Abstract 5541: Proteasome inhibitor synergizes with histone deacetylase inhibitor to trigger ROS- and ER stress-induced apoptosis of nasopharyngeal carcinoma independent of aggresome disruption

Author(s):  
Alan K. Chiang ◽  
Kwai Fung Hui
2012 ◽  
Author(s):  
Benjamin Verillaud ◽  
Mélanie Gressette ◽  
Marie-Paule Sablin ◽  
Hélène Lelièvre ◽  
Anne Jacquet-Bescond ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2526-2526
Author(s):  
Weili Zhao ◽  
Lan Wang ◽  
Yuanhua Liu ◽  
Jinsong Yan ◽  
Christophe Leboeuf ◽  
...  

Abstract The anti-CD20 monoclonal antibody rituximab has shown promising results in the clinical treatment of patients with B-cell non-Hodgkin’s lymphoma (B-NHL). However, its therapeutic effect could still be improved. This study examined the anti-tumor activity of rituximab combined with histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) in CD20-positivie B-NHL cell lines, as well as in primary B-NHL cells and a murine B-NHL model. The combination treatment sensitized B-NHL cells to apoptosis in a synergistic manner, concomitant with Bcl-2/Bcl-XL downregulation, mitochondrial instability, and caspase activation. Particularly in Daudi cells relatively insensitive to rituximab, these events were associated with nuclear factor-kB (NF-kB) inactivation. SAHA presented functional complementation with rituximab, through decreasing IKKa/b and IkBa phosphorylation, thus preventing NF-kB nuclear translocation. In addition, SAHA induced IkBa cleavage to a stable inhibitory form and caused NF-kB degradation in response to caspase-3 activation. As an independent approach, co-administration of rituximab and SAHA resulted in a clear decline in levels of ERK cascade members, including extracellular-signal-regulated kinase (erk) itself, upstream Raf-1, mitogen-activated protein kinase/ERK Kinase Kinase-1 (mekk1), and mekk2, as well as their downstream transcription factor target c-myc. By western blot, c-Myc protein was subsequently downregulated when treated with rituximab or SAHA, and the degradation was most significant in combination group. More importantly, rituximab-SAHA combination significantly promoted primary B-NHL cells apoptosis and improved the survival time of a SCID mouse lymphoma model established with intravenous injection of Daudi cells. Terminal deoxytransferase-catalyzed DNA nick-end labeling and ultrastructural study revealed increased apoptotic lymphoma cells on mice spleen sections of combination group. Taken together, these findings emphasized the value of targeting apoptosis signaling pathway in lymphoma therapy. Rituximab in conjunction with histone deacetylase inhibitor may represent a novel strategy in treating patients with B-NHL.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4389-4389
Author(s):  
Sara Galimberti ◽  
Martina Canestraro ◽  
Simone Pacini ◽  
Rita Fazzi ◽  
Enrico Orciuolo ◽  
...  

Abstract PS-341 (Bortezomib) is a novel dipeptide boronic acid proteasome inhibitor with in vitro and in vivo antitumor activity that induces apoptosis in different human cancer cell lines. Suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, has been reported to induce apoptosis of leukemic cells by increasing the cyclin-dependent kinase inhibitor p21 or generating reactive oxygen species (ROS). Co-exposure of BCR/ABL-positive cells both sensitive and resistant to imatinib to these compounds has been previously reported resulting in an increased apoptotic rate. To extend this observation, we examined the effect of treatment with bortezomib or/and SAHA of a megakaryoblastic cell line (MO7-e). Cell proliferation, ROS production, cell cycle progression, induction of apoptosis and differentiation has been investigated. Bortezomib was shown to retain NF-kB in the cytoplasm and inhibit cell growth (IC50=20nM), in a dose/time-dependent way. This anti-proliferative activity resulted to be lineage-specific, because other leukemic cell lines were unaffected by the bortezomib treatment. Moreover, bortezomib in MO7-e cells increased ROS production and induced a significant pro-apoptotic effect (50% vs 5% in control). Finally, cell cycle was blocked in the G2 phase and bortezomib was able to down-regulate WT1 expression, gene that could play a relevant prognostic role in myeloproliferative disorders. Moreover, any significant effect on cell differentiation was found. SAHA also resulted able to inactivate NF-kB and to inhibit cell proliferation, at 1.5 uM. It did not increase significantly ROS production, blocked cell cycle in the G1 phase and down-regulated WT1 expression (10 fold minus than bortezomib). Neverthelles, SAHA also did not induce differentiation of megakaryoblatic cells. Co-exposure of this cell line to minimally toxic concentrations of bortezomib (5 nM) and SAHA (0.5 uM), resulted in a significant increase of anti-proliferative (50% of growth inhibition vs 15% with bortezomib and 10% with SAHA alone) and pro-apoptotic effect (45% vs 20% of bortezomib and 15% of SAHA alone). Interestingly, immunocitochemistry assays detecting the NF-kB p65 subunit showed that the co-exposure to bortezomib and SAHA resulted in a minor NF-kB inactivation than that achieved with single compounds. This finding was confirmed by the pre-incubation of MO7-e cells with SAHA in respect of the pre-incubation with bortezomib or the simultaneously addition of the two drugs. In fact, pre-incubating megakaryoblasts with SAHA, the anti-proliferative effect of bortezomib significantly decreased. In conclusion, this study supports the association of a proteasome with a histone deacetylase inhibitor, in a time-sequence-related way, especially in chronic myeloproliferative disorders where a spontaneous NF-kB activation and a WT1 over-expression have been reported.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2856-2856 ◽  
Author(s):  
Rekha Rao ◽  
Warren Fiskus ◽  
Ramesh Balusu ◽  
Hongwei Ma ◽  
James Bradner ◽  
...  

Abstract Abstract 2856 The proteasome inhibitor bortezpmib has been shown to markedly increase the intracellular levels of misfolded proteins, induce aggresome formation and cause endoplasmic reticulum (ER) stress, resulting in apoptosis of human Mantle Cell Lymphoma (MCL) cells. Consistent with this, Bortezomib displays clinical efficacy in patients with relapsed and refractory MCL. We have recently reported that the pan-histone deacetylase (HDAC) inhibitor panobinostat, by also inhibiting HDAC6, abrogates aggresome formation and induces Endoplasmic Stress (ER) stress, as well as potentiates bortezomib-induced apoptosis of MCL cells. Here, we determined the anti-MCL cell activity of an HDAC6-specific inhibitor, WT-161 alone and in combination with the novel, orally bio-available, proteasome inhibitor carfilzomib (Proteolix Inc.) against human, cultured and primary, patient-derived MCL cells. Treatment with WT-161 (0.1 to 1.0 uM) resulted in a dose-dependent increase in the acetylation of alpha-tubulin and heat shock protein (hsp) 90, without any appreciable increase in the levels of acetylated histone (H) 3. Consistent with WT-161 mediated hyperacetylation and inhibition of hsp90 chaperone function, treatment with WT-161 increased the intracellular levels of polyubiuitylated proteins in the cultured MCL JeKo-1 and Z138 cells. WT-161 was also noted to dose-dependently deplete the levels of cyclin D1 in the cultured MCL cells. Treatment with WT-161 also induced ER stress response in the MCL cells, demonstrated by increase in the protein levels of Glucose regulated protein (GRP) 78, phosphorylated eIF2 (eukaryotic initation factor 2) α, and induction of the pro-apoptotic transcription factor CHOP (CAAT/Enhancer Binding Protein Homologous Protein). We next determined the effects of co-treatment with WT-161 on carfilzomib-induced aggresome formation, ER stress response and apoptosis of the cultured and primary MCL cells. Co-treatment with WT-161 (0.25 uM) abrogated carfilzomib-induced aggresome formation in MCL cells, as evidenced by confocal immunofluorescent staining of aggresomes with anti-HDAC6 and anti-ubiquitin antibodies. Compared to each agent alone, co-treatment with WT-161 and carfilzomib induced more intracellular polyubiquitylated proteins and induced higher levels of CHOP in the cultured MCL cells. Co-treatment with WT-161 and carfilzomib also synergistically induced apoptosis of the cultured MCL cells (combination indices < 1.0). Notably, co-treatment with WT-161 and carfilzomib also synergistically induced apoptosis of primary MCL cells (combination indices < 1.0). These findings strongly support the in vivo testing of the combination of an HDAC6-specific inhibitor such as WT-161 with the proteasome inhibitor carfilzomib against human MCL cells. Disclosures: No relevant conflicts of interest to declare.


PLoS ONE ◽  
2014 ◽  
Vol 9 (3) ◽  
pp. e91325 ◽  
Author(s):  
Mélanie Gressette ◽  
Benjamin Vérillaud ◽  
Anne-Sophie Jimenez-Pailhès ◽  
Hélène Lelièvre ◽  
Kwok-Wai Lo ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document