scholarly journals Dihydroartemisinin Induces Growth Arrest and Overcomes Dexamethasone Resistance in Multiple Myeloma

2020 ◽  
Vol 10 ◽  
Author(s):  
Ying Chen ◽  
Rui Li ◽  
Yuqi Zhu ◽  
Sixia Zhong ◽  
Jinjun Qian ◽  
...  
2021 ◽  
Vol 11 ◽  
Author(s):  
Ying Chen ◽  
Rui Li ◽  
Yuqi Zhu ◽  
Sixia Zhong ◽  
Jinjun Qian ◽  
...  

Blood ◽  
2000 ◽  
Vol 95 (3) ◽  
pp. 1039-1046 ◽  
Author(s):  
G. Teoh ◽  
Y.-T. Tai ◽  
M. Urashima ◽  
S. Shirahama ◽  
M. Matsuzaki ◽  
...  

It has been reported that the activation of multiple myeloma (MM) cells by CD40 induces proliferation, growth arrest, and apoptosis. To determine whether the biologic sequelae of CD40 activation in MM cells depends on p53 function, we identified temperature-sensitive p53 mutations in the RPMI 8226 (tsp53E285K) and the HS Sultan (tsp53Y163H) MM cell lines. These cells were then used as a model system of inducible wtp53-like function because wild-type-like p53 is induced at permissive (30°C) but not at restrictive (37°C) temperatures. Using p21-luciferase reporter assays, we confirmed that CD40 induces p53 transactivation in RPMI 8226 and HS Sultan cells cultured under permissive, but not restrictive, conditions. Furthermore, CD40 activation of these MM cells under permissive, but not restrictive, temperatures increased the expression of p53 and p21 mRNA and protein. Importantly, CD40 activation induced the proliferation of RPMI 8226 and HS Sultan cells at restrictive temperatures and growth arrest and increased subG1 phase cells at permissive temperatures. These data confirmed that CD40 activation might have distinct biologic sequelae in MM cells, depending on their p53 status.


Blood ◽  
1996 ◽  
Vol 88 (6) ◽  
pp. 2219-2227 ◽  
Author(s):  
M Urashima ◽  
A Ogata ◽  
D Chauhan ◽  
MB Vidriales ◽  
G Teoh ◽  
...  

Interleukin-6 (IL-6) mediates autocrine and paracrine growth of multiple myeloma (MM) cells and inhibits tumor cell apoptosis. Abnormalities of retinoblastoma protein (pRB) and mutations of RB gene have been reported in up to 70% of MM patients and 80% of MM-derived cell lines. Because dephosphorylated (activated) pRB blocks transition from G1 to S phase of the cell cycle whereas phosphorylated (inactivated) pRB releases this growth arrest, we characterized the role of pRB in IL-6-mediated MM cell growth. Both phosphorylated and dephosphorylated pRB were expressed in all serum-starved MM patient cells and MM-derived cell lines, but pRB was predominantly in its phosphorylated form. In MM cells that proliferated in response to IL-6, exogenous IL-6 downregulated dephosphorylated pRB and decreased dephosphorylated pRB-E2F complexes. Importantly, culture of MM cells with RB antisense, but not RB sense, oligonucleotide (ODN) triggered IL- 6 secretion and proliferation in MM cells; however, proliferation was only partially inhibited by neutralizing anti-IL-6 monoclonal antibody (MoAb). In contrast to MM cells, normal splenic B cells express dephosphorylated pRB. Although CD40 ligand (CD40L) triggers a shift from dephosphorylated to phosphorylated pRB and proliferation of B cells, the addition of exogenous IL-6 to CD40L-treated B cells does not alter either pRB or proliferation, as observed in MM cells. These results suggest that phosphorylated pRB is constitutively expressed in MM cells and that IL-6 further shifts pRB from its dephosphorylated to its phosphorylated form, thereby promoting MM cell growth via two mechanisms; by decreasing the amount of E2F bound by dephosphorylated pRB due to reduced dephosphorylated pRB, thereby releasing growth arrest; and by upregulating IL-6 secretion by MM cells and related IL-6- mediated autocrine tumor cell growth.


2006 ◽  
Vol 72 (11) ◽  
pp. 1423-1431 ◽  
Author(s):  
Michal Hayun ◽  
Yaniv Naor ◽  
Merav Weil ◽  
Michael Albeck ◽  
Alpha Peled ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3417-3417
Author(s):  
Yutaka Okuno ◽  
Hiro Tatetsu ◽  
Shikiko Ueno ◽  
Hiroyuki Hata ◽  
Yasuhiro Yamada ◽  
...  

Abstract It has been reported that disruption of transcription factors critical for hematopoiesis, such as C/EBPa and AML1, is involved in leukemogenesis. PU.1 is a transcription factor important for both myeloid and lymphoid development. We reported that mice in which the levels of PU.1 were 20% of that of wild-type developed acute myeloid leukemia, T cell lymphoma, and a CLL-like disease. These findings strongly suggest that PU.1 has tumor suppressive activity in multiple hematopoietic lineages. Last year, we reported that PU.1 is downregulated in a majority of multiple myeloma cell lines and and freshly isolated CD138 positive myeloma cells from certain number of myeloma patients, and that tet-off inducible exogenous expression of PU.1 in PU.1 negative myeloma cell lines induced cell growth arrest and apoptosis. Based on their PU.1 expression levels, we divided the myeloma patients into two groups, namely PU.1 high and PU.1 low-to-negative, (cutoff index of 25th percentile of the PU.1 expression level distribution among all patients). The PU.1 low-to-negative patients had a significantly poorer prognosis than the PU.1 high patients. To elucidate the mechanisms of downregulation of PU.1, we performed sequence and epigenetic analysis of the promoter region and the -17 kb upstream region that is conserved among mammalians and important for proper expression of PU.1. There are no mutations in these regions of all five myeloma cell lines. In contrast, the -17 kb upstream region was highly methylated in 3 of 4 PU.1 negative myeloma cell lines, while the promoter region was also methylated to various levels in all five myeloma cell lines including one PU.1 positive cell line. These data suggested that the downregulation of PU.1 in myeloma cell lines might be dependent on the methylation of both regulatory regions of PU.1 gene, especially the -17 kb upstream region. We also evaluated the mechanisms of cell growth arrest and apoptosis of myeloma cell lines induced by PU.1. Among apoptosis-related genes, we identified that TRAIL was upregulated after PU.1 induction. To evaluate the effect of upregulation of TRAIL, we stably introduced siRNA for TRAIL into myeloma cell lines expressing PU.1, and we found that apoptosis of these cells was partially suppressed by siRNA for TRAIL, suggesting that apoptosis of myeloma cells induced by PU.1 might be at least partially due to TRAIL upregulation. We are currently performing DNA microarray analysis to compare the expression levels of genes between before and after PU.1 induction, in order to further elucidate the mechanisms of cell growth arrest and apoptosis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3065-3065 ◽  
Author(s):  
David Samuel diCapua Siegel ◽  
Laura McBride ◽  
Elizabeth Bilotti ◽  
Linda Schmidt ◽  
Zhijie Gao ◽  
...  

Abstract Abstract 3065 Introduction: Significant improvements have been made in the treatment of relapsed/refractory (RR) multiple myeloma (MM), although the disease remains incurable. Recently, a number of clinical trials have evaluated the efficacy of vorinostat (Zolinza®), an oral inhibitor of Class I and II histone deacetylases. Vorinostat has been evaluated as a single agent, showing minimal activity. In vitro studies demonstrated synergy between vorinostat and other pro-apoptotic agents. This led to Phase I and II trials of vorinostat in combination with both proteasome inhibitors and IMiDs. Based upon the positive phase I data, large, multinational phase II and III trials combining vorinostat (Z) and bortezomib (V) are ongoing. Previously presented Phase I data on the combination of lenalidomide (R), dexamethasone and vorinostat has been encouraging. To date, no large experience with this combination in patients previously found to be refractory to lenalidomide and dexamethasone (RD) has been reported. Here we report our single institution experience of 28 consecutive patients with RD refractory myeloma or VRD refractory myeloma treated with the RDZ or VRDZ respectively. Materials and Methods: This is a retrospective chart review of patients who received commercially available oral vorinostat 300 mg or 400 mg once daily (days 1–7 and days 15–21) and lenalidomide 10–25mg (days 1–21) in a 28-day cycle. Ten patients also received bortezomib 1.3 mg/m2 as an intravenous bolus on days 1, 4, 8, and 11. Subjects: All patients were refractory to prior RD. Most of the patients were relapsed and refractory not only to RD, but also to VRD. (Please refer to the table below.) All of the patients treated with VRDZ were R/R to prior VRD. 23/28 of these patients had previous autologous peripheral blood stem cell transplants (ASCT). 11 had two transplants, 1 had three and 5 had previous allogeneic transplants. The median prior lines of therapy were 4 (2-10) and median prior regimens was 5 (2-11). Results: An overall response rate (ORR) of 43% was noted. This included 8 partial responses (PRs) and 4 very good partial responses (VGPRs) or better. An additional 5 showed minimal responses (MRs) and 8 showed stable disease (SD). The overall clinical benefit rate (including MRs and SD) was 89%. The duration of response ranged from two months to 23+ months. The most common toxicities were GI, mostly diarrhea and cramping. Cytopenias were also experienced, but were not different from those expected for this population treated with lenalidomide-based therapy alone. We will report on additional patients, more complete toxicity data, event-free (EFS) and overall survival (OS), as well as a limited subgroup analysis. Conclusions: These results suggest that this convenient oral regimen of vorinostat combined with lenalidomide and dexamethasone is well tolerated in patients with heavily pretreated, RD relapsed/refractory MM. These results further demonstrate the ability of vorinostat to overcome resistance to RD and VRD. Disclosures: Siegel: Celgene: Advisory board, Speakers Bureau; Merck: Advisory board; Millennium: Advisory Board, Speakers Bureau. Off Label Use: vorinostat for multiple myeloma. Bilotti:Celgene: Advisory Board, Speakers Bureau; Merck: Honoraria; Millennium: Advisory Board, Speakers Bureau. McNeill:Celgene: Advisory Board, Speakers Bureau; Millennium: Advisory Board, Speakers Bureau. Graef:Merck Research Laboratories: Employment. Vesole:Celgene: Speakers Bureau; Millennium Pharmaceuticals, Inc.: Speakers Bureau.


2012 ◽  
Vol 2012 ◽  
pp. 1-10 ◽  
Author(s):  
Douaa Sayed ◽  
Mohamed K. Al-Sadoon ◽  
Gamal Badr

Background. Multiple myeloma (MM), an almost incurable disease, is the second most common blood cancer. Initial chemotherapeutic treatment could be successful; however, resistance development urges the use of higher toxic doses accompanied by hematopoietic stem cell transplantation. The establishment of more effective treatments that can overcome or circumvent chemoresistance has become a priority. We recently demonstrated that venom extracted fromWalterinnesia aegyptia(WEV) either alone or in combination with silica nanoparticles (WEV+NPs) mediated the growth arrest and apoptosis of prostate cancer cells. In the present study, we evaluated the impact of WEV alone and WEV+NP on proliferation and apoptosis of MM cells.Methods. The impacts of WEV alone and WEV+NP were monitored in MM cells from 70 diagnosed patients. The influences of WEV and WEV+NP were assessed with flow cytometry analysis.Results. WEV alone and WEV+NP decreased the viability of MM cells. Using a CFSE proliferation assay, we found that WEV+NP strongly inhibited MM cell proliferation. Furthermore, analysis of the cell cycle using the propidium iodide (PI) staining method indicated that WEV+NP strongly altered the cell cycle of MM cells and enhanced the induction of apoptosis.Conclusions. Our data reveal the biological effects of WEV and WEV+NP on MM cells that enable these compounds to function as effective treatments for MM.


Blood ◽  
2002 ◽  
Vol 100 (6) ◽  
pp. 2187-2194 ◽  
Author(s):  
Dharminder Chauhan ◽  
Laurence Catley ◽  
Teru Hideshima ◽  
Guilan Li ◽  
Richard Leblanc ◽  
...  

Abstract 2-Methoxyestradiol (2ME2) an estrogen derivative, induces growth arrest and apoptosis in leukemic cells and is also antiangiogenic. In this study, we demonstrate that 2ME2 inhibits growth and induces apoptosis in multiple myeloma (MM) cell lines and patient cells. Significantly, 2ME2 also inhibits growth and induces apoptosis in MM cells resistant to conventional therapies including melphalan (LR-5), doxorubicin (Dox-40 and Dox-6), and dexamethasone (MM.1R). In contrast to its effects on MM cells, 2ME2 does not reduce the survival of normal peripheral blood lymphocytes. Moreover, 2ME2 enhances Dex-induced apoptosis, and its effect is not blocked by interleukin-6 (IL-6). We next examined the effect of 2ME2 on MM cells in the bone marrow (BM) milieu. 2ME2 decreases survival of BM stromal cells (BMSCs), as well as secretion of vascular endothelial growth factor (VEGF), and IL-6 triggered by the adhesion of MM cells to BMSCs. We show that apoptosis induced by 2ME2 is mediated by the release of mitochondrial cytochrome-c (cyto-c) and Smac, followed by the activation of caspases-8, -9, and -3. Finally, 2ME2 inhibits MM cell growth, prolongs survival, and decreases angiogenesis in a murine model. These studies, therefore, demonstrate that 2ME2 mediates anti-MM activity directly on MM cells and in the BM microenvironment. They provide a framework for the use of 2ME2, either alone or in combination with Dex, to overcome drug resistance and to improve outcome in MM.


2008 ◽  
Vol 26 (15_suppl) ◽  
pp. 8581-8581
Author(s):  
P. Baumann ◽  
K. Müller ◽  
S. Mandl-Weber ◽  
R. Doblhofer ◽  
A. Ammendola ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3966-3966
Author(s):  
David K Edwards ◽  
Venkata D Yellapantula ◽  
Kristi Allen ◽  
Wen Yu Wong ◽  
Jessica Albanese ◽  
...  

Abstract Abstract 3966 The drug treatments currently available for multiple myeloma patients are dramatic improvements over historical regimens, stopping or slowing cancer growth in 80–90% of patients and leading to complete remission in approximately 40% of patients. Many of the new treatment regimens include “novel agents” in combination with dexamethasone, one of the most effective agents used to treat myeloma. The direct mechanism by which dexamethasone works in myeloma is not well characterized but it is assumed that it activates glucocorticoid receptors which results in gene expression changes that promote apoptosis in lymphoid cells. However, often the disease becomes resistant to dexamethasone, and the mechanism for this resistance is not entirely known. To study the mechanism of resistance, two isogenic cell lines, MM.1R and MM.1S, were independently created from the parental cell line MM.1 to represent models of resistance and sensitivity, respectively, to dexamethasone. This model system was created by Steve Rosen and colleagues in the 1990s and was recently deposited in ATCC. Previous studies have demonstrated differential expression of the glucocorticoid receptor NR3C1 but have not precisely identified the genetic difference between MM.1R and MM.1S across the whole genome. To better understand the mechanism behind the differences in drug sensitivity between these isogenic cell lines, we performed extensive characterization of MM.1R and MM.1S. We purchased both lines from ATCC and analyzed each using flow cytometry, CGH, CGH-SNP, mRNA sequencing, and exome sequencing. First, we broadly examined both cell lines, demonstrating a 300,000-fold difference in IC50 of MM.1R to MM.1S after 6 days of dexamethasone treatment. No significant ploidy difference was found between the two lines by flow cytometry analysis. Our CGH results identified 4 copy number differences unique to MM.1R (chr2:p37.1–37.3 deletion, chr4:q32.3–33 deletion, chr5:31.3 deletion, and chr7:q36.3 amplification), the third of which suggested a possible homozygous deletion within NR3C1. To confirm this deletion, we designed primer sets at ∼1kb intervals spanning the entire NR3C1 gene and performed PCR on MM.1R and MM.1S. Our results indicate the presence of a ∼5–8kb deletion of NR3C1 in MM.1R. Additionally, we analyzed our mRNA sequencing data using TopHat-Fusion and identified an inverted fusion between NR3C1 and ARHGAP26, which we confirmed through PCR amplification and Sanger sequencing. From mRNA sequencing, we identified 63 genes with differential expression between MM.1R and MM.1S (FPKM > 5 in either cell line and greater than fourfold change between them). These results demonstrate a reduction in expression of NR3C1 caused by the two independent deletions identified by CGH. The gene with the larges fold change was MGST1, which is associated with drug resistance and thus may be associated with dexamethasone resistance in this model system based on its expression profile. We analyzed our exome sequencing results for high-confidence (called by both SAMtools and GATK) non-synonymous mutations not present in the 1000 Genomes Project and filtered them for expression (FPKM > 5). We identified 218 mutations in MM.1R, 208 mutations which were also expressed in MM.1S and 10 mutations which were not expressed in MM.1S. The 10 genes with these mutations—PDIA5, TCERG1, RANBP9, MMS22L, PHF19, RNMTL1, AURKB, ERN1, GPCPD1, PIGT—present potential additional contributors to dexamethasone resistance. Specifically, for example, overexpression of RANBPM (the protein from RANBP9) results in increased glucocorticoid activity, suggesting that it may work in concert with NR3C1 to mediate the effects of dexamethasone. Ultimately, our results indicate that, unlike previous assumptions, there are several contributors to dexamethasone resistance in this model system and likely even more in the general patient populations, not just differential expression of NR3C1. Furthermore, we have discovered that this differential expression is due to biallelic inactivation of NR3C1 in MM.1R. Future studies will test the relative contribution of each factor to the differential sensitivity to dexamethasone observed in this model system and a broader understanding of this problem in multiple myeloma. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document