Skip to main content
Top
Published in: Targeted Oncology 4/2017

01-08-2017 | Original Research Article

Synthesis and Characterization of Novel BMI1 Inhibitors Targeting Cellular Self-Renewal in Hepatocellular Carcinoma

Authors: Monica Bartucci, Mohamed S. Hussein, Eric Huselid, Kathleen Flaherty, Michele Patrizii, Saurabh V. Laddha, Cindy Kui, Rachel A. Bigos, John A. Gilleran, Mervat M. S. El Ansary, Mona A. M. Awad, S. David Kimball, David J. Augeri, Hatem E. Sabaawy

Published in: Targeted Oncology | Issue 4/2017

Login to get access

Abstract

Background

Hepatocellular carcinoma (HCC) represents one of the most lethal cancers worldwide due to therapy resistance and disease recurrence. Tumor relapse following treatment could be driven by the persistence of liver cancer stem-like cells (CSCs). The protein BMI1 is a member of the polycomb epigenetic factors governing cellular self-renewal, proliferation, and stemness maintenance. BMI1 expression also correlates with poor patient survival in various cancer types.

Objective

We aimed to elucidate the extent to which BMI1 can be used as a potential therapeutic target for CSC eradication in HCC.

Methods

We have recently participated in characterizing the first known pharmacological small molecule inhibitor of BMI1. Here, we synthesized a panel of novel BMI1 inhibitors and examined their ability to alter cellular growth and eliminate cancer progenitor/stem-like cells in HCC with different p53 backgrounds.

Results

Among various molecules examined, RU-A1 particularly downregulated BMI1 expression, impaired cell viability, reduced cell migration, and sensitized HCC cells to 5-fluorouracil (5-FU) in vitro. Notably, long-term analysis of HCC survival showed that, unlike chemotherapy, RU-A1 effectively reduced CSC content, even as monotherapy. BMI1 inhibition with RU-A1 diminished the number of stem-like cells in vitro more efficiently than the model compound C-209, as demonstrated by clonogenic assays and impairment of CSC marker expression. Furthermore, xenograft assays in zebrafish showed that RU-A1 abrogated tumor growth in vivo.

Conclusions

This study demonstrates the ability to identify agents with the propensity for targeting CSCs in HCC that could be explored as novel treatments in the clinical setting.
Appendix
Available only for authorised users
Literature
2.
3.
go back to reference Yilmaz G, Akyol G, Cakir A, Ilhan M. Investigation of diagnostic utility and expression profiles of stem cell markers (CD133 and CD90) in hepatocellular carcinoma, small cell dysplasia, and cirrhosis. Pathol Res Pract. 2014;210(7):419–25.CrossRefPubMed Yilmaz G, Akyol G, Cakir A, Ilhan M. Investigation of diagnostic utility and expression profiles of stem cell markers (CD133 and CD90) in hepatocellular carcinoma, small cell dysplasia, and cirrhosis. Pathol Res Pract. 2014;210(7):419–25.CrossRefPubMed
4.
go back to reference Lee TK, Castilho A, Ma S, Ng IO. Liver cancer stem cells: implications for a new therapeutic target. Liver Int. 2009;29(7):955–65.CrossRefPubMed Lee TK, Castilho A, Ma S, Ng IO. Liver cancer stem cells: implications for a new therapeutic target. Liver Int. 2009;29(7):955–65.CrossRefPubMed
6.
go back to reference Smith LL, Yeung J, Zeisig BB, Popov N, Huijbers I, Barnes J, et al. Functional crosstalk between Bmi1 and MLL/Hoxa9 axis in establishment of normal hematopoietic and leukemic stem cells. Cell Stem Cell. 2011;8(6):649–62.CrossRefPubMed Smith LL, Yeung J, Zeisig BB, Popov N, Huijbers I, Barnes J, et al. Functional crosstalk between Bmi1 and MLL/Hoxa9 axis in establishment of normal hematopoietic and leukemic stem cells. Cell Stem Cell. 2011;8(6):649–62.CrossRefPubMed
7.
go back to reference Chiba T, Miyagi S, Saraya A, Aoki R, Seki A, Morita Y, et al. The polycomb gene product BMI1 contributes to the maintenance of tumor-initiating side population cells in hepatocellular carcinoma. Cancer Res. 2008;68(19):7742–9.CrossRefPubMed Chiba T, Miyagi S, Saraya A, Aoki R, Seki A, Morita Y, et al. The polycomb gene product BMI1 contributes to the maintenance of tumor-initiating side population cells in hepatocellular carcinoma. Cancer Res. 2008;68(19):7742–9.CrossRefPubMed
8.
go back to reference Lukacs RU, Memarzadeh S, Wu H, Witte ON. Bmi-1 is a crucial regulator of prostate stem cell self-renewal and malignant transformation. Cell Stem Cell. 2010;7(6):682–93.CrossRefPubMedPubMedCentral Lukacs RU, Memarzadeh S, Wu H, Witte ON. Bmi-1 is a crucial regulator of prostate stem cell self-renewal and malignant transformation. Cell Stem Cell. 2010;7(6):682–93.CrossRefPubMedPubMedCentral
9.
go back to reference Effendi K, Mori T, Komuta M, Masugi Y, Du W, Sakamoto M. Bmi-1 gene is upregulated in early-stage hepatocellular carcinoma and correlates with ATP-binding cassette transporter B1 expression. Cancer Sci. 2010;101(3):666–72.CrossRefPubMed Effendi K, Mori T, Komuta M, Masugi Y, Du W, Sakamoto M. Bmi-1 gene is upregulated in early-stage hepatocellular carcinoma and correlates with ATP-binding cassette transporter B1 expression. Cancer Sci. 2010;101(3):666–72.CrossRefPubMed
10.
go back to reference Wang H, Pan K, Zhang HK, Weng DS, Zhou J, Li JJ, et al. Increased polycomb-group oncogene Bmi-1 expression correlates with poor prognosis in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134(5):535–41.CrossRefPubMed Wang H, Pan K, Zhang HK, Weng DS, Zhou J, Li JJ, et al. Increased polycomb-group oncogene Bmi-1 expression correlates with poor prognosis in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2008;134(5):535–41.CrossRefPubMed
11.
go back to reference Bansal N, Campbell N, Medina D, Dipaola RS, Bertino JR, Sabaawy HE. Targeting Bmi1 in human prostate tumor initiating cells. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research. 2010, Philadelphia: AACR: Washington, DC. p. Abstract nr 4279. Bansal N, Campbell N, Medina D, Dipaola RS, Bertino JR, Sabaawy HE. Targeting Bmi1 in human prostate tumor initiating cells. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research. 2010, Philadelphia: AACR: Washington, DC. p. Abstract nr 4279.
12.
go back to reference Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C, Davis T, et al. Self-renewal as a therapeutic target in human colorectal cancer. Nat Med. 2014;20(1):29–36.CrossRefPubMed Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C, Davis T, et al. Self-renewal as a therapeutic target in human colorectal cancer. Nat Med. 2014;20(1):29–36.CrossRefPubMed
13.
go back to reference Bansal N, Bartucci M, Yusuff S, Davis S, Flaherty K, Huselid E, et al. BMI-1 targeting interferes with patient-derived tumor-initiating cell survival and tumor growth in prostate cancer. Clin Cancer Res. 2016;22(24):6176–91.CrossRefPubMed Bansal N, Bartucci M, Yusuff S, Davis S, Flaherty K, Huselid E, et al. BMI-1 targeting interferes with patient-derived tumor-initiating cell survival and tumor growth in prostate cancer. Clin Cancer Res. 2016;22(24):6176–91.CrossRefPubMed
14.
go back to reference Nishida Y, Maeda A, Chachad D, Ishizawa J, Qiu YH, Kornblau SM, et al. Preclinical activity of the novel B-cell-specific Moloney murine leukemia virus integration site 1 inhibitor PTC-209 in acute myeloid leukemia: implications for leukemia therapy. Cancer Sci. 2015;106(12):1705–13.CrossRefPubMedPubMedCentral Nishida Y, Maeda A, Chachad D, Ishizawa J, Qiu YH, Kornblau SM, et al. Preclinical activity of the novel B-cell-specific Moloney murine leukemia virus integration site 1 inhibitor PTC-209 in acute myeloid leukemia: implications for leukemia therapy. Cancer Sci. 2015;106(12):1705–13.CrossRefPubMedPubMedCentral
15.
go back to reference Mayr C, Wagner A, Loeffelberger M, Bruckner D, Jakab M, Berr F, et al. The BMI1 inhibitor PTC-209 is a potential compound to halt cellular growth in biliary tract cancer cells. Oncotarget. 2016;7(1):745–58.CrossRefPubMed Mayr C, Wagner A, Loeffelberger M, Bruckner D, Jakab M, Berr F, et al. The BMI1 inhibitor PTC-209 is a potential compound to halt cellular growth in biliary tract cancer cells. Oncotarget. 2016;7(1):745–58.CrossRefPubMed
16.
go back to reference Bolomsky A, Schlangen K, Schreiner W, Zojer N, Ludwig H. Targeting of BMI-1 with PTC-209 shows potent anti-myeloma activity and impairs the tumour microenvironment. J Hematol Oncol. 2016;9:17.CrossRefPubMedPubMedCentral Bolomsky A, Schlangen K, Schreiner W, Zojer N, Ludwig H. Targeting of BMI-1 with PTC-209 shows potent anti-myeloma activity and impairs the tumour microenvironment. J Hematol Oncol. 2016;9:17.CrossRefPubMedPubMedCentral
17.
go back to reference Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X, et al. Glutaminase 2 negatively regulates the PI3K/AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget. 2014;5(9):2635–47.CrossRefPubMedPubMedCentral Liu J, Zhang C, Lin M, Zhu W, Liang Y, Hong X, et al. Glutaminase 2 negatively regulates the PI3K/AKT signaling and shows tumor suppression activity in human hepatocellular carcinoma. Oncotarget. 2014;5(9):2635–47.CrossRefPubMedPubMedCentral
18.
go back to reference Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347(1–2):70–8.PubMed Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods. 2009;347(1–2):70–8.PubMed
19.
go back to reference Bansal N, Davis S, Tereshchenko I, Budak-Alpdogan T, Zhong H, Stein MN, et al. Enrichment of human prostate cancer cells with tumor initiating properties in mouse and zebrafish xenografts by differential adhesion. Prostate. 2014;74(2):187–200.CrossRefPubMed Bansal N, Davis S, Tereshchenko I, Budak-Alpdogan T, Zhong H, Stein MN, et al. Enrichment of human prostate cancer cells with tumor initiating properties in mouse and zebrafish xenografts by differential adhesion. Prostate. 2014;74(2):187–200.CrossRefPubMed
21.
go back to reference Perola E, Walters WP, Charifson PS. A detailed comparison of current docking and scoring methods on systems of pharmaceutical relevance. Proteins. 2004;56(2):235–49.CrossRefPubMed Perola E, Walters WP, Charifson PS. A detailed comparison of current docking and scoring methods on systems of pharmaceutical relevance. Proteins. 2004;56(2):235–49.CrossRefPubMed
22.
go back to reference Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26(15):2166–76.CrossRefPubMed Hussain SP, Schwank J, Staib F, Wang XW, Harris CC. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26(15):2166–76.CrossRefPubMed
23.
go back to reference Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397(6715):164–8.CrossRefPubMed Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M. The oncogene and Polycomb-group gene bmi-1 regulates cell proliferation and senescence through the ink4a locus. Nature. 1999;397(6715):164–8.CrossRefPubMed
24.
go back to reference Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, et al. Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat Cell Biol. 2010;12(10):982–92.CrossRefPubMed Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, et al. Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat Cell Biol. 2010;12(10):982–92.CrossRefPubMed
25.
go back to reference Grinstein E, Mahotka C. Stem cell divisions controlled by the proto-oncogene BMI-1. J Stem Cells. 2009;4(3):141–6.PubMed Grinstein E, Mahotka C. Stem cell divisions controlled by the proto-oncogene BMI-1. J Stem Cells. 2009;4(3):141–6.PubMed
26.
go back to reference Cayrol C, Knibiehler M, Ducommun B. p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene. 1998;16(3):311–20.CrossRefPubMed Cayrol C, Knibiehler M, Ducommun B. p21 binding to PCNA causes G1 and G2 cell cycle arrest in p53-deficient cells. Oncogene. 1998;16(3):311–20.CrossRefPubMed
28.
go back to reference Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.CrossRefPubMed Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.CrossRefPubMed
29.
go back to reference Sun JH, Luo Q, Liu LL, Song GB. Liver cancer stem cell markers: progression and therapeutic implications. World J Gastroenterol. 2016;22(13):3547–57.CrossRefPubMedPubMedCentral Sun JH, Luo Q, Liu LL, Song GB. Liver cancer stem cell markers: progression and therapeutic implications. World J Gastroenterol. 2016;22(13):3547–57.CrossRefPubMedPubMedCentral
31.
go back to reference Mihic-Probst D, Kuster A, Kilgus S, Bode-Lesniewska B, Ingold-Heppner B, Leung C, et al. Consistent expression of the stem cell renewal factor BMI-1 in primary and metastatic melanoma. Int J Cancer. 2007;121(8):1764–70.CrossRefPubMed Mihic-Probst D, Kuster A, Kilgus S, Bode-Lesniewska B, Ingold-Heppner B, Leung C, et al. Consistent expression of the stem cell renewal factor BMI-1 in primary and metastatic melanoma. Int J Cancer. 2007;121(8):1764–70.CrossRefPubMed
32.
go back to reference Glinsky GV, Berezovska O, Glinskii AB. Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest. 2005;115(6):1503–21.CrossRefPubMedPubMedCentral Glinsky GV, Berezovska O, Glinskii AB. Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest. 2005;115(6):1503–21.CrossRefPubMedPubMedCentral
33.
go back to reference Hayry V, Tynninen O, Haapasalo HK, Wolfer J, Paulus W, Hasselblatt M, et al. Stem cell protein BMI-1 is an independent marker for poor prognosis in oligodendroglial tumours. Neuropathol Appl Neurobiol. 2008;34(5):555–63.CrossRefPubMed Hayry V, Tynninen O, Haapasalo HK, Wolfer J, Paulus W, Hasselblatt M, et al. Stem cell protein BMI-1 is an independent marker for poor prognosis in oligodendroglial tumours. Neuropathol Appl Neurobiol. 2008;34(5):555–63.CrossRefPubMed
34.
go back to reference Martin-Padura I, Marighetti P, Agliano A, Colombo F, Larzabal L, Redrado M, et al. Residual dormant cancer stem-cell foci are responsible for tumor relapse after antiangiogenic metronomic therapy in hepatocellular carcinoma xenografts. Lab Investig. 2012;92(7):952–66.CrossRefPubMed Martin-Padura I, Marighetti P, Agliano A, Colombo F, Larzabal L, Redrado M, et al. Residual dormant cancer stem-cell foci are responsible for tumor relapse after antiangiogenic metronomic therapy in hepatocellular carcinoma xenografts. Lab Investig. 2012;92(7):952–66.CrossRefPubMed
35.
go back to reference Katoh M, Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer Res. 2007;13(14):4042–5.CrossRefPubMed Katoh M, Katoh M. WNT signaling pathway and stem cell signaling network. Clin Cancer Res. 2007;13(14):4042–5.CrossRefPubMed
36.
go back to reference Mehta M, Khan A, Danish S, Haffty BG, Sabaawy HE. Radiosensitization of primary human glioblastoma stem-like cells with low-dose AKT inhibition. Mol Cancer Ther. 2015;14(5):1171–80.CrossRefPubMedPubMedCentral Mehta M, Khan A, Danish S, Haffty BG, Sabaawy HE. Radiosensitization of primary human glioblastoma stem-like cells with low-dose AKT inhibition. Mol Cancer Ther. 2015;14(5):1171–80.CrossRefPubMedPubMedCentral
37.
go back to reference Kim JH, Yoon SY, Jeong SH, Kim SY, Moon SK, Joo JH, et al. Overexpression of Bmi-1 oncoprotein correlates with axillary lymph node metastases in invasive ductal breast cancer. Breast. 2004;13(5):383–8.CrossRefPubMed Kim JH, Yoon SY, Jeong SH, Kim SY, Moon SK, Joo JH, et al. Overexpression of Bmi-1 oncoprotein correlates with axillary lymph node metastases in invasive ductal breast cancer. Breast. 2004;13(5):383–8.CrossRefPubMed
38.
go back to reference Leung C, Lingbeek M, Shakhova O, Liu J, Tanger E, Saremaslani P, et al. Bmi1 is essential for cerebellar development and is overexpressed in human medulloblastomas. Nature. 2004;428(6980):337–41.CrossRefPubMed Leung C, Lingbeek M, Shakhova O, Liu J, Tanger E, Saremaslani P, et al. Bmi1 is essential for cerebellar development and is overexpressed in human medulloblastomas. Nature. 2004;428(6980):337–41.CrossRefPubMed
39.
go back to reference Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60.CrossRefPubMed Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60.CrossRefPubMed
40.
go back to reference Berezovska OP, Glinskii AB, Yang Z, Li XM, Hoffman RM, Glinsky GV. Essential role for activation of the Polycomb group (PcG) protein chromatin silencing pathway in metastatic prostate cancer. Cell Cycle. 2006;5(16):1886–901.CrossRefPubMed Berezovska OP, Glinskii AB, Yang Z, Li XM, Hoffman RM, Glinsky GV. Essential role for activation of the Polycomb group (PcG) protein chromatin silencing pathway in metastatic prostate cancer. Cell Cycle. 2006;5(16):1886–901.CrossRefPubMed
41.
go back to reference Ruan ZP, Xu R, Lv Y, Tian T, Wang WJ, Guo H, et al. Bmi1 knockdown inhibits hepatocarcinogenesis. Int J Oncol. 2013;42(1):261–8.PubMed Ruan ZP, Xu R, Lv Y, Tian T, Wang WJ, Guo H, et al. Bmi1 knockdown inhibits hepatocarcinogenesis. Int J Oncol. 2013;42(1):261–8.PubMed
42.
go back to reference Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer. 2006;6(11):846–56.CrossRefPubMed Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer. 2006;6(11):846–56.CrossRefPubMed
43.
go back to reference Fan L, Xu C, Wang C, Tao J, Ho C, Jiang L, et al. Bmi1 is required for hepatic progenitor cell expansion and liver tumor development. PLoS One. 2012;7(9):e46472.CrossRefPubMedPubMedCentral Fan L, Xu C, Wang C, Tao J, Ho C, Jiang L, et al. Bmi1 is required for hepatic progenitor cell expansion and liver tumor development. PLoS One. 2012;7(9):e46472.CrossRefPubMedPubMedCentral
44.
go back to reference Cao L, Bombard J, Cintron K, Sheedy J, Weetall ML, Davis TW. BMI1 as a novel target for drug discovery in cancer. J Cell Biochem. 2011;112(10):2729–41.CrossRefPubMed Cao L, Bombard J, Cintron K, Sheedy J, Weetall ML, Davis TW. BMI1 as a novel target for drug discovery in cancer. J Cell Biochem. 2011;112(10):2729–41.CrossRefPubMed
45.
go back to reference Siddique HR, Saleem M. Role of BMI1, a stem cell factor, in cancer recurrence and chemoresistance: preclinical and clinical evidences. Stem Cells. 2012;30(3):372–8.CrossRefPubMed Siddique HR, Saleem M. Role of BMI1, a stem cell factor, in cancer recurrence and chemoresistance: preclinical and clinical evidences. Stem Cells. 2012;30(3):372–8.CrossRefPubMed
46.
go back to reference Jiang L, Li J, Song L. Bmi-1, stem cells and cancer. Acta Biochim Biophys Sin Shanghai. 2009;41(7):527–34.CrossRefPubMed Jiang L, Li J, Song L. Bmi-1, stem cells and cancer. Acta Biochim Biophys Sin Shanghai. 2009;41(7):527–34.CrossRefPubMed
47.
go back to reference Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 1999;13(20):2678–90.CrossRefPubMedPubMedCentral Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev. 1999;13(20):2678–90.CrossRefPubMedPubMedCentral
48.
go back to reference Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425(6961):962–7.CrossRefPubMedPubMedCentral Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425(6961):962–7.CrossRefPubMedPubMedCentral
50.
go back to reference Blackledge NP, Farcas AM, Kondo T, King HW, McGouran JF, Hanssen LL, et al. Variant PRC1 complex-dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell. 2014;157(6):1445–59.CrossRefPubMedPubMedCentral Blackledge NP, Farcas AM, Kondo T, King HW, McGouran JF, Hanssen LL, et al. Variant PRC1 complex-dependent H2A ubiquitylation drives PRC2 recruitment and polycomb domain formation. Cell. 2014;157(6):1445–59.CrossRefPubMedPubMedCentral
51.
go back to reference Fasano CA, Dimos JT, Ivanova NB, Lowry N, Lemischka IR, Temple S. shRNA knockdown of Bmi-1 reveals a critical role for p21-Rb pathway in NSC self-renewal during development. Cell Stem Cell. 2007;1(1):87–99.CrossRefPubMed Fasano CA, Dimos JT, Ivanova NB, Lowry N, Lemischka IR, Temple S. shRNA knockdown of Bmi-1 reveals a critical role for p21-Rb pathway in NSC self-renewal during development. Cell Stem Cell. 2007;1(1):87–99.CrossRefPubMed
52.
go back to reference Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 2009;8(10):806–23.CrossRefPubMed Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks PB. Tumour-initiating cells: challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 2009;8(10):806–23.CrossRefPubMed
Metadata
Title
Synthesis and Characterization of Novel BMI1 Inhibitors Targeting Cellular Self-Renewal in Hepatocellular Carcinoma
Authors
Monica Bartucci
Mohamed S. Hussein
Eric Huselid
Kathleen Flaherty
Michele Patrizii
Saurabh V. Laddha
Cindy Kui
Rachel A. Bigos
John A. Gilleran
Mervat M. S. El Ansary
Mona A. M. Awad
S. David Kimball
David J. Augeri
Hatem E. Sabaawy
Publication date
01-08-2017
Publisher
Springer International Publishing
Published in
Targeted Oncology / Issue 4/2017
Print ISSN: 1776-2596
Electronic ISSN: 1776-260X
DOI
https://doi.org/10.1007/s11523-017-0501-x

Other articles of this Issue 4/2017

Targeted Oncology 4/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine