Ahn; Virginia K. Bond; Rui Wang; Angela F. Drew; Francisca C. Gushiken; Donald M. Lamkin; Katherine Collins; Koen DeGeest; Susan K. Lutgendorf

BACKGROUND The mechanisms of paraneoplastic thrombocytosis in ovarian cancer and the role that platelets play in abetting cancer growth are unclear. METHODS We analyzed clinical data on 619 patients with epithelial ovarian cancer to test associations between platelet counts and disease outcome. Human samples and mouse models of epithelial ovarian cancer were used to explore the underlying mechanisms of paraneoplastic thrombocytosis. The effects of platelets on tumor growth and angiogenesis were ascertained. RESULTS Thrombocytosis was significantly associated with advanced disease and shortened survival. Plasma levels of thrombopoietin and interleukin-6 were significantly elevated in patients who had thrombocytosis as compared with those who did not. In mouse models, increased hepatic thrombopoietin synthesis in response to tumor-derived interleukin-6 was an underlying mechanism of paraneoplastic thrombocytosis. Tumor-derived interleukin-6 and hepatic thrombopoietin were also linked to thrombocytosis in patients. Silencing thrombopoietin and interleukin-6 abrogated thrombocytosis in tumor-bearing mice. Anti-interleukin-6 antibody treatment significantly reduced platelet counts in tumor-bearing mice and in patients with epithelial ovarian cancer. In addition, neutralizing interleukin-6 significantly enhanced the therapeutic efficacy of paclitaxel in mouse models of epithelial ovarian cancer. The use of an antiplatelet antibody to halve platelet counts in tumor-bearing mice significantly reduced tumor growth and angiogenesis. CONCLUSIONS These findings support the existence of a paracrine circuit wherein increased production of thrombopoietic cytokines in tumor and host tissue leads to paraneoplastic thrombocytosis, which fuels tumor growth. We speculate that countering paraneoplastic thrombocytosis either directly or indirectly by targeting these cytokines may have therapeutic potential. (Funded by the National Cancer Institute and others.).


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.


Journal of Clinical Oncology | 2012

Resistance and Escape From Antiangiogenesis Therapy: Clinical Implications and Future Strategies

Justin Bottsford-Miller; Robert L. Coleman; Anil K. Sood

Angiogenesis has long been considered an important target for cancer therapy. Initial efforts have primarily focused on targeting of endothelial and tumor-derived vascular endothelial growth factor signaling. As evidence emerges that angiogenesis has significant mechanistic complexity, therapeutic resistance and escape have become practical limitations to drug development. Here, we review the mechanisms by which dynamic changes occur in the tumor microenvironment in response to antiangiogenic therapy, leading to drug resistance. These mechanisms include direct selection of clonal cell populations with the capacity to rapidly upregulate alternative proangiogenic pathways, increased invasive capacity, and intrinsic resistance to hypoxia. The implications of normalization of vasculature with subsequently improved vascular function as a result of antiangiogenic therapy are explored, as are the implications of the ability to incorporate and co-opt otherwise normal vasculature. Finally, we consider the extent to which a better understanding of the biology of hypoxia and reoxygenation, as well as the depth and breadth of systems invested in angiogenesis, may offer putative biomarkers and novel therapeutic targets. Insights gained through this work may offer solutions for personalizing antiangiogenesis approaches and improving the outcome of patients with cancer.


Cancer Discovery | 2011

A Novel Platform for Detection of CK+ and CK− CTCs

Chad V. Pecot; Farideh Z. Bischoff; Julie Ann Mayer; Karina L. Wong; Tam Pham; Justin Bottsford-Miller; Rebecca L. Stone; Yvonne G. Lin; Padmavathi Jaladurgam; Ju Won Roh; Blake W. Goodman; William M. Merritt; Tony J. Pircher; Stephen D. Mikolajczyk; Alpa M. Nick; Joseph Celestino; Cathy Eng; Lee M. Ellis; Michael T. Deavers; Anil K. Sood

UNLABELLED Metastasis is a complex, multistep process that begins with the epithelial-mesenchymal transition (EMT). Circulating tumor cells (CTC) are believed to have undergone EMT and thus lack or express low levels of epithelial markers commonly used for enrichment and/or detection of such cells. However, most current CTC detection methods target only EpCAM and/or cytokeratin (CK) to enrich epithelial CTCs, resulting in failure to recognize other, perhaps more important, CTC phenotypes that lack expression of these markers. Here, we describe a population of complex aneuploid CTCs that do not express CK or CD45 antigen in patients with breast, ovarian, or colorectal cancer. These cells were not observed in healthy subjects. We show that the primary epithelial tumors were characterized by similar complex aneuploidy, indicating conversion to an EMT phenotype in the captured cells. Collectively, our study provides a new method for highly efficient capture of previously unrecognized populations of CTCs. SIGNIFICANCE Current assays for CTC capture likely miss populations of cells that have undergone EMT. Capture and study of CTCs that have undergone EMT would allow a better understanding of the mechanisms driving metastasis.


Journal of Biological Chemistry | 2010

Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis.

Mian M.K. Shahzad; Jesusa M.G. Arevalo; Guillermo N. Armaiz-Pena; Chunhua Lu; Rebecca L. Stone; Myrthala Moreno-Smith; Masato Nishimura; Jeong Won Lee; Nicholas B. Jennings; Justin Bottsford-Miller; Pablo Vivas-Mejia; Susan K. Lutgendorf; Gabriel Lopez-Berestein; Menashe Bar-Eli; Steven W. Cole; Anil K. Sood

A growing number of studies indicate that chronic stress can accelerate tumor growth due to sustained sympathetic nervous system activation. Our recent findings suggest that chronic stress is associated with increased IL8 levels. Here, we examined the molecular and biological significance of IL8 in stress-induced tumor growth. Norepinephrine (NE) treatment of ovarian cancer cells resulted in a 250–300% increase in IL8 protein and 240–320% increase in its mRNA levels. Epinephrine treatment resulted in similar increases. Moreover, NE treatment resulted in a 3.5–4-fold increase in IL8 promoter activity. These effects were blocked by propranolol. Promoter deletion analyses suggested that AP1 transcription factors might mediate catecholamine-stimulated up-regulation of IL8. siRNA inhibition studies identified FosB as the pivotal component responsible for IL8 regulation by NE. In vivo chronic stress resulted in increased tumor growth (by 221 and 235%; p < 0.01) in orthotopic xenograft models involving SKOV3ip1 and HeyA8 ovarian carcinoma cells. This enhanced tumor growth was completely blocked by IL8 or FosB gene silencing using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine nanoliposomes. IL8 and FosB silencing reduced microvessel density (based on CD31 staining) by 2.5- and 3.5-fold, respectively (p < 0.001). Our findings indicate that neurobehavioral stress leads to FosB-driven increases in IL8, which is associated with increased tumor growth and metastases. These findings may have implications for ovarian cancer management.


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.


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.


Clinical Cancer Research | 2011

Functional Roles of Src and Fgr in Ovarian Carcinoma

Hye Sun Kim; Hee Dong Han; Guillermo N. Armaiz-Pena; Rebecca L. Stone; Eun Ji Nam; Jeong Won Lee; Mian M.K. Shahzad; Alpa M. Nick; Sun Joo Lee; Ju Won Roh; Masato Nishimura; Lingegowda S. Mangala; Justin Bottsford-Miller; Gary E. Gallick; Gabriel Lopez-Berestein; Anil K. Sood

Purpose:Src is an attractive target because it is overexpressed in a number of malignancies, including ovarian cancer. However, the effect of Src silencing on other Src family kinases (SFKs) is not known. We hypothesized that other SFK members could compensate for the lack of Src activity. Experimental Design: Cell viability after either Src or Fgr silencing was examined in ovarian cancer cell lines by MTT assay. Expression of SFKs after Src silencing in ovarian cancer cells was examined by real-time reverse transcriptase (RT)-PCR. Therapeutic effect of in vivo Src and/or Fgr silencing was examined using siRNA incorporated into chitosan nanoparticles (siRNA/CH-NP). Microvessel density, cell proliferation, and apoptosis markers were determined by immunohistochemical staining in ovarian tumor tissues. Results:Src silencing enhanced cytotoxicity of docetaxel in both SKOV3ip1 and HeyA8 cells. In addition, Src silencing using siRNA/CH-NP in combination with docetaxel resulted in significant inhibition of tumor growth compared with control siRNA/CH-NP (81.8% reduction in SKOV3ip1, P = 0.017; 84.3% reduction in HeyA8, P < 0.005). These effects were mediated by decreased tumor cell proliferation and angiogenesis, and increased tumor cell apoptosis. Next, we assessed the effects of Src silencing on other SFK members in ovarian cancer cell lines. Src silencing resulted in significantly increased Fgr levels. Dual Src and Fgr silencing in vitro resulted in increased apoptosis that was mediated by increased caspase and AKT activity. In addition, dual silencing of Src and Fgr in vivo using siRNA/CH-NP resulted in the greatest reduction in tumor growth compared with silencing of either Src or Fgr alone in the HeyA8 model (68.8%, P < 0.05). Conclusions: This study demonstrates that, in addition to Src, Fgr plays a biologically significant role in ovarian cancer growth and might represent an important target. Clin Cancer Res; 17(7); 1713–21. ©2011 AACR.


Journal of the National Cancer Institute | 2011

Silencing of p130Cas in Ovarian Carcinoma: A Novel Mechanism for Tumor Cell Death

Alpa M. Nick; Rebecca L. Stone; Guillermo N. Armaiz-Pena; Bulent Ozpolat; Ibrahim Tekedereli; Whitney S. Graybill; Charles N. Landen; Gabriel J. Villares; Pablo Vivas-Mejia; Justin Bottsford-Miller; Hye Sun Kim; Ju Seog Lee; Soo Mi Kim; Keith A. Baggerly; Prahlad T. Ram; Michael T. Deavers; Robert L. Coleman; Gabriel Lopez-Berestein; Anil K. Sood

BACKGROUND We investigated the clinical and biological significance of p130cas, an important cell signaling molecule, in ovarian carcinoma. METHODS Expression of p130cas in ovarian tumors, as assessed by immunohistochemistry, was associated with tumor characteristics and patient survival. The effects of p130cas gene silencing with small interfering RNAs incorporated into neutral nanoliposomes (siRNA-DOPC), alone and in combination with docetaxel, on in vivo tumor growth and on tumor cell proliferation (proliferating cell nuclear antigen) and apoptosis (terminal deoxynucleotidyl transferase dUTP nick-end labeling) were examined in mice bearing orthotopic taxane-sensitive (HeyA8 and SKOV3ip1) or taxane-resistant (HeyA8-MDR) ovarian tumors (n = 10 per group). To determine the specific mechanisms by which p130cas gene silencing abrogates tumor growth, we measured cell viability (MTT assay), apoptosis (fluorescence-activated cell sorting), autophagy (immunoblotting, fluorescence, and transmission electron microscopy), and cell signaling (immunoblotting) in vitro. All statistical tests were two-sided. RESULTS Of 91 ovarian cancer specimens, 70 (76%) had high p130cas expression; and 21 (24%) had low p130cas expression. High p130cas expression was associated with advanced tumor stage (P < .001) and higher residual disease (>1 cm) following primary cytoreduction surgery (P = .007) and inversely associated with overall survival and progression-free survival (median overall survival: high p130cas expression vs low expression, 2.14 vs 9.1 years, difference = 6.96 years, 95% confidence interval = 1.69 to 9.48 years, P < .001; median progression-free survival: high p130cas expression vs low expression, 1.04 vs 2.13 years, difference = 1.09 years, 95% confidence interval = 0.47 to 2.60 years, P = .01). In mice bearing orthotopically implanted HeyA8 or SKOV3ip1 ovarian tumors, treatment with p130cas siRNA-DOPC in combination with docetaxel chemotherapy resulted in the greatest reduction in tumor growth compared with control siRNA therapy (92%-95% reduction in tumor growth; P < .001 for all). Compared with control siRNA therapy, p130cas siRNA-DOPC reduced SKOV3ip1 cell proliferation (31% reduction, P < .001) and increased apoptosis (143% increase, P < .001) in vivo. Increased tumor cell apoptosis may have persisted despite pan-caspase inhibition by the induction of autophagy and related signaling pathways. CONCLUSIONS Increased p130cas expression is associated with poor clinical outcome in human ovarian carcinoma, and p130cas gene silencing decreases tumor growth through stimulation of apoptotic and autophagic cell death.


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.

Collaboration


Dive into the Justin Bottsford-Miller's collaboration.

Top Co-Authors

Avatar

Anil K. Sood

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

Alpa M. Nick

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

Chad V. Pecot

University of North Carolina at Chapel Hill

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

Cristina Ivan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gabriel Lopez-Berestein

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge