686
Views
15
CrossRef citations to date
0
Altmetric
Review

Therapeutic inhibition of BCL-2 and related family members

&
Pages 293-301 | Received 07 Sep 2016, Accepted 30 Jan 2017, Published online: 09 Feb 2017

References

  • Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988 Sep 29;335:440–442.
  • Tsujimoto Y, Cossman J, Jaffe E, et al. Involvement of the bcl-2 gene in human follicular lymphoma. Science. 1985 Jun 21;228:1440–1443.
  • Reed JC, Alford SE, Kothari A, et al. Bcl-2-family proteins and hematologic malignancies: history and future prospects. Blood. 2008 Apr 1;111:3322–3330.
  • Delbridge AR, Grabow S, Strasser A, et al. Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nat Rev Cancer. 2016;16:99–109.
  • Miyashita T, Reed JC. Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell line. Blood. 1993 Jan 1;81:151–157.
  • Vaux DL, Weissman IL, Kim SK. Prevention of programmed cell death in Caenorhabditis elegans by human bcl-2. Science. 1992 Dec 18;258:1955–1957.
  • Hengartner MO, Horvitz C HR. elegans cell survival gene ced-9 encodes a functional homolog of the mammalian proto-oncogene bcl-2. Cell. 1994 Feb 25;76:665–676.
  • Vela L, Marzo I. Bcl-2 family of proteins as drug targets for cancer chemotherapy: the long way of BH3 mimetics from bench to bedside. Curr Opin Pharmacol. 2015 Aug;23:74–81.
  • Hogg SJ, Newbold A, Vervoort SJ, et al. BET inhibition induces apoptosis in aggressive B-cell lymphoma via epigenetic regulation of BCL-2 family members. Mol Cancer Ther. 2016;15:2030–2041.
  • May WS, Tyler PG, Ito T, et al. Interleukin-3 and bryostatin-1 mediate hyperphosphorylation of Bcl-2 alpha in association with suppression of apoptosis. J Biol Chem. 1994 Oct 28;269:26865–26870.
  • Ito T, Deng X, Carr B, et al. Bcl-2 phosphorylation required for anti-apoptosis function. J Biol Chem. 1997 May 02;272:11671–11673.
  • Basu A, Haldar S. Identification of a novel Bcl-xL phosphorylation site regulating the sensitivity of taxol- or 2-methoxyestradiol-induced apoptosis. FEBS Lett. 2003 Mar 13;538:41–47.
  • Maurer U, Charvet C, Wagman AS, et al. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell. 2006 Mar 17;21:749–760.
  • Nifoussi SK, Vrana JA, Domina AM, et al. Thr 163 phosphorylation causes Mcl-1 stabilization when degradation is independent of the adjacent GSK3-targeted phosphodegron, promoting drug resistance in cancer. Plos One. 2012;7:e47060.
  • Helmreich EJM. The biochemistry of cell signalling. Oxford; New York: Oxford University Press; 2001.
  • Coultas L, Strasser A, Coultas L, et al. The role of the Bcl-2 protein family in cancer. [Review] [83 refs]. Semin Cancer Biol. 2003;13:115–123.
  • Linette GP, Hess JL, Sentman CL, et al. Peripheral T-cell lymphoma in lckpr-bcl-2 transgenic mice. Blood. 1995 Aug 15;86:1255–1260.
  • Jager R, Herzer U, Schenkel J, et al. Overexpression of Bcl-2 inhibits alveolar cell apoptosis during involution and accelerates c-myc-induced tumorigenesis of the mammary gland in transgenic mice. Oncogene. 1997 Oct 9;15:1787–1795.
  • Alford SE, Kothari A, Loeff FC, et al. BH3 inhibitor sensitivity and Bcl-2 dependence in primary acute lymphoblastic leukemia cells. Cancer Res. 2015;75:1366–1375.
  • Fresquet V, Rieger M, Carolis C, et al. Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma. Blood. 2014;123:4111–4119.
  • 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.
  • Brunelle JK, Letai A. Control of mitochondrial apoptosis by the Bcl-2 family. J Cell Sci. 2009;122:437–441.
  • van Delft MF, Wei AH, Mason KD, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10:389–399.
  • O’Brien SM, Cunningham CC, Golenkov AK, et al. Phase I to II multicenter study of oblimersen sodium, a Bcl-2 antisense oligonucleotide, in patients with advanced chronic lymphocytic leukemia. J Clin Oncol. 2005 Oct 20;23:7697–7702.
  • Frantz S. Lessons learnt from Genasense’s failure. Nat Rev Drug Discov. 2004 Jul;3:542–543.
  • Oltersdorf T, Elmore SW, Shoemaker AR, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005 06/02/print;435:677–681.
  • 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.
  • Wojciechowski S, Tripathi P, Bourdeau T, et al. Bim/Bcl-2 balance is critical for maintaining naive and memory T cell homeostasis. J Exp Med. 2007 Jul 09;204:1665–1675.
  • Suryani S, Carol H, Chonghaile TN, et al. Cell and molecular determinants of in vivo efficacy of the BH3 mimetic ABT-263 against pediatric acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2014 Sep 1;20:4520–4531.
  • 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 Feb 10;30:488–496.
  • Cleary JM, Lima CM, Hurwitz HI, et al. A phase I clinical trial of navitoclax, a targeted high-affinity Bcl-2 family inhibitor, in combination with gemcitabine in patients with solid tumors. Invest New Drugs. 2014;32:937–945.
  • Nguyen M, Marcellus RC, Roulston A, et al. Small molecule obatoclax (GX15-070) antagonizes MCL-1 and overcomes MCL-1-mediated resistance to apoptosis. Proc Natl Acad Sci U S A. 2007;104:19512–19517.
  • Koh GS, Jiang N, Dong DF, et al. BH3-mimetics, ABT-737 and obatoclax, work synergistically to induce cell death in leukemic cell lines. Blood (ASH Annual Meeting Abstracts). 2010;116:1850.
  • Shore GC, Viallet J. Modulating the bcl-2 family of apoptosis suppressors for potential therapeutic benefit in cancer. Hematology Am Soc Hematol Educ Program. 2005;226–230.
  • Schimmer AD, O’Brien S, Kantarjian H, et al. A phase I study of the pan bcl-2 family inhibitor obatoclax mesylate in patients with advanced hematologic malignancies. Clin Cancer Res. 2008;14:8295–8301.
  • O’Brien SM, Claxton DF, Crump M, et al. Phase I study of obatoclax mesylate (GX15-070), a small molecule pan-Bcl-2 family antagonist, in patients with advanced chronic lymphocytic leukemia. Blood. 2009;113:299–305.
  • Balakrishnan K, Burger JA, Wierda WG, et al. AT-101 induces apoptosis in CLL B cells and overcomes stromal cell-mediated Mcl-1 induction and drug resistance. Blood. 2009;113:149–153.
  • Zerp SF, Stoter R, Kuipers G, et al. AT-101, a small molecule inhibitor of anti-apoptotic Bcl-2 family members, activates the SAPK/JNK pathway and enhances radiation-induced apoptosis. Radiat Oncol. 2009;4:47.
  • McGregor N, Patel L, Craig M, et al. AT-101 (R-(-)-gossypol acetic acid) enhances the effectiveness of androgen deprivation therapy in the VCaP prostate cancer model. J Cell Biochem. 2010;110:1187–1194.
  • Liu G, Wk K, Wilding G, et al. An open-label, multicenter, phase I/II study of single-agent AT-101 in men with castrate-resistant prostate cancer. Clin Cancer Res. 2009;15:3172–3176.
  • 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.
  • Ham J, Costa C, Sano R, et al. Exploitation of the apoptosis-primed state of MYCN-amplified neuroblastoma to develop a potent and specific targeted therapy combination. Cancer Cell. 2016 Feb;8(29):159–172.
  • Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016 Jan 28;374:311–322.
  • Ma S, Brander DM, Seymour JF, et al. Deep and durable responses following venetoclax (ABT-199 /GDC-0199) combined with rituximab in patients with relapsed/refractory chronic lymphocytic leukemia: results from a phase 1b study. Blood. 2015;126:830.
  • Flinn IW, Brunvand M, Choi MY, et al. Safety and efficacy of a combination of venetoclax (GDC-0199/ABT-199) and obinutuzumab in patients with relapsed/refractory or previously untreated chronic lymphocytic leukemia - results from a phase 1b study (GP28331). Blood. 2015;126:494.
  • Deng J, Isik E, Fernandes SM, et al. Ibrutinib therapy increases BCL-2 dependence and enhances sensitivity to venetoclax in CLL. Blood. 2015;126:490.
  • Kumar SKVR, Kaufman JL, Mikhael JR, et al. Safety and efficacy of venetoclax (ABT-199/GDC-0199) monotherapy for relapsed/refractory multiple myeloma: phase 1 preliminary results. Blood. 2015;126:4219.
  • Moreau PC-KA, Roberts AW, Agarwal AB, et al. Safety and efficacy of venetoclax (ABT-199/GDC-0199) in combination with bortezomib and dexamethasone in relapsed/refractory multiple myeloma: phase 1b results. Blood. 2015;126:3038.
  • Gerecitano JF, Roberts AW, Seymour JF, et al. A phase 1 study of venetoclax (ABT-199 /GDC-0199) monotherapy in patients with relapsed/refractory non-hodgkin lymphoma. Blood. 2015;126:254.
  • De Vos S, Swinnen L, Kozloff M, et al. A dose-escalation study of venetoclax (ABT-199/GDC-0199) in combination with bendamustine and rituximab in patients with relapsed or refractory non-hodgkin’s lymphoma. Blood. 2015;126:255.
  • Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6:1106–1117.
  • Chan SM, Thomas D, Corces-Zimmerman MR, et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat Med. 2015;21:178–184.
  • DiNardo C, Pollyea D, Pratz K, et al. A phase 1b study of venetoclax (ABT-199/GDC-0199) in combination with decitabine or azacitidine in treatment-naive patients with acute myelogenous leukemia who are ≥to 65 years and not eligible for standard induction therapy. Blood. 2015;126:327.
  • Amundson SA, Myers TG, Scudiero D, et al. An informatics approach identifying markers of chemosensitivity in human cancer cell lines. Cancer Res. 2000 Nov;1(60):6101–6110.
  • Baranski Z, De Jong Y, Ilkova T, et al. Pharmacological inhibition of Bcl-xL sensitizes osteosarcoma to doxorubicin. Oncotarget. 2015;6:36113–36125.
  • Lessene G, Czabotar PE, Sleebs BE, et al. Structure-guided design of a selective BCL-X(L) inhibitor. Nat Chem Biol. 2013 Jun;9:390–397.
  • Tao ZF, Hasvold L, Wang L, et al. Discovery of a potent and selective BCL-XL inhibitor with in vivo activity. ACS Med Chem Lett. 2014 Oct;9(5):1088–1093.
  • Kozopas KM, Yang T, Buchan HL, et al. MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2. Proc Natl Acad Sci U S A. 1993 Apr 15;90:3516–3520.
  • Thomas RL, Roberts DJ, Kubli DA, et al. Loss of MCL-1 leads to impaired autophagy and rapid development of heart failure. Genes Dev. 2013 Jun 15;27:1365–1377.
  • Wang X, Bathina M, Lynch J, et al. Deletion of MCL-1 causes lethal cardiac failure and mitochondrial dysfunction. Genes Dev. 2013 Jun 15;27:1351–1364.
  • 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.
  • Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010 Feb;18(463):899–905.
  • Goodwin CM, Rossanese OW, Olejniczak ET, et al. Myeloid cell leukemia-1 is an important apoptotic survival factor in triple-negative breast cancer. Cell Death Differ. 2015;22:2098–2106.
  • Nakajima W, Hicks MA, Tanaka N, et al. Noxa determines localization and stability of MCL-1 and consequently ABT-737 sensitivity in small cell lung cancer. Cell Death Dis. 2014;5:e1052.
  • Varadarajan S, Vogler M, Butterworth M, et al. Evaluation and critical assessment of putative MCL-1 inhibitors. Cell Death Differ. 2013;20:1475–1484.
  • Besbes S, Mirshahi M, Pocard M, et al. New dimension in therapeutic targeting of BCL-2 family proteins. Oncotarget. 2015 May 30;6:12862–12871.
  • Doi K, Li R, Sung SS, et al. Discovery of marinopyrrole A (maritoclax) as a selective Mcl-1 antagonist that overcomes ABT-737 resistance by binding to and targeting Mcl-1 for proteasomal degradation. J Biol Chem. 2012;287:10224–10235.
  • Shellman YG, Pandey MK, Gowda K, et al. Proteasomal degradation of Mcl-1 by maritoclax induces apoptosis and enhances the efficacy of ABT-737 in melanoma cells. Plos ONE. 2013;8:e78570.
  • Varadarajan S, Poornima P, Milani M, et al. Maritoclax and dinaciclib inhibit MCL-1 activity and induce apoptosis in both a MCL-1-dependent and -independent manner. Oncotarget. 2015 May 20;6:12668–12681.
  • Kotschy A, Szlavik Z, Murray J, et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature. 2016 Oct 19;538:477–482.

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.