scholarly journals Combined Targeting of Distinct c-Myc and JunB Transcriptional Programs for Multiple Myelioma Therapy

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 4415-4415
Author(s):  
Judith Lind ◽  
Felix Czernilofsky ◽  
Sonia Vallet ◽  
Fengjuan Fan ◽  
Martin Sattler ◽  
...  

c-Myc plays a pivotal role in multiple myeloma (MM) pathogenesis; and the BET protein BRD4 is a key regulator of c-Myc transcription. Recently, a pathophysiologic role in MM has also been attributed to the AP-1 family member JunB. Approaches to target transcription factors (TFs), such as c-Myc and JunB, currently emerge among the most promising novel anti-MM strategies, with a potentially high therapeutic index. However, redundancy phenomena represent a major challenge for targeting c-Myc- or JunB-mediated tumor proliferation programs. MZ-1 is a novel proteolysis-targeting chimera combining the recognition sequence for the E3-ligase Von-Hippel-Lindau with JQ1, a moiety that targets BRD4. Indeed, beyond direct inhibition by JQ1, MZ-1 significantly decreased BRD4 as well as c-Myc protein levels in MM cell lines and primary cells. Consequently, MZ-1 inhibited MM cell growth through G0/G1 arrest; and induced tumor cell death. Moreover, patient-derived BMSC- or exogenous IL-6- induced BRD4/c-Myc upregulation in MM cells was inhibited by MZ-1 treatment, indicating that targeting BRD4, at least in part, overcomes the protective effect of the microenvironment. Of note, MZ-1 did not have an impact on BMSCs/IL-6- induced upregulation of JunB RNA or protein levels. Conversely, dox-induced knockdown of BMSC/IL-6-triggered JunB upregulation in TetshJunB/MM.1S cells did not decrease BRD4/c-Myc RNA or protein levels. Similar data were obtained in other MM cell lines and primary cells. RNAseq, unbiased GSEA, and luciferase reporter assays of representative target genes further supported the co-existence of c-Myc- and JunB-mediated proliferative programs, which are initiated by the same stimuli. Importantly, MZ-1 in combination with dox-induced knockdown of BMSC/IL-6-triggered JunB upregulation in TetshJunB/MM.1S cells significantly decreased both BRD4/c-Myc as well as JunB protein levels; synergistically inhibited tumor cell proliferation and induced cell death. Similar results were obtained in other MM cell line and primary cells upon treatment with MZ-1 and transient siJunB. Finally, our preliminary results show the synergistic in vivo activity of MZ-1 in BMSC:TetshJunB/MM.1S versus BMSC:TetshSCR/MM.1S-carrying doxycyclin- treated NSG mice. Taken together, these data for the first time delineate the existence of distinct c-Myc and JunB transcriptional programs that contribute to MM proliferation. They thereby provide the preclinical rationale for the development of therapeutic strategies that combine targeting of distinct transcriptional programs to improve outcome in MM. Disclosures Vallet: Roche Pharmaceuticals: Consultancy; Pfizer: Honoraria; MSD: Honoraria. Podar:Celgene: Consultancy, Honoraria; Amgen Inc.: Honoraria; Roche Pharmaceuticals: Research Funding; Takeda: Consultancy; Janssen Pharmaceuticals: Consultancy, Honoraria.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2644-2644
Author(s):  
Judith Lind ◽  
Sonia Vallet ◽  
Karoline Kollmann ◽  
Osman Aksoy ◽  
Vincent Sunder-Plassmann ◽  
...  

Abstract INTRODUCTION Transcription factors (TFs) are convergence points of signaling cascades that coordinate cell differentiation, proliferation and survival and are commonly deregulated in cancer, including multiple myeloma (MM). They contribute to the initiation of MM and promote tumor cell growth and drug resistance. Both cMyc, a merging point of the PI3K-, and JunB, a merging point of the MEK/MAPK-signaling pathway, play pivotal roles in MM. Exciting novel approaches to inhibit TFs like proteolysis-targeting-chimera (PROTAC) promise to lead to selective tumor cell death with little/no consequence for normal cells. However, redundancy phenomena of transcriptional programs are likely to challenge their efficacy. Here, we report our final results on combined targeting of distinct c-Myc & JunB transcriptional programs for MM therapy. METHODS MM cell lines and patient MM cells were analyzed. Following CRISPR-loss-of-function screens for cMyc & JunB across MM cell lines and correlation analyses in MM patient datasets, the functional relevance of BRD4/c-Myc- and MEK/JunB-induced TF programs was delineated using genomic and chemical approaches in 2D and 3D models of the bone marrow (BM) microenvironment. Specifically, effects of single or combined targeting of cMyc- and JunB-induced TF-programs were analyzed by flow cytometry, western blot, RNAseq, qPCR and luciferase assays. In vitro and ex vivo results were finally verified in a MM xenograft mouse model. RESULTS While CRISPR loss-of-function screens across various MM cell lines confirmed their growth dependency on cMyc and JunB, we did not observe correlative expression levels among these TFs, neither in the publicly available GSE6477 nor in the CoMMpass dataset. In contrast, a significant positive correlation was observed between Brd4 and cMyc, and MEK and JunB expression levels, respectively. The existence of two distinct Brd4/cMyc and MEK/JunB transcriptional programs in MM cells was subsequently supported by a lack of changes in cMyc mRNA/protein levels and resultant transcriptional activity upon JunB knockdown, and vice versa. Likewise, MZ-1, a novel PROTAC which targets Brd4, resulted in the inhibition of BMSC/IL-6- induced cMyc- but not JunB- upregulation. Conversely, neither the MEK inhibitor trametinib nor doxycycline-induced knockdown of BMSC/IL-6- induced JunB upregulation in TetshJunB/MM.1S cells reduced Brd4/c-Myc mRNA/protein levels. Importantly, the activity of MZ-1 and trametinib was predicted by Brd4 and JunB expression levels using mathematical models, respectively. Further, combination of MZ-1 with trametinib or JunB knockdown synergistically inhibited tumor cell proliferation, and induced cell death in a 2D and a dynamic 3D model of the MM-BM milieu. Finally, our in vitro and ex vivo results were confirmed in vivo, utilizing BMSC:TetshJunB/MM.1S vs. BMSC:TetshSCR/MM.1S-carrying NSG mice treated with MZ-1 with/without doxycycline or trametinib. CONCLUSION In summary, our data demonstrate for the first time the existence of non-overlapping cMyc and JunB-regulated TF programs providing a rationale for combined cMyc:JunB targeting treatment strategies in MM. Disclosures Vallet: Pfizer: Honoraria; MSD: Honoraria; Roche Pharmaceuticals: Consultancy. Podar: Celgene: Consultancy, Honoraria; Roche Pharmaceuticals: Research Funding; Janssen Pharmaceuticals: Consultancy, Honoraria; Amgen Inc.: Consultancy, Honoraria.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5239-5239
Author(s):  
Wenge Wang ◽  
Amriti R. Lulla ◽  
Liz J. Hernandez-Borrero ◽  
David T. Dicker ◽  
Emmanuel K. Teye ◽  
...  

Abstract Quinacrine is a bioactive acridine derivative which has been used for treatment of malaria, giardiasis, systemic lupus erythematosus, and rheumatoid arthritis. In searching for p53 pathway activating agents for cancer therapy, we found that quinacrine stabilizes p53 and induces p53-dependent and p53-independent tumor cell death. Quinacrine also induces expression of TRAIL Death Receptor 5 (DR5) and reduces expression of anti-apoptotic Mcl-1 in tumor cells. These activities predict synergies with TRAIL (tumor necrosis factor-related apoptosis inducing ligand) and chemotherapeutic agents in inducing extrinsic and intrinsic pathway mediated apoptosis. In addition, quinacrine suppresses NFkB activity in tumor cells. Clinical trials have been ongoing for treatment of solid tumors including colon cancer, renal cancer, prostate cancer, and non-small cell lung cancer with quinacrine in combination with chemotherapy or tyrosine-kinase inhibitors, however, the therapeutic potential of quinacrine in blood cancer cells has not been established. We tested quinacrine on hematopoietic malignant cells, which included cell lines of myeloid leukemia, lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, and multiple myeloma. We found that quinacrine induces massive cell death in the cell lines tested, at concentrations from as less as 1 microM to 5 microM, 2-10 times lower than required to induce solid tumor cell death. Quinacrine synergizes with TRAIL in inducing cell death of TRAIL-sensitive cells and reverses resistance in TRAIL-resistant cells. Quinacrine also synergizes with chemotherapeutic agents, such as antimetabolites, alkylating agents, and tyrosine kinase inhibitors, in inducing apoptosis of hematopoietic cancer cell lines. Our work supports translational efforts to advance the use of quinacrine from bench to clinic and provides rationale for combination chemotherapeutic regimes for treatment of hematopoietic malignancies. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 17 (10) ◽  
pp. 1094-1106 ◽  
Author(s):  
Maria Cosenza ◽  
Monica Civallero ◽  
Stefania Fiorcari ◽  
Samantha Pozzi ◽  
Luigi Marcheselli ◽  
...  

2019 ◽  
Vol 19 (1) ◽  
pp. 29-37 ◽  
Author(s):  
Vivian M. Rumjanek ◽  
Raquel C. Maia ◽  
Eduardo J. Salustiano ◽  
Paulo R.R. Costa

In an attempt to find anticancer agents that could overcome multidrug resistance (MDR), two new classes of modified isoflavonoids were designed and synthesized, and their effectiveness evaluated against a vast array of tumor cell lines. Pterocarpanquinone (LQB-118) and 11a-aza-5-carbapterocarpan (LQB-223) were the most promising. LQB-118 induced cell death, in vitro, in the µM range, to a number of human cancer cell lines as well as to fresh tumor cells obtained from patients with acute or chronic myeloid leukemia, independent on whether they exhibit the MDR phenotype or not. Furthermore, leukemic cells were more sensitive to LQB- 118 compared to cells from solid tumors. Given to mice, in vivo, LQB-118 affected the growth of melanoma, Ehrlich carcinoma and prostate cancer cells. Conversely, no general toxicity was observed in vivo, by biochemical, hematological, anatomical or histological parameters and toxicity in vitro against normal cells was low. The process involved in tumor cell death seemed to vary according to cell type. Apoptosis was studied by externalization of phosphatidylserine, DNA fragmentation, caspase-3 activation, reduced expression of XIAP and survivin, ER stress, cytosolic calcium increase and mitochondrial membrane depolarization. Autophagy was also evaluated inhibiting caspase-9, with no effect observed in beclin 1, whereas pre-treatment with rapamycin increased cytotoxicity induced by LQB-118. In addition, LQB-118 increased ROS, inhibited NFκB nuclear translocation and secretion of TNF-α, modulated microRNAs miR-9 and miR-21 and modified the cell cycle. Despite being less studied, the cytotoxic effect of the 11a-aza-5-carbapterocarpan LQB-223 was present against several tumor cell lines, including those with the MDR phenotype.


2015 ◽  
Vol 14 (1) ◽  
pp. 15-18
Author(s):  
D. A. Afanasieva ◽  
M. A. Baryshnikova ◽  
Yu. A. Khochenkova ◽  
P. V. Golyshko ◽  
D. A. Khochenkov ◽  
...  

Previous studies have shown that aranosa-lio initiate cell death by activation of CD95-receptor of apoptosis. The mechanism of liposomal aranosa action is still unknown. Anticancer drugs may cause tumor cell death by the activation of autophagy. The purpose of this study was to determine the influence of liposomal aranosa on autophagy. The study was assessed on human melanoma cell lines mel Mtp, mel Ibr, mel Kor, mel Z and mel Mtp clone X. The cells were treated with aranosa-lio or liposomal aranosa at concentration 450 μg/ml for 24 hours. The expression of autophagy were analyzed the expression of mRNA Beclin 1. Expression of mRNA Beclin 1 was reduced on the majority of cell lines, which might be due to the fact that the liposomal form of the drug caused cell death by intrinsic apoptosis which is suppose to suppress autophagy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4926-4926
Author(s):  
Kelly J Walkovich ◽  
Xuwen Liu ◽  
Anne L McCollom ◽  
Colin S Duckett

Abstract Abstract 4926 CD30 is a member of the tumor necrosis factor (TNF) receptor family that is normally found on the cell surface of a small subset of activated lymphocytes but is overexpressed on the surface of anaplastic large cell lymphoma (ALCL) and Hodgkins lymphoma (HL) cells. Although many drugs exist that treat lymphomas by triggering the intrinsic cell death pathway, current chemotherapeutic regimens are limited by unwanted side effects, including secondary malignancies that limit event-free survival. The tumor-restricted overexpression of CD30 makes it an attractive target for therapeutic intervention. Depending on the cellular context, CD30 stimulation has been linked to cell death, cell cycle arrest, or paradoxically, proliferation. In ALCL tumor cell lines, CD30 stimulation activates both the canonical and noncanonical NF-kB pathways while in HL tumor cell lines, CD30 stimulation only slightly enhances NF-kB activity above constitutive levels, implying a role for NF-kB in determining the sensitivity or resistance of lymphoma cells to CD30-induced apoptosis. In addition, IAP antagonists, small synthetic compounds that mimic the structure of the second mitochondrial activator of caspase (Smac) and target IAP molecules that affect the activation of the non-canonical NF-kB pathway, induce apoptosis and/or sensitize cells to death via secondary signals such as TNF. This suggests that the modulation of IAP levels, and consequently regulation of the non-canonical NF-kB pathways, may also have a role in determining tumor cell death. Using representative ALCL and HL tumor cell lines, we have found that CD30 stimulation via its physiologically ligand in combination with standard chemotherapeutic agents results in increased efficacy in tumor cell death in the majority of ALCL cell lines but not HL cell lines. Similarly, IAP antagonists in combination with standard chemotherapeutic agents also resulted in enhanced tumor cell death in most ALCL but not HL cell lines. This augmentation of tumor cell death suggests that CD30-induced apoptosis and IAP antagonist-induced killing may have important consequences in the clinical treatment of CD30 positive malignancies. Currently, we are further investigating the role of both CD30 stimulation via its physiological ligand and IAP antagonists in impacting the activation of the canonical and noncanonical NF-kB pathways alone and in combination with currently utilized chemotherapeutic agents to modulate the apoptotic threshold in CD30 positive lymphoma cells. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document