Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Pan-Bcl-2 inhibitor Obatoclax is a potent late stage autophagy inhibitor in colorectal cancer cells independent of canonical autophagy signaling

Authors: Bruno Christian Koehler, Adam Jassowicz, Anna-Lena Scherr, Stephan Lorenz, Praveen Radhakrishnan, Nicole Kautz, Christin Elssner, Johanna Weiss, Dirk Jaeger, Martin Schneider, Henning Schulze-Bergkamen

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Colorectal cancer is the third most common malignancy in humans and novel therapeutic approaches are urgently needed. Autophagy is an evolutionarily highly conserved cellular process by which cells collect unnecessary organelles or misfolded proteins and subsequently degrade them in vesicular structures in order to refuel cells with energy. Dysregulation of the complex autophagy signaling network has been shown to contribute to the onset and progression of cancer in various models. The Bcl-2 family of proteins comprises central regulators of apoptosis signaling and has been linked to processes involved in autophagy. The antiapoptotic members of the Bcl-2 family of proteins have been identified as promising anticancer drug targets and small molecules inhibiting those proteins are in clinical trials.

Methods

Flow cytometry and colorimetric assays were used to assess cell growth and cell death. Long term 3D cell culture was used to assess autophagy in a tissue mimicking environment in vitro. RNA interference was applied to modulate autophagy signaling. Immunoblotting and q-RT PCR were used to investigate autophagy signaling. Immunohistochemistry and fluorescence microscopy were used to detect autophagosome formation and autophagy flux.

Results

This study demonstrates that autophagy inhibition by obatoclax induces cell death in colorectal cancer (CRC) cells in an autophagy prone environment. Here, we demonstrate that pan-Bcl-2 inhibition by obatoclax causes a striking, late stage inhibition of autophagy in CRC cells. In contrast, ABT-737, a Mcl-1 sparing Bcl-2 inhibitor, failed to interfere with autophagy signaling. Accumulation of p62 as well as Light Chain 3 (LC3) was observed in cells treated with obatoclax. Autophagy inhibition caused by obatoclax is further augmented in stressful conditions such as starvation. Furthermore, our data demonstrate that inhibition of autophagy caused by obatoclax is independent of the essential pro-autophagy proteins Beclin-1, Atg7 and Atg12.

Conclusions

The objective of this study was to dissect the contribution of Bcl-2 proteins to autophagy in CRC cells and to explore the potential of Bcl-2 inhibitors for autophagy modulation. Collectively, our data argue for a Beclin-1 independent autophagy inhibition by obatoclax. Based on this study, we recommend the concept of autophagy inhibition as therapeutic strategy for CRC.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917.CrossRefPubMed Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–917.CrossRefPubMed
2.
go back to reference Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368(19):1845–6.CrossRefPubMed Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. N Engl J Med. 2013;368(19):1845–6.CrossRefPubMed
3.
go back to reference Huett A, Xavier RJ. Autophagy at the gut interface: mucosal responses to stress and the consequences for inflammatory bowel diseases. Inflamm Bowel Dis. 2010;16(1):152–74.PubMedCentralCrossRefPubMed Huett A, Xavier RJ. Autophagy at the gut interface: mucosal responses to stress and the consequences for inflammatory bowel diseases. Inflamm Bowel Dis. 2010;16(1):152–74.PubMedCentralCrossRefPubMed
4.
go back to reference Lai K, Killingsworth MC, Lee CS. The significance of autophagy in colorectal cancer pathogenesis and implications for therapy. J Clin Pathol. 2014;67(10):854–8.CrossRefPubMed Lai K, Killingsworth MC, Lee CS. The significance of autophagy in colorectal cancer pathogenesis and implications for therapy. J Clin Pathol. 2014;67(10):854–8.CrossRefPubMed
6.
go back to reference Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest. 2015;125(1):1–4.CrossRefPubMed Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest. 2015;125(1):1–4.CrossRefPubMed
7.
go back to reference Jiang X, Overholtzer M, Thompson CB. Autophagy in cellular metabolism and cancer. J Clin Invest. 2015;125(1):47–54.CrossRefPubMed Jiang X, Overholtzer M, Thompson CB. Autophagy in cellular metabolism and cancer. J Clin Invest. 2015;125(1):47–54.CrossRefPubMed
8.
go back to reference Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, et al. Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (−)-Gossypol (AT-101). BMC Cancer. 2015;15:224.PubMedCentralCrossRefPubMed Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, et al. Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (−)-Gossypol (AT-101). BMC Cancer. 2015;15:224.PubMedCentralCrossRefPubMed
10.
go back to reference Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell death & disease. 2013;4:e838.CrossRef Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell death & disease. 2013;4:e838.CrossRef
11.
go back to reference Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–26.CrossRefPubMed Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13(10):714–26.CrossRefPubMed
12.
go back to reference Goldberg SB, Supko JG, Neal JW, Muzikansky A, Digumarthy S, Fidias P, et al. A phase I study of erlotinib and hydroxychloroquine in advanced non-small-cell lung cancer. J Thorac Oncol. 2012;7(10):1602–8.PubMedCentralCrossRefPubMed Goldberg SB, Supko JG, Neal JW, Muzikansky A, Digumarthy S, Fidias P, et al. A phase I study of erlotinib and hydroxychloroquine in advanced non-small-cell lung cancer. J Thorac Oncol. 2012;7(10):1602–8.PubMedCentralCrossRefPubMed
13.
go back to reference Tang Y, Li M, Wang YL, Threadgill MD, Xiao M, Mou CF, et al. ART1 promotes starvation-induced autophagy: a possible protective role in the development of colon carcinoma. Am J Cancer Res. 2015;5(2):498–513.PubMedCentralPubMed Tang Y, Li M, Wang YL, Threadgill MD, Xiao M, Mou CF, et al. ART1 promotes starvation-induced autophagy: a possible protective role in the development of colon carcinoma. Am J Cancer Res. 2015;5(2):498–513.PubMedCentralPubMed
14.
go back to reference Hu F, Wei F, Wang Y, Wu B, Fang Y, Xiong B. EGCG synergizes the therapeutic effect of cisplatin and oxaliplatin through autophagic pathway in human colorectal cancer cells. J Pharmacol Sci. 2015;128(1):27–34.CrossRefPubMed Hu F, Wei F, Wang Y, Wu B, Fang Y, Xiong B. EGCG synergizes the therapeutic effect of cisplatin and oxaliplatin through autophagic pathway in human colorectal cancer cells. J Pharmacol Sci. 2015;128(1):27–34.CrossRefPubMed
15.
go back to reference Zhai H, Fesler A, Ba Y, Wu S, Ju J. Inhibition of colorectal cancer stem cell survival and invasive potential by hsa-miR-140-5p mediated suppression of Smad2 and autophagy. Oncotarget. 2015. Zhai H, Fesler A, Ba Y, Wu S, Ju J. Inhibition of colorectal cancer stem cell survival and invasive potential by hsa-miR-140-5p mediated suppression of Smad2 and autophagy. Oncotarget. 2015.
16.
17.
go back to reference Koehler BC, Jager D, Schulze-Bergkamen H. Targeting cell death signaling in colorectal cancer: current strategies and future perspectives. World J Gastroenterol. 2014;20(8):1923–34.PubMedCentralCrossRefPubMed Koehler BC, Jager D, Schulze-Bergkamen H. Targeting cell death signaling in colorectal cancer: current strategies and future perspectives. World J Gastroenterol. 2014;20(8):1923–34.PubMedCentralCrossRefPubMed
18.
go back to reference 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;23:74–81.CrossRefPubMed 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;23:74–81.CrossRefPubMed
19.
go back to reference Kang YJ, Lu MK, Guan KL. The TSC1 and TSC2 tumor suppressors are required for proper ER stress response and protect cells from ER stress-induced apoptosis. Cell Death Differ. 2011;18(1):133–44.PubMedCentralCrossRefPubMed Kang YJ, Lu MK, Guan KL. The TSC1 and TSC2 tumor suppressors are required for proper ER stress response and protect cells from ER stress-induced apoptosis. Cell Death Differ. 2011;18(1):133–44.PubMedCentralCrossRefPubMed
20.
go back to reference Fu LL, Cheng Y, Liu B. Beclin-1: autophagic regulator and therapeutic target in cancer. Int J Biochem Cell Biol. 2013;45(5):921–4.CrossRefPubMed Fu LL, Cheng Y, Liu B. Beclin-1: autophagic regulator and therapeutic target in cancer. Int J Biochem Cell Biol. 2013;45(5):921–4.CrossRefPubMed
21.
go back to reference Koehler BC, Scherr AL, Lorenz S, Elssner C, Kautz N, Welte S, et al. Pan-Bcl-2 inhibitor obatoclax delays cell cycle progression and blocks migration of colorectal cancer cells. PLoS ONE. 2014;9(9):e106571.PubMedCentralCrossRefPubMed Koehler BC, Scherr AL, Lorenz S, Elssner C, Kautz N, Welte S, et al. Pan-Bcl-2 inhibitor obatoclax delays cell cycle progression and blocks migration of colorectal cancer cells. PLoS ONE. 2014;9(9):e106571.PubMedCentralCrossRefPubMed
22.
go back to reference Basit F, Cristofanon S, Fulda S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013;20(9):1161–73.PubMedCentralCrossRefPubMed Basit F, Cristofanon S, Fulda S. Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 2013;20(9):1161–73.PubMedCentralCrossRefPubMed
23.
go back to reference Yazbeck VY, Li C, Grandis JR, Zang Y, Johnson DE. Single-agent obatoclax (GX15-070) potently induces apoptosis and pro-survival autophagy in head and neck squamous cell carcinoma cells. Oral Oncol. 2014;50(2):120–7.PubMedCentralCrossRefPubMed Yazbeck VY, Li C, Grandis JR, Zang Y, Johnson DE. Single-agent obatoclax (GX15-070) potently induces apoptosis and pro-survival autophagy in head and neck squamous cell carcinoma cells. Oral Oncol. 2014;50(2):120–7.PubMedCentralCrossRefPubMed
24.
go back to reference Koehler BC, Scherr AL, Lorenz S, Urbanik T, Kautz N, Elssner C, et al. Beyond cell death - antiapoptotic bcl-2 proteins regulate migration and invasion of colorectal cancer cells in vitro. PLoS ONE. 2013;8(10):e76446.PubMedCentralCrossRefPubMed Koehler BC, Scherr AL, Lorenz S, Urbanik T, Kautz N, Elssner C, et al. Beyond cell death - antiapoptotic bcl-2 proteins regulate migration and invasion of colorectal cancer cells in vitro. PLoS ONE. 2013;8(10):e76446.PubMedCentralCrossRefPubMed
25.
go back to reference Koehler BC, Urbanik T, Vick B, Boger RJ, Heeger S, Galle PR, et al. TRAIL-induced apoptosis of hepatocellular carcinoma cells is augmented by targeted therapies. World J Gastroenterol. 2009;15(47):5924–35.PubMedCentralCrossRefPubMed Koehler BC, Urbanik T, Vick B, Boger RJ, Heeger S, Galle PR, et al. TRAIL-induced apoptosis of hepatocellular carcinoma cells is augmented by targeted therapies. World J Gastroenterol. 2009;15(47):5924–35.PubMedCentralCrossRefPubMed
26.
go back to reference Guo S, Liang Y, Murphy SF, Huang A, Shen H, Kelly DF, et al. A rapid and high content assay that measures cyto-ID-stained autophagic compartments and estimates autophagy flux with potential clinical applications. Autophagy. 2015;11(3):560–72.CrossRefPubMed Guo S, Liang Y, Murphy SF, Huang A, Shen H, Kelly DF, et al. A rapid and high content assay that measures cyto-ID-stained autophagic compartments and estimates autophagy flux with potential clinical applications. Autophagy. 2015;11(3):560–72.CrossRefPubMed
27.
go back to reference Grimaldi A, Santini D, Zappavigna S, Lombardi A, Misso G, Boccellino M, et al. Antagonistic effects of chloroquine on autophagy occurrence potentiate the anticancer effects of everolimus on renal cancer cells. Cancer Biol Ther. 2015;16(4):567–79.CrossRefPubMed Grimaldi A, Santini D, Zappavigna S, Lombardi A, Misso G, Boccellino M, et al. Antagonistic effects of chloroquine on autophagy occurrence potentiate the anticancer effects of everolimus on renal cancer cells. Cancer Biol Ther. 2015;16(4):567–79.CrossRefPubMed
28.
go back to reference Fukuda T, Oda K, Wada-Hiraike O, Sone K, Inaba K, Ikeda Y, et al. The anti-malarial chloroquine suppresses proliferation and overcomes cisplatin resistance of endometrial cancer cells via autophagy inhibition. Gynecol Oncol. 2015;137(3):538–45.CrossRefPubMed Fukuda T, Oda K, Wada-Hiraike O, Sone K, Inaba K, Ikeda Y, et al. The anti-malarial chloroquine suppresses proliferation and overcomes cisplatin resistance of endometrial cancer cells via autophagy inhibition. Gynecol Oncol. 2015;137(3):538–45.CrossRefPubMed
29.
30.
32.
go back to reference Komatsu M, Ichimura Y. Physiological significance of selective degradation of p62 by autophagy. FEBS Lett. 2010;584(7):1374–8.CrossRefPubMed Komatsu M, Ichimura Y. Physiological significance of selective degradation of p62 by autophagy. FEBS Lett. 2010;584(7):1374–8.CrossRefPubMed
33.
go back to reference Pierzynska-Mach A, Janowski PA, Dobrucki JW. Evaluation of acridine orange, LysoTracker Red, and quinacrine as fluorescent probes for long-term tracking of acidic vesicles. Cytometry A. 2014;85(8):729–37.CrossRefPubMed Pierzynska-Mach A, Janowski PA, Dobrucki JW. Evaluation of acridine orange, LysoTracker Red, and quinacrine as fluorescent probes for long-term tracking of acidic vesicles. Cytometry A. 2014;85(8):729–37.CrossRefPubMed
34.
go back to reference Moreno A, SantoDomingo J, Fonteriz RI, Lobaton CD, Montero M, Alvarez J. A confocal study on the visualization of chromaffin cell secretory vesicles with fluorescent targeted probes and acidic dyes. J Struct Biol. 2010;172(3):261–9.CrossRefPubMed Moreno A, SantoDomingo J, Fonteriz RI, Lobaton CD, Montero M, Alvarez J. A confocal study on the visualization of chromaffin cell secretory vesicles with fluorescent targeted probes and acidic dyes. J Struct Biol. 2010;172(3):261–9.CrossRefPubMed
35.
go back to reference Millot C, Millot JM, Morjani H, Desplaces A, Manfait M. Characterization of acidic vesicles in multidrug-resistant and sensitive cancer cells by acridine orange staining and confocal microspectrofluorometry. J Histochem Cytochem. 1997;45(9):1255–64.CrossRefPubMed Millot C, Millot JM, Morjani H, Desplaces A, Manfait M. Characterization of acidic vesicles in multidrug-resistant and sensitive cancer cells by acridine orange staining and confocal microspectrofluorometry. J Histochem Cytochem. 1997;45(9):1255–64.CrossRefPubMed
37.
go back to reference Mukhopadhyay S, Panda PK, Sinha N, Das DN, Bhutia SK. Autophagy and apoptosis: where do they meet? Apoptosis. 2014;19(4):555–66.CrossRefPubMed Mukhopadhyay S, Panda PK, Sinha N, Das DN, Bhutia SK. Autophagy and apoptosis: where do they meet? Apoptosis. 2014;19(4):555–66.CrossRefPubMed
38.
go back to reference Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta. 2013;1833(12):3448–59.CrossRefPubMed Nikoletopoulou V, Markaki M, Palikaras K, Tavernarakis N. Crosstalk between apoptosis, necrosis and autophagy. Biochim Biophys Acta. 2013;1833(12):3448–59.CrossRefPubMed
40.
go back to reference Kimura T, Takabatake Y, Takahashi A, Isaka Y. Chloroquine in cancer therapy: a double-edged sword of autophagy. Cancer Res. 2013;73(1):3–7.CrossRefPubMed Kimura T, Takabatake Y, Takahashi A, Isaka Y. Chloroquine in cancer therapy: a double-edged sword of autophagy. Cancer Res. 2013;73(1):3–7.CrossRefPubMed
41.
go back to reference Wilson CA, Browning JL. Death of HT29 adenocarcinoma cells induced by TNF family receptor activation is caspase-independent and displays features of both apoptosis and necrosis. Cell Death Differ. 2002;9(12):1321–33.CrossRefPubMed Wilson CA, Browning JL. Death of HT29 adenocarcinoma cells induced by TNF family receptor activation is caspase-independent and displays features of both apoptosis and necrosis. Cell Death Differ. 2002;9(12):1321–33.CrossRefPubMed
42.
go back to reference Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, et al. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature. 2015;524(7565):361–5.CrossRefPubMed Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, et al. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature. 2015;524(7565):361–5.CrossRefPubMed
43.
go back to reference Zhang Z, Guo M, Zhao S, Xu W, Shao J, Zhang F, et al. The update on transcriptional regulation of autophagy in normal and pathologic cells: A novel therapeutic target. Biomed Pharmacother. 2015;74:17–29.CrossRefPubMed Zhang Z, Guo M, Zhao S, Xu W, Shao J, Zhang F, et al. The update on transcriptional regulation of autophagy in normal and pathologic cells: A novel therapeutic target. Biomed Pharmacother. 2015;74:17–29.CrossRefPubMed
44.
go back to reference Polager S, Ofir M, Ginsberg D. E2F1 regulates autophagy and the transcription of autophagy genes. Oncogene. 2008;27(35):4860–4.CrossRefPubMed Polager S, Ofir M, Ginsberg D. E2F1 regulates autophagy and the transcription of autophagy genes. Oncogene. 2008;27(35):4860–4.CrossRefPubMed
45.
go back to reference Sun T, Li D, Wang L, Xia L, Ma J, Guan Z, et al. c-Jun NH2-terminal kinase activation is essential for up-regulation of LC3 during ceramide-induced autophagy in human nasopharyngeal carcinoma cells. J Transl Med. 2011;9:161.PubMedCentralCrossRefPubMed Sun T, Li D, Wang L, Xia L, Ma J, Guan Z, et al. c-Jun NH2-terminal kinase activation is essential for up-regulation of LC3 during ceramide-induced autophagy in human nasopharyngeal carcinoma cells. J Transl Med. 2011;9:161.PubMedCentralCrossRefPubMed
46.
go back to reference Kadowaki M, Karim MR. Cytosolic LC3 ratio as a quantitative index of macroautophagy. Methods Enzymol. 2009;452:199–213.CrossRefPubMed Kadowaki M, Karim MR. Cytosolic LC3 ratio as a quantitative index of macroautophagy. Methods Enzymol. 2009;452:199–213.CrossRefPubMed
47.
48.
go back to reference Bai H, Inoue J, Kawano T, Inazawa J. A transcriptional variant of the LC3A gene is involved in autophagy and frequently inactivated in human cancers. Oncogene. 2012;31(40):4397–408.CrossRefPubMed Bai H, Inoue J, Kawano T, Inazawa J. A transcriptional variant of the LC3A gene is involved in autophagy and frequently inactivated in human cancers. Oncogene. 2012;31(40):4397–408.CrossRefPubMed
49.
go back to reference Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4(2):151–75.PubMedCentralCrossRefPubMed Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4(2):151–75.PubMedCentralCrossRefPubMed
50.
go back to reference Su J, Liu F, Xia M, Xu Y, Li X, Kang J, et al. p62 participates in the inhibition of NF-kappaB signaling and apoptosis induced by sulfasalazine in human glioma U251 cells. Oncol Rep. 2015;34(1):235–43.PubMed Su J, Liu F, Xia M, Xu Y, Li X, Kang J, et al. p62 participates in the inhibition of NF-kappaB signaling and apoptosis induced by sulfasalazine in human glioma U251 cells. Oncol Rep. 2015;34(1):235–43.PubMed
51.
go back to reference Jain A, Rusten TE, Katheder N, Elvenes J, Bruun JA, Sjottem E, et al. p62/Sequestosome-1, Autophagy-related Gene 8, and Autophagy in Drosophila Are Regulated by Nuclear Factor Erythroid 2-related Factor 2 (NRF2), Independent of Transcription Factor TFEB. J Biol Chem. 2015;290(24):14945–62.CrossRefPubMed Jain A, Rusten TE, Katheder N, Elvenes J, Bruun JA, Sjottem E, et al. p62/Sequestosome-1, Autophagy-related Gene 8, and Autophagy in Drosophila Are Regulated by Nuclear Factor Erythroid 2-related Factor 2 (NRF2), Independent of Transcription Factor TFEB. J Biol Chem. 2015;290(24):14945–62.CrossRefPubMed
52.
go back to reference Mohamed A, Ayman A, Deniece J, Wang T, Kovach C, Siddiqui MT, et al. P62/Ubiquitin IHC Expression Correlated with Clinicopathologic Parameters and Outcome in Gastrointestinal Carcinomas. Frontiers in Oncology. 2015;5:70.PubMedCentralCrossRefPubMed Mohamed A, Ayman A, Deniece J, Wang T, Kovach C, Siddiqui MT, et al. P62/Ubiquitin IHC Expression Correlated with Clinicopathologic Parameters and Outcome in Gastrointestinal Carcinomas. Frontiers in Oncology. 2015;5:70.PubMedCentralCrossRefPubMed
53.
go back to reference Burdelski C, Reiswich V, Hube-Magg C, Kluth M, Minner S, Koop C, et al. Cytoplasmic Accumulation of Sequestosome 1 (p62) Is a Predictor of Biochemical Recurrence, Rapid Tumor Cell Proliferation, and Genomic Instability in Prostate Cancer. Clin Cancer Res. 2015;15(21):3471–9.CrossRef Burdelski C, Reiswich V, Hube-Magg C, Kluth M, Minner S, Koop C, et al. Cytoplasmic Accumulation of Sequestosome 1 (p62) Is a Predictor of Biochemical Recurrence, Rapid Tumor Cell Proliferation, and Genomic Instability in Prostate Cancer. Clin Cancer Res. 2015;15(21):3471–9.CrossRef
54.
go back to reference Shen Y, Yang J, Zhao J, Xiao C, Xu C, Xiang Y. The switch from ER stress-induced apoptosis to autophagy via ROS-mediated JNK/p62 signals: A survival mechanism in methotrexate-resistant choriocarcinoma cells. Exp Cell Res. 2015;334(2):207–18.CrossRefPubMed Shen Y, Yang J, Zhao J, Xiao C, Xu C, Xiang Y. The switch from ER stress-induced apoptosis to autophagy via ROS-mediated JNK/p62 signals: A survival mechanism in methotrexate-resistant choriocarcinoma cells. Exp Cell Res. 2015;334(2):207–18.CrossRefPubMed
55.
56.
go back to reference Erlich S, Mizrachy L, Segev O, Lindenboim L, Zmira O, Adi-Harel S, et al. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy. 2007;3(6):561–8.CrossRefPubMed Erlich S, Mizrachy L, Segev O, Lindenboim L, Zmira O, Adi-Harel S, et al. Differential interactions between Beclin 1 and Bcl-2 family members. Autophagy. 2007;3(6):561–8.CrossRefPubMed
57.
go back to reference Maiuri MC, Criollo A, Tasdemir E, Vicencio JM, Tajeddine N, Hickman JA, et al. BH3-only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy. 2007;3(4):374–6.CrossRefPubMed Maiuri MC, Criollo A, Tasdemir E, Vicencio JM, Tajeddine N, Hickman JA, et al. BH3-only proteins and BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin 1 and Bcl-2/Bcl-X(L). Autophagy. 2007;3(4):374–6.CrossRefPubMed
58.
go back to reference Mehrpour M, Esclatine A, Beau I, Codogno P. Overview of macroautophagy regulation in mammalian cells. Cell Res. 2010;20(7):748–62.CrossRefPubMed Mehrpour M, Esclatine A, Beau I, Codogno P. Overview of macroautophagy regulation in mammalian cells. Cell Res. 2010;20(7):748–62.CrossRefPubMed
60.
go back to reference Schwartz-Roberts JL, Shajahan AN, Cook KL, Warri A, Abu-Asab M, Clarke R. GX15-070 (obatoclax) induces apoptosis and inhibits cathepsin D- and L-mediated autophagosomal lysis in antiestrogen-resistant breast cancer cells. Mol Cancer Ther. 2013;12(4):448–59.PubMedCentralCrossRefPubMed Schwartz-Roberts JL, Shajahan AN, Cook KL, Warri A, Abu-Asab M, Clarke R. GX15-070 (obatoclax) induces apoptosis and inhibits cathepsin D- and L-mediated autophagosomal lysis in antiestrogen-resistant breast cancer cells. Mol Cancer Ther. 2013;12(4):448–59.PubMedCentralCrossRefPubMed
61.
go back to reference McCoy F, Hurwitz J, McTavish N, Paul I, Barnes C, O’Hagan B, et al. Obatoclax induces Atg7-dependent autophagy independent of beclin-1 and BAX/BAK. Cell death & disease. 2010;1:e108.CrossRef McCoy F, Hurwitz J, McTavish N, Paul I, Barnes C, O’Hagan B, et al. Obatoclax induces Atg7-dependent autophagy independent of beclin-1 and BAX/BAK. Cell death & disease. 2010;1:e108.CrossRef
62.
go back to reference Bonapace L, Bornhauser BC, Schmitz M, Cario G, Ziegler U, Niggli FK, et al. Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance. J Clin Invest. 2010;120(4):1310–23.PubMedCentralCrossRefPubMed Bonapace L, Bornhauser BC, Schmitz M, Cario G, Ziegler U, Niggli FK, et al. Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance. J Clin Invest. 2010;120(4):1310–23.PubMedCentralCrossRefPubMed
63.
go back to reference Wroblewski D, Jiang CC, Croft A, Farrelly ML, Zhang XD, Hersey P. OBATOCLAX and ABT-737 induce ER stress responses in human melanoma cells that limit induction of apoptosis. PLoS ONE. 2013;8(12), e84073.PubMedCentralCrossRefPubMed Wroblewski D, Jiang CC, Croft A, Farrelly ML, Zhang XD, Hersey P. OBATOCLAX and ABT-737 induce ER stress responses in human melanoma cells that limit induction of apoptosis. PLoS ONE. 2013;8(12), e84073.PubMedCentralCrossRefPubMed
64.
go back to reference Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ, Minhajuddin M, et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 2013;12(3):329–41.PubMedCentralCrossRefPubMed Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ, Minhajuddin M, et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 2013;12(3):329–41.PubMedCentralCrossRefPubMed
65.
go back to reference Sharma A, Singh K, Mazumder S, Hill BT, Kalaycio M, Almasan A. BECN1 and BIM interactions with MCL-1 determine fludarabine resistance in leukemic B cells. Cell death & disease. 2013;4:e628.CrossRef Sharma A, Singh K, Mazumder S, Hill BT, Kalaycio M, Almasan A. BECN1 and BIM interactions with MCL-1 determine fludarabine resistance in leukemic B cells. Cell death & disease. 2013;4:e628.CrossRef
66.
go back to reference Cruickshanks N, Hamed HA, Bareford MD, Poklepovic A, Fisher PB, Grant S, et al. Lapatinib and obatoclax kill tumor cells through blockade of ERBB1/3/4 and through inhibition of BCL-XL and MCL-1. Mol Pharmacol. 2012;81(5):748–58.PubMedCentralCrossRefPubMed Cruickshanks N, Hamed HA, Bareford MD, Poklepovic A, Fisher PB, Grant S, et al. Lapatinib and obatoclax kill tumor cells through blockade of ERBB1/3/4 and through inhibition of BCL-XL and MCL-1. Mol Pharmacol. 2012;81(5):748–58.PubMedCentralCrossRefPubMed
Metadata
Title
Pan-Bcl-2 inhibitor Obatoclax is a potent late stage autophagy inhibitor in colorectal cancer cells independent of canonical autophagy signaling
Authors
Bruno Christian Koehler
Adam Jassowicz
Anna-Lena Scherr
Stephan Lorenz
Praveen Radhakrishnan
Nicole Kautz
Christin Elssner
Johanna Weiss
Dirk Jaeger
Martin Schneider
Henning Schulze-Bergkamen
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1929-y

Other articles of this Issue 1/2015

BMC Cancer 1/2015 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