Network


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

Hotspot


Dive into the research topics where Jean M. Hansen is active.

Publication


Featured researches published by Jean M. Hansen.


European Journal of Cancer | 2016

Targeting the tumour microenvironment in ovarian cancer.

Jean M. Hansen; Robert L. Coleman; Anil K. Sood

The study of cancer initiation, growth, and metastasis has traditionally been focused on cancer cells, and the view that they proliferate due to uncontrolled growth signalling owing to genetic derangements. However, uncontrolled growth in tumours cannot be explained solely by aberrations in cancer cells themselves. To fully understand the biological behaviour of tumours, it is essential to understand the microenvironment in which cancer cells exist, and how they manipulate the surrounding stroma to promote the malignant phenotype. Ovarian cancer is the leading cause of death from gynaecologic cancer worldwide. The majority of patients will have objective responses to standard tumour debulking surgery and platinum-taxane doublet chemotherapy, but most will experience disease recurrence and chemotherapy resistance. As such, a great deal of effort has been put forth to develop therapies that target the tumour microenvironment in ovarian cancer. Herein, we review the key components of the tumour microenvironment as they pertain to this disease, outline targeting opportunities and supporting evidence thus far, and discuss resistance to therapy.


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.


Journal of Clinical Investigation | 2016

FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal

Monika Haemmerle; Justin Bottsford-Miller; Sunila Pradeep; Morgan Taylor; Hyun Jin Choi; Jean M. Hansen; Heather J. Dalton; Rebecca L. Stone; Min Soon Cho; Alpa M. Nick; Archana S. Nagaraja; Tony Gutschner; Kshipra M. Gharpure; Lingegowda S. Mangala; Rajesha Rupaimoole; Hee Dong Han; Behrouz Zand; Guillermo N. Armaiz-Pena; Sherry Y. Wu; Chad V. Pecot; Alan R. Burns; Gabriel Lopez-Berestein; Vahid Afshar-Kharghan; Anil K. Sood

Recent studies in patients with ovarian cancer suggest that tumor growth may be accelerated following cessation of antiangiogenesis therapy; however, the underlying mechanisms are not well understood. In this study, we aimed to compare the effects of therapy withdrawal to those of continuous treatment with various antiangiogenic agents. Cessation of therapy with pazopanib, bevacizumab, and the human and murine anti-VEGF antibody B20 was associated with substantial tumor growth in mouse models of ovarian cancer. Increased tumor growth was accompanied by tumor hypoxia, increased tumor angiogenesis, and vascular leakage. Moreover, we found hypoxia-induced ADP production and platelet infiltration into tumors after withdrawal of antiangiogenic therapy, and lowering platelet counts markedly inhibited tumor rebound after withdrawal of antiangiogenic therapy. Focal adhesion kinase (FAK) in platelets regulated their migration into the tumor microenvironment, and FAK-deficient platelets completely prevented the rebound tumor growth. Additionally, combined therapy with a FAK inhibitor and the antiangiogenic agents pazopanib and bevacizumab reduced tumor growth and inhibited negative effects following withdrawal of antiangiogenic therapy. In summary, these results suggest that FAK may be a unique target in situations in which antiangiogenic agents are withdrawn, and dual targeting of FAK and VEGF could have therapeutic implications for ovarian cancer management.


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.


Nature Communications | 2017

Platelets reduce anoikis and promote metastasis by activating YAP1 signaling

Monika Haemmerle; Morgan Taylor; Tony Gutschner; Sunila Pradeep; Min Soon Cho; Jianting Sheng; Yasmin M. Lyons; Archana S. Nagaraja; Robert L. Dood; Yunfei Wen; Lingegowda S. Mangala; Jean M. Hansen; Rajesha Rupaimoole; Kshipra M. Gharpure; Cristian Rodriguez-Aguayo; Sun Young Yim; Ju Seog Lee; Cristina Ivan; Wei Hu; Gabriel Lopez-Berestein; Stephen T. C. Wong; Beth Y. Karlan; Douglas A. Levine; Jinsong Liu; Vahid Afshar-Kharghan; Anil K. Sood

Thrombocytosis is present in more than 30% of patients with solid malignancies and correlates with worsened patient survival. Tumor cell interaction with various cellular components of the tumor microenvironment including platelets is crucial for tumor growth and metastasis. Although it is known that platelets can infiltrate into tumor tissue, secrete pro-angiogenic and pro-tumorigenic factors and thereby increase tumor growth, the precise molecular interactions between platelets and metastatic cancer cells are not well understood. Here we demonstrate that platelets induce resistance to anoikis in vitro and are critical for metastasis in vivo. We further show that platelets activate RhoA-MYPT1-PP1-mediated YAP1 dephosphorylation and promote its nuclear translocation which induces a pro-survival gene expression signature and inhibits apoptosis. Reduction of YAP1 in cancer cells in vivo protects against thrombocytosis-induced increase in metastasis. Collectively, our results indicate that cancer cells depend on platelets to avoid anoikis and succeed in the metastatic process.Platelets have been associated with increased tumor growth and metastasis but the mechanistic details of this interaction are still unclear. Here the authors show that platelets improve anoikis resistance of cancer cells and increase metastasis by activating Yap through a RhoA/MYPT-PP1 pathway.


European Journal of Cancer | 2015

Venous thromboembolism, interleukin-6 and survival outcomes in patients with advanced ovarian clear cell carcinoma

Koji Matsuo; Kosei Hasegawa; Kiyoshi Yoshino; Ryusuke Murakami; Takeshi Hisamatsu; Rebecca L. Stone; Rebecca A. Previs; Jean M. Hansen; Yuji Ikeda; Akiko Miyara; Kosuke Hiramatsu; Takayuki Enomoto; Keiichi Fujiwara; Noriomi Matsumura; Ikuo Konishi; Lynda D. Roman; Hani Gabra; Christina Fotopoulou; Anil K. Sood

BACKGROUND We compared survival outcomes and risk of venous thromboembolism (VTE) among patients with advanced and early-stage ovarian clear cell carcinoma (OCCC) and serous ovarian carcinoma (SOC), as well as potential links with interleukin-6 (IL-6) levels. METHODS A multicenter case-control study was conducted in 370 patients with OCCC and 938 with SOC. In a subset of 200 cases, pretreatment plasma IL-6 levels were examined. FINDINGS Patients with advanced OCCC had the highest 2-year cumulative VTE rates (advanced OCCC 43.1%, advanced SOC 16.2%, early-stage OCCC 11.9% and early-stage SOC 6.4%, P<0.0001) and the highest median levels of IL-6 (advanced OCCC 17.8 pg/mL, advanced SOC 9.0 pg/mL, early-stage OCCC 4.2 pg/mL and early-stage SOC 5.0 pg/mL, P=0.006). Advanced OCCC (hazard ratio [HR] 3.38, P<0.0001), thrombocytosis (HR 1.42, P=0.032) and elevated IL-6 (HR 8.90, P=0.046) were independent predictors of VTE. In multivariate analysis, patients with advanced OCCC had significantly poorer 5-year progression-free and overall survival rates than those with advanced SOC (P<0.01), and thrombocytosis was an independent predictor of decreased survival outcomes (P<0.01). Elevated IL-6 levels led to poorer 2-year progression-free survival rates in patients with OCCC (50% versus 87.5%, HR 4.89, P=0.016) than in those with SOC (24.9% versus 40.8%, HR 1.40, P=0.07). INTERPRETATION Advanced OCCC is associated with an increased incidence of VTE and decreased survival outcomes, which has major implications for clinical management of OCCC.


Clinical Cancer Research | 2016

Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer

Yu Kang; Archana S. Nagaraja; Guillermo N. Armaiz-Pena; Piotr L. Dorniak; Wei Hu; Rajesha Rupaimoole; Tao Liu; Kshipra M. Gharpure; Rebecca A. Previs; Jean M. Hansen; Cristian Rodriguez-Aguayo; Cristina Ivan; Prahlad T. Ram; Vasudha Sehgal; Gabriel Lopez-Berestein; Susan K. Lutgendorf; Steve W. Cole; Anil K. Sood

Purpose: Chronic adrenergic activation has been shown to associate with adverse clinical outcomes in cancer patients, but the underlying mechanisms are not well understood. The focus of the current study was to determine the functional and biologic effects of adrenergic pathways on response to chemotherapy in the context of ovarian cancer. Experimental Design: Increased DUSP1 production by sympathetic nervous system mediators (e.g., norepinephrine) was analyzed by real-time quantitative RT-PCR and by Western blotting. In vitro chemotherapy-induced cell apoptosis was examined by flow cytometry. For in vivo therapy, a well-characterized model of chronic stress was used. Results: Catecholamines significantly inhibited paclitaxel- and cisplatin-induced apoptosis in ovarian cancer cells. Genomic analyses of cells treated with norepinephrine identified DUSP1 as a potential mediator. DUSP1 overexpression resulted in reduced paclitaxel-induced apoptosis in ovarian cancer cells compared with control; conversely, DUSP1 gene silencing resulted in increased apoptosis compared with control cells. DUSP1 gene silencing in vivo significantly enhanced response to paclitaxel and increased apoptosis. In vitro analyses indicated that norepinephrine-induced DUSP1 gene expression was mediated through ADRB2 activation of cAMP–PLC–PKC–CREB signaling, which inhibits JNK-mediated phosphorylation of c-Jun and protects ovarian cancer cells from apoptosis. Moreover, analysis of The Cancer Genome Atlas data showed that increased DUSP1 expression was associated with decreased overall (P = 0.049) and progression-free (P = 0.0005) survival. Conclusions: These findings provide a new understanding of the mechanisms by which adrenergic pathways can impair response to chemotherapy and have implications for cancer management. Clin Cancer Res; 22(7); 1713–24. ©2015 AACR.


Clinical Cancer Research | 2017

Macrophages Facilitate Resistance to Anti-VEGF Therapy by Altered VEGFR Expression

Heather J. Dalton; Sunila Pradeep; Michael McGuire; Yared Hailemichael; Shaolin Ma; Yasmin A. Lyons; Guillermo N. Armaiz-Pena; Rebecca A. Previs; Jean M. Hansen; Rajesha Rupaimoole; Vianey Gonzalez-Villasana; Min Soon Cho; Sherry Y. Wu; Lingegowda S. Mangala; Nicholas B. Jennings; Wei Hu; Robert R. Langley; Hong Mu; Michael Andreeff; Menashe Bar-Eli; Willem W. Overwijk; Prahlad T. Ram; Gabriel Lopez-Berestein; Robert L. Coleman; Anil K. Sood

Purpose: VEGF-targeted therapies have modest efficacy in cancer patients, but acquired resistance is common. The mechanisms underlying such resistance are poorly understood. Experimental Design: To evaluate the potential role of immune cells in the development of resistance to VEGF blockade, we first established a preclinical model of adaptive resistance to anti-VEGF therapy. Additional in vitro and in vivo studies were carried out to characterize the role of macrophages in such resistance. Results: Using murine cancer models of adaptive resistance to anti-VEGF antibody (AVA), we found a previously unrecognized role of macrophages in such resistance. Macrophages were actively recruited to the tumor microenvironment and were responsible for the emergence of AVA resistance. Depletion of macrophages following emergence of resistance halted tumor growth and prolonged survival of tumor-bearing mice. In a macrophage-deficient mouse model, resistance to AVA failed to develop, but could be induced by injection of macrophages. Downregulation of macrophage VEGFR-1 and VEGFR-3 expression accompanied upregulation of alternative angiogenic pathways, facilitating escape from anti-VEGF therapy. Conclusions: These findings provide a new understanding of the mechanisms underlying the modest efficacy of current antiangiogenesis therapies and identify new opportunities for combination approaches for ovarian and other cancers. Clin Cancer Res; 23(22); 7034–46. ©2017 AACR.


Molecular Cancer Therapeutics | 2016

Antitumor and antiangiogenic effects of aspirin-PC in ovarian cancer

Yan Huang; Lenard M. Lichtenberger; Morgan Taylor; Justin Bottsford-Miller; Monika Haemmerle; Michael J. Wagner; Yasmin A. Lyons; Sunila Pradeep; Wei Hu; Rebecca A. Previs; Jean M. Hansen; Dexing Fang; Piotr L. Dorniak; Justyna Filant; Elizabeth J. Dial; Fangrong Shen; Hiroto Hatakeyama; Anil K. Sood

To determine the efficacy of a novel and safer (for gastrointestinal tract) aspirin (aspirin-PC) in preclinical models of ovarian cancer, in vitro dose–response studies were performed to compare the growth-inhibitory effect of aspirin-PC versus aspirin on three human (A2780, SKOV3ip1, and HeyA8) and a mouse (ID8) ovarian cancer cell line over an 8-day culture period. In the in vivo studies, the aspirin test drugs were studied alone and in the presence of a VEGF-A inhibitor (bevacizumab or B20), due to an emerging role for platelets in tumor growth following antiangiogenic therapy, and we examined their underlying mechanisms. Aspirin-PC was more potent (vs. aspirin) in blocking the growth of both human and mouse ovarian cancer cells in monolayer culture. Using in vivo model systems of ovarian cancer, we found that aspirin-PC significantly reduced ovarian cancer growth by 50% to 90% (depending on the ovarian cell line). The efficacy was further enhanced in combination with Bevacizumab or B20. The growth-inhibitory effect on ovarian tumor mass and number of tumor nodules was evident, but less pronounced for aspirin and the VEGF inhibitors alone. There was no detectable gastrointestinal toxicity. Both aspirin and aspirin-PC also inhibited cell proliferation, angiogenesis, and increased apoptosis of ovarian cancer cells. In conclusion, PC-associated aspirin markedly inhibits the growth of ovarian cancer cells, which exceeds that of the parent drug, in both cell culture and in mouse model systems. We also found that both aspirin-PC and aspirin have robust antineoplastic action in the presence of VEGF-blocking drugs. Mol Cancer Ther; 15(12); 2894–904. ©2016 AACR.


Molecular Cancer Therapeutics | 2015

Dual Metronomic Chemotherapy with Nab-Paclitaxel and Topotecan Has Potent Antiangiogenic Activity in Ovarian Cancer

Rebecca A. Previs; Guillermo N. Armaiz-Pena; Yvonne G. Lin; Ashley Davis; Sunila Pradeep; Heather J. Dalton; Jean M. Hansen; William M. Merritt; Alpa M. Nick; Robert R. Langley; Robert L. Coleman; Anil K. Sood

There is growing recognition of the important role of metronomic chemotherapy in cancer treatment. On the basis of their unique antiangiogenic effects, we tested the efficacy of nab-paclitaxel, which stimulates thrombospondin-1, and topotecan, which inhibits hypoxia-inducible factor 1-α, at metronomic dosing for the treatment of ovarian carcinoma. In vitro and in vivo SKOV3ip1, HeyA8, and HeyA8-MDR (taxane-resistant) orthotopic models were used to examine the effects of metronomic nab-paclitaxel and metronomic topotecan. We examined cell proliferation (Ki-67), apoptosis (cleaved caspase-3), and angiogenesis (microvessel density, MVD) in tumors obtained at necropsy. In vivo therapy experiments demonstrated treatment with metronomic nab-paclitaxel alone and in combination with metronomic topotecan resulted in significant reductions in tumor weight (62% in the SKOV3ip1 model, P < 0.01 and 96% in the HeyA8 model, P < 0.03) compared with vehicle (P < 0.01). In the HeyA8-MDR model, metronomic monotherapy with either cytotoxic agent had modest effects on tumor growth, but combination therapy decreased tumor burden by 61% compared with vehicle (P < 0.03). The greatest reduction in MVD (P < 0.05) and proliferation was seen in combination metronomic therapy groups. Combination metronomic therapy resulted in prolonged overall survival in vivo compared with other groups (P < 0.001). Tube formation was significantly inhibited in RF-24 endothelial cells exposed to media conditioned with metronomic nab-paclitaxel alone and media conditioned with combination metronomic nab-paclitaxel and metronomic topotecan. The combination of metronomic nab-paclitaxel and metronomic topotecan offers a novel, highly effective therapeutic approach for ovarian carcinoma that merits further clinical development. Mol Cancer Ther; 14(12); 2677–86. ©2015 AACR.

Collaboration


Dive into the Jean M. Hansen's collaboration.

Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

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

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

Sunila Pradeep

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

Guillermo N. Armaiz-Pena

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sherry Y. Wu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yasmin A. Lyons

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Archana S. Nagaraja

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge