1,170
Views
32
CrossRef citations to date
0
Altmetric
Review

Mcl-1 inhibitors: a patent review

&
Pages 163-178 | Received 10 Apr 2016, Accepted 14 Oct 2016, Published online: 17 Nov 2016

References

  • Moreira IS, Fernandes PA, Ramos MJ. Hot spots–a review of the protein-protein interface determinant amino-acid residues. Proteins. 2007;68:803–812.
  • Jones S, Thornton JM. Principles of protein-protein interactions. Proc Natl Acad Sci U S A. 1996;93:13–20.
  • Wells JA, McClendon CL. Reaching for high-hanging fruit in drug discovery at protein-protein interfaces. Nature. 2007;450:1001–1009.
  • Arkin MR, Wells JA. Small-molecule inhibitors of protein-protein interactions: progressing towards the dream. Nat Rev Drug Discov. 2004;3:301–317.
  • Fletcher S, Hamilton AD. Protein surface recognition and proteomimetics: mimics of protein surface structure and function. Curr Opin Chem Biol. 2005;9:632–638.
  • Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495–516.
  • Favaloro B, Allocati N, Graziano V, et al. Role of apoptosis in disease. Aging (Albany. NY). 2012;4:330–349.
  • Brunelle JK, Letai A. Control of mitochondrial apoptosis by the Bcl-2 family. J Cell Sci. 2009;122:437–441.
  • Adams JM, Cory S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 2007;26:1324–1337.
  • Chen L, Willis SN, Wei A, et al. Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005;17:393–403.
  • Czabotar PE, Lee EF, van Delft MF, et al. Structural insights into the degradation of Mcl-1 induced by BH3 domains. Proc Natl Acad Sci U S A. 2007;104:6217–6222.
  • Fadeel B, Orrenius S. Apoptosis: a basic biological phenomenon with wide-ranging implications in human disease. J Intern Med. 2005;258:479–517.
  • Yip KW, Reed JC. Bcl-2 family proteins and cancer. Oncogene. 2008;27:6398–6406.
  • Khan SB, Maududi T, Barton K, et al. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol. 2004;125:156–161.
  • Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res. 2003;63:3637–3645.
  • Mori N, Matsuda T, Tadano M, et al. Apoptosis induced by the histone deacetylase inhibitor FR901228 in human t-cell leukemia virus type 1-infected t-cell Lines and primary adult t-cell leukemia cells. J Virol. 2004;78:4582–4590.
  • Yokoyama Y. Induction of apoptosis by mono(2-ethylhexyl)phthalate (MEHP) in U937 cells. Toxicol Lett. 2003;144:371–381.
  • Gregory GP, Hogg SJ, Kats LM, et al. CDK9 inhibition by dinaciclib potently suppresses Mcl-1 to induce durable apoptotic responses in aggressive MYC-driven B-cell lymphoma in vivo. Leukemia. 2015;29:1437–1441.
  • Ma Y, Cress WD, Haura EB. Flavopiridol-induced apoptosis is mediated through up-regulation of E2F1 and repression of Mcl-1. Mol Cancer Ther. 2003;2:73–81.
  • MacCallum DE, Melville J, Frame S, et al. Seliciclib (CYC202, R-Roscovitine) induces cell death in multiple myeloma cells by inhibition of RNA polymerase II-dependent transcription and down-regulation of Mcl-1. Cancer Res. 2005;65:5399–5407.
  • Leitch AE, Riley NA, Sheldrake TA, et al. The cyclin-dependent kinase inhibitor R-roscovitine down-regulates Mcl-1 to override pro-inflammatory signalling and drive neutrophil apoptosis. Eur J Immunol. 2010;40:1127–1138.
  • Chao S-H, Price DH. Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. J Biol Chem. 2001;276:31793–31799.
  • Rahmani M, Davis EM, Bauer C, et al. Apoptosis induced by the kinase inhibitor BAY 43-9006 in human leukemia cells involves down-regulation of Mcl-1 through inhibition of translation. J Biol Chem. 2005;280:35217–35227.
  • Letai A, Bassik MC, Walensky LD, et al. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2:183–192.
  • Stewart ML, Fire E, Keating AE, et al. The MCL-1 BH3 helix is an exclusive MCL-1 inhibitor and apoptosis sensitizer. Nat Chem Biol. 2010;6:595–601.
  • Walensky LD, Kung AL, Escher I, et al. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science. 2004;305:1466–1470.
  • Cao X, Yap JL, Newell-Rogers MK, et al. The novel BH3 α-helix mimetic JY-1-106 induces apoptosis in a subset of cancer cells (lung cancer, colon cancer and mesothelioma) by disrupting Bcl-xL and Mcl-1 protein-protein interactions with Bak. Mol Cancer. 2013;12:42.
  • Kutzki O, Park HS, Ernst JT, et al. Development of a potent Bcl-x(L) antagonist based on alpha-helix mimicry. J Am Chem Soc. 2002;124:11838–11839.
  • Yin H, Hamilton AD. Terephthalamide derivatives as mimetics of the helical region of Bak peptide target Bcl-xL protein. Bioorg Med Chem Lett. 2004;14:1375–1379.
  • Sebti SM, Hamilton AD, Sill K, et al. Compounds and methods for inducing apoptosis in cancer cells using a BH3 alpha-helical mimetic. WO2012097133. 2012.
  • Wang S, Wang G, Tang G, et al. Small molecule inhibitors of anti-apoptotic BCL-2 family members and the uses thereof. WO2006023778. 2006.
  • Pellecchia M Naphthalene-based inhibitors of anti-apoptotic proteins. WO2010120943. 2010.
  • Pellecchia M, Reed JC Naphthalene-based inhibitors of anti-apoptotic proteins. WO2009052443. 2009.
  • Wang S, Nikolovska-Coleska Z, Yang C-Y, et al. Apogossypolone and the uses thereof. WO2006050447. 2006.
  • Guo M, Shuguang Z, Navarre L, et al. Process for preparing r-gossypol l-phenylalaninol dienamine. WO2009045410. 2009.
  • Holmlund JT, Sorensen M, Leopold L, et al. Pulsatile dosing of gossypol for treatment of disease. WO2008150506. 2008.
  • Wang S, Chen J Gossypol co-crystals and the use thereof. WO 2005094804. 2005.
  • Wang S, Yang D Small molecule antagonists of BCL-2 family proteins. WO 2005069771. 2005.
  • Wang S, Yang D Small molecule antagonists of BCL2 family proteins. WO 2002097053. 2002.
  • Li R, Sebti S, Liu Y, et al. Marinopyrrole derivatives and methods of making and using same. WO2015126912. 2015.
  • Bajwa N, Liao C, Nikolovska-Coleska Z. Inhibitors of the anti-apoptotic Bcl-2 proteins: a patent review. Expert Opin Ther Pat. 2012;22:37–55.
  • Reed JC, Pellecchia M. Apoptosis-based therapies for hematologic malignancies. Blood. 2005;106:408–418.
  • Oltersdorf T, Elmore SW, Shoemaker AR, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435:677–681.
  • Del V, Moore G, Schlis KD, et al. BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia. Leukemia. 2008;111:2300–2309.
  • Kline MP, Rajkumar SV, Timm MM, et al. ABT-737, an inhibitor of Bcl-2 family proteins, is a potent inducer of apoptosis in multiple myeloma cells. Leukemia. 2007;21:1549–1560.
  • Buron N, Porceddu M, Brabant M, et al. Use of human cancer cell lines mitochondria to explore the mechanisms of BH3 peptides and ABT-737-induced mitochondrial membrane permeabilization. PLoS One. 2010;5:e9924.
  • Konopleva M, Contractor R, Tsao T, et al. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell. 2006;10:375–388.
  • Del Gaizo Moore V, Brown JR, Certo M, et al. Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737. J Clin Invest. 2007;117:112–121.
  • High LM, Szymanska B, Wilczynska-Kalak U, et al. The Bcl-2 homology domain 3 mimetic ABT-737 targets the apoptotic machinery in acute lymphoblastic leukemia resulting in synergistic in vitro and in vivo interactions with established drugs. Mol Pharmacol. 2010;77:483–494.
  • Tse C, Shoemaker AR, Adickes J, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–3428.
  • Shoemaker AR, Mitten MJ, Adickes J, et al. Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models. Clin Cancer Res. 2008;14:3268–3277.
  • Wilson WH, O’Connor OA, Czuczman MS, et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol. 2010;11:1149–1159.
  • Roberts AW, Seymour JF, Brown JR, et al. Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease. J Clin Oncol. 2012;30:488–496.
  • Rudin CM, Hann CL, Garon EB, et al. Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in patients with relapsed small cell lung cancer. Clin Cancer Res. 2012;18:3163–3169.
  • Gandhi L, Camidge DR, Ribeiro De Oliveira M, et al. Phase I study of Navitoclax (ABT-263), a novel Bcl-2 family inhibitor, in patients with small-cell lung cancer and other solid tumors. J Clin Oncol. 2011;29:909–916.
  • Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013;19:202–208.
  • ClinicalTrials.gov. [cited 2016 Mar 11] Available from: https://clinicaltrials.gov/ct2/results?term=ABT-199&Search=Search
  • Roche. US FDA grants breakthrough therapy designation for investigational Bcl-2 inhibitor venetoclax in 17p deletion relapsed-refractory chronic lymphocytic leukemia. [cited 2016 Mar 11]. Available from: http://www.roche.com/investors/updates/inv-update-2015-05-07.htm
  • Genentech: Press Releases |FDA Grants Priority Review for Venetoclax New Drug Application. [cited 2016 Mar 11]. Available from: http://www.gene.com/media/press-releases/14616/2016-01-12/fda-grants-priority-review-for-venetocla
  • FDA approves new drug for chronic lymphocytic leukemia in patients with a specific chromosomal abnormality. [cited 2016 June 16]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm495253.htm
  • Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–313.
  • Aichberger KJ, Mayerhofer M, Krauth M-T, et al. Identification of mcl-1 as a BCR/ABL-dependent target in chronic myeloid leukemia (CML): evidence for cooperative antileukemic effects of imatinib and mcl-1 antisense oligonucleotides. Blood. 2005;105:3303–3311.
  • Brotin E, Meryet-Figuière M, Simonin K, et al. Bcl-XL and MCL-1 constitute pertinent targets in ovarian carcinoma and their concomitant inhibition is sufficient to induce apoptosis. Int J Cancer. 2010;126:885–895.
  • Chetoui N, Sylla K, Gagnon-Houde J-V, et al. Down-regulation of mcl-1 by small interfering RNA sensitizes resistant melanoma cells to fas-mediated apoptosis. Mol Cancer Res. 2008;6:42–52.
  • Cho-Vega JH, Rassidakis GZ, Admirand JH, et al. MCL-1 expression in B-cell non-Hodgkin’s lymphomas. Hum Pathol. 2004;35:1095–1100.
  • Glaser SP, Lee EF, Trounson E, et al. Anti-apoptotic Mcl-1 is essential for the development and sustained growth of acute myeloid leukemia. Genes Dev. 2012;26:120–125.
  • Sieghart W, Losert D, Strommer S, et al. Mcl-1 overexpression in hepatocellular carcinoma: a potential target for antisense therapy. J Hepatol. 2006;44:151–157.
  • Zhang H, Guttikonda S, Roberts L, et al. Mcl-1 is critical for survival in a subgroup of non-small-cell lung cancer cell lines. Oncogene. 2011;30:1963–1968.
  • Wei S-H, Dong K, Lin F, et al. Inducing apoptosis and enhancing chemosensitivity to gemcitabine via RNA interference targeting Mcl-1 gene in pancreatic carcinoma cell. Cancer Chemother Pharmacol. 2008;62:1055–1064.
  • Krajewski S, Krajewska M, Ehrmann J, et al. Immunohistochemical analysis of Bcl-2, Bcl-X, Mcl-1, and Bax in tumors of central and peripheral nervous system origin. Am J Pathol. 1997;150:805–814.
  • Krajewska M, Krajewski S, Epstein JI, et al. Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers. Am J Pathol. 1996;148:1567–1576.
  • Krajewska M, Fenoglio-Preiser CM, Krajewski S, et al. Immunohistochemical analysis of Bcl-2 family proteins in adenocarcinomas of the stomach. Am J Pathol. 1996;149:1449–1457.
  • Wertz IE, Kusam S, Lam C, et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature. 2011;471:110–114.
  • Taniai M, Grambihler A, Higuchi H, et al. Mcl-1 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in human cholangiocarcinoma cells. Cancer Res. 2004;64:3517–3524.
  • Lin X, Morgan-Lappe S, Huang X, et al. “Seed” analysis of off-target siRNAs reveals an essential role of Mcl-1 in resistance to the small-molecule Bcl-2/Bcl-XL inhibitor ABT-737. Oncogene. 2007;26:3972–3979.
  • Akgul C. Mcl-1 is a potential therapeutic target in multiple types of cancer. Cell Mol Life Sci. 2009;66:1326–1336.
  • Keuling AM, Felton KEA, Parker AAM, et al. RNA silencing of Mcl-1 enhances ABT-737-mediated apoptosis in melanoma: role for a caspase-8-dependent pathway. PLoS One. 2009;4:e6651.
  • Thallinger C, Wolschek MF, Wacheck V, et al. Mcl-1 antisense therapy chemosensitizes human melanoma in a SCID mouse xenotransplantation model. J Invest Dermatol. 2003;120:1081–1086.
  • Park C-M, Bruncko M, Adickes J, et al. Discovery of an orally bioavailable small molecule inhibitor of prosurvival B-cell lymphoma 2 proteins. J Med Chem. 2008;51:6902–6915.
  • Wendt MD. Discovery of ABT-263, a Bcl-family protein inhibitor: observations on targeting a large protein-protein interaction. Expert Opin Drug Discov. 2008;3:1123–1143.
  • Seymour JF, Gerecitano JF, Kahl BS, et al. The single-agent Bcl-2 inhibitor ABT-199 (GDC-0199) in patients with Relapsed/Refractory (R/R) Non-Hodgkin Lymphoma (NHL): responses observed in all Mantle Cell Lymphoma (MCL) patients. Blood. 2013;122:1789.
  • Paper: preliminary results of a phase 2, open-label study of venetoclax (ABT-199 /GDC-0199) monotherapy in patients with chronic lymphocytic leukemia relapsed after or refractory to ibrutinib or idelalisib therapy. [cited 2016 Mar 29]. Available from: https://ash.confex.com/ash/2015/webprogram/Paper81347.html
  • Wendt MD, Shen W, Dickman DA, et al. Apoptosis promoters. WO2007008627. 2007.
  • Petros AM, Swann SL, Song D, et al. Fragment-based discovery of potent inhibitors of the anti-apoptotic MCL-1 protein. Bioorg Med Chem Lett. 2014;24:1484–1488.
  • Bruncko M, Song X, Ding H, et al. 7-Nonsubstituted indole MCL-1 inhibitors. WO2008130970. 2008.
  • Elmore SW, Souers AJ, Bruncko M, et al. 7-Substituted indole MCL-1 inhibitors. WO2008131000. 2008.
  • Tanaka Y, Aikawa K, Nishida G, et al. Discovery of potent Mcl-1/Bcl-xL dual inhibitors by using a hybridization strategy based on structural analysis of target proteins. J Med Chem. 2013;56:9635–9645.
  • Bruncko M, Wang L, Sheppard GS, et al. Structure-guided design of a series of MCL-1 inhibitors with high affinity and selectivity. J Med Chem. 2015;58:2180–2194.
  • Leverson JD, Zhang H, Chen J, et al. Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax). Cell Death Dis. 2015;6:e1590.
  • Lee T, Pelz NF, Belmar J, et al. Substituted benzofuran, benzothiophene and indole MCL-1 inhibitors. WO2014047427. 2014.
  • Friberg A, Vigil D, Zhao B, et al. Discovery of Potent Myeloid Cell Leukemia 1 (Mcl-1) inhibitors using fragment-based methods and structure-based design. J Med Chem. 2013;56:15–30.
  • Burke JP, Bian Z, Shaw S, et al. Discovery of tricyclic indoles that potently inhibit Mcl-1 using fragment-based methods and structure-based design. J Med Chem. 2015;58:3794–3805.
  • Pelz NF, Bian Z, Zhao B, et al. Discovery of 2-Indole-acylsulfonamide Myeloid Cell Leukemia 1 (Mcl-1) inhibitors using fragment-based methods. J Med Chem. 2016;59:2054-2066.
  • Mao H, Hajduk PJ, Craig R, et al. Rational design of diflunisal analogues with reduced affinity for human serum albumin. J Am Chem Soc. 2001;123:10429–10435.
  • Lee T, Kim K, Christov PP, et al. Substituted indole MCL-1 inhibitors. WO2015031608. 2015.
  • Lee T, Bian Z, Belmar J, et al. Substituted indole MCL-1 inhibitors. WO2015148854. 2015.
  • Nicolovska-Coleska Z, Showalter HD, Liao C, et al. Small molecule inhibitors of MCL-1 and uses thereof. WO2013052943. 2013.
  • Abulwerdi FA, Liao C, Mady AS, et al. 3-substituted-N-(4-hydroxynaphthalen-1-yl)arylsulfonamides as a novel class of selective Mcl-1 inhibitors: structure-based design, synthesis, SAR, and biological evaluation. J Med Chem. 2014;57:4111–4133.
  • Nikolovska-Coleska Z, Bajwa N, Liao C, et al. Small molecule inhibitors of mcl-1 and uses thereof. WO2013149124. 2013.
  • Nikolovska-Coleska Z, Abulwerdi F, Showalter H, et al. Small molecule inhibitors of mcl-1 and uses thereof. WO2015153959. 2015.
  • Walensky LD, Stewart ML, Cohen N Small molecules for the modulation of mcl-1 and methods of modulatiing cell death, cell division, cell differentiation and methods of treating disorders. WO2011094708. 2011.
  • Walensky LD Inhibition of MCL-1 and/or BFL-1/A1. WO2013142281. 2013.
  • Wang G, Nikolovska-Coleska Z, Yang C-Y, et al. Structure-based design of potent small-molecule inhibitors of anti-apoptotic Bcl-2 proteins. J Med Chem. 2006;49:6139–6142.
  • Cohen NA, Stewart ML, Gavathiotis E, et al. A competitive stapled peptide screen identifies a selective small molecule that overcomes MCL-1-dependent leukemia cell survival. Chem Biol. 2012;19:1175–1186.
  • Lee S, Wales TE, Escudero S, et al. Allosteric inhibition of antiapoptotic mcl-1. Nat Struct Mol Biol. 2016 Jun;23(6):600–607.
  • Cardone MH, Kolodziej AF, Richard D Compositions and methods useful for treating diseases. WO2012122370. 2012.
  • Richard DJ, Lena R, Bannister T, et al. Hydroxyquinoline-derived compounds and analoguing of selective Mcl-1 inhibitors using a functional biomarker. Bioorg Med Chem. 2013;21:6642–6649.
  • Chai CL, Bermardo L, Paul H, et al. Biarylrhodanine and pyridylrhodanine compounds and their use. WO2010024783. 2010.
  • Kotschy A, Szlávik Z, Csékei M, et al. New thienopyrimidine derivatives, a process for their preparation and pharmaceutical compositions containing them. WO2015097123. 2015.
  • Servier announces expansion of collaboration for the development and commercialization of anticancer drug candidates targeting apoptosis. [cited 2016 June 16]. Available from: http://www.servier.com/content/servier-announces-expansion-collaboration-development-and-commercialization-anticancer-drug
  • Brown SP, Li Y, Lizarzaburu ME, et al. Tetrahydronaphthalene derivatives that inhibit mcl-1 protein. WO2016033486. 2016.
  • Drennen B, Scheenstra JA, Yap JL, et al. Structural re-engineering of the α-helix mimetic JY-1-106 into small molecules: disruption of the Mcl-1-Bak-BH3 protein-protein interaction with 2,6-di-substituted nicotinates. Chem Med Chem. 2016;11:827-833.
  • Lanning ME, Yu W, Yap JL, et al. Structure-based design of N-substituted 1-Hydroxy-4-sulfamoyl-2-naphthoates as selective inhibitors of the Mcl-1 oncoprotein. Eur J Med Chem. 2016;113:273-292.
  • Chen L, Wilder PT, Drennen B, et al. Structure-based design of 3-carboxy-substituted 1,2,3,4-tetrahydroquinolines as inhibitors of myeloid cell leukemia-1 (Mcl-1). Org Biomol Chem. 2016;14:5505-5510.
  • Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:311–322.
  • A study to compare the efficacy and safety of Obinutuzumab + GDC-0199 versus obinutuzumab + chlorambucil in patients with chronic lymphocytic leukemia - Full Text View - ClinicalTrials.gov. [cited 2016 Mar 29]. Available from: https://clinicaltrials.gov/ct2/show/NCT02242942
  • A study of GDC-0199 (ABT-199) plus mabthera/rituxan (rituximab) compared with bendamustine plus mabthera/rituxan (rituximab) in patients with relapsed or refractory chronic lymphocytic leukemia - Full Text View - ClinicalTrials.gov. [cited 2016 Mar 29]. Available from: https://clinicaltrials.gov/ct2/show/NCT02005471
  • Vo -T-T, Ryan J, Carrasco R, et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell. 2012;151:344–355.
  • Ni Chonghaile T, Sarosiek KA, Vo -T-T, et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science. 2011;334:1129–1133.
  • Touzeau C, Ryan J, Guerriero J, et al. BH3 profiling identifies heterogeneous dependency on Bcl-2 family members in multiple myeloma and predicts sensitivity to BH3 mimetics. Leukemia. 2015;30:761–764.
  • Deng J, Carlson N, Takeyama K, et al. BH3 profiling identifies three distinct classes of apoptotic blocks to predict response to ABT-737 and conventional chemotherapeutic agents. Cancer Cell. 2007;12:171–185.
  • La Thangue NB, Kerr DJ. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat Rev Clin Oncol. 2011;8:587–596.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.