Zoledronate Can Overcome Drug Resistance in a Dexamethasone Resistant Myeloma Cell Line.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4894-4894
Author(s):  
Masayuki Koizumi ◽  
Chiaki Nakaseko ◽  
Chikako Ohwada ◽  
Ryuko Cho ◽  
Miki Nishimura ◽  
...  

Abstract <Introduction> Multiple myeloma (MM) is almost invariably fatal despite all available chemotherapeutic and supportive treatment. Initial treatment of MM with dexamethasone (Dex), a key drug for MM chemotherapy, effectively induces myeloma cell death. However, prolonged drug exposure results in the development of Dex-resistance and clinical failures. Therefore, one of the most important issues in myeloma therapy is to overcome resistance to Dex at the stage of clinical refractoriness. Several recent in-vitro studies have demonstrated an antitumor effect of nitrogen-containing amino-bisphosphonates (N-BPs) in some tumor cell lines, including myeloma cell lines. However, few data are available concerning the effects of N-BPs on Dex resistant myeloma cell lines. In this study, we have established a Dex-resistant human myeloma cell line and investigated the antitumor effect of the third generation bisphosphonate Zoledronate (ZOL) on the Dex-resistant subline. <Methods> 1) Development of de novo resistance through continuous exposure to Dex. A Dex-resistant human myeloma cell line (Dex-R) was selected from the Dex-sensitive myeloma cell line RPMI8226 by continuously exposing cells to gradually increasing doses of Dex. Resistance was validated by flow cytometry. 2) Apoptosis assay. Dual-color FACS with propidium iodide (PI) and Annexin V was used to detect Dex-induced apoptosis in RPMI8226 and Dex-R. 3) Surface markers. Phenotypes of sensitive and resistant cells were compared by FACS analysis. 4) Cell Proliferation and DNA synthesis assay. RPMI8226 and Dex-R cells were cultured in 96 wells plates for 72 hours in the absence or presence of Dex (1 μM) or increasing concentrations of ZOL (2.5 to 50 μM). Cell growth was assessed by WST-8 assay kit. DNA synthesis was measured according to thymidine uptake. 5) Actin staining. RPMI8226 and Dex-R cells were cultured for 48 hours in the absence or presence of Dex (2 μM) or ZOL (40 μM). The distribution of f-actin stained by fluorescent phalloidin was investigated by confocal fluorescence microscopy. <Results> The developed subline Dex-R showed reduced apoptotic and antiproliferative responses to Dex treatment. Flow cytometry on 24 hours Dex exposure revealed significantly low percentage of apoptotic (Annexin V positive and PI negative) population in Dex-R cells compared with RPMI8226 cells (6.7% vs. 29.9%). Our data demonstrate that Dex-R cells showed increased CD38, CD44, CD49d (VLA-4) expression, and decreased CD45, CD95 (Fas), CD138 (Syndecan-I) expression as compared to native RPMI8226 cells. ZOL induced apoptosis and inhibited DNA synthesis and cell proliferation in both RPMI8226 and Dex-R cell lines in a dose-dependant manner. Interestingly, some modifications of the cellular morphology were observed in ZOL-treated cells. Rhodamine-phalloidin staining of the f-actin cytoskeleton showed disruption of the cytoskeleton in these deformed cells. This fibroblast-like cellular morphology was common to Dex-R and RPMI8226 cells treated with ZOL, but was not detected under Dex treatment. This result might reflect different mechanism of antitumor effect between Dex and ZOL.. <Conclusion> Our results suggest that ZOL can induce myeloma cell death in vitro in a different mechanistic way than Dex. ZOL might be effective as an antitumor drug for Dex resistant myeloma.

Blood ◽  
1999 ◽  
Vol 94 (10) ◽  
pp. 3551-3558 ◽  
Author(s):  
Maged S. Mahmoud ◽  
Ryuichi Fujii ◽  
Hideaki Ishikawa ◽  
Michio M. Kawano

In multiple myeloma (MM), the cell surface protein, CD19, is specifically lost while it continues to be expressed on normal plasma cells. To examine the biological significance of loss of CD19 in human myeloma, we have generated CD19 transfectants of a tumorigenic human myeloma cell line (KMS-5). The CD19 transfectants showed slower growth rate in vitro than that of control transfectants. They also showed a lower capability for colony formation as evaluated by anchorage-independent growth in soft agar assay. The CD19 transfectants also had reduced tumorigenicity in vivo when subcutaneously implanted into severe combined immunodeficiency (SCID)-human interleukin-6 (hIL-6) transgenic mice. The growth-inhibitory effect was CD19-specific and probably due to CD19 signaling because this effect was not observed in cells transfected with a truncated form of CD19 that lacks the cytoplasmic signaling domain. The in vitro growth-inhibitory effect was confirmed in a nontumorigenic human myeloma cell line (U-266). However, introduction of the CD19 gene into a human erythroleukemia cell line (K-562) also induced growth inhibition, suggesting that this effect is CD19-specific, but not restricted to myeloma cells. These data suggest that the specific and generalized loss of CD19 in human myeloma cells could be an important factor contributing to the proliferation of the malignant plasma cell clones in this disease.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5169-5169
Author(s):  
Paul Neeson ◽  
Karen Chen ◽  
Tsin Tai ◽  
Amanda Shin ◽  
Joanne Davis ◽  
...  

Abstract The proteasome inhibitor Bortezomib (Bz) is a potent inducer of plasma cell apoptosis via suppression of NFkB activity. Bz is also associated with the dose limiting induction of thrombocytopenia as well as inhibition of dendritic cell (DC) function, which may limit the induction of autologous T cell responses to myeloma antigens in vivo. We hypothesized that the dose, and therefore the adverse effects of Bz, may be reduced by the addition of the agonistic anti-TRAIL-R1 antibody Mapatumumab (Mp). A dose of Bz that did not result in detectable apoptosis was determined for each of the cultured human plasma cell lines (RPMI8226, U266, LP-1, NCI-H929, OPM-2 and JJN3) via titration experiments in which each cell line was cultured for 24–48 hrs in Bz concentrations from 0.1–10nM. Cells were stained with annexin V-FITC and the viability dye 7-AAD and analyzed on a BD LSRII flow cytometer. Plasma cell surface expression of TRAIL-R-1 and -2 were also assessed and correlated with sensitivity to Mp induced apoptosis. Bz monotherapy at 10nM produced 85–98% apoptosis in all plasma cell lines. In contrast 4/6 cell lines were sensitive to Mp alone, RPMI8226 at 0.06ug/ml, U266 and OPM-2 at 1ug/ml and LP-1 at 10ug/ml. When non-apoptosis inducing Bz doses were used in combination with titrated doses of Mp (from 0.01 to 50ug/ml) apoptosis of RPMI8226, U266 and OPM-2 was enhanced by 32%, 30% and 15% respectively compared to Mp alone (p=<0.05 for each cell line). The particular sensitivity of RPMI8226 to Bz/Mp therapy correlated with this cell line’s higher level of TRAIL-R 1 surface expression in comparison to U266. In contrast, the addition of Bz to Mp did not result in any additional apoptosis induction over Mp alone in LP-1, JJN3 and NCI-H929. Conversely, 24 hrs of Bz pretreatment resulted in subsequent enhanced sensitivity of LP-1 to Mp but not NCI-H929. In conclusion, the effective dose of Bz may be significantly reduced when used in combination with Mp. The Bz+Mp combination therapy may be more efficacious than Bz alone and allow dose modification of Bz. Reduction in Bz dose may in-turn reduce side-effects whilst allowing the preservation or promotion of endogenous immune responses. Figure 1. Combination bortezomib and Mapatumamab therapy induce synergistic death of the human myeloma cell line RPMI8226. A dose response study for (A) bortezomib (B) Mapatumamab was performed on RPMI8226 cells by treating the cells with titrated doses of the monotherapy for 48 hours in triplicate wells. Subsequently RPMI8226 cells were treated with combination bortezomib and Mapatumamab therapy (C). Bortezomib dose (1nM) did not induce apoptosis above untreated controls, Mp was added at doses from 0.03 to 0.15ug/ml. A statistical comparison was made between apoptosis with Mp monotherapy versus Bz plus Mp using student t-test, * represents data points where combination therapy is statistically different to Mp alone, p<0.05. Results are representative of two separate experiments. Figure 1. Combination bortezomib and Mapatumamab therapy induce synergistic death of the human myeloma cell line RPMI8226. A dose response study for (A) bortezomib (B) Mapatumamab was performed on RPMI8226 cells by treating the cells with titrated doses of the monotherapy for 48 hours in triplicate wells. Subsequently RPMI8226 cells were treated with combination bortezomib and Mapatumamab therapy (C). Bortezomib dose (1nM) did not induce apoptosis above untreated controls, Mp was added at doses from 0.03 to 0.15ug/ml. A statistical comparison was made between apoptosis with Mp monotherapy versus Bz plus Mp using student t-test, * represents data points where combination therapy is statistically different to Mp alone, p<0.05. Results are representative of two separate experiments.


2019 ◽  
Vol 71 (2) ◽  
pp. 573-581 ◽  
Author(s):  
Akira Iwamoto ◽  
Yuichi Inoue ◽  
Hirofumi Tachibana ◽  
Hiroharu Kawahara

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1568-1568 ◽  
Author(s):  
Rajni Sinha ◽  
Ebenezer David ◽  
Emily Zeilter ◽  
Claire Torre ◽  
Jonathan L. Kaufman ◽  
...  

Abstract Introduction Multiple myeloma is a clonal plasma cell malignancy characterized by proliferation and accumulation of plasma cells in the bone marrow. Most patients are incurable with the current treatment modalities. Clearly novel agents are needed to improve the outcome for patients with myeloma. We have previously shown that the combination of bortezomib and tipifarnib results in synergistic myeloma cell death. This increase in apoptosis is associated with down regulation of phosphorylated AKT, a potent anti-apoptotic signaling molecule. Therefore, agents that target AKT represent ideal compounds for further study in myeloma. Perifosine is a novel, oral bioavailable alkylphospholipid. Perifosine has displayed apoptotic and antipropliferative activity in vitro and in vivo in several human cancer models including leukemia. Perifosine exerts its actions by interfering with key intracellular pathways including AKT, MAPK, JNK, p21waf1. Our hypothesis is that targeting AKT via multiple upstream pathways will result in increased myeloma cell apoptosis. Therefore, we assessed the effects of single agent perifosine with and without tipifarnib on multiple myeloma cell lines. Method The myeloma cell line RPMI8226 was used. Cell viability and proliferation were assessed using MTT assays. Cells were incubated with increasing concentrations of both agents alone and in combination. Cell proliferation was assayed at 24, 48 and 72 hours. Western blots were then carried out to evaluate the effects of the intracellular protein PDK1, one of the critical signaling molecules that phosphorylates and activates AKT. Results As we and others have previously shown, tipifarnib at concentrations that can be achieved clinically is associated with minimal cytotoxicity. At 5 μM, tipifarnib decrease proliferation by only 20%. In contrast, there is a potent dose response effect of single agent perifosine (Fig. 1). These results were apparent as early as 24 hours. When tipifarnib at 5 μM is used in combination with a subtherapeutic dose of perifosine (2 μM), there is a marked decrease in cell proliferation (Fig. 2). In addition, combination therapy resulted in a reduction in the phosphorylated form of PDK1, a critical finding that was not seen with either drug alone. Conclusion Combination therapy with tipifarnib and perifosine results in less cell proliferation compared to either agent used alone in the RPMI8226 myeloma cell line. The dosages employed in these in-vitro studies are lower than those used in previously published data and are clinically achievable. Studies targeting other cell lines including MM.1R, MM.1S, and U266 are in progress. Analysis of AKT, Caspase 3, 8 and 9 are being explored to help delineate the mechanism of this novel combination. The goal is to develop further effective treatment options for patients with myeloma. Figure 1 Figure 1. Figure 2 Figure 2.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5728-5728
Author(s):  
Ferit Avcu ◽  
Mustafa Guner ◽  
Muammer Misirci ◽  
Pinar Elci ◽  
Mukerrem Safali ◽  
...  

Abstract Ankaferd Blood Stopper (ABS), a unique traditional herbal mixture, has been used topically to stop bleeding for centuries in Anatolia. As well as ABS has been used as a blood-stopping agent, it may also have a considerable therapeutic benefit, because of its anti-infective, anti-neoplastic, and wound healing properties. The aim of this study is to investigate the anti-neoplastic effects of the ABS on myeloma cell line, in vitro and on the plasmocytoma development in Balb/c mice by intraperitoneal injection of pristane, in vivo. We therefore sought to evaluate the efficacy of ABS on MM cells and to study the modulation of cell-death pathways. The cytotoxicity of ABS against the MM cell lines (RPMI-8226, and ARH-77) was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide-dye reduction assay. Responses to ABS by RPMI-8226 and ARH-77 cell lines were dose dependent but not time dependent. The IC50 values for RPMI 8226 and ARH 77 myeloma cell lines in 24h were 12,84 μL/mL and 13,86 μL/mL, respectively. Various cell-death characteristics such as caspase-3, Bcl-2, Bax were studied in response to ABS, but we couldn’t demonstrate specific features of apoptotic cell death, in vitro. We have also investigated the effect of the ABS on the pristane (2.6.10.14-tetramethylpentadecane)-induced plasmacytoma (PCT) development on six-week-old BALB/c mice. Three groups of mice were treated with intraperitoneal ABS (1 mg/kg, 0.5mg/kg, and 0,1mg/kg) per-week for eight weeks after pristane-induced PCT development. The study was stopped at twelfth week, the remaining mice were autopsied, and peritoneal tissues were examined histologically for PCTs. A database of different groups’ mice was analyzed using Kaplan-Meier and Cox regression statistics based on variables. Kaplan-Meier analysis revealed a difference of the survival of pristane-induced alone between the groups of pristane-induced plus ABS 1 mg/kg, 0.5mg/kg, and 0.1mg/kg. (Log-rank, p=0.016; p<0.001 and p<0.001; respectively). The present results indicate that direct anti-tumor effect of ABS on pristane-induced PCT and significantly increased survival. This hypothesis needs to now be further investigated in clinical trials. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 63 (1) ◽  
pp. 141-145
Author(s):  
Yuliya Khochenkova ◽  
Eliso Solomko ◽  
Oksana Ryabaya ◽  
Yevgeniya Stepanova ◽  
Dmitriy Khochenkov

The discovery for effective combinations of anticancer drugs for treatment for breast cancer is the actual problem in the experimental chemotherapy. In this paper we conducted a study of antitumor effect of the combination of sunitinib and bortezomib against MDA-MB-231 and SKBR-3 breast cancer cell lines in vitro. We found that bortezomib in non-toxic concentrations can potentiate the antitumor activity of sunitinib. MDA-MB-231 cell line has showed great sensitivity to the combination of bortezomib and sunitinib in vitro. Bortezomib and sunitinib caused reduced expression of receptor tyrosine kinases VEGFR1, VEGFR2, PDGFRa, PDGFRß and c-Kit on HER2- and HER2+ breast cancer cell lines


Blood ◽  
1993 ◽  
Vol 82 (12) ◽  
pp. 3721-3729 ◽  
Author(s):  
N Huang ◽  
MM Kawano ◽  
H Harada ◽  
Y Harada ◽  
A Sakai ◽  
...  

Abstract Recent immunophenotypic analysis has shown that the heterogeneous expression of the adhesion molecule VLA-5 classifies myeloma cells into VLA-5+ mature and VLA-5- immature subpopulations. To further clarify the two myeloma subpopulations, we generated a monoclonal antibody, MPC- 1, by immunizing mice with an adherent human myeloma cell line, KMS-5. The MPC-1 antibody recognized a 48-Kd surface antigen on KMS-5 but not on U-266, a nonadherent human myeloma cell line. Specificity characterization showed that MPC-1 antigen was expressed on mature myeloma cells, normal plasma cells, and mature B cells, whereas pre-B cells and germinal center B cells lacked its expression. Monocytes and a human bone marrow stromal cell line, KM102, also expressed this antigen. Two subclones of MPC-1+ VLA-5+ (KMS-5Ad) and MPC-1-VLA-5+ (KMS- 5NAd) were separated from the KMS-5 cell line. The KMS-5NAd adhered to KM102 more tightly than did the KMS-5NAd, and the U-266 (MPC-1-VLA-5-) displayed almost no adherence to the KM102. The adhesion of the KMS-5Ad was partially inhibited by the MPC-1 antibody. These results, taken together, suggest that the MPC-1 antigen serves as a differentiation marker for B-lineage cells, including plasma cells, and may function as an adhesion molecule involved in the interaction of mature myeloma cells with bone marrow stromal cells.


PLoS ONE ◽  
2017 ◽  
Vol 12 (8) ◽  
pp. e0182152 ◽  
Author(s):  
Etsuko Tokunaga ◽  
Hidehiko Akiyama ◽  
Vadim A. Soloshonok ◽  
Yuki Inoue ◽  
Hideaki Hara ◽  
...  

Science ◽  
1982 ◽  
Vol 216 (4549) ◽  
pp. 997-999 ◽  
Author(s):  
A Karpas ◽  
P Fischer ◽  
D Swirsky

Sign in / Sign up

Export Citation Format

Share Document