Targeting the Over-Expressed Transcription Factor Yin-Yang 1 (YY1) Sensitizes Resistant Multiple Myeloma (MM) Cell Lines to Apoptosis by Bortezomib or Melphalan,

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3991-3991
Author(s):  
Hong Liu ◽  
Sara Huerta-Yepez ◽  
Clara M. Rivera-Pazos ◽  
Ma de Lourdes Cabera-Muñoz ◽  
Stavroula Baritaki ◽  
...  

Abstract Abstract 3991 The transcription factor Yin-Yang 1 (YY1) is a multi-functional DNA-binding protein which can activate, repress, or initiate transcription depending on the context in which it binds. We have previously reported that YY1 is a repressor for the death receptors Fas and DR5 as well as it regulates both chemo- and immuno- resistance in many tumors such as prostate carcinoma and lymphoma. YY1 has also been reported to be a prognostic factor for some cancers. Due to the inherent resistance of multiple myeloma (MM) cell lines to various cytotoxic drugs, we examined the expression levels of YY1 in both MM cell lines and bone marrow (BM)-derived MM tissues. Analysis for YY1 was performed by immunohistochemistry (IHC) and western blot. We found that, compared to normal peripheral blood mononuclear cells and normal bone marrow, the expression of YY1 was significantly upregulated in both the cell lines and patient-derived MM tissues. The expression of YY1 was distributed in both the cytoplasm and the nucleus. Further, the intensity and frequency of cells expressing YY1 in the nuclei of MM tissues were significantly higher among patients with progressive disease as compared to patients with stable disease. In order to ascertain the role of YY1 in the maintenance of drug resistance in MM, we hypothesized that, due to the overexpression of YY1 in MM, inhibition of YY1 expression and activity should lower the threshold of resistance and render the MM cells more sensitive to drug-induced apoptosis. This hypothesis was tested using the MM U266 cell line and the drugs bortezomib and melphalan as models. The inhibition of YY1 was done by knocking down YY1 following transfection with YY1 siRNA. Analysis of the transfected cells with YY1 siRNA, but not with control siRNA, inhibited YY1 expression as analyzed by IHC and western blot. The U266 cells were first transfected with YY1 siRNA or control siRNA for 48 hours and then treated with various concentrations of bortezomib (2.5 and 5.0 nM) or melphalan (10 and 20 μM) that were not toxic to the cells for 48 hours. The cells were analyzed for apoptosis by activation of the effector caspase 3. The findings demonstrate that YY1/siRNA-treated cells were sensitized to apoptosis by bortezomib or melphalan. Analysis of the underlying mechanism by which the inhibition of YY1 sensitizes the MM cells via regulation of the apoptotic pathways will be presented. We are also currently testing specific chemical inhibitors for YY1 that are being tested both in vitro and pre-clinically in mice. The present findings demonstrate that YY1 is a potential target for therapeutic intervention and the combination of YY1 inhibitors with non-toxic doses of bortezomib, melphalan or other cytotoxic drugs may be beneficial for the treatment of MM patients who are no longer responding to current treatments. Further, we propose that, based on our findings that the level and nuclear localization of YY1 dictates disease progression in a subset of patients, inhibition of YY1 with treatment may reduce the frequency of patients with progressive disease. *Contributed equally Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5083-5083
Author(s):  
Mehran S. Neshat ◽  
Haiming Chen ◽  
Melinda S. Gordon ◽  
James R. Berenson ◽  
Benjamin Bonavida

Abstract The transcription factor Yin Yang 1 (YY1) regulates cellular differentiation and response to apoptotic stimuli. YY1 exerts its pleiotropic effects through regulation of promoter activity of critical genes, as well as association and direct modulation of stability and function of a subset of proteins. Genes that are regulated by YY1 include those that control the cell cycle, development, differentiation and tumor suppression. For example, it has been reported that YY1 inhibits the proto-oncoprotein c-Myc (Austen, et al., Oncogene, 1998, 17:511) and negatively regulates the tumor suppressor gene p53 (Sui, et al., 2004, Cell 117: 859). Thus, expression and activity of YY1 in tumor cells may be involved in the pathogenesis of disease, as well as controlling response to drug stimuli. YY1 is regulated at transcriptional and post-translational levels in response to intra and extracellular signals. It has been reported that YY1 undergoes proteolytic cleavage. Caspase-dependent N-terminal cleavage of YY1 has been reported in response to physiological (Fas, TNF, L-glutamate) and chemical (staurosporine, etoposide, okadaic acid) death promoting factors. Similar presence of truncated YY1 is observed in in vitro models of skeletal and cardiac muscle differentiation. N-terminal truncated YY1 lacks its transactivation domain, while DNA binding remains unaltered. Hence, YY1 function may be altered by truncated forms. We hypothesized that post-translational processing of YY1 occurs in bone marrow and may be important in tumor progression and response to therapeutic agents. This study thereby aimed to determine whether altered levels and/or forms of YY1 are expressed in the bone marrow of multiple myeloma patients and to identify their potential downstream effectors. YY1 expression in protein lysates of bone marrow aspirates from nine patients was determined by Western blot analysis. Truncated species of YY1 were present in 6/8 samples. In contrast to myeloma bone marrow, one plasma cell leukemia sample showed high levels of YY1 and no truncated forms. Similar high levels of YY1 expression was observed in established tumor xenografts of a plasma cell leukemia tumor. We are presently extending the pool of analyzed normal and cancer harboring tissues and examining potential correlation of YY1 and its altered forms with disease status and prior therapeutic history. Identification and purification of cell populations that generate altered forms of the protein and its effect on expression and function of YY1 interacting proteins are under investigation.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2892-2892
Author(s):  
Sara Huerta-Yepez ◽  
Stavroula Baritaki ◽  
Hong Liu ◽  
Karla MC Mendez-Maldonado ◽  
Manuel L Penichet ◽  
...  

Abstract Abstract 2892 Multiple myeloma (MM) is an incurable hematological malignancy. Thalidomide, bortezomib and lenalidomide have emerged as highly active agents in the treatment of MM. Although initial clinical responses to drug therapy are achieved, a significant number of MM patients relapses and no longer responds to further treatments. Hence, the mechanism of resistance of MM to current therapeutic regimens remains elusive. It will be useful to identify prognostic markers that can predict the poor response of patients to therapies and to develop novel therapeutics for these patients. Our studies and those of others have recently identified a gene product, Raf-1 kinase inhibitory protein (RKIP) that has been shown to be involved in the regulation of tumor sensitivity to drugs and that has been found to be poorly expressed in different histological resistant tumors studied. RKIP is a member of the phosphatidylethanolamine-binding protein (PEBP) that functions as a potent inhibitor of different kinases of the Raf/MAPK (Raf-1 kinase) and NF-k B (TAK-1, NIK) activation pathways, and, thereby, antagonizing both cell survival and the expression of anti-apoptotic gene products. RKIP activity undergoes a post translational modification involving PKC–meditated phosphorylation at Ser 153 that inhibits its association with RAF/MAPK and NF-k B. The objective of this study was to examine the expression and function of RKIP and phospho-RKIP (p-RKIP) in MM and examined their clinical significance. MM cell lines and bone marrow-derived MM tissues were analyzed by both immunohistochemistry (IHC) and western blot for the expression of RKIP and p-RKIP. The antibody to RKIP recognizes both non-phospho and p-RKIP whereas the antibody against p-RKIP is specific. We demonstrate the unusual overexpression of RKIP in MM cell lines and tissues and the majority was in its inactive phosphorylated form. The expression of both forms in normal bone marrow was very low. Analysis of the ratio of RKIP/p-RKIP suggested its clinical significance wereby a higher ratio correlated with poor progression and a low ratio correlated with progression. The overexpression of p-RKIP suggested that it maintains the survival and the resistance of MM to cytotoxic drugs. We hypothesized that the dephosphorylation of RKIP may render RKIP active and modify the resistance to cytotoxic drugs. This hypothesis was tested in MM cell lines. Tumor cells were treated with the PKC inhibitor bisindolylmalemide (BIM) (5–10 μg/ml) for different times and complete inhibition of p-RKIP as detected as early as 4 h, while non phospho-RKIP was preserved, as detected by IHC and western blot. Treatment with BIM and followed with bortezomib (2.5-nM) resulted in significant cytotoxicity of the bortezomib-resistant MM cells. Overall, the findings demonstrate the unusual overexpression of p-RKIP in MM cells that plays an important role in survival and maintenance of tumor cell-resistance to cytotoxic drugs. The reversal of the active non-phosphorylated form of RKIP by the use of the specific PKC inhibitor rendered the tumor cells sensitive to bortezomib-induced cytotoxicity. These findings support the clinical application of specific PKC inhibitors in combination with bortezomib or other drugs in the treatment of patients who are resistant to conventional therapies. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 45 (3) ◽  
pp. 1184-1192 ◽  
Author(s):  
SARA HUERTA-YEPEZ ◽  
HONG LIU ◽  
STAVROULA BARITAKI ◽  
MARIA DEL LOURDES CEBRERA-MUÑOZ ◽  
CLARA RIVERA-PAZOS ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2827-2827
Author(s):  
Massimo Libra ◽  
Giancarlo Castellano ◽  
Stavroula Baritaki ◽  
Sara Huerta-Yepez ◽  
Giovanni Ligresti ◽  
...  

Abstract Objective and Rationale Non-Hodgkin lymphomas (NHL) are a heterogeneous group of lymphoproliferative malignancies with variable patterns of behavior and responses to treatment. Although at present a significant number of high grade lymphoma patients can be cured with intensive regimens of cytotoxic and immunotherapeutic drugs, still these patients may relapse or become resistant to treatment. Therefore, there is a need to identify novel factors involved in lymphoma development and progression and hopefully to be used as targeted therapies. Thus, the objective of the present study was to explore the potential involvement of Yin Yang 1 (YY1) in the development as well as in the prognosis of NHL. Hypothesis Our previous studies showed that YY1 is overexpressed in several human cancer cell lines and its expression correlates with resistance to chemo- immune-mediated apoptosis. Prognostic and diagnostic significance of YY1 has been recently shown in prostate cancer. Thus, we hypothesized that YY1 overexpression in NHL may be involved in transformation and act as a prognostic biomarker. Methods and Designs Overexpression of YY1 has been shown by western blot and RT-PCR in NHL cell lines compared to normal B-cells. Validation of in vitro results has been performed by immunohistochemistry (IHC) and bioinformatics in NHL tissues. IHC analyses were performed in two commercial NHL tissue arrays containing 104 samples. Results Gene expression analysis and association with clinical features of the transcription factor YY1 were evaluated using three different datasets of publicly available microarray data from lymphoma tumor-biopsy specimens. By analyzing the data set (221 NHL and 25 normal B-cells) by Basso et al., (Nat Genet 37:382, 2005) higher YY1 transcript levels were found in lymphoma tissues (BL and DLBCL) in comparison to normal B-cells. No significant differences were found with the other lymphoma histotypes. This class comparison analysis revealed that YY1 could be involved in the transformation of B cells giving rise to high-grade lymphomas. Expression levels of YY1 were analyzed in association with the available main clinical and biological parameters using the expression data of two different high grade lymphoma studies with a total of approximately 400 samples (Rosenwald et al., N Engl J Med.346:1937, 2002; Hummel et al., N Engl J Med.354:2419, 2006). Upregulated expression of YY1 was found in the subgroups of patients with positive proliferation signature scores with respect to those with negative proliferation signature scores. A significant correlation was found between BCL6, post-germinal center (GC) marker and YY1 expression. The presence of BCL6 protein in the tumors is associated with high levels of YY1 gene transcription. These data confirm that YY1 is mostly overexpressed among high-grade lymphomas that usually are post-GC transit. Survival analysis in both datasets reveal that higher levels of YY1 gene transcription are associated with poor outcome. Finally, biological network analysis, using the Proprietary Ingenuity Pathway analysis software, was performed to better understand the biological significance of these findings. Among all genes present in the data set by Hummel et al. (2006) a significant positive correlation with YY1 expression was found in 374 genes. In this data set, Top Networks Functions associated with YY1 expression levels were cellular movement, cell morphology, cell cycle, and cell-to-cell adhesion. These analyses indicate the potential direct involvement of YY1 in the regulation of cell cycle pathways and in the regulation of cellular motility. Conclusions and Implications Overall, the findings reveal that YY1 is involved in lymphoma development and may be useful as a biomarker of NHL transformation as well as a potential target for therapeutic interventions.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5545-5545 ◽  
Author(s):  
Elizabeth D. Lightbody ◽  
Mairead Reidy ◽  
Michael P. Agius ◽  
Salma El-Behaedi ◽  
Romanos Sklavenitis-Pistofidis ◽  
...  

Introduction Multiple myeloma (MM) is an incurable hematological malignancy characterized by the clonal expansion of malignant plasma cells (PCs) within the bone marrow. MM is genetically heterogeneous with aberrations including hyperdiploidy and chromosomal translocations commonly involving the immunoglobulin heavy chain (IgH) region. Many transcription factors can revoke their normal processes and act as oncogenes when they are brought under the control of IgH regulatory regions by a chromosomal translocation. Interferon Regulatory Factor 4 (IRF4) is a transcription factor which controls plasma cell differentiation and possesses many regulatory roles including interferon response, immune cell response, cell proliferation, apoptosis, and metabolism. IRF4 has proven to be a genetic vulnerability in MM as silencing studies in a large panel of MM cell lines with various genetic etiologies have demonstrated IRF4 expression is essential for MM cell survival. Standard of care treatments that indirectly suppress IRF4 including Proteasome inhibitors and Cereblon modulators have provided the greatest clinical outcomes for patients. However, like many other transcription factors, IRF4 has been notoriously difficult to target due to the protein's lack of amenable binding pockets favored for small molecule inhibitor development. Thus, identifying novel mechanisms and compounds to target IRF4 (directly or indirectly) can provide significant clinical impacts for MM patients. Methods To discover compounds capable of depleting IRF4 levels, we performed a high-throughput drug screen utilizing the Selleckchem Drug Repurposing Library on a widely accepted IRF4-dependent cell line. This library consists of over 2,000 diverse compounds that have well validated mechanisms of actions and have additionally passed clinical phase 1 safety trials for accelerated translational use. MM.1S cells were treated for 48 hours in duplicate (n = 2) with 10 μM compound. Following treatment, the cells were fixed, permeabilized, and stained for viability and IRF4 levels. IRF4 expression and viability was acquired by using flow cytometry, with high dose lenalidomide and shRNA for IRF4 as positive controls. Compounds that reduced IRF4 levels and cell viability across both experimental runs were ranked and selected with a cutoff of 40% as promising candidate compounds for further validation. Results Our drug screen results revealed 20 compounds (undisclosed) which met our cutoff of a decrease of IRF4 levels by 40% or greater. Ten hits were selected as having greater or equal to IRF4 depleting properties of lenalidomide and moved forward to be validated by western blot. Six drugs were shown to deplete IRF4 by western blot in MM.1S and KMS-18 cells at 10 μM doses. Interestingly, 4 out of the 10 hits all belong to same compound class that selectively bind to the same target receptor (undisclosed). Additional experiments confirmed these class of compounds deplete IRF4 levels in a dose dependent manner (EC50 = 1 μM). A time course revealed that IRF4 levels decrease shortly after the binding of these drugs to their widely reported target receptor, suggesting this is a selective drug/target receptor-mediated mechanism directly altering levels of IRF4. In vitro studies demonstrated the ability to both halt cell growth and decrease the viability of a panel of 8 MM cell lines, with IC50's ranging from 1.6 - 8.5 μM. Synergy studies with Lenolidomide and Bortezomib are underway to determine any synergistic combinations with standard of care therapies. In vivo studies and RNA-sequencing are also currently underway to determine the impact of these compounds on MM tumor growth and overall survival, as well as better define the mechanism of action driving this novel class of IRF4 targeting compounds. Conclusions Despite knowledge that IRF4 is a biologically potent target in MM there have been no extensive studies highlighting drugs capable of targeting this transcription factor and its oncogenic signaling network. This screen has revealed novel compounds, some of which are clinically used, that are capable of depleting a highly dependent gene in MM. Notably, these compounds are able to deplete IRF4 in a novel mechanism which is capable of affecting survival of MM cell lines that represent the heterogeneity of myeloma, and thus holds potential for significant clinical impact. Disclosures Ghobrial: Amgen: Consultancy; Janssen: Consultancy; Celgene: Consultancy; Takeda: Consultancy; Sanofi: Consultancy; BMS: Consultancy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3394-3394
Author(s):  
Sara Huerta-Yepez ◽  
Stravoula Baritaki ◽  
Angeles Hernandez-Cuecto ◽  
Yu-Mei Anguino-Hernandez ◽  
Mehran Neshat ◽  
...  

Abstract Yin Yang 1 (YY1) is a ubiquitously expressed zinc-finger transcriptional regulator that can function as an activator, a repressor, or an initiator binding protein. YY1 has been implicated in the control of cell growth, development, differentiation and tumor suppression. We have recently reported that YY1 regulates the transcription of death receptors in carcinoma and lymphoma and thus, YY1 regulates tumor sensitivity to immune-mediated apoptosis (Huerta-Yepez, et al., 2004, Oncogene23: 4993–5003; Vega, M., et al., 2005, Journal of Immunology175: 2174–2183). In addition, we have also reported that human prostate tumor tissues overexpress YY1 (Seligson, et al., 2005, International Journal of Oncology 27:131). This study investigated the expression of YY1 in multiple myeloma (MM) with the objective of determining whether YY1 plays a role in the progression and drug- resistance of MM. We have initiated these studies by examining nine bone marrow (BM) samples derived from patients with MM. Immunohistochemical studies were performed for the detection and cytoplasmic or nuclear localization of YY1 in the MM cells and also in adjacent normal mature and immature cells. The intensity of staining by the anti-YY1 antibody was scored and the relative intensity was calculated. Two slides from each patient were analyzed and 200 cells per slide were scored. Mean intensities of all samples were calculated and the data were subjected to statistical analysis. YY1 expression in normal BM was low and primarily of cytoplasmic origin. In contrast, YY1 was significantly overexpressed in MM cells. The mean intensity in the MM was approximately three-four fold higher than that of the normal cells and was primarily of nuclear origin (p-value < 0.05). The signals that control shuttling YY1 are undefined. The expression of YY1 in normal mature and immature cells was low and there was comparable staining in the cytoplasm and the nucleus. Analysis of the cell distribution expressing YY1 by flow cytometry revealed that greater than 50% of the cells in the CD38+ subset expressed YY1. In addition, the MM tumor cells also expressed high level of pleiotrophin (PTN) and the patients had high levels of circulating PTN. PTN is a growth factor for MM and PTN transcription is regulated by an initiator element and could thus be a target of YY1-mediated transcriptional control. These findings suggest that YY1 overexpression may be involved in the pathogenesis of MM and is a potential target for therapeutic intervention. Current studies are aimed at examining the prognostic and diagnostic roles of YY1 and PTN in MM and molecular mechanisms that underlie their overexpression in MM.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3445-3445
Author(s):  
Ayman A. Saad ◽  
James Fortney ◽  
Lin Wang ◽  
Heather O’Leary ◽  
Laura Gibson ◽  
...  

Abstract INTRODUCTION: Multiple myeloma cells display functional CXCR4 chemokine receptor that stimulates the migration of these cells toward their natural ligand, CXCL12 (stromal-derived factor, SDF-1a). CXCL12 is secreted by bone marrow stroma. Consistent with their CXCR4 expression, myeloma cells home to the marrow microenvironment, where adhesive interactions promote growth, survival, and confer cell adhesion-mediated drug resistance. METHODS U266-B1 cells (ATCC myeloma cell line) were pre-treated with recombinant CXCL12 for 30 minutes prior to the addition of melphalan for up to 72 hours. Both melphalan and CXCL12 were added at 24 hour intervals. Cell lines alone and cell lines with only melphalan or only CXCL12 were used as controls. We have also tested the influence of adding AMD3100, a reversible inhibitor of CXCR4, on myeloma cell survival. U266-B1 cells in media alone were pre-treated with AMD3100 for 24 hours prior to treatment with melphalan for 16 hours. Cell viability following treatment was quantified by flow cytometry assay using Annexin-V-FITC staining. Western blot analysis was used to quantify the apoptotic activity of the cell lines using 4 apoptotic markers: PARP (poly ADP-ribose polymerase), caspase-3, Bcl-2, and Mcl-1. RESULTS: Recombinant CXCL12 conferred a protective effect to myeloma cell lines during melphalan treatment. This effect was more pronounced at 72 hours of treatment. Western blot analysis showed diminished expression of the apoptotic markers, cleaved PARP (poly ADP-ribose polymerase) and active caspase-3 in the melphalan-treated cell lines with prior exposure to CXCL12. Additionally, pretreatment with AMD3100 resulted in enhanced apoptosis following melphalan treatment. CONCLUSION: Our data showed that CXCL12, a naturally occurring cytokine secreted by bone marrow stromal cells confers a protective effect on myeloma cells against apoptosis. Disruption of this effect by AMD3100 resulted in enhanced melphalan-induced apoptosis of myeloma cells. We are continuing to study this effect with the potential future utility of AMD3100 as a melphalan chemo-sensitizer in the treatment of multiple myeloma.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1139-1139
Author(s):  
Reeder McNeil Robinson ◽  
Leticia Reyes ◽  
Ravyn M. Thompson ◽  
Yefim Manevich ◽  
Jesse McClure ◽  
...  

Abstract Proteasome inhibitors (PIs) are cornerstone agents in the treatment of Multiple Myeloma (MM). Although initially effective, resistance to PIs inevitably emerges, presenting an obstacle to sustained and durable treatment responses in the clinic. To address this limitation, we set out to discover new small molecules that are able to restore PI sensitivity in resistant MM cells. We screened multiple chemical libraries using a cell-based screening method that identifies synergistic combinations with the PI bortezomib (Btz). This method uncovered compound E61 that, in subsequent rounds of screening, demonstrated potent PI re-sensitizing activity. E61 synergistically enhanced the activity of multiple PIs, including Btz, carfilzomib, ixazomib, and oprozomib by 3-15 fold in a genetically diverse panel of PI sensitive and resistant MM cells. In addition, E61 exhibited strong anti-MM activity as a single agent after extended treatment times (48 hours) and demonstrated >10-fold selectivity for MM cell lines over normal peripheral blood mononuclear cells (PBMCs), normal lymphocytes, and a panel of normal fibroblast cell lines. Importantly, the PI sensitizing activity of E61 was also limited to MM cells, as the drug failed to enhance the activity of PIs in normal cells. For a hit stage molecule, E61 showed exceptional tolerability and activity in vivo, significantly improving animal survival and reducing the number of CD138+ MM plasma cells in the bone marrow of mice. We used a xenotransplant model where NOD-SCID IL2Rgamma-/- (NSG) mice were injected via the lateral tail vein with PI resistant MM cells. Using this model, mice reliably reached the survival endpoint between weeks six and seven, with death being caused by the infiltration of mouse bone marrow by MM plasma cells and the development of bone lesions that closely resemble the human MM pathology. Continuous dosing with E61 (50 mg/kg, i.p., daily) was able to cure 38% of mice, with surviving mice showing only minimal residual disease (i.e., 1.0-1.5% CD138+ MM cells in the bone marrow) at the termination of the experiment. The molecular effects of E61 which lead to its anti-MM activity are characterized primarily by oxidative and endoplasmic stress responses. E61 induces reactive oxygen species (ROS) formation and oxidative damage to proteins, effects that are synergistically potentiated by the addition of PIs. This oxidative burst is critical to the anti-MM activity of E61, as the neutralizing of ROS with various molecular scavengers blocks the pro-apoptotic effects of E61. E61 triggers a robust induction of canonical ER stress markers including phospho-eIF2a, ATF4, XBP-1s, and CHOP. In order to identify the direct molecular target of E61, we chemically modified the molecule to enable copper-catalyzed azide-alkyne click chemistry coupling to fluorescent dyes and immobilizing agents. This strategy was used in tandem with peptide mass fingerprinting, which identified a small set of E61 protein binding partners that are principally involved in the proper folding of nascent polypeptide chains. Additionally, we have synthesized a small library of derivatives which retain the PI re-sensitization phenotype, that have increased metabolic stability against human liver microsomes by at least 5-fold. Ongoing efforts by our group are aimed at further validating and confirming this mechanism of action of E61 as well as further optimizing the chemical structure of E61 for enhanced potency and pharmaceutical properties toward the goal of clinical development. Overall, this work demonstrates the potential of developmental compound E61, a new class of small molecule with an apparent novel mechanism of action, as a new drug candidate for the treatment of refractory MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2730-2730 ◽  
Author(s):  
Sara Huerta-Yepez ◽  
Clara Rivera-Pazos ◽  
Massimo Libra ◽  
Stavroula Baritaki ◽  
Haiming Chen ◽  
...  

Abstract Objective and Rationale Yin-Yang 1 (YY1) is a multi-functional DNA-binding protein, which can activate, repress or initiate transcription depending on the context in which it binds. In addition, YY1 can modulate protein levels or activity through protein-protein interaction. YY1 has been identified as a potential repressor factor for several genes. We have reported that YY1 can act as a transcription repressor for both Fas and TRAIL DR5. In addition to YY1-mediated regulation of tumor cell resistance to cytotoxic immunotherapy, it also has been shown to regulate resistance to chemotherapy [Baritaki et al., J Immunol 80:6199,2008]. Overexpression of YY1 has been shown to be of prognostic significance in prostate cancer [Seligson et al., Int J Oncol 27:131,2005]. Hypothesis In this study, we have examined the expression of YY1 in MM (cell lines and patients’ bone marrow [BM]) by hypothesizing that the resistance of MM cells to various cytotoxic agents may be, in part, regulated by overexpression of YY1 and that YY1 may also be of prognostic significance. Designs and Methods MM cell lines and fresh BM samples from MM patients (n=21) were examined for YY1 expression, cytoplasmic and nuclear, by immunohistochemistry and by Western. The specificity of the anti-YY1 antibody was demonstrated by the competitive inhibition with a peptide used for immunization. Control isotype IgG did not show any staining on the cells. Results First, we found that various MM cell lines (RMPI 8226, IM-9, U266) overexpress YY1 both in the cytoplasm and the nucleus. Next, we examined the expression of YY1 in BM derived from MM patients by immunohistochemistry. BM from MM patients showed overexpression of YY1 as compared to normal bone marrow samples. Furthermore, analysis of both the intensity and frequency of cells expressing YY1 in both the cytoplasm and nucleus was shown to be significantly higher among patients with progressive disease as compared to patients with stable or responsive disease. Conclusions and Implications These findings show that the expression of YY1 among patients with MM may correlate with progression and also suggest the prognostic significance of YY1 in MM patients. Inhibition of YY1 by various agents (example: low-dose chemotherapy, proteasome inhibitors, NO donors, NF-kB inhibitors, etc.) all result in the reversal of resistance to various cytotoxic agents (eg CDDP, TRAIL) therefore, the findings also imply that agents that can inhibit YY1 expression in MM patients may be of therapeutic potential when used in combination with conventional therapeutics.


2010 ◽  
Vol 107 (12) ◽  
pp. 1490-1497 ◽  
Author(s):  
Konstanze Beck ◽  
Ben J. Wu ◽  
Jun Ni ◽  
Fernando S. Santiago ◽  
Kristine P. Malabanan ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document