99 results on '"Diane Palmieri"'
Search Results
2. Supplementary Figure Legend from Paclitaxel–Hyaluronic NanoConjugates Prolong Overall Survival in a Preclinical Brain Metastases of Breast Cancer Model
- Author
-
Paul R. Lockman, Quentin R. Smith, Patricia S. Steeg, Diane Palmieri, Werner J. Geldenhuys, Hussaini S. Qhattal, Tori B. Terrell, Kaci A. Bohn, Mohamed I. Nounou, Chris E. Adkins, Xinli Liu, and Rajendar K. Mittapalli
- Abstract
Supplementary Figure Legends
- Published
- 2023
3. Data from Notch1 Inhibition Alters the CD44hi/CD24lo Population and Reduces the Formation of Brain Metastases from Breast Cancer
- Author
-
Ann F. Chambers, Alison L. Allan, Patricia S. Steeg, Diane Palmieri, Paula J. Foster, Emeline J. Ribot, Carmen Simedrea, and Patricia M. McGowan
- Abstract
Brain metastasis from breast cancer is an increasingly important clinical problem. Here we assessed the role of CD44hi/CD24lo cells and pathways that regulate them, in an experimental model of brain metastasis. Notch signaling (mediated by γ-secretase) has been shown to contribute to maintenance of the cancer stem cell (CSC) phenotype. Cells sorted for a reduced stem-like phenotype had a reduced ability to form brain metastases compared with unsorted or CD44hi/CD24lo cells (P < 0.05; Kruskal–Wallis). To assess the effect of γ-secretase inhibition, cells were cultured with DAPT and the CD44/CD24 phenotypes quantified. 231-BR cells with a CD44hi/CD24lo phenotype was reduced by about 15% in cells treated with DAPT compared with DMSO-treated or untreated cells (P = 0.001, ANOVA). In vivo, mice treated with DAPT developed significantly fewer micro- and macrometastases compared with vehicle treated or untreated mice (P = 0.011, Kruskal–Wallis). Notch1 knockdown reduced the expression of CD44hi/CD24lo phenotype by about 20%. In vitro, Notch1 shRNA resulted in a reduction in cellular growth at 24, 48, and 72 hours time points (P = 0.033, P = 0.002, and P = 0.009, ANOVA) and about 60% reduction in Matrigel invasion was observed (P < 0.001, ANOVA). Cells transfected with shNotch1 formed significantly fewer macrometastases and micrometastases compared with scrambled shRNA or untransfected cells (P < 0.001; Kruskal–Wallis). These data suggest that the CSC phenotype contributes to the development of brain metastases from breast cancer, and this may arise in part from increased Notch activity. Mol Cancer Res; 9(7); 834–44. ©2011 AACR.
- Published
- 2023
4. Supplementary Figures 1 through 3 from Paclitaxel–Hyaluronic NanoConjugates Prolong Overall Survival in a Preclinical Brain Metastases of Breast Cancer Model
- Author
-
Paul R. Lockman, Quentin R. Smith, Patricia S. Steeg, Diane Palmieri, Werner J. Geldenhuys, Hussaini S. Qhattal, Tori B. Terrell, Kaci A. Bohn, Mohamed I. Nounou, Chris E. Adkins, Xinli Liu, and Rajendar K. Mittapalli
- Abstract
Supplementary Figure S1: NMR Spectra of HA-Paclitaxel. 1H-NMR spectrum of HA-paclitaxel. Specific groups of HA-paclitaxel are shown with arrows; Supplementary Figure S2: Representative bioluminescent images of animals 24 hrs after intracardiac injection of the 231Br cells; Supplementary Figure S3: Representative image demonstrating that bioluminescence correlates well with histological location and burden of metastatic lesions in brain.
- Published
- 2023
5. Supplementary Table 1, Figures 1-4 from Notch1 Inhibition Alters the CD44hi/CD24lo Population and Reduces the Formation of Brain Metastases from Breast Cancer
- Author
-
Ann F. Chambers, Alison L. Allan, Patricia S. Steeg, Diane Palmieri, Paula J. Foster, Emeline J. Ribot, Carmen Simedrea, and Patricia M. McGowan
- Abstract
Supplementary Table 1, Figures 1-4 from Notch1 Inhibition Alters the CD44hi/CD24lo Population and Reduces the Formation of Brain Metastases from Breast Cancer
- Published
- 2023
6. Supplementary Data from Vorinostat Inhibits Brain Metastatic Colonization in a Model of Triple-Negative Breast Cancer and Induces DNA Double-Strand Breaks
- Author
-
Patricia S. Steeg, Quentin R. Smith, Victoria M. Richon, Paul S. Meltzer, Kevin Camphausen, Seth M. Steinberg, Lionel Feigenbaum, Julie L. Bronder, Robert Walker, John F. Reilly, Sean Davis, David J. Liewehr, Helen R. Thorsheim, Kaci A. Bohn, Julie A. Gaasch, Rajendar K. Mittapalli, Vinay Rudraraju, Kunal S. Taskar, Eleazar Vega-Valle, Yongzhen Qian, Natasha Flores, Matthew Johnson, Elizabeth Hargrave, Jeanne Herring, Emily Hua, Fancy C. Thomas, Paul R. Lockman, and Diane Palmieri
- Abstract
Supplementary Data from Vorinostat Inhibits Brain Metastatic Colonization in a Model of Triple-Negative Breast Cancer and Induces DNA Double-Strand Breaks
- Published
- 2023
7. Supplementary Figure 2 from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
PDF file - 113KB, MGMT expression status in Jimt-1-Br3 cells predicts sensitivity to temozolomide.
- Published
- 2023
8. Supplementary Figure Legends from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
PDF file - 96KB
- Published
- 2023
9. Data from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
Purpose: Brain metastases of breast cancer cause neurocognitive damage and are incurable. We evaluated a role for temozolomide in the prevention of brain metastases of breast cancer in experimental brain metastasis models.Experimental Design: Temozolomide was administered in mice following earlier injection of brain-tropic HER2–positive JIMT-1-BR3 and triple-negative 231-BR-EGFP sublines, the latter with and without expression of O6-methylguanine-DNA methyltransferase (MGMT). In addition, the percentage of MGMT-positive tumor cells in 62 patient-matched sets of breast cancer primary tumors and resected brain metastases was determined immunohistochemically.Results: Temozolomide, when dosed at 50, 25, 10, or 5 mg/kg, 5 days per week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cells. At a 1 mg/kg dose, temozolomide prevented 68% of large brain metastases, and was ineffective at a dose of 0.5 mg/kg. When the 50 mg/kg dose was administered beginning on days 18 or 24, temozolomide efficacy was reduced or absent. Temozolomide was ineffective at preventing brain metastases in MGMT-transduced 231-BR-EGFP and MGMT-expressing JIMT-1-BR3 sublines. In 62 patient-matched sets of primary breast tumors and resected brain metastases, 43.5% of the specimens had concordant low MGMT expression, whereas in another 14.5% of sets high MGMT staining in the primary tumor corresponded with low staining in the brain metastasis.Conclusions: Temozolomide profoundly prevented the outgrowth of experimental brain metastases of breast cancer in an MGMT-dependent manner. These data provide compelling rationale for investigating the preventive efficacy of temozolomide in a clinical setting. Clin Cancer Res; 20(10); 2727–39. ©2014 AACR.
- Published
- 2023
10. Data from Alterations in Pericyte Subpopulations Are Associated with Elevated Blood–Tumor Barrier Permeability in Experimental Brain Metastasis of Breast Cancer
- Author
-
Brunilde Gril, Patricia S. Steeg, Priscilla K. Brastianos, Naema Nayyar, Renata Duchnowska, Ewa Izycka-Swieszewska, Wojciech Kloc, Seth M. Steinberg, David J. Liewehr, Diane Palmieri, Emily Hua, Emily Sechrest, Afroz Shareef Mohammad, Chris E. Adkins, Paul R. Lockman, and L. Tiffany Lyle
- Abstract
Purpose: The blood–brain barrier (BBB) is modified to a blood–tumor barrier (BTB) as a brain metastasis develops from breast or other cancers. We (i) quantified the permeability of experimental brain metastases, (ii) determined the composition of the BTB, and (iii) identified which elements of the BTB distinguished metastases of lower permeability from those with higher permeability.Experimental Design: A SUM190-BR3 experimental inflammatory breast cancer brain metastasis subline was established. Experimental brain metastases from this model system and two previously reported models (triple-negative MDA-231-BR6, HER2+ JIMT-1-BR3) were serially sectioned; low- and high-permeability lesions were identified with systemic 3-kDa Texas Red dextran dye. Adjoining sections were used for quantitative immunofluorescence to known BBB and neuroinflammatory components. One-sample comparisons against a hypothesized value of one were performed with the Wilcoxon signed-rank test.Results: When uninvolved brain was compared with any brain metastasis, alterations in endothelial, pericytic, astrocytic, and microglial components were observed. When metastases with relatively low and high permeability were compared, increased expression of a desmin+ subpopulation of pericytes was associated with higher permeability (231-BR6 P = 0.0002; JIMT-1-BR3 P = 0.004; SUM190-BR3 P = 0.008); desmin+ pericytes were also identified in human craniotomy specimens. Trends of reduced CD13+ pericytes (231-BR6 P = 0.014; JIMT-1-BR3 P = 0.002, SUM190-BR3, NS) and laminin α2 (231-BR6 P = 0.001; JIMT-1-BR3 P = 0.049; SUM190-BR3 P = 0.023) were also observed with increased permeability.Conclusions: We provide the first account of the composition of the BTB in experimental brain metastasis. Desmin+ pericytes and laminin α2 are potential targets for the development of novel approaches to increase chemotherapeutic efficacy. Clin Cancer Res; 22(21); 5287–99. ©2016 AACR.
- Published
- 2023
11. Supplementary Data from Pazopanib Reveals a Role for Tumor Cell B-Raf in the Prevention of HER2+ Breast Cancer Brain Metastasis
- Author
-
Patricia S. Steeg, Seth M. Steinberg, David J. Liewehr, Lilia Ileva, DeeDee Smart, Yong Qian, Diane Palmieri, and Brunilde Gril
- Abstract
Supplementary Figures S1-S5; Supplementary Materials and Methods.
- Published
- 2023
12. Supplementary Figure 1 from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
PDF file - 67KB, MGMT overexpression in 231-BR-EGFP cells increases sensitivity to temozolomide in vitro.
- Published
- 2023
13. Supplementary Figure Legends from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
PDF file - 96KB
- Published
- 2023
14. Data from Heterogeneous Blood–Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer
- Author
-
Quentin R. Smith, Patricia S. Steeg, Diane Palmieri, Suyun Huang, Julie A. Gaasch, Helen R. Thorsheim, Amanda Roberts, Chris E. Adkins, Kaci A. Bohn, Brunilde Gril, Vinay Rudraraju, Kunal S. Taskar, Rajendar K. Mittapalli, and Paul R. Lockman
- Abstract
Purpose: Brain metastases of breast cancer appear to be increasing in incidence, confer significant morbidity, and threaten to compromise gains made in systemic chemotherapy. The blood–tumor barrier (BTB) is compromised in many brain metastases; however, the extent to which this influences chemotherapeutic delivery and efficacy is unknown. Herein, we answer this question by measuring BTB passive integrity, chemotherapeutic drug uptake, and anticancer efficacy in vivo in two breast cancer models that metastasize preferentially to brain.Experimental Design:Experimental brain metastasis drug uptake and BTB permeability were simultaneously measured using novel fluorescent and phosphorescent imaging techniques in immune-compromised mice. Drug-induced apoptosis and vascular characteristics were assessed using immunofluorescent microscopy.Results: Analysis of over 2,000 brain metastases from two models (human 231-BR-Her2 and murine 4T1-BR5) showed partial BTB permeability compromise in greater than 89% of lesions, varying in magnitude within and between metastases. Brain metastasis uptake of 14C-paclitaxel and 14C-doxorubicin was generally greater than normal brain but less than 15% of that of other tissues or peripheral metastases, and only reached cytotoxic concentrations in a small subset (∼10%) of the most permeable metastases. Neither drug significantly decreased the experimental brain metastatic ability of 231-BR-Her2 tumor cells. BTB permeability was associated with vascular remodeling and correlated with overexpression of the pericyte protein desmin.Conclusions: This work shows that the BTB remains a significant impediment to standard chemotherapeutic delivery and efficacy in experimental brain metastases of breast cancer. New brain permeable drugs will be needed. Evidence is presented for vascular remodeling in BTB permeability alterations. Clin Cancer Res; 16(23); 5664–78. ©2010 AACR.
- Published
- 2023
15. Supplementary Figure 3 from Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Patricia S. Steeg, Jacek Jassem, Seth M. Steinberg, David J. Liewehr, Stephen M. Hewitt, Andreas M. Stark, Brunilde Gril, Katarzyna Sosińska-Mielcarek, Wojciech Biernat, Yongzhen Qian, Emily Hua, Stephan Woditschka, Renata Duchnowska, and Diane Palmieri
- Abstract
PDF file - 96KB, MGMT expression in matched sets of human breast primary tumors and resected brain metastases does not affect overall patient survival.
- Published
- 2023
16. CCR Translation for This Article from Heterogeneous Blood–Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer
- Author
-
Quentin R. Smith, Patricia S. Steeg, Diane Palmieri, Suyun Huang, Julie A. Gaasch, Helen R. Thorsheim, Amanda Roberts, Chris E. Adkins, Kaci A. Bohn, Brunilde Gril, Vinay Rudraraju, Kunal S. Taskar, Rajendar K. Mittapalli, and Paul R. Lockman
- Abstract
CCR Translation for This Article from Heterogeneous Blood–Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer
- Published
- 2023
17. Supplementary Data File from Alterations in Pericyte Subpopulations Are Associated with Elevated Blood–Tumor Barrier Permeability in Experimental Brain Metastasis of Breast Cancer
- Author
-
Brunilde Gril, Patricia S. Steeg, Priscilla K. Brastianos, Naema Nayyar, Renata Duchnowska, Ewa Izycka-Swieszewska, Wojciech Kloc, Seth M. Steinberg, David J. Liewehr, Diane Palmieri, Emily Hua, Emily Sechrest, Afroz Shareef Mohammad, Chris E. Adkins, Paul R. Lockman, and L. Tiffany Lyle
- Abstract
Supplemental Tables. Supplemental Tables S1-6. Table S1. Antibodies used for quantification and concentrations. Table S2. Antibodies that could not be quantified in the model systems under fixation conditions used to identify Texas Red dextran permeability. Table S3. Summary statistics for analysis of brain metastases vs. uninvolved brain for each analyzed immunofluorescence marker. Table S4. Summary statistics for analysis of highly permeable vs. poorly permeable brain metastases for each analyzed immunofluorescence marker. Table S5. Antibodies that could not be quantified in human specimens. Table S6: Summary of immunofluorescence analysis of human brain metastasis specimens Supplemental Figures. Supplemental Figures S1-9. Figure S1. Homogenous HER2 staining in a SUM190-BR3 brain metastasis. Figure S2. No significant trend in Claudin-5 expression in three models of brain metastasis. Figure S3. Increased VEGF expression in brain metastasis. Figure S4. Decreased ZO-1 expression in brain metastasis. Figure S5. Decreased AQP4 expression and an alteration in the AQP4 expression pattern in brain metastasis. Figure S6: Heterogeneous trends in Collagen IV expression in three models of brain metastasis. Figure S7: Colocalization of Desmin+ pericytes and CD13+ pericytes with PDGFR-β. Figure S8: Desmin immunofluorescent staining in human specimens. Figure S9. Examples of immunofluorescent staining in human specimen #5 Supplemental Experimental Procedures. Detailed experimental procedures that were not included in the body of the manuscript. These include SUM190-BR3 cell line derivation, animal experiments, immunofluorescence, image analysis, statistical analysis, and human craniotomy specimen preparation.
- Published
- 2023
18. Data from Vorinostat Inhibits Brain Metastatic Colonization in a Model of Triple-Negative Breast Cancer and Induces DNA Double-Strand Breaks
- Author
-
Patricia S. Steeg, Quentin R. Smith, Victoria M. Richon, Paul S. Meltzer, Kevin Camphausen, Seth M. Steinberg, Lionel Feigenbaum, Julie L. Bronder, Robert Walker, John F. Reilly, Sean Davis, David J. Liewehr, Helen R. Thorsheim, Kaci A. Bohn, Julie A. Gaasch, Rajendar K. Mittapalli, Vinay Rudraraju, Kunal S. Taskar, Eleazar Vega-Valle, Yongzhen Qian, Natasha Flores, Matthew Johnson, Elizabeth Hargrave, Jeanne Herring, Emily Hua, Fancy C. Thomas, Paul R. Lockman, and Diane Palmieri
- Abstract
Purpose: As chemotherapy and molecular therapy improve the systemic survival of breast cancer patients, the incidence of brain metastases increases. Few therapeutic strategies exist for the treatment of brain metastases because the blood-brain barrier severely limits drug access. We report the pharmacokinetic, efficacy, and mechanism of action studies for the histone deactylase inhibitor vorinostat (suberoylanilide hydroxamic acid) in a preclinical model of brain metastasis of triple-negative breast cancer.Experimental Design: The 231-BR brain trophic subline of the MDA-MB-231 human breast cancer cell line was injected into immunocompromised mice for pharmacokinetic and metastasis studies. Pharmacodynamic studies compared histone acetylation, apoptosis, proliferation, and DNA damage in vitro and in vivo.Results: Following systemic administration, uptake of [14C]vorinostat was significant into normal rodent brain and accumulation was up to 3-fold higher in a proportion of metastases formed by 231-BR cells. Vorinostat prevented the development of 231-BR micrometastases by 28% (P = 0.017) and large metastases by 62% (P < 0.0001) compared with vehicle-treated mice when treatment was initiated on day 3 post-injection. The inhibitory activity of vorinostat as a single agent was linked to a novel function in vivo: induction of DNA double-strand breaks associated with the down-regulation of the DNA repair gene Rad52.Conclusions: We report the first preclinical data for the prevention of brain metastasis of triple-negative breast cancer. Vorinostat is brain permeable and can prevent the formation of brain metastases by 62%. Its mechanism of action involves the induction of DNA double-strand breaks, suggesting rational combinations with DNA active drugs or radiation. (Clin Cancer Res 2009;15(19):6148–57)
- Published
- 2023
19. Data from Pazopanib Reveals a Role for Tumor Cell B-Raf in the Prevention of HER2+ Breast Cancer Brain Metastasis
- Author
-
Patricia S. Steeg, Seth M. Steinberg, David J. Liewehr, Lilia Ileva, DeeDee Smart, Yong Qian, Diane Palmieri, and Brunilde Gril
- Abstract
Purpose: Brain metastases of breast cancer contribute significantly to patient morbidity and mortality. We have tested pazopanib, a recently approved antiangiogenic drug that targets VEGFR1, VEGFR2, VEGFR3, PDGFRβ, PDGFRα, and c-kit, for prevention of experimental brain metastases and mechanism of action.Experimental Design:In vitro assays included B-Raf enzymatic assays, Western blots, and angiogenesis assays. For in vivo assays, HER2 transfectants of the brain seeking sublines of MDA-MB-231 cells (231-BR-HER2) and MCF7 cells (MCF7-HER2-BR3, derived herein) were injected into the left cardiac ventricle of mice and treated with vehicle or pazopanib beginning on day 3 postinjection. Brain metastases were counted histologically, imaged, and immunostained.Results: Treatment with 100 mg/kg of pazopanib resulted in a 73% decline in large 231-BR-HER2 metastases (P < 0.0001) and a 39% decline in micrometastases (P = 0.004). In vitro, pazopanib was directly antiproliferative to 231-BR-HER2 breast cancer cells and inhibited MEK and ERK activation in vitro despite B-Raf and Ras mutations. Enzymatic assays demonstrated that pazopanib directly inhibited the wild type and exon 11 oncogenic mutant, but not the V600E mutant forms of B-Raf. Activation of the B-Raf targets pERK1/2 and pMEK1/2 was decreased in pazopanib-treated brain metastases whereas blood vessel density was unaltered. In the MCF7-HER2-BR3 experimental brain metastasis model, pazopanib reduced overall brain metastasis volume upon magnetic resonance imaging (MRI) by 55% (P = 0.067), without affecting brain metastasis vascular density.Conclusions: The data identify a new activity for pazopanib directly on tumor cells as a pan-Raf inhibitor and suggest its potential for prevention of brain metastatic colonization of HER2+ breast cancer. Clin Cancer Res; 17(1); 142–53. ©2010 AACR.
- Published
- 2023
20. Supplementary Figures S1-S9 from Heterogeneous Blood–Tumor Barrier Permeability Determines Drug Efficacy in Experimental Brain Metastases of Breast Cancer
- Author
-
Quentin R. Smith, Patricia S. Steeg, Diane Palmieri, Suyun Huang, Julie A. Gaasch, Helen R. Thorsheim, Amanda Roberts, Chris E. Adkins, Kaci A. Bohn, Brunilde Gril, Vinay Rudraraju, Kunal S. Taskar, Rajendar K. Mittapalli, and Paul R. Lockman
- Abstract
Supplementary Figures S1-S9.
- Published
- 2023
21. Data from Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Author
-
Patricia S. Steeg, Kenneth Aldape, Seth M. Steinberg, Alexander O. Vortmeyer, Douglas Halverson, Lionel Feigenbaum, Eleazar Vega-Valle, Raffael Kurek, Andreas M. Stark, Robert J. Weil, Toshiyuki Yoneda, Jeanne M. Herring, Julie L. Bronder, and Diane Palmieri
- Abstract
Retrospective studies of breast cancer patients suggest that primary tumor Her-2 overexpression or trastuzumab therapy is associated with a devastating complication: the development of central nervous system (brain) metastases. Herein, we present Her-2 expression trends from resected human brain metastases and data from an experimental brain metastasis assay, both indicative of a functional contribution of Her-2 to brain metastatic colonization. Of 124 archival resected brain metastases from breast cancer patients, 36.2% overexpressed Her-2, indicating an enrichment in the frequency of tumor Her-2 overexpression at this metastatic site. Using quantitative real-time PCR of laser capture microdissected epithelial cells, Her-2 and epidermal growth factor receptor (EGFR) mRNA levels in a cohort of 12 frozen brain metastases were increased up to 5- and 9-fold, respectively, over those of Her-2–amplified primary tumors. Co-overexpression of Her-2 and EGFR was also observed in a subset of brain metastases. We then tested the hypothesis that overexpression of Her-2 increases the colonization of breast cancer cells in the brain in vivo. A subclone of MDA-MB-231 human breast carcinoma cells that selectively metastasizes to brain (231-BR) overexpressed EGFR; 231-BR cells were transfected with low (4- to 8-fold) or high (22- to 28-fold) levels of Her-2. In vivo, in a model of brain metastasis, low or high Her-2–overexpressing 231-BR clones produced comparable numbers of micrometastases in the brain as control transfectants; however, the Her-2 transfectants yielded 3-fold greater large metastases (>50 μm2; P < 0.001). Our data indicate that Her-2 overexpression increases the outgrowth of metastatic tumor cells in the brain in this model system. [Cancer Res 2007;67(9):4190–8]
- Published
- 2023
22. Supplementary Tables 1-4, Figure Legends 1-2 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Author
-
Patricia S. Steeg, Paul S. Meltzer, Marie-Lise Lacombe, William G. Stetler-Stevenson, Diane Palmieri, Sandrine Arnaud-Dabernat, Tara K. Maga, Sylvie Dumont, Mathieu Boissan, Abdel G. Elkahloun, Jong Heun Lee, and Christine E. Horak
- Abstract
Supplementary Tables 1-4, Figure Legends 1-2 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Published
- 2023
23. Data from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Author
-
Patricia S. Steeg, Paul S. Meltzer, Marie-Lise Lacombe, William G. Stetler-Stevenson, Diane Palmieri, Sandrine Arnaud-Dabernat, Tara K. Maga, Sylvie Dumont, Mathieu Boissan, Abdel G. Elkahloun, Jong Heun Lee, and Christine E. Horak
- Abstract
Exogenous overexpression of the metastasis suppressor gene Nm23-H1 reduces the metastatic potential of multiple types of cancer cells and suppresses in vitro tumor cell motility and invasion. Mutational analysis of Nm23-H1 revealed that substitution mutants P96S and S120G did not inhibit motility and invasion. To elucidate the molecular mechanism of Nm23-H1 motility suppression, expression microarray analysis of an MDA-MB-435 cancer cell line overexpressing wild-type Nm23-H1 was done and cross-compared with expression profiles from lines expressing the P96S and S120G mutants. Nine genes, MET, PTN, SMO, FZD1, L1CAM, MMP2, NETO2, CTGF, and EDG2, were down-regulated by wild-type but not by mutant Nm23-H1 expression. Reduced expression of these genes coincident with elevated Nm23-H1 expression was observed in human breast tumor cohorts, a panel of breast carcinoma cell lines, and hepatocellular carcinomas from control versus Nm23-M1 knockout mice. The functional significance of the down-regulated genes was assessed by transfection and in vitro motility assays. Only EDG2 overexpression significantly restored motility to Nm23-H1–suppressed cancer cells, enhancing motility by 60-fold in these cells. In addition, silencing EDG2 expression with small interfering RNA reduced the motile phenotype of metastatic breast cancer cells. These data suggest that Nm23-H1 suppresses metastasis, at least in part, through down-regulation of EDG2 expression. [Cancer Res 2007;67(15):7238–46]
- Published
- 2023
24. Supplementary Table 1 from Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Author
-
Patricia S. Steeg, Kenneth Aldape, Seth M. Steinberg, Alexander O. Vortmeyer, Douglas Halverson, Lionel Feigenbaum, Eleazar Vega-Valle, Raffael Kurek, Andreas M. Stark, Robert J. Weil, Toshiyuki Yoneda, Jeanne M. Herring, Julie L. Bronder, and Diane Palmieri
- Abstract
Supplementary Table 1 from Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Published
- 2023
25. Supplementary Methods from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 75K
- Published
- 2023
26. Supplementary Figure 2 from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 5.8MB
- Published
- 2023
27. Supplementary Table 2 from Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Author
-
Patricia S. Steeg, Kenneth Aldape, Seth M. Steinberg, Alexander O. Vortmeyer, Douglas Halverson, Lionel Feigenbaum, Eleazar Vega-Valle, Raffael Kurek, Andreas M. Stark, Robert J. Weil, Toshiyuki Yoneda, Jeanne M. Herring, Julie L. Bronder, and Diane Palmieri
- Abstract
Supplementary Table 2 from Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Published
- 2023
28. Supplementary Figure 5 from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 1.7MB
- Published
- 2023
29. Supplementary Figure 2 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Author
-
Patricia S. Steeg, Paul S. Meltzer, Marie-Lise Lacombe, William G. Stetler-Stevenson, Diane Palmieri, Sandrine Arnaud-Dabernat, Tara K. Maga, Sylvie Dumont, Mathieu Boissan, Abdel G. Elkahloun, Jong Heun Lee, and Christine E. Horak
- Abstract
Supplementary Figure 2 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Published
- 2023
30. Supplementary Figure 1 from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 954K
- Published
- 2023
31. Supplementary Figure 1 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Author
-
Patricia S. Steeg, Paul S. Meltzer, Marie-Lise Lacombe, William G. Stetler-Stevenson, Diane Palmieri, Sandrine Arnaud-Dabernat, Tara K. Maga, Sylvie Dumont, Mathieu Boissan, Abdel G. Elkahloun, Jong Heun Lee, and Christine E. Horak
- Abstract
Supplementary Figure 1 from Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Published
- 2023
32. Supplementary Figure 4 from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 4.9MB
- Published
- 2023
33. Supplementary Figure 3 from Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, S. Patricia Becerra, Diane Palmieri, Kevin Camphausen, Mary M. Herman, José D. Paltán-Ortiz, Seth M. Steinberg, David J. Liewehr, Yeva Snitkovsky, Yongzhen Qian, Ira Gordon, Christian Graves, Monika Deshpande, Preeti Subramanian, and Daniel P. Fitzgerald
- Abstract
PDF file - 1.3MB
- Published
- 2023
34. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth
- Author
-
Kenneth Ellis, Min Li, Joseph H. McCarty, Dihua Yu, Chenyu Zhang, Qingling Zhang, Hai Wang, Mujeeburahiman Cheerathodi, Aysegul A. Sahin, Sunil R. Lakhani, Xiao Wang, Wen Chien Huang, Diane Palmieri, Suyun Huang, Patricia S. Steeg, Siyuan Zhang, Kenneth Aldape, Jodi M. Saunus, Frank J. Lowery, Lin Zhang, Ping Li, and Jun Yao
- Subjects
Male ,Down-Regulation ,Exosomes ,Exosome ,Article ,Metastasis ,Evolution, Molecular ,Mice ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Gene silencing ,PTEN ,Genes, Tumor Suppressor ,Gene Silencing ,RNA, Messenger ,Chemokine CCL2 ,Cell Proliferation ,Tumor microenvironment ,Multidisciplinary ,biology ,Brain Neoplasms ,Tumor Suppressor Proteins ,Calcium-Binding Proteins ,Microfilament Proteins ,PTEN Phosphohydrolase ,Brain ,medicine.disease ,Adaptation, Physiological ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Astrocytes ,Immunology ,Cancer cell ,Cancer research ,biology.protein ,Female ,Brain metastasis ,Astrocyte - Abstract
The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.
- Published
- 2015
35. Analysis of radiation therapy in a model of triple-negative breast cancer brain metastasis
- Author
-
Emily Hua, Kevin Camphausen, Sudhanshu Shukla, DeeDee Smart, Alejandra Garcia-Glaessner, Patricia S. Steeg, Brunilde Gril, Heather A. Cameron, Sarah J.E. Wong-Goodrich, Tiffany Lyle, Diane Palmieri, and Tamalee Kramp
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Fluorescent Antibody Technique ,Mice, Nude ,Triple Negative Breast Neoplasms ,Radiation Tolerance ,Article ,Metastasis ,Mice ,Breast cancer ,Cell Line, Tumor ,Radioresistance ,medicine ,Animals ,Humans ,Radiosensitivity ,Brain Neoplasms ,business.industry ,Cancer ,Radiotherapy Dosage ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Radiation therapy ,Oncology ,Cancer research ,Female ,Cranial Irradiation ,business ,Ex vivo ,Brain metastasis - Abstract
Most cancer patients with brain metastases are treated with radiation therapy, yet this modality has not yet been meaningfully incorporated into preclinical experimental brain metastasis models. We applied two forms of whole brain radiation therapy (WBRT) to the brain-tropic 231-BR experimental brain metastasis model of triple-negative breast cancer. When compared to sham controls, WBRT as 3 Gy × 10 fractions (3 × 10) reduced the number of micrometastases and large metastases by 87.7 and 54.5 %, respectively (both p < 0.01); whereas a single radiation dose of 15 Gy × 1 (15 × 1) was less effective, reducing metastases by 58.4 % (p < 0.01) and 47.1 % (p = 0.41), respectively. Neuroinflammation in the adjacent brain parenchyma was due solely to a reaction from metastases, and not radiotherapy, while adult neurogenesis in brains was adversely affected following both radiation regimens. The nature of radiation resistance was investigated by ex vivo culture of tumor cells that survived initial WBRT (“Surviving” cultures). The Surviving cultures surprisingly demonstrated increased radiosensitivity ex vivo. In contrast, re-injection of Surviving cultures and re-treatment with a 3 × 10 WBRT regimen significantly reduced the number of large and micrometastases that developed in vivo, suggesting a role for the microenvironment. Micrometastases derived from tumor cells surviving initial 3 × 10 WBRT demonstrated a trend toward radioresistance upon repeat treatment (p = 0.09). The data confirm the potency of a fractionated 3 × 10 WBRT regimen and identify the brain microenvironment as a potential determinant of radiation efficacy. The data also nominate the Surviving cultures as a potential new translational model for radiotherapy.
- Published
- 2015
36. Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner
- Author
-
Brunilde Gril, Stephan Woditschka, Diane Palmieri, Katarzyna Sosińska-Mielcarek, P. S. Steeg, David J. Liewehr, Stephen M. Hewitt, Emily Hua, Seth M. Steinberg, W. Biernat, Andreas M. Stark, Renata Duchnowska, Jacek Jassem, and Yongzhen Qian
- Subjects
Oncology ,Cancer Research ,Pathology ,medicine.medical_specialty ,Dacarbazine ,Mice, Nude ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Kaplan-Meier Estimate ,Article ,Drug Administration Schedule ,Mice ,Breast cancer ,Cell Line, Tumor ,Internal medicine ,Temozolomide ,medicine ,Animals ,Humans ,Antineoplastic Agents, Alkylating ,DNA Modification Methylases ,Dose-Response Relationship, Drug ,Brain Neoplasms ,business.industry ,Tumor Suppressor Proteins ,Cancer ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Primary tumor ,DNA Repair Enzymes ,Treatment Outcome ,MCF-7 Cells ,Female ,RNA Interference ,business ,Brain metastasis ,medicine.drug - Abstract
Purpose: Brain metastases of breast cancer cause neurocognitive damage and are incurable. We evaluated a role for temozolomide in the prevention of brain metastases of breast cancer in experimental brain metastasis models. Experimental Design: Temozolomide was administered in mice following earlier injection of brain-tropic HER2–positive JIMT-1-BR3 and triple-negative 231-BR-EGFP sublines, the latter with and without expression of O6-methylguanine-DNA methyltransferase (MGMT). In addition, the percentage of MGMT-positive tumor cells in 62 patient-matched sets of breast cancer primary tumors and resected brain metastases was determined immunohistochemically. Results: Temozolomide, when dosed at 50, 25, 10, or 5 mg/kg, 5 days per week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cells. At a 1 mg/kg dose, temozolomide prevented 68% of large brain metastases, and was ineffective at a dose of 0.5 mg/kg. When the 50 mg/kg dose was administered beginning on days 18 or 24, temozolomide efficacy was reduced or absent. Temozolomide was ineffective at preventing brain metastases in MGMT-transduced 231-BR-EGFP and MGMT-expressing JIMT-1-BR3 sublines. In 62 patient-matched sets of primary breast tumors and resected brain metastases, 43.5% of the specimens had concordant low MGMT expression, whereas in another 14.5% of sets high MGMT staining in the primary tumor corresponded with low staining in the brain metastasis. Conclusions: Temozolomide profoundly prevented the outgrowth of experimental brain metastases of breast cancer in an MGMT-dependent manner. These data provide compelling rationale for investigating the preventive efficacy of temozolomide in a clinical setting. Clin Cancer Res; 20(10); 2727–39. ©2014 AACR.
- Published
- 2014
37. Abstract P6-11-04: Profound prevention of experimental brain metastases of breast cancer by temozolomide in a MGMT-dependent manner
- Author
-
Diane Palmieri, Wojciech Biernat, Yongzhen Qian, Seth M. Steinberg, P. S. Steeg, Stephen M. Hewitt, Andreas M. Stark, Jacek Jassem, K Sosinska-Mielcarek, Renata Duchnowska, Brunilde Gril, Emily Hua, and David J. Liewehr
- Subjects
Oncology ,Cancer Research ,Pathology ,medicine.medical_specialty ,Temozolomide ,Dependent manner ,business.industry ,medicine.medical_treatment ,Central nervous system ,Cancer ,medicine.disease ,medicine.anatomical_structure ,Breast cancer ,Cell culture ,Internal medicine ,medicine ,business ,Saline ,Brain metastasis ,medicine.drug - Abstract
Purpose: Brain metastases of breast cancer cause neurocognitive damage and are incurable. We evaluated in experimental brain metastasis model a role of temozolomide, an oral brain permeable alkylating agent characterized by significant uptake in the central nervous system, in the prevention of brain metastases of breast cancer. Material and methods: To assess preventive role of temozolomide, mice were inoculated with 175,000 triple-negative 231-BR-EGFP cells in 0.1 mL PBS in the left ventricle of the heart. Three days after tumor cell inoculation, mice were randomized to temozolomide at the dose of 50 mg/kg delivered by oral gavage in saline, 5 days a week for 4 weeks, or vehicle. Subsequent experiments used temozolomide doses of 25, 10, 5, 1 and 0.5 mg/kg. To evaluate the efficacy of temozolomide in treating established BM, mice received temozolomide (50 mg/kg) beginning on either day 18 or day 24 post-injection of 231-BR-EGFR cells, 5 days a week for two and one week, respectively. To investigate the impact of temozolomide on survival, mice injected with 231-BR-EGFP cells were randomized to vehicle, temozolomide on days 3-14, or temozolomide on days 17-28 post-injection, per the schedule described above. To determine the functional contribution of MGMT expression in the BM preventive model, similar experiments were performed using 231-BR-EGFP cells with induced MGMT expression, and MGMT-positive Jimt-1 cells. Metastases were counted in step sections of one hemisphere of each brain. Additionally, the percentage of MGMT-positive tumor cells in 62 patient-matched sets of breast cancer primary tumors and resected brain metastases was determined immunohistochemically. Results: Temozolomide, when dosed at 50, 25, 10 or 5 mg/kg, 5 days/week, beginning 3 days after inoculation, completely prevented the formation of experimental brain metastases from MGMT-negative 231-BR-EGFP cell line. At a 1 mg/kg dose, temozolomide prevented 68% of large brain metastases, and was ineffective at a dose of 0.5 mg/kg. When the 50 mg/kg dose was administered beginning on days 18 or 24, temozolomide efficacy was reduced or absent. Both schedules of temozolomide (days 3-14 and days 17-28) significantly increased survival (P = .0003 by long-rank test). Earlier administration of temozolomide resulted in long term survival of 6 and 2 out of 10 mice, respectively; a significant difference compared to vehicle (P < .0001 and .0003, respectively).Temozolomide was ineffective at preventing brain metastases in the MGMT-positive 231-BR-EGFP and Jimt-BR3 sublines. In 62 patient-matched sets of primary breast tumors and resected brain metastases 43.5% of the specimens had concordant low MGMT expression, while in another 14.5% sets high MGMT staining in the primary tumor corresponded with low staining in the brain metastasis. Conclusions: Temozolomide profoundly prevents the outgrowth of experimental brain metastases of breast cancer in a MGMT-dependent manner. The majority of patients had low MGMT expressing brain metastases. These data provide a compelling rationale for investigating preventive efficacy of temozolomide in high-risk advanced breast cancer patients. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P6-11-04.
- Published
- 2013
38. Effect of Inhibition of the Lysophosphatidic Acid Receptor 1 on Metastasis and Metastatic Dormancy in Breast Cancer
- Author
-
Joji Nakayama, Patricia S. Steeg, Diane Palmieri, Seth M. Steinberg, Maximilien Murone, Fernando Vidal-Vanaclocha, Mary Albaugh, Jean-Claude Marshall, Maryse Barbier, David J. Liewehr, Joshua Collins, and Christine E. Horak
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,MAP Kinase Signaling System ,Blotting, Western ,Mice, Nude ,Antineoplastic Agents ,Biology ,Article ,Gene Expression Regulation, Enzymologic ,Metastasis ,Small hairpin RNA ,Mice ,Random Allocation ,eIF-2 Kinase ,Western blot ,Cell Line, Tumor ,medicine ,Animals ,RNA, Small Interfering ,Receptors, Lysophosphatidic Acid ,Receptor ,Cell Proliferation ,Analysis of Variance ,Mice, Inbred BALB C ,medicine.diagnostic_test ,Cell growth ,Kinase ,Liver Neoplasms ,Mammary Neoplasms, Experimental ,Isoxazoles ,NM23 Nucleoside Diphosphate Kinases ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Blot ,Ki-67 Antigen ,Oncology ,Cancer research ,Female ,Propionates - Abstract
Background Previous studies identified the human nonmetastatic gene 23 (NME1, hereafter Nm23-H1) as the first metastasis suppressor gene. An inverse relationship between Nm23-H1 and expression of lysophosphatidic acid receptor 1 gene (LPAR1, also known as EDG2 or hereafter LPA1) has also been reported. However, the effects of LPA1 inhibition on primary tumor size, metastasis, and metastatic dormancy have not been investigated. Methods The LPA1 inhibitor Debio-0719 or LPA1 short hairpinned RNA (shRNA) was used. Primary tumor size and metastasis were investigated using the 4T1 spontaneous metastasis mouse model and the MDA-MB-231T experimental metastasis mouse model (n = 13 mice per group). Proliferation and p38 intracellular signaling in tumors and cell lines were determined by immunohistochemistry and western blot to investigate the effects of LPA1 inhibition on metastatic dormancy. An analysis of variance-based two-tailed t test was used to determine a statistically significant difference between treatment groups. Results In the 4T1 spontaneous metastasis mouse model, Debio-0719 inhibited the metastasis of 4T1 cells to the liver (mean = 25.2 liver metastases per histologic section for vehicle-treated mice vs 6.8 for Debio-0719-treated mice, 73.0% reduction, P < .001) and lungs (mean = 6.37 lesions per histologic section for vehicle-treated mice vs 0.73 for Debio-0719-treated mice, 88.5% reduction, P < .001), with no effect on primary tumor size. Similar results were observed using the MDA-MB-231T experimental pulmonary metastasis mouse model. LPA1 shRNA also inhibited metastasis but did not affect primary tumor size. In 4T1 metastases, but not primary tumors, expression of the proliferative markers Ki67 and pErk was reduced by Debio-0719, and phosphorylation of the p38 stress kinase was increased, indicative of metastatic dormancy. Conclusion The data identify Debio-0719 as a drug candidate with metastasis suppressor activity, inducing dormancy at secondary tumor sites
- Published
- 2012
39. Opposing Effects of Pigment Epithelium–Derived Factor on Breast Cancer Cell versus Neuronal Survival: Implication for Brain Metastasis and Metastasis-Induced Brain Damage
- Author
-
Patricia S. Steeg, Mary M. Herman, Yeva Snitkovsky, S. Patricia Becerra, Seth M. Steinberg, Preeti Subramanian, Ira K. Gordon, Monika Deshpande, Christian A. Graves, Yongzhen Qian, Daniel P. Fitzgerald, Kevin Camphausen, Diane Palmieri, David J. Liewehr, and Jose D. Paltán-Ortiz
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Fluorescent Antibody Technique ,Apoptosis ,Breast Neoplasms ,Brain damage ,Biology ,Article ,Metastasis ,Mice ,Breast cancer ,PEDF ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Nerve Growth Factors ,Neoplasm Metastasis ,Eye Proteins ,Serpins ,Cell Proliferation ,Neurons ,Cancer ,Human brain ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Cancer research ,Female ,medicine.symptom ,Brain metastasis - Abstract
Brain metastases are a significant cause of morbidity and mortality for patients with cancer, yet preventative and therapeutic options remain an unmet need. The cytokine pigment epithelium–derived factor (PEDF) is downregulated in resected human brain metastases of breast cancer compared with primary breast tumors, suggesting that restoring its expression might limit metastatic spread. Here, we show that outgrowth of large experimental brain metastases from human 231-BR or murine 4T1-BR breast cancer cells was suppressed by PEDF expression, as supported by in vitro analyses as well as direct intracranial implantation. Notably, the suppressive effects of PEDF were not only rapid but independent of the effects of this factor on angiogenesis. Paralleling its cytotoxic effects on breast cancer cells, PEDF also exerted a prosurvival effect on neurons that shielded the brain from tumor-induced damage, as indicated by a relative 3.5-fold reduction in the number of dying neurons adjacent to tumors expressing PEDF. Our findings establish PEDF as both a metastatic suppressor and a neuroprotectant in the brain, highlighting its role as a double agent in limiting brain metastasis and its local consequences. Cancer Res; 72(1); 144–53. ©2012 AACR.
- Published
- 2012
40. Lapatinib Distribution in HER2 Overexpressing Experimental Brain Metastases of Breast Cancer
- Author
-
Stephen D. Rubin, Vinay Rudraraju, Kunal S Taskar, Emily Hua, Diane Palmieri, Ramakrishna Samala, Brunilde Gril, Stephen Castellino, Paul R. Lockman, Julie A. Lockman, Patricia S. Steeg, Quentin R. Smith, Joseph W. Polli, Helen R. Thorsheim, and Rajendar K. Mittapalli
- Subjects
Oncology ,medicine.medical_specialty ,Receptor, ErbB-2 ,Administration, Oral ,Pharmaceutical Science ,Antineoplastic Agents ,Breast Neoplasms ,Lapatinib ,Article ,Mice ,Breast cancer ,Oral administration ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Distribution (pharmacology) ,Neoplasm ,Pharmacology (medical) ,skin and connective tissue diseases ,Receptor ,Pharmacology ,Brain Neoplasms ,business.industry ,Organic Chemistry ,Brain ,medicine.disease ,Up-Regulation ,Drug Resistance, Neoplasm ,Injections, Intravenous ,Quinazolines ,Molecular Medicine ,Female ,business ,Ex vivo ,Biotechnology ,Brain metastasis ,medicine.drug - Abstract
Lapatinib, a small molecule EGFR/HER2 inhibitor, partially inhibits the outgrowth of HER2+ brain metastases in preclinical models and in a subset of CNS lesions in clinical trials of HER2+ breast cancer. We investigated the ability of lapatinib to reach therapeutic concentrations in the CNS following 14C-lapatinib administration (100 mg/kg p.o. or 10 mg/kg, i.v.) to mice with MDA-MD-231-BR-HER2 brain metastases of breast cancer. Drug concentrations were determined at differing times after administration by quantitative autoradiography and chromatography. 14C-Lapatinib concentration varied among brain metastases and correlated with altered blood-tumor barrier permeability. On average, brain metastasis concentration was 7–9-fold greater than surrounding brain tissue at 2 and 12 h after oral administration. However, average lapatinib concentration in brain metastases was still only 10–20% of those in peripheral metastases. Only in a subset of brain lesions (17%) did lapatinib concentration approach that of systemic metastases. No evidence was found of lapatinib resistance in tumor cells cultured ex vivo from treated brains. Results show that lapatinib distribution to brain metastases of breast cancer is partially restricted and blood-tumor barrier permeability is a key component of lapatinib therapeutic efficacy which varies between tumors.
- Published
- 2011
41. Notch1 Inhibition Alters the CD44hi/CD24lo Population and Reduces the Formation of Brain Metastases from Breast Cancer
- Author
-
Carmen Simedrea, Diane Palmieri, Patricia M. McGowan, Paula J. Foster, Emeline J. Ribot, Patricia S. Steeg, Alison L. Allan, and Ann F. Chambers
- Subjects
Cancer Research ,Population ,Mice, Nude ,Breast Neoplasms ,Biology ,Article ,Mice ,Breast cancer ,Cancer stem cell ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Receptor, Notch1 ,education ,Molecular Biology ,education.field_of_study ,Brain Neoplasms ,CD24 ,CD44 ,CD24 Antigen ,Cancer ,Transfection ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Hyaluronan Receptors ,Oncology ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Female ,Amyloid Precursor Protein Secretases ,Brain metastasis - Abstract
Brain metastasis from breast cancer is an increasingly important clinical problem. Here we assessed the role of CD44hi/CD24lo cells and pathways that regulate them, in an experimental model of brain metastasis. Notch signaling (mediated by γ-secretase) has been shown to contribute to maintenance of the cancer stem cell (CSC) phenotype. Cells sorted for a reduced stem-like phenotype had a reduced ability to form brain metastases compared with unsorted or CD44hi/CD24lo cells (P < 0.05; Kruskal–Wallis). To assess the effect of γ-secretase inhibition, cells were cultured with DAPT and the CD44/CD24 phenotypes quantified. 231-BR cells with a CD44hi/CD24lo phenotype was reduced by about 15% in cells treated with DAPT compared with DMSO-treated or untreated cells (P = 0.001, ANOVA). In vivo, mice treated with DAPT developed significantly fewer micro- and macrometastases compared with vehicle treated or untreated mice (P = 0.011, Kruskal–Wallis). Notch1 knockdown reduced the expression of CD44hi/CD24lo phenotype by about 20%. In vitro, Notch1 shRNA resulted in a reduction in cellular growth at 24, 48, and 72 hours time points (P = 0.033, P = 0.002, and P = 0.009, ANOVA) and about 60% reduction in Matrigel invasion was observed (P < 0.001, ANOVA). Cells transfected with shNotch1 formed significantly fewer macrometastases and micrometastases compared with scrambled shRNA or untransfected cells (P < 0.001; Kruskal–Wallis). These data suggest that the CSC phenotype contributes to the development of brain metastases from breast cancer, and this may arise in part from increased Notch activity. Mol Cancer Res; 9(7); 834–44. ©2011 AACR.
- Published
- 2011
42. Abstract B51: Advancing cancer health disparities research through the Beau Biden Cancer MoonshotSM
- Author
-
L. Michelle Bennett, Amy E. Kennedy, Diane Palmieri, and Sonia Rosenfield
- Subjects
Government ,medicine.medical_specialty ,Cancer prevention ,020205 medical informatics ,Epidemiology ,02 engineering and technology ,Biobank ,Pediatric cancer ,Health equity ,Transformative research ,Underserved Population ,Oncology ,Family medicine ,Political science ,0202 electrical engineering, electronic engineering, information engineering ,medicine ,Socioeconomic status - Abstract
Established in 2016, the Beau Biden Cancer MoonshotSM was created with the goal of accelerating ten years' worth of progress in cancer prevention, diagnosis, and treatment in just five years. A group of research experts, patient advocates, and government and private sector representatives was convened to form a Blue Ribbon Panel (BRP) to develop recommendations for accelerating research progress. Working groups in seven key areas focused on identifying opportunities and impediments with input from stakeholders. The BRP released a report detailing 10 transformative research recommendations that, if implemented, could reduce the burden of cancer in the United States. These included the areas of clinical trials, data sharing, cancer immunology, implementation science, pediatric cancer, precision prevention and early detection, and tumor evolution and progression. Reducing cancer disparities is a cross-cutting theme at the forefront of the BRP recommendations. The BRP recommendations consider disparities across a range of research areas, including reducing barriers to participation in and access to clinical trials through the engagement of diverse populations; expanding the use of proven prevention and early-detection strategies to all populations, including urban poor and rural communities; and educating and increasing access to genetic testing and counseling for those at high risk for inherited cancers regardless of socioeconomic or insurance status. Recommendations involving the collection of biospecimens and the expansion of biobanks and databases also have emphasis on including as many diverse patients and samples as possible. When Congress passed the 21st Century Cures Act in December 2016, $1.8 billion in funding was authorized for the Cancer Moonshot over the next 7 years. An initial $300 million was appropriated for fiscal year 2017. NCI is implementing the recommendations through new research initiatives designed to accelerate our understanding of cancer. The Moonshot Funding Opportunity Announcements (FOAs) will encourage applicants to consider integrating health disparities into their research efforts. Some examples include access to care through an emerging technologies effort, dissemination and implementation of effective strategies to reduce health disparities and improve quality in underserved populations, and improving smoking-cessation efforts in socioeconomically disadvantaged populations. By integrating health disparities into the objectives of the Cancer Moonshot, there is an opportunity to reduce cancer disparities across the cancer continuum in an accelerated time frame. Citation Format: Sonia Rosenfield, Amy E. Kennedy, Diane Palmieri, L. Michelle Bennett. Advancing cancer health disparities research through the Beau Biden Cancer MoonshotSM [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B51.
- Published
- 2018
43. Vorinostat Inhibits Brain Metastatic Colonization in a Model of Triple-Negative Breast Cancer and Induces DNA Double-Strand Breaks
- Author
-
Julie A. Gaasch, Emily Hua, Lionel Feigenbaum, Patricia S. Steeg, Quentin R. Smith, Natasha M. Flores, Eleazar Vega-Valle, Kunal S Taskar, Kaci A. Bohn, Diane Palmieri, Jeanne M. Herring, Helen R. Thorsheim, Seth M. Steinberg, Paul R. Lockman, Paul S. Meltzer, David J. Liewehr, Elizabeth Hargrave, Vinay Rudraraju, Matthew Johnson, John F. Reilly, Kevin Camphausen, Victoria M. Richon, Robert L. Walker, Yongzhen Qian, Sean Davis, Rajendar K. Mittapalli, Fancy Thomas, and Julie L. Bronder
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.drug_class ,Mice, Nude ,Antineoplastic Agents ,Breast Neoplasms ,Biology ,Hydroxamic Acids ,Histone Deacetylases ,Article ,Metastasis ,Rats, Sprague-Dawley ,Mice ,Breast cancer ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,DNA Breaks, Double-Stranded ,Enzyme Inhibitors ,Vorinostat ,Triple-negative breast cancer ,Mice, Inbred BALB C ,Brain Neoplasms ,Carcinoma ,Histone deacetylase inhibitor ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,Histone Deacetylase Inhibitors ,Oncology ,Blood-Brain Barrier ,Cancer research ,Female ,Breast disease ,medicine.drug ,Brain metastasis - Abstract
Purpose: As chemotherapy and molecular therapy improve the systemic survival of breast cancer patients, the incidence of brain metastases increases. Few therapeutic strategies exist for the treatment of brain metastases because the blood-brain barrier severely limits drug access. We report the pharmacokinetic, efficacy, and mechanism of action studies for the histone deactylase inhibitor vorinostat (suberoylanilide hydroxamic acid) in a preclinical model of brain metastasis of triple-negative breast cancer. Experimental Design: The 231-BR brain trophic subline of the MDA-MB-231 human breast cancer cell line was injected into immunocompromised mice for pharmacokinetic and metastasis studies. Pharmacodynamic studies compared histone acetylation, apoptosis, proliferation, and DNA damage in vitro and in vivo. Results: Following systemic administration, uptake of [14C]vorinostat was significant into normal rodent brain and accumulation was up to 3-fold higher in a proportion of metastases formed by 231-BR cells. Vorinostat prevented the development of 231-BR micrometastases by 28% (P = 0.017) and large metastases by 62% (P < 0.0001) compared with vehicle-treated mice when treatment was initiated on day 3 post-injection. The inhibitory activity of vorinostat as a single agent was linked to a novel function in vivo: induction of DNA double-strand breaks associated with the down-regulation of the DNA repair gene Rad52. Conclusions: We report the first preclinical data for the prevention of brain metastasis of triple-negative breast cancer. Vorinostat is brain permeable and can prevent the formation of brain metastases by 62%. Its mechanism of action involves the induction of DNA double-strand breaks, suggesting rational combinations with DNA active drugs or radiation. (Clin Cancer Res 2009;15(19):6148–57)
- Published
- 2009
44. Uptake of ANG1005, A Novel Paclitaxel Derivative, Through the Blood-Brain Barrier into Brain and Experimental Brain Metastases of Breast Cancer
- Author
-
Julie A. Gaasch, Diane Palmieri, Paul R. Lockman, Helen R. Thorsheim, Patricia S. Steeg, Satyanarayana Goda, Kunal S Taskar, Vinay Rudraraju, Quentin R. Smith, Rajendar K. Mittapalli, and Fancy Thomas
- Subjects
Pathology ,medicine.medical_specialty ,Paclitaxel ,Brain tumor ,Mice, Nude ,Pharmaceutical Science ,Breast Neoplasms ,Blood–brain barrier ,Article ,Metastasis ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,Pharmacology (medical) ,Chromatography, High Pressure Liquid ,Pharmacology ,Brain Neoplasms ,business.industry ,Organic Chemistry ,Cancer ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Rats ,medicine.anatomical_structure ,ANG1005 ,chemistry ,Blood-Brain Barrier ,Nanoparticles for drug delivery to the brain ,Cancer research ,Molecular Medicine ,Female ,Breast disease ,Peptides ,business ,Biotechnology - Abstract
We evaluated the uptake of angiopep-2 paclitaxel conjugate, ANG1005, into brain and brain metastases of breast cancer in rodents. Most anticancer drugs show poor delivery to brain tumors due to limited transport across the blood-brain barrier (BBB). To overcome this, a 19-amino acid peptide (angiopep-2) was developed that binds to low density lipoprotein receptor-related protein (LRP) receptors at the BBB and has the potential to deliver drugs to brain by receptor-mediated transport.The transfer coefficient (K(in)) for brain influx was measured by in situ rat brain perfusion. Drug distribution was determined at 30 min after i.v. injection in mice bearing intracerebral MDA-MB-231BR metastases of breast cancer.The BBB K(in) for (125)I-ANG1005 uptake (7.3 +/- 0.2 x 10(-3) mL/s/g) exceeded that for (3)H-paclitaxel (8.5 +/- 0.5 x 10(-5)) by 86-fold. Over 70% of (125)I-ANG1005 tracer stayed in brain after capillary depletion or vascular washout. Brain (125)I-ANG1005 uptake was reduced by unlabeled angiopep-2 vector and by LRP ligands, consistent with receptor transport. In vivo uptake of (125)I-ANG1005 into vascularly corrected brain and brain metastases exceeded that of (14)C-paclitaxel by 4-54-fold.The results demonstrate that ANG1005 shows significantly improved delivery to brain and brain metastases of breast cancer compared to free paclitaxel.
- Published
- 2009
45. Analyses of Resected Human Brain Metastases of Breast Cancer Reveal the Association between Up-Regulation of Hexokinase 2 and Poor Prognosis
- Author
-
Gary Davis, Emily Hua, Julie L. Bronder, Sean Davis, Patricia S. Steeg, Paul S. Meltzer, Raffael Kurek, Robert J. Weil, Kenneth Aldape, S. Martin Shreeve, Daniel P. Fitzgerald, Maria J. Merino, Andreas M. Stark, Diane Palmieri, Seth M. Steinberg, and H. Maximilian Mehdorn
- Subjects
Cancer Research ,Cell Survival ,Breast Neoplasms ,Biology ,Polymerase Chain Reaction ,Article ,Metastasis ,Small hairpin RNA ,Adenosine Triphosphate ,PEDF ,Breast cancer ,Cell Line, Tumor ,Hexokinase ,Gene expression ,medicine ,Humans ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Proportional Hazards Models ,Gene knockdown ,Brain Neoplasms ,Reproducibility of Results ,Cancer ,Human brain ,Prognosis ,medicine.disease ,Up-Regulation ,Glucose ,medicine.anatomical_structure ,Oncology ,Gene Knockdown Techniques ,Cancer research ,Female - Abstract
Brain metastases of breast cancer seem to be increasingin incidence as systemic therapy improves. Metastatic disease in the brain is associated with high morbidity and mortality. We present the first gene expression analysis of laser-captured epithelial cells from resected human brain metastases of breast cancer compared with unlinked primary breast tumors. The tumors were matched for histology, tumor-node-metastasis stage, and hormone receptor status. Most differentially expressed genes were down-regulated in the brain metastases, which included, surprisingly, many genes associated with metastasis. Quantitative real-time PCR analysis confirmed statistically significant differences or strong trends in the expression of six genes: BMP1, PEDF, LAMγ3, SIAH, STHMN3, and TSPD2. Hexokinase 2 (HK2) was also of interest because of its increased expression in brain metastases. HK2 is important in glucose metabolism and apoptosis. In agreement with our microarray results, HK2 levels (both mRNA and protein) were elevated in a brain metastatic derivative (231-BR) of the human breast carcinoma cell line MDA-MB-231 relative to the parental cell line (231-P) in vitro. Knockdown of HK2 expression in 231-BR cells using short hairpin RNA reduced cell proliferation when cultures were maintained in glucose-limiting conditions. Finally, HK2 expression was analyzed in a cohort of 123 resected brain metastases of breast cancer. High HK2 expression was significantly associated with poor patient survival after craniotomy (P = 0.028). The data suggest that HK2 overexpression is associated with metastasis to the brain in breast cancer and it may be a therapeutic target. (Mol Cancer Res 2009;7(9):1438–45)
- Published
- 2009
46. Effect of Lapatinib on the Outgrowth of Metastatic Breast Cancer Cells to the Brain
- Author
-
Julie L. Bronder, Brunilde Gril, David J. Liewehr, Maria Merino, Diane Palmieri, Stephen D. Rubin, Patricia S. Steeg, Eleazar Vega-Valle, Jeanne M. Herring, Seth M. Steinberg, and Lionel Feigenbaum
- Subjects
Cancer Research ,Pathology ,Receptor, ErbB-2 ,Mice ,Cell Movement ,Trastuzumab ,Epidermal growth factor receptor ,Phosphorylation ,RNA, Small Interfering ,skin and connective tissue diseases ,Mice, Inbred BALB C ,biology ,Brain Neoplasms ,Immunochemistry ,Articles ,Metastatic breast cancer ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Oncology ,Research Design ,Female ,Breast disease ,Signal Transduction ,medicine.drug ,medicine.medical_specialty ,Immunoblotting ,Antineoplastic Agents ,Breast Neoplasms ,Transfection ,Lapatinib ,Breast cancer ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Gene Silencing ,Protein Kinase Inhibitors ,Cell Proliferation ,Analysis of Variance ,business.industry ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Quinazolines ,Cancer research ,biology.protein ,business ,Brain metastasis - Abstract
The brain is increasingly being recognized as a sanctuary site for metastatic tumor cells in women with HER2-overexpressing breast cancer who receive trastuzumab therapy. There are no approved or widely accepted treatments for brain metastases other than steroids, cranial radiotherapy, and surgical resection. We examined the efficacy of lapatinib, an inhibitor of the epidermal growth factor receptor (EGFR) and HER2 kinases, for preventing the outgrowth of breast cancer cells in the brain in a mouse xenograft model of brain metastasis.EGFR-overexpressing MDA-MB-231-BR (231-BR) brain-seeking breast cancer cells were transfected with an expression vector that contained or lacked the HER2 cDNA and used to examine the effect of lapatinib on the activation (ie, phosphorylation) of cell signaling proteins by immunoblotting, on cell growth by the tetrazolium salt 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, and on cell migration using a Boyden chamber assay. The outgrowth of large (ie,50 microm(2)) and micrometastases was counted in brain sections from nude mice that had been injected into the left cardiac ventricle with 231-BR cells and, beginning 5 days later, treated by oral gavage with lapatinib or vehicle (n = 22-26 mice per treatment group). All statistical tests were two-sided.In vitro, lapatinib inhibited the phosphorylation of EGFR, HER2, and downstream signaling proteins; cell proliferation; and migration in 231-BR cells (both with and without HER2). Among mice injected with 231-BR-vector cells, those treated with 100 mg lapatinib/kg body weight had 54% fewer large metastases 24 days after starting treatment than those treated with vehicle (mean number of large metastases per brain section: 1.56 vs 3.36, difference = 1.80, 95% confidence interval [CI] = 0.92 to 2.68, P.001), whereas treatment with 30 mg lapatinib/kg body weight had no effect. Among mice injected with 231-BR-HER2 cells, those treated with either dose of lapatinib had 50%-53% fewer large metastases than those treated with vehicle (mean number of large metastases per brain section, 30 mg/kg vs vehicle: 3.21 vs 6.83, difference = 3.62, 95% CI = 2.30 to 4.94, P.001; 100 mg/kg vs vehicle: 3.44 vs 6.83, difference = 3.39, 95% CI = 2.08 to 4.70, P.001). Immunohistochemical analysis revealed reduced phosphorylation of HER2 in 231-BR-HER2 cell-derived brain metastases from mice treated with the higher dose of lapatinib compared with 231-BR-HER2 cell-derived brain metastases from vehicle-treated mice (P.001).Lapatinib is the first HER2-directed drug to be validated in a preclinical model for activity against brain metastases of breast cancer.
- Published
- 2008
47. Nm23-H1 Suppresses Tumor Cell Motility by Down-regulating the Lysophosphatidic Acid Receptor EDG2
- Author
-
Tara Maga, William G. Stetler-Stevenson, Diane Palmieri, Christine E. Horak, Mathieu Boissan, Patricia S. Steeg, Marie-Lise Lacombe, Jong Heun Lee, Abdel G. Elkahloun, Paul S. Meltzer, Sylvie Dumont, and Sandrine Arnaud-Dabernat
- Subjects
Cancer Research ,Small interfering RNA ,Immunoblotting ,Down-Regulation ,Motility ,Breast Neoplasms ,Biology ,Metastasis ,Cohort Studies ,Immunoenzyme Techniques ,Mice ,Cell Movement ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,RNA, Messenger ,Receptors, Lysophosphatidic Acid ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Regulation of gene expression ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Transfection ,NM23 Nucleoside Diphosphate Kinases ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Drug Combinations ,Metastasis Suppressor Gene ,Oncology ,Nucleoside-Diphosphate Kinase ,Cancer cell ,Cancer research ,Proteoglycans ,Collagen ,Laminin - Abstract
Exogenous overexpression of the metastasis suppressor gene Nm23-H1 reduces the metastatic potential of multiple types of cancer cells and suppresses in vitro tumor cell motility and invasion. Mutational analysis of Nm23-H1 revealed that substitution mutants P96S and S120G did not inhibit motility and invasion. To elucidate the molecular mechanism of Nm23-H1 motility suppression, expression microarray analysis of an MDA-MB-435 cancer cell line overexpressing wild-type Nm23-H1 was done and cross-compared with expression profiles from lines expressing the P96S and S120G mutants. Nine genes, MET, PTN, SMO, FZD1, L1CAM, MMP2, NETO2, CTGF, and EDG2, were down-regulated by wild-type but not by mutant Nm23-H1 expression. Reduced expression of these genes coincident with elevated Nm23-H1 expression was observed in human breast tumor cohorts, a panel of breast carcinoma cell lines, and hepatocellular carcinomas from control versus Nm23-M1 knockout mice. The functional significance of the down-regulated genes was assessed by transfection and in vitro motility assays. Only EDG2 overexpression significantly restored motility to Nm23-H1–suppressed cancer cells, enhancing motility by 60-fold in these cells. In addition, silencing EDG2 expression with small interfering RNA reduced the motile phenotype of metastatic breast cancer cells. These data suggest that Nm23-H1 suppresses metastasis, at least in part, through down-regulation of EDG2 expression. [Cancer Res 2007;67(15):7238–46]
- Published
- 2007
48. Her-2 Overexpression Increases the Metastatic Outgrowth of Breast Cancer Cells in the Brain
- Author
-
Diane Palmieri, Patricia S. Steeg, Kenneth Aldape, Alexander O. Vortmeyer, Toshiyuki Yoneda, Raffael Kurek, Andreas M. Stark, Eleazar Vega-Valle, Jeanne M. Herring, Lionel Feigenbaum, Douglas Halverson, Robert J. Weil, Julie L. Bronder, and Seth M. Steinberg
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Receptor, ErbB-2 ,Transplantation, Heterologous ,Breast Neoplasms ,Transfection ,Metastasis ,Mice ,Breast cancer ,Trastuzumab ,Cell Line, Tumor ,Cell Adhesion ,medicine ,Carcinoma ,Animals ,Humans ,RNA, Messenger ,Epidermal growth factor receptor ,Mice, Inbred BALB C ,biology ,Brain Neoplasms ,business.industry ,Human brain ,medicine.disease ,Primary tumor ,medicine.anatomical_structure ,Oncology ,biology.protein ,Female ,business ,Neoplasm Transplantation ,Brain metastasis ,medicine.drug - Abstract
Retrospective studies of breast cancer patients suggest that primary tumor Her-2 overexpression or trastuzumab therapy is associated with a devastating complication: the development of central nervous system (brain) metastases. Herein, we present Her-2 expression trends from resected human brain metastases and data from an experimental brain metastasis assay, both indicative of a functional contribution of Her-2 to brain metastatic colonization. Of 124 archival resected brain metastases from breast cancer patients, 36.2% overexpressed Her-2, indicating an enrichment in the frequency of tumor Her-2 overexpression at this metastatic site. Using quantitative real-time PCR of laser capture microdissected epithelial cells, Her-2 and epidermal growth factor receptor (EGFR) mRNA levels in a cohort of 12 frozen brain metastases were increased up to 5- and 9-fold, respectively, over those of Her-2–amplified primary tumors. Co-overexpression of Her-2 and EGFR was also observed in a subset of brain metastases. We then tested the hypothesis that overexpression of Her-2 increases the colonization of breast cancer cells in the brain in vivo. A subclone of MDA-MB-231 human breast carcinoma cells that selectively metastasizes to brain (231-BR) overexpressed EGFR; 231-BR cells were transfected with low (4- to 8-fold) or high (22- to 28-fold) levels of Her-2. In vivo, in a model of brain metastasis, low or high Her-2–overexpressing 231-BR clones produced comparable numbers of micrometastases in the brain as control transfectants; however, the Her-2 transfectants yielded 3-fold greater large metastases (>50 μm2; P < 0.001). Our data indicate that Her-2 overexpression increases the outgrowth of metastatic tumor cells in the brain in this model system. [Cancer Res 2007;67(9):4190–8]
- Published
- 2007
49. Alterations in Pericyte Subpopulations Are Associated with Elevated Blood-Tumor Barrier Permeability in Experimental Brain Metastasis of Breast Cancer
- Author
-
Afroz S. Mohammad, Priscilla K. Brastianos, David J. Liewehr, Diane Palmieri, Ewa Izycka-Swieszewska, Naema Nayyar, Seth M. Steinberg, L. Tiffany Lyle, Patricia S. Steeg, Paul R. Lockman, Renata Duchnowska, Emily Hua, Wojciech Kloc, Brunilde Gril, Emily Sechrest, and Chris E. Adkins
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Texas Red ,Breast Neoplasms ,Biology ,Blood–brain barrier ,Inflammatory breast cancer ,Permeability ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Brain Neoplasms ,Brain ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Permeability (electromagnetism) ,Blood-Brain Barrier ,030220 oncology & carcinogenesis ,Desmin ,Female ,Pericyte ,Pericytes ,Brain metastasis - Abstract
Purpose: The blood–brain barrier (BBB) is modified to a blood–tumor barrier (BTB) as a brain metastasis develops from breast or other cancers. We (i) quantified the permeability of experimental brain metastases, (ii) determined the composition of the BTB, and (iii) identified which elements of the BTB distinguished metastases of lower permeability from those with higher permeability. Experimental Design: A SUM190-BR3 experimental inflammatory breast cancer brain metastasis subline was established. Experimental brain metastases from this model system and two previously reported models (triple-negative MDA-231-BR6, HER2+ JIMT-1-BR3) were serially sectioned; low- and high-permeability lesions were identified with systemic 3-kDa Texas Red dextran dye. Adjoining sections were used for quantitative immunofluorescence to known BBB and neuroinflammatory components. One-sample comparisons against a hypothesized value of one were performed with the Wilcoxon signed-rank test. Results: When uninvolved brain was compared with any brain metastasis, alterations in endothelial, pericytic, astrocytic, and microglial components were observed. When metastases with relatively low and high permeability were compared, increased expression of a desmin+ subpopulation of pericytes was associated with higher permeability (231-BR6 P = 0.0002; JIMT-1-BR3 P = 0.004; SUM190-BR3 P = 0.008); desmin+ pericytes were also identified in human craniotomy specimens. Trends of reduced CD13+ pericytes (231-BR6 P = 0.014; JIMT-1-BR3 P = 0.002, SUM190-BR3, NS) and laminin α2 (231-BR6 P = 0.001; JIMT-1-BR3 P = 0.049; SUM190-BR3 P = 0.023) were also observed with increased permeability. Conclusions: We provide the first account of the composition of the BTB in experimental brain metastasis. Desmin+ pericytes and laminin α2 are potential targets for the development of novel approaches to increase chemotherapeutic efficacy. Clin Cancer Res; 22(21); 5287–99. ©2016 AACR.
- Published
- 2015
50. Translational approaches using metastasis suppressor genes
- Author
-
Jong Heun Lee, Patricia S. Steeg, Diane Palmieri, Christine E. Horak, and Douglas Halverson
- Subjects
Histidine Kinase ,Physiology ,Medroxyprogesterone Acetate ,Biology ,Metastasis ,Metastasis Suppression ,Mice ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Metastasis suppressor ,Neoplasm Metastasis ,Mice, Knockout ,Regulation of gene expression ,Cancer ,Cell Biology ,NM23 Nucleoside Diphosphate Kinases ,medicine.disease ,Primary tumor ,Gene Expression Regulation, Neoplastic ,Metastasis Suppressor Gene ,Nucleoside-Diphosphate Kinase ,Mutagenesis, Site-Directed ,Cancer research ,Protein Kinases - Abstract
Cancer metastasis is a significant contributor to breast cancer patient morbidity and mortality. In order to develop new anti-metastatic therapies, we need to understand the biological and biochemical mechanisms of metastasis. Toward these efforts, we and others have studied metastasis suppressor genes, which halt metastasis in vivo without affecting primary tumor growth. The first metastasis suppressor gene identified was nm23, also known as NDP kinase. Nm23 represents the most widely validated metastasis suppressor gene, based on transfection and knock-out mouse strategies. The biochemical mechanism of metastasis suppression via Nm23 is unknown and likely complex. Two potential mechanisms include binding proteins and a histidine kinase activity. Elevation of Nm23 expression in micrometastatic tumor cells may constitute a translational strategy for the limitation of metastatic colonization in high risk cancer patients. To date, medroxyprogesterone acetate (MPA) has been identified as a candidate compound for clinical testing.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.