Sulindac sulfide as a non-immune suppressive γ-secretase modulator to target triple-negative breast cancer
暂无分享,去创建一个
B. Collins-Burow | D. Wyczechowska | C. Hicks | S. Shanthalingam | Fokhrul M. Hossain | Samarpan Majumder | M. Matossian | J. Zabaleta | Lucio Miele | Keli Xu | Kristina Larter | Giulia Monticone | Barbara A. Osborne | Deniz A. Ucar | Yong Ran | Hanh H. Luu | Soroor Heidari | Yaguang Xi | Matthew R. Burow | Luis Del Valle | Jovanny Zabaleta | Todd Golde
[1] Sung-Hak Kim,et al. The oncogenic JAG1 intracellular domain is a transcriptional cofactor that acts in concert with DDX17/SMAD3/TGIF2. , 2022, Cell reports.
[2] A. Boulares,et al. Targeting the Cbl-b-Notch1 axis as a novel immunotherapeutic strategy to boost CD8+ T-cell responses , 2022, Frontiers in Immunology.
[3] P. Marcato,et al. The Prostaglandin E2 Pathway and Breast Cancer Stem Cells: Evidence of Increased Signaling and Potential Targeting , 2022, Frontiers in Oncology.
[4] Jie Xie,et al. Blockade of Cycloxygenase-2 ameliorates sepsis induced immune-suppression by regulating myeloid-derived suppressor cells. , 2022, International immunopharmacology.
[5] N. Donato,et al. Therapeutic inhibition of USP9x-mediated Notch signaling in triple-negative breast cancer , 2021, Proceedings of the National Academy of Sciences.
[6] H. Wathieu,et al. In-depth characterization of a new patient-derived xenograft model for metaplastic breast carcinoma to identify viable biologic targets and patterns of matrix evolution within rare tumor types , 2021, Clinical and Translational Oncology.
[7] S. Beer-Hammer,et al. Myeloid-Derived Suppressor Cells Dampen Airway Inflammation Through Prostaglandin E2 Receptor 4 , 2021, Frontiers in Immunology.
[8] K. Brennan,et al. Notch Signalling in Breast Development and Cancer , 2021, Frontiers in Cell and Developmental Biology.
[9] T. Lee,et al. Cancer Stem Cells: Emerging Key Players in Immune Evasion of Cancers , 2021, Frontiers in Cell and Developmental Biology.
[10] R. Rad,et al. Anti-inflammatory chemoprevention attenuates the phenotype in a mouse model of esophageal adenocarcinoma. , 2021, Carcinogenesis.
[11] F. Feng,et al. γ-Secretase inhibitors for breast cancer and hepatocellular carcinoma: From mechanism to treatment. , 2021, Life sciences.
[12] Xu Zhou,et al. Single-cell RNA-seq dissects the intratumoral heterogeneity of triple-negative breast cancer based on gene regulatory networks , 2021, Molecular therapy. Nucleic acids.
[13] J. Bibb,et al. Gamma secretase inhibitors in cancer: a current perspective on clinical performance. , 2020, The oncologist.
[14] J. Fernández-Piqueras,et al. More Insights on the Use of γ‐Secretase Inhibitors in Cancer Treatment , 2020, The oncologist.
[15] R. Chakrabarti,et al. The many facets of Notch signaling in breast cancer: toward overcoming therapeutic resistance , 2020, Genes & development.
[16] Medhat A. Haroun,et al. Dual targeting of Notch and Wnt/β-catenin pathways: Potential approach in triple-negative breast cancer treatment , 2020, Naunyn-Schmiedeberg's Archives of Pharmacology.
[17] C. Osipo,et al. Notch Signaling in Breast Cancer: A Role in Drug Resistance , 2020, Cells.
[18] E. Martin,et al. Patient-Derived Xenografts as an Innovative Surrogate Tumor Model for the Investigation of Health Disparities in Triple Negative Breast Cancer , 2020, Women's health reports.
[19] X. Bian,et al. Triple-negative breast cancer molecular subtyping and treatment progress , 2020, Breast Cancer Research.
[20] V. Rotello,et al. Triple-Negative Breast Cancer: A Review of Conventional and Advanced Therapeutic Strategies , 2020, International journal of environmental research and public health.
[21] H. Wathieu,et al. Evaluation of deacetylase inhibition in metaplastic breast carcinoma using multiple derivations of preclinical models of a new patient-derived tumor , 2019, bioRxiv.
[22] A. Damjanović,et al. Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer—How We Can Rise to the Challenge , 2019, Cells.
[23] S. Loi,et al. Triple-negative breast cancer: recent treatment advances , 2019, F1000Research.
[24] M. V. Giuli,et al. Notch Signaling Activation as a Hallmark for Triple-Negative Breast Cancer Subtype , 2019, Journal of oncology.
[25] Jeong-Seok Nam,et al. Targeting Cancer Stem Cells in Triple-Negative Breast Cancer , 2019, Cancers.
[26] T. Golde,et al. Individual and combined presenilin 1 and 2 knockouts reveal that both have highly overlapping functions in HEK293T cells , 2019, The Journal of Biological Chemistry.
[27] N. Gassler,et al. Targeting Wnt Signaling via Notch in Intestinal Carcinogenesis , 2019, Cancers.
[28] B. Rowan,et al. Drug resistance profiling of a new triple negative breast cancer patient-derived xenograft model , 2019, BMC Cancer.
[29] Yangqiu Li,et al. Notch inhibition enhances graft‐versus‐leukemia while reducing graft‐versus‐host disease , 2019, European journal of pharmacology.
[30] J. Crabtree,et al. Notch Signaling Regulates Mitochondrial Metabolism and NF-κB Activity in Triple-Negative Breast Cancer Cells via IKKα-Dependent Non-canonical Pathways , 2018, Front. Oncol..
[31] C. Konaka,et al. NSAIDs may prevent EGFR-TKI-related skin rash in non-small cell lung cancer patients . , 2018, International journal of clinical pharmacology and therapeutics.
[32] J. Bluestone,et al. Revisiting IL-2: Biology and therapeutic prospects , 2018, Science Immunology.
[33] S. Winandy,et al. A Review of Notch Processing With New Insights Into Ligand-Independent Notch Signaling in T-Cells , 2018, Front. Immunol..
[34] Johan Hartman,et al. Chemoresistance Evolution in Triple-Negative Breast Cancer Delineated by Single-Cell Sequencing , 2018, Cell.
[35] Yun-jiang Zhou,et al. Effects of sulindac sulfide on proliferation and apoptosis of human breast cancer cell. , 2018, Oncology letters.
[36] Hans Clevers,et al. Organoids in cancer research , 2018, Nature Reviews Cancer.
[37] A. Omar,et al. EP4 as a Therapeutic Target for Aggressive Human Breast Cancer , 2018, International journal of molecular sciences.
[38] E. Martin,et al. Panobinostat suppresses the mesenchymal phenotype in a novel claudin-low triple negative patient-derived breast cancer model , 2018, Oncoscience.
[39] S. Winandy,et al. Novel TCR-Mediated Mechanisms of Notch Activation and Signaling , 2018, The Journal of Immunology.
[40] E. Martin,et al. A novel patient-derived xenograft model for claudin-low triple-negative breast cancer , 2018, Breast Cancer Research and Treatment.
[41] Wentao Yang,et al. Differential expression and clinical significance of epithelial-mesenchymal transition markers among different histological types of triple-negative breast cancer , 2018, Journal of Cancer.
[42] Dorte Nielsen,et al. Triple negative breast cancer – prognostic role of immune-related factors: a systematic review , 2018, Acta oncologica.
[43] G. Turashvili,et al. Tumor Heterogeneity in Breast Cancer , 2017, Front. Med..
[44] Robert W. Hsieh,et al. Role of epithelial to mesenchymal transition associated genes in mammary gland regeneration and breast tumorigenesis , 2017, Nature Communications.
[45] L. Newman,et al. Triple-Negative Breast Cancer, Stem Cells, and African Ancestry. , 2017, The American journal of pathology.
[46] G. Curigliano,et al. Notch inhibitors and their role in the treatment of triple negative breast cancer: promises and failures , 2017, Current opinion in oncology.
[47] B. Osborne,et al. Anti-Jagged Immunotherapy Inhibits MDSCs and Overcomes Tumor-Induced Tolerance. , 2017, Cancer research.
[48] A. Mortazavi,et al. Prostaglandin E2 Leads to the Acquisition of DNMT3A-Dependent Tolerogenic Functions in Human Myeloid-Derived Suppressor Cells. , 2017, Cell reports.
[49] A. Rishi,et al. Myc mediates cancer stem-like cells and EMT changes in triple negative breast cancers cells , 2017, PloS one.
[50] H. Berman,et al. Notch Shapes the Innate Immunophenotype in Breast Cancer. , 2017, Cancer discovery.
[51] L. Del Valle,et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells , 2017, Oncoimmunology.
[52] T. Golde,et al. γ‐Secretase inhibitors in cancer clinical trials are pharmacologically and functionally distinct , 2017, EMBO molecular medicine.
[53] S. Ferrone,et al. Immunomodulating and Immunoresistance Properties of Cancer-Initiating Cells: Implications for the Clinical Success of Immunotherapy , 2017, Immunological investigations.
[54] M. Götte,et al. Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways , 2017, Molecular Cancer.
[55] D. Curiel,et al. COX2/mPGES1/PGE2 pathway regulates PD-L1 expression in tumor-associated macrophages and myeloid-derived suppressor cells , 2017, Proceedings of the National Academy of Sciences.
[56] Z. Shao,et al. Clinical and molecular relevance of mutant-allele tumor heterogeneity in breast cancer , 2017, Breast Cancer Research and Treatment.
[57] J. Lebrun,et al. Cyclooxygenase-2 regulates TGFβ-induced cancer stemness in triple-negative breast cancer , 2017, Scientific Reports.
[58] L. Pusztai,et al. Predictors of Chemosensitivity in Triple Negative Breast Cancer: An Integrated Genomic Analysis , 2016, PLoS medicine.
[59] G. Curigliano,et al. Phase I study of the gamma secretase inhibitor PF-03084014 in combination with docetaxel in patients with advanced triple-negative breast cancer , 2016, Oncotarget.
[60] Q. Sun,et al. NOTCH1 is a poor prognostic factor for breast cancer and is associated with breast cancer stem cells , 2016, OncoTargets and therapy.
[61] J. Balko,et al. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease , 2016, Nature Reviews Clinical Oncology.
[62] I. Fentiman,et al. The Role of NSAIDs in Breast Cancer Prevention and Relapse: Current Evidence and Future Perspectives , 2016, Breast Care.
[63] X. Shao,et al. NUMB negatively regulates the epithelial-mesenchymal transition of triple-negative breast cancer by antagonizing Notch signaling , 2016, Oncotarget.
[64] Harold L. Moses,et al. Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection , 2016, PloS one.
[65] D. Salomon,et al. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer , 2016, Breast Cancer Research and Treatment.
[66] Yongsheng Wang,et al. Sulindac, a non-steroidal anti-inflammatory drug, mediates breast cancer inhibition as an immune modulator , 2016, Scientific Reports.
[67] J. Grandis,et al. Treatment of Triple-Negative Breast Cancer with TORC1/2 Inhibitors Sustains a Drug-Resistant and Notch-Dependent Cancer Stem Cell Population. , 2016, Cancer research.
[68] K. Gogineni,et al. A Review of Systemic Treatment in Metastatic Triple-Negative Breast Cancer , 2016, Breast cancer : basic and clinical research.
[69] S. Safe,et al. Sulindac sulfide inhibits colon cancer cell growth and downregulates specificity protein transcription factors , 2015, BMC Cancer.
[70] Bing Liu,et al. Cyclooxygenase-2 promotes tumor growth and suppresses tumor immunity , 2015, Cancer Cell International.
[71] Samira Mohammadi-Yeganeh,et al. Targeting Of miR9/NOTCH1 Interaction Reduces Metastatic Behavior in Triple‐negative Breast Cancer , 2015, Chemical biology & drug design.
[72] J. Pietenpol,et al. Clinical implications of molecular heterogeneity in triple negative breast cancer. , 2015, Breast.
[73] L. Di Francesco,et al. New Insights into the Mechanism of Action of Aspirin in the Prevention of Colorectal Neoplasia. , 2015, Current pharmaceutical design.
[74] B. Bedogni,et al. Notch1 Autoactivation via Transcriptional Regulation of Furin, Which Sustains Notch1 Signaling by Processing Notch1-Activating Proteases ADAM10 and Membrane Type 1 Matrix Metalloproteinase , 2015, Molecular and Cellular Biology.
[75] Michael Kahn,et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update , 2015, Nature Reviews Clinical Oncology.
[76] Shiying Yu,et al. Expression of Notch1 Correlates with Breast Cancer Progression and Prognosis , 2015, PloS one.
[77] Annie A Wu,et al. Reprogramming the tumor microenvironment: tumor-induced immunosuppressive factors paralyze T cells , 2015, Oncoimmunology.
[78] L. Castellano,et al. Mammosphere formation assay from human breast cancer tissues and cell lines. , 2015, Journal of visualized experiments : JoVE.
[79] B. Fowlkes,et al. Notch signaling regulates antigen sensitivity of naive CD4+ T cells by tuning co-stimulation. , 2015, Immunity.
[80] Matthew E. Ritchie,et al. limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.
[81] P. Rejto,et al. PEST Domain Mutations in Notch Receptors Comprise an Oncogenic Driver Segment in Triple-Negative Breast Cancer Sensitive to a γ-Secretase Inhibitor , 2015, Clinical Cancer Research.
[82] R. Kumar,et al. Companion Diagnostics and Cancer Biomarkers Coordinate Hyperactivation of Notch 1 and Ras / MAPK Pathways Correlates with Poor Patient Survival : Novel Therapeutic Strategy for Aggressive Breast Cancers , 2014 .
[83] Vandana G Abramson,et al. Molecular Heterogeneity of Triple-Negative Breast Cancer , 2014, Current Breast Cancer Reports.
[84] C. Moskaluk,et al. Discovery of biomarkers predictive of GSI response in triple-negative breast cancer and adenoid cystic carcinoma. , 2014, Cancer discovery.
[85] Kathleen A Cronin,et al. US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status. , 2014, Journal of the National Cancer Institute.
[86] Keli Xu,et al. Targeting Met and Notch in the Lfng-deficient, Met-amplified triple-negative breast cancer , 2014, Cancer biology & therapy.
[87] T. Golde,et al. Non-Canonical Notch Signaling Drives Activation and Differentiation of Peripheral CD4+ T Cells , 2014, Front. Immunol..
[88] T. Golde,et al. γ-Secretase inhibitors and modulators. , 2013, Biochimica et biophysica acta.
[89] S. Grivennikov,et al. Top Notch cancer stem cells by paracrine NF-κB signaling in breast cancer , 2013, Breast Cancer Research.
[90] X. Chen,et al. Sulindac Selectively Inhibits Colon Tumor Cell Growth by Activating the cGMP/PKG Pathway to Suppress Wnt/β-Catenin Signaling , 2013, Molecular Cancer Therapeutics.
[91] G. Piazza,et al. COX-Independent Mechanisms of Cancer Chemoprevention by Anti-Inflammatory Drugs , 2013, Front. Oncol..
[92] J. Lewin,et al. Abstract 410: Elevated Jagged-1 and Notch-1 expression in high grade and metastatic prostate cancers. , 2013 .
[93] L. Miele,et al. Correlation of Notch1, pAKT and nuclear NF-κB expression in triple negative breast cancer. , 2013, American journal of cancer research.
[94] Stephen T. C. Wong,et al. Identification of Prognosis-Relevant Subgroups in Patients with Chemoresistant Triple-Negative Breast Cancer , 2013, Clinical Cancer Research.
[95] S. Mandrekar,et al. Population Pharmacokinetic Model for Cancer Chemoprevention With Sulindac in Healthy Subjects , 2013, Journal of clinical pharmacology.
[96] J. Christensen,et al. Synergistic Effect of the γ‐Secretase Inhibitor PF‐03084014 and Docetaxel in Breast Cancer Models , 2013, Stem cells translational medicine.
[97] K. Gajiwala,et al. Specific inhibition of Notch1 signaling enhances the antitumor efficacy of chemotherapy in triple negative breast cancer through reduction of cancer stem cells. , 2013, Cancer letters.
[98] Ian Krop,et al. Preclinical and Clinical Studies of Gamma Secretase Inhibitors with Docetaxel on Human Breast Tumors , 2013, Clinical Cancer Research.
[99] P. Kalinski,et al. Generation of myeloid-derived suppressor cells using prostaglandin E2 , 2012, Transplantation research.
[100] A. Frey,et al. Myeloid-Derived Suppressor Cells and anti-tumor T cells: a complex relationship , 2012, Immunological investigations.
[101] K. Blennow,et al. Second Generation γ-Secretase Modulators Exhibit Different Modulation of Notch β and Aβ Production* , 2012, The Journal of Biological Chemistry.
[102] T. Maruyama,et al. Targeting COX-2 and EP4 to control tumor growth, angiogenesis, lymphangiogenesis and metastasis to the lungs and lymph nodes in a breast cancer model , 2012, Laboratory Investigation.
[103] C. Perou,et al. Lunatic fringe deficiency cooperates with the Met/Caveolin gene amplicon to induce basal-like breast cancer. , 2012, Cancer cell.
[104] Q. Cui,et al. Sulindac inhibits tumor cell invasion by suppressing NF-κB mediated transcription of microRNAs , 2012, Oncogene.
[105] P. Kalinski. Regulation of Immune Responses by Prostaglandin E2 , 2012, The Journal of Immunology.
[106] J. B. Jordan,et al. NSAID-based γ-secretase modulators do not bind to the amyloid-β polypeptide. , 2011, Biochemistry.
[107] Damian Smedley,et al. BioMart Central Portal: an open database network for the biological community , 2011, Database J. Biol. Databases Curation.
[108] X. Chen,et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. , 2011, The Journal of clinical investigation.
[109] J. Allalunis-Turner,et al. Notch signaling as a therapeutic target for breast cancer treatment? , 2011, Breast Cancer Research.
[110] Garret A. FitzGerald,et al. Prostaglandins and Inflammation , 2011, Arteriosclerosis, thrombosis, and vascular biology.
[111] Jorge S Reis-Filho,et al. Triple-negative breast cancer. , 2010, The New England journal of medicine.
[112] A. Statnikov,et al. The Notch/Hes1 pathway sustains NF-κB activation through CYLD repression in T cell leukemia. , 2010, Cancer cell.
[113] B. Reif,et al. Amyloid beta 42 peptide (Aβ42)-lowering compounds directly bind to Aβ and interfere with amyloid precursor protein (APP) transmembrane dimerization , 2010, Proceedings of the National Academy of Sciences.
[114] Wei Zhang,et al. Sulindac sulfide selectively inhibits growth and induces apoptosis of human breast tumor cells by phosphodiesterase 5 inhibition, elevation of cyclic GMP, and activation of protein kinase G , 2009, Molecular Cancer Therapeutics.
[115] N Harbeck,et al. Triple-negative breast cancer--current status and future directions. , 2009, Annals of oncology : official journal of the European Society for Medical Oncology.
[116] Seung-Woo Hong,et al. Sulindac induces apoptotic cell death in susceptible human breast cancer cells through, at least in part, inhibition of IKKβ , 2009, Apoptosis.
[117] Ramón Díaz-Uriarte,et al. Pomelo II: finding differentially expressed genes , 2009, Nucleic Acids Res..
[118] P. Reddanna,et al. Eicosanoids in inflammation and cancer: the role of COX-2 , 2009, Expert review of clinical immunology.
[119] M. Wolfe,et al. Substrate-targeting γ-secretase modulators , 2008, Nature.
[120] C. Behl,et al. Effects of sulindac sulfide on the membrane architecture and the activity of γ-secretase , 2008, Neuropharmacology.
[121] S. Mandrekar,et al. Comparative Bioavailability of Sulindac in Capsule and Tablet Formulations , 2008, Cancer Epidemiology Biomarkers & Prevention.
[122] S. Narod,et al. Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence , 2007, Clinical Cancer Research.
[123] L. Habel,et al. NSAIDs and breast cancer recurrence in a prospective cohort study , 2007, Cancer Causes & Control.
[124] L. O’Driscoll,et al. A phase I clinical and pharmacokinetic study of the multi-drug resistance protein-1 (MRP-1) inhibitor sulindac, in combination with epirubicin in patients with advanced cancer , 2006, Cancer Chemotherapy and Pharmacology.
[125] B. Leibovich,et al. Exisulind in the treatment of prostate cancer , 2005, Expert review of anticancer therapy.
[126] B. Aggarwal,et al. Nonsteroidal anti-inflammatory agents differ in their ability to suppress NF-κB activation, inhibition of expression of cyclooxygenase-2 and cyclin D1, and abrogation of tumor cell proliferation , 2004, Oncogene.
[127] G. Hortobagyi,et al. Phase I and II study of exisulind in combination with capecitabine in patients with metastatic breast cancer. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.
[128] G. Anderson,et al. Breast cancer and nonsteroidal anti-inflammatory drugs: prospective results from the Women's Health Initiative. , 2003, Cancer research.
[129] S. Weggen,et al. Evidence That Nonsteroidal Anti-inflammatory Drugs Decrease Amyloid β42 Production by Direct Modulation of γ-Secretase Activity* , 2003, Journal of Biological Chemistry.
[130] Pritam Das,et al. NSAIDs and enantiomers of flurbiprofen target γ-secretase and lower Aβ42 in vivo , 2003 .
[131] T. Iwatsubo,et al. Sulindac Sulfide Is a Noncompetitive γ-Secretase Inhibitor That Preferentially Reduces Aβ42 Generation* , 2003, The Journal of Biological Chemistry.
[132] M. Thun,et al. Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues. , 2002, Journal of the National Cancer Institute.
[133] G. Piazza,et al. Preclinical and clinical studies of docetaxel and exisulind in the treatment of human lung cancer. , 2002, Seminars in oncology.
[134] Rong Wang,et al. A subset of NSAIDs lower amyloidogenic Aβ42 independently of cyclooxygenase activity , 2001, Nature.
[135] E. T. Goluboff,et al. Exisulind, a selective apoptotic antineoplastic drug , 2001, Expert opinion on investigational drugs.
[136] A. B. Lyons,et al. Analysing cell division in vivo and in vitro using flow cytometric measurement of CFSE dye dilution. , 2000, Journal of immunological methods.
[137] M. Ashburner,et al. Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.
[138] R. Gaynor,et al. Sulindac Inhibits Activation of the NF-κB Pathway* , 1999, The Journal of Biological Chemistry.
[139] J. Morrow,et al. Sulindac sulfide, but not sulindac sulfone, inhibits colorectal cancer growth. , 1999, Neoplasia.
[140] W. Ray,et al. Use of nonsteroidal anti-inflammatory drugs and incidence of colorectal cancer: a population-based study. , 1999, Archives of internal medicine.
[141] M. Bevan,et al. Correlating notch signaling with thymocyte maturation. , 1998, Immunity.
[142] J. Vane,et al. Mechanism of action of nonsteroidal anti-inflammatory drugs. , 1998, The American journal of medicine.
[143] N. Davies,et al. Clinical Pharmacokinetics of Sulindac , 1997, Clinical pharmacokinetics.
[144] K. Namboodiri,et al. Nonsteroidal antiinflammatory drugs and breast cancer. , 1996, Epidemiology.
[145] P. Pépin,et al. Effects of sulindac and oltipraz on the tumorigenicity of 4-(methylnitrosamino)1-(3-pyridyl)-1-butanone in A/J mouse lung. , 1992, Carcinogenesis.
[146] J. Jacquemier,et al. Prostaglandin in human breast cancer: Evidence suggesting that an elevated prostaglandin production is a marker of high metastatic potential for neoplastic cells. , 1980, Journal of the National Cancer Institute.
[147] C. G. Arman,et al. Identification of the biologically active form of sulindac. , 1977, The Journal of pharmacology and experimental therapeutics.
[148] F. Rojo,et al. Breast Cancer Immunology and Immunotherapy: Current Status and Future Perspectives. , 2017, International review of cell and molecular biology.
[149] A. Tan,et al. Triple-negative breast cancer: molecular subtypes and new targets for therapy. , 2015, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.
[150] C. Osipo,et al. The functional role of Notch signaling in triple-negative breast cancer. , 2013, Vitamins and hormones.
[151] B. de Strooper,et al. Presenilins and γ-secretase: structure, function, and role in Alzheimer Disease. , 2012, Cold Spring Harbor perspectives in medicine.
[152] M. Reedijk. Notch signaling and breast cancer. , 2012, Advances in experimental medicine and biology.
[153] L. Minter,et al. Canonical and non-canonical Notch signaling in CD4⁺ T cells. , 2012, Current topics in microbiology and immunology.
[154] B. Imbimbo. Therapeutic Potential of γ -Secretase Inhibitors and Modulators , 2008 .
[155] Malay Mandal,et al. Targeting the NF-κB signaling pathway in Notch1-induced T-cell leukemia , 2007, Nature Medicine.
[156] L. Minter,et al. Notch signalling during peripheral T-cell activation and differentiation , 2007, Nature Reviews Immunology.
[157] Exisulind: Aptosyn, FGN 1, Prevatac, sulindac sulfone. , 2004, Drugs in R&D.
[158] G. Griffiths. Exisulind Cell Pathways. , 2000, Current opinion in investigational drugs.
[159] J. Vane,et al. Mechanism of action of antiinflammatory drugs. , 1998, International journal of tissue reactions.
[160] Y. Benjamini,et al. Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .
[161] K. Seibert,et al. Role of inducible cyclooxygenase (COX-2) in inflammation. , 1994, Receptor.