Hypoxia Induces Up-Regulation Of P-Glycoprotein and Promotes Resistance To Carfilzomib In Multiple Myeloma

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4907-4907
Author(s):  
Joseph Abraham ◽  
Salama N Noha ◽  
Abdel Kareem Azab

Abstract Introduction Multiple myeloma (MM) is a malignant neoplastic cancer of plasma cells that involves the bone marrow. Generally, patients will respond to treatment initially, but they later become resistant to therapy, and this is ultimately due to a change in the biology of the tumor. Multi-drug-resistance transporter proteins were shown to play a role in drug resistance in MM patients; P-glyco-protein (P-gp) is the most studied of the multi-drug resistance proteins, and it becomes up-regulated in response to many chemotheries. Hypoxia was shown to develop in the BM niche during progression of MM and to play a major role in the dissemination of MM cells to the new BM niches. Tumor-hypoxia was shown todevelop many kinds of solid tumors and hematologic malignancies. Specifically, hypoxia was shown to develop in the BM niche during progression of MM and to play a major role in the dissemination of MM cells to the new BM niches. In this study, we examinned the effect of hypoxia on the expression and activity of P-gp in MM and its contributing to drug resistance to therapies used in MM. Methods and Results We tested the effect of hypoxia on the activity of P-gp in MM lines. We incubated MM cells under hypoxic and normoxic conditions, and we tested their ability to pump out Rhodamine (Rh) by measuring Rh content in the cells by fluorescent reader. First, we optimized the concentration of Rh and the time of incubation with the cells. We found that at all concentrations tested (0.1, 0.5, 1, 5 and 10 ug/ml) and at all incubation time of cells with Rh with MM cells (0.25, 0.5, 1, 2, 4, 6, 8 and 24hrs) , hypoxia increased the efflux of Rh. The most significant efflux was achieved when incubating the cells for 1hr with Rh 1ug/ml. We found that hypoxia increased the efflux of Rh in all MM cell lines tested. Incubation of RPMI cells under hypoxic for 24hrs and 48hrs decreased the Rh content of the cells by about 40% and 65%, respectively. Carfilzomib was previously reported to be a substrate of P-gp, we tested the effect of carfilzomib on the efflux of Rd in the MM cells. Hypoxic and normoxic MM cells were treated for 5hrs with carfilzomib (5 nM) and then incubated for 1hrs with Rh (1ug/ml). We tested the Rh content of the cells by fluorescent reader and found that carfilzomib competed with Rh on the P-gp and decreased the efflux of Rh induced by hypoxic. We tested the effect of carfilzomib on induction of P-gp in hypoxic and normoxic MM cells by treating RPMI cells with a low dose of carfilzomib (0.25nM) for 48hrs under hypoxic or normoxic conditions, and tested the cells ability to efflux Rh. We found that carfilzomib increased P-gp expression and induced efflux of about 30% of the Rh in non-treated normoxic cells.  Hypoxia induced efflux of about 65% of normoxic cells, but no effect was observed with the treatment of carfilzomib. Furthermore, we tested the hypoxia-induced P-gp expression in MM on the sensitivity of MM cells to carfilzomib. We incubated MM cells for 24hrs in hypoxic and normoxic conditions, and cells were treated with carfilzomib (0 or 5nM) for additional 24hrs. We found that while carfilzomib induced the death of about 40% of the cells under normoxic condition, it had no significant effect on the survival of MM cell under hypoxic conditions. Conclusion Hypoxia induced a significant up-regulation of P-gp in MM cells, and increased MM drug resistance to carfilzomib. These results provide mechanistic evidence for drug resistance to carfilzomib in MM, and suggest hypoxia as a novel therapeutic to prevent upregulation of P-gp and drug resistance. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 9-10
Author(s):  
Ye Yang ◽  
Chunyan Gu ◽  
Wang Wang ◽  
Xiaozhu Tang

Key findings CHEK1 and circCHEK1_246aa induce multiple myeloma cell proliferation, drug resistance, and bone lesion formation CHEK1 and circCHEK1_246aa evoke myeloma chromosomal instability, partially through CEP170 activation Abstract Multiple myeloma (MM) is characterized by clonal expansion of plasma cells in the bone marrow (BM). Therefore, effective therapeutic interventions must target both myeloma cells and the BM niche. In the present study, we first demonstrated that CHEK1 expression was significantly increased in human MM samples relative to normal plasma cells, and that in MM patients, high CHEK1 expression was associated with poor outcomes. CHEK1 overexpression increased cellular proliferation in MM cells and evoked drug resistance in vitro, while CHEK1 knockdown abrogated this effect. Moreover, CHEK1 was a high-risk gene for poor outcome in MM patients, and, in paired samples from MM patients taken from newly diagnosed and relapsed MM, CHEK1 expression was upregulated. CHEK1-mediated increases in cell proliferation and drug resistance were due in part to CHEK1-induced chromosomal instability (CIN), as demonstrated by Giemsa staining, exon sequencing, and immunofluorescence. CHEK1 activated CIN, partly by phosphorylating CEP170. Interestingly, CHEK1 promoted osteoclast differentiation by direct phosphorylation and activation of NFATc1, indicating that CHEK1 inhibition could target both MM cell proliferation and macrophage osteoclast differentiation in the BM niche. Intriguingly, we also discovered that MM cells expressed circCHEK1_246aa, a circular CHEK1 RNA, which encoded and was translated to the CHEK1 kinase catalytic center. Transfection of circCHEK1_246aa increased MM CIN and osteoclast differentiation similarly to CHEK1 overexpression, suggesting that MM cells could secrete circCHEK1_246aa in the BM niche to increase the invasive potential of MM cells and promote osteoclast differentiation. Finally, we demonstrated in vivo in xenograft models that CHEK1 overexpression prompted MM proliferation and drug resistance, while CHEK1 knockdown conversely inhibited MM growth. Together, these findings suggest that targeting the enzymatic catalytic center encoded by CHEK1 mRNA and circCHEK1_246aa is a promising therapeutic modality to target both MM cells and the BM niche. Figure Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Jihane Khalife ◽  
James F. Sanchez ◽  
Flavia Pichiorri

Multiple myeloma (MM) is a cancer of terminally differentiated plasma cells (PCs) that develop at multiple sites within the bone marrow (BM). MM is treatable but rarely curable because of the frequent emergence of drug resistance and relapse. Increasing evidence indicates that the BM microenvironment plays a major role in supporting MM-PC survival and resistance to therapy. The BM microenvironment is a complex milieu containing hematopoietic cells, stromal cells, endothelial cells, immune cells, osteoclasts and osteoblasts, all contributing to the pathobiology of MM, including PC proliferation, escape from immune surveillance, angiogenesis and bone disease development. Small extracellular vesicles (EVs) are heterogenous lipid structures released by all cell types and mediate local and distal cellular communication. In MM, EVs are key mediators of the cross-talk between PCs and the surrounding microenvironment because of their ability to deliver bioactive cargo molecules such as lipids, mRNAs, non-coding regulatory RNA and proteins. Hence, MM-EVs highly contribute to establish a tumor-supportive BM niche that impacts MM pathogenesis and disease progression. In this review, we will first highlight the effects of RNA-containing, MM-derived EVs on the several cellular compartments within the BM microenvironment that play a role in the different aspects of MM pathology. We will also touch on the prospective use of MM-EV-associated non-coding RNAs as clinical biomarkers in the context of “liquid biopsy” in light of their importance as a promising tool in MM diagnosis, prognosis and prediction of drug resistance.


2021 ◽  
Vol 20 (1) ◽  
Author(s):  
Chunyan Gu ◽  
Wang Wang ◽  
Xiaozhu Tang ◽  
Tingting Xu ◽  
Yanxin Zhang ◽  
...  

Abstract Background Multiple myeloma (MM) is still incurable and characterized by clonal expansion of plasma cells in the bone marrow (BM). Therefore, effective therapeutic interventions must target both myeloma cells and the BM niche. Methods Cell proliferation, drug resistance, and chromosomal instability (CIN) induced by CHEK1 were confirmed by Giemsa staining, exon sequencing, immunofluorescence and xenograft model in vivo. Bone lesion was evaluated by Tartrate-resistant acid phosphatase (TRAP) staining. The existence of circCHEK1_246aa was evaluated by qPCR, Sanger sequencing and Mass Spectrometer. Results We demonstrated that CHEK1 expression was significantly increased in human MM samples relative to normal plasma cells, and that in MM patients, high CHEK1 expression was associated with poor outcomes. Increased CHEK1 expression induced MM cellular proliferation and evoked drug-resistance in vitro and in vivo. CHEK1-mediated increases in cell proliferation and drug resistance were due in part to CHEK1-induced CIN. CHEK1 activated CIN, partly by phosphorylating CEP170. Interestingly, CHEK1 promoted osteoclast differentiation by upregulating NFATc1 expression. Intriguingly, we discovered that MM cells expressed circCHEK1_246aa, a circular CHEK1 RNA, which encoded and was translated to the CHEK1 kinase catalytic center. Transfection of circCHEK1_246aa increased MM CIN and osteoclast differentiation similarly to CHEK1 overexpression, suggesting that MM cells could secrete circCHEK1_246aa in the BM niche to increase the invasive potential of MM cells and promote osteoclast differentiation. Conclusions Our findings suggest that targeting the enzymatic catalytic center encoded by CHEK1 mRNA and circCHEK1_246aa is a promising therapeutic modality to target both MM cells and BM niche.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1931-1931 ◽  
Author(s):  
Karthik Ramasamy ◽  
Hazera Khatun ◽  
Lee Macpherson ◽  
Ghulam J. Mufti ◽  
Stephen Schey ◽  
...  

Abstract Abstract 1931 Dexamethasone is a conventional anti-multiple myeloma (MM) drug that effectively induces MM cell death at presentation and in relapsed patients that continues to be used against MM alone or in combination with the new therapeutic agents. However, MM cells develop resistance to Dexamethasone and several changes in MM cells have been proposed to explain the lack of response. Long exposure to Dexamethasone can induce genetic changes in MM cells resulting in downregulation or expression of mutated or truncated forms of the glucocorticoid receptor. Additionally, overexpression of members of the ABC transporter will extrude Dexamethasone and reduce intracellular levels, leading to resistance. Resistance to Dexamethasone can also arise from the interaction of MM cells with the bone marrow (BM) microenvironment. More specifically, the pro-apoptotic effect of Dexamethasone can be blocked by cytokines secreted in the BM microenvironment or the contact of MM cells to the BM stroma leading to cell adhesion mediated drug resistance. We now report new mechanisms of resistance involving a direct impact of Dexamethasone on the BM microenvironment independently of the presence of MM cells. We show using a novel co-culture experimental platform that resistance to Dexamethasone in MM results from the direct pro-proliferative effect of Dexamethasone on fibroblasts in the BM stroma, which involves the Src family of kinases. Using both MM cell lines and BM aspirates from MM patients, we found that MM cells in the presence of fibronectin accelerate Dexamethasone-induced proliferation of fibroblasts. Additionally, we found that the integrity and functionality of osteoclasts is not affected by treatment with Dexamethasone. These results suggest that resistance of MM cells to Dexamethasone can arise from the direct effect of the drug on fibroblasts as well as on the lack effect on osteoclasts. This would result in formation of BM niches that prevent apoptosis of MM induced by Dexamethasone. Our data show that adhesion of MM cells on stromal cells or osteoclasts rather than adhesion on fibronectin promote MM cell adhesion drug resistance to Dexamethasone. The Src and c-Abl kinase inhibitor Dasatinib used at clinically achievable doses (150 nM) blocked adhesion of MM cells on fibroblasts and osteoclasts. Additionally, both fibroblast proliferation and osteoclasts activity can be blocked with Dasatinib resulting in sensitisation of MM cells to treatment with Dexamethasone. However, Dasatinib alone did not affect MM cell proliferation or viability. Our data support the therapeutic use of Dexamethasone in combination with Src inhibitors against MM to target MM cell viability and the protective role of the BM microenvironment, respectively. Our results also show a new mechanism that promotes drug resistance through the direct effect of anti-tumour therapeutic drugs on the stroma independently of the presence of tumour cells. Our data support the use of co-culture experimental platforms of tumour cells with the tumour microenvironment to evaluate and predict more accurately the effect of possible anti-tumoral activity of therapeutic drugs on both tumour cells and stroma as well as to understand the mechanisms of drug resistance. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5404-5404 ◽  
Author(s):  
Guido J. Tricot ◽  
Ye Yang ◽  
Zhimin Gu ◽  
Hongwei Xu ◽  
Junwei Huang ◽  
...  

Abstract Background Despite the major advances in the therapy of MM, many patients ultimately relapse and die of this disease. This is most likely due to the persistence of multiple myeloma stem cells (MMSCs), which survive even after the most intensive treatments and are insensitive to non-transplant therapy. We have previously demonstrated that RARα2 expression increased in CD138 selected plasma cells of relapsed myelomas (MM) and that increased expression was linked to poor prognosis in newly diagnosed MM patients. In this study, we further explore the relationship between RARα2 and MMSCs. Materials and Methods RT-PCR was used to detect RARα2, induced pluripotent stem cell (iPS) gene, Wnt and Hedgehog (Hh) gene expression in MMSCs and bulk cells. Clonogenic formation assays were performed by plating RARα2 over-expressing ARP1 and OCI-MY5 MM cell lines in soft agar, treated with bortezomib, doxorubicin, etoposide, and MK-571. Side population (SP) fraction, ALDH activity and cell apoptosis in MM cells were detected by flow cytometry. Western blot were utilized to dissect RARα2-related signaling pathway. Effect of RARα2 on MM progression in vivo was also analyzed in 5TGM1 myeloma mice. Results Higher expression of RARα2 was identified in the multiple myeloma stem cell (MMSC) fraction. Over-expression of RARα2 in MM cell lines resulted in 1) increased drug-resistance 2) increased clonogenic potential 3) activation of both Wnt and Hedgehog (Hh) pathways 4) increased side population (SP) and aldehyde dehydrogenase1 (ALDH1) levels and 5) increased expression of iPS genes. The opposite effects were seen with RARα2 knockdown. RARα2 induced drug resistance by activating the drug efflux pump gene ABCC3 and anti-apoptotic Bcl-2 family members. Inhibition of Wnt signaling or ABCC3 function could overcome drug resistance in RARα2 over-expressing MM cells. Our In vivo study indicated that targeting Wnt and Hh pathways using CAY10404, cyclopamine or itraconazole could significantly reduce tumor burden and increase survival time in the 5TGM1 mouse model. We confirmed these findings in primary myeloma stem cells. Finally, we showed that primary myelomas with high RARα2 expression possess similar features as myeloma stem cells. Conclusion This study demonstrates that RARα2 plays a crucial role in MMSCs by activating Wnt and Hh pathways, regulating the transcription of iPS genes and conferring drug resistance. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (12) ◽  
pp. 3881-3889 ◽  
Author(s):  
Joel G. Turner ◽  
Jana L. Gump ◽  
Chunchun Zhang ◽  
James M. Cook ◽  
Douglas Marchion ◽  
...  

AbstractWe investigated the role of the breast cancer resistance protein (BCRP/ABCG2) in drug resistance in multiple myeloma (MM). Human MM cell lines, and MM patient plasma cells isolated from bone marrow, were evaluated for ABCG2 mRNA expression by quantitative polymerase chain reaction (PCR) and ABCG2 protein, by Western blot analysis, immunofluorescence microscopy, and flow cytometry. ABCG2 function was determined by measuring topotecan and doxorubicin efflux using flow cytometry, in the presence and absence of the specific ABCG2 inhibitor, tryprostatin A. The methylation of the ABCG2 promoter was determined using bisulfite sequencing. We found that ABCG2 expression in myeloma cell lines increased after exposure to topotecan and doxorubicin, and was greater in logphase cells when compared with quiescent cells. Myeloma patients treated with topotecan had an increase in ABCG2 mRNA and protein expression after treatment with topotecan, and at relapse. Expression of ABCG2 is regulated, at least in part, by promoter methylation both in cell lines and in patient plasma cells. Demethylation of the promoter increased ABCG2 mRNA and protein expression. These findings suggest that ABCG2 is expressed and functional in human myeloma cells, regulated by promoter methylation, affected by cell density, up-regulated in response to chemotherapy, and may contribute to intrinsic drug resistance.


2020 ◽  
Vol 0 (0) ◽  
Author(s):  
Emine Öksüzoğlu ◽  
Gül Kozalak

AbstractBackgroundMultiple myeloma (MM), a malignancy of plasma cells, is the second most prevalent hematological cancer. Bortezomib is the most effective chemotherapeutic drug used in treatment. However, drug-resistance prevents success of chemotherapy. One of the factors causing drug-resistance is dysfunction of apoptotic-pathways. This study aimed to evaluate the relationship between expression levels of Bcl-2, Bax, caspase-3 and p-53 genes involved in apoptosis and the development of bortezomib-resistance in MM cell lines.Materials and methodsMultiple myeloma KMS20 (bortezomib-resistant) and KMS28 (bortezomib-sensitive) cell lines were used. 3-[4,5-Dimethylthiazol-2-yl] 1-2,5-diphenyltetrazolium bromide (MTT) assay was performed to determine IC50 values of bortezomib. RNAs were isolated from bortezomib-treated cell lines, followed by cDNA synthesis. Expression levels of the genes were analyzed by using q-Realtime-PCR.ResultsAs a result, Bcl-2/Bax ratio was higher in KMS20 (resistant) cells than in KMS28 (sensitive) cells. Expression of caspase-3 decreased in KMS20-cells, whereas increased in KMS28-cells. The results indicate that apoptosis was suppressed in resistant cells.ConclusionThese findings will enable us to understand the molecular mechanisms leading to drug-resistance in MM cells and to develop new methods to prevent the resistance. Consequently, preventing the development of bortezomib resistance by eliminating the factors which suppress apoptosis may be a new hope for MM treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1737-1737
Author(s):  
David M. Lucas ◽  
Ellen J. Sass ◽  
Ryan B. Edwards ◽  
Li Pan ◽  
Gerard Lozanski ◽  
...  

Abstract Abstract 1737 Poster Board I-763 We previously reported the efficacy and B-cell selectivity of the natural product silvestrol in acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL), using both primary cells and B-cell lines. We also showed that silvestrol inhibits translation, resulting in rapid depletion of the short half-life protein Mcl-1 followed by mitochondrial damage and apoptosis. Cencic et al. reported that silvestrol directly blocks translation initiation by aberrantly promoting interaction of eIF4A with capped mRNA (PLoS One 2009; 4(4):e5223). However, the loss of Mcl-1 in breast and prostate cancer cell lines is delayed relative to what we observe in B-leukemias (48 hr vs. 4-6 hr in CLL and ALL cells). Additionally, silvestrol does not reduce Mcl-1 expression in normal T-cells to the same extent that it does in B-cells, potentially explaining in part the relative resistance of T-cells to this agent. We therefore investigated cell-type differences, as well as the importance of Mcl-1, in silvestrol-mediated cytotoxicity. We incubated the ALL cell line 697 with gradually increasing concentrations of silvestrol to generate a cell line (697-R) with resistance to 30 nM silvestrol (IC50 of parental 697 < 5 nM). No differences between 697-R and the parental line were detected upon detailed immunophenotyping. However, cytogenetic analysis revealed a balanced 7q;9p translocation in 697-R not present in the parental 697 cell line that may be related to the emergence of a resistant clone. We also detected no difference in expression of multi-drug resistance proteins MDR-1 and MRP, which can contribute to resistance to complex amphipathic molecules such as silvestrol. In contrast, we found that baseline Mcl-1 protein expression is strikingly increased in 697-R cells relative to the parental line, although these cells still show similar percent-wise reduction in Mcl-1 upon re-exposure to 80 nM silvestrol. To investigate whether this resistance to silvestrol is reversible, 697-R cells were maintained without silvestrol for 6 weeks (∼18 passages). During this time, viability remained near 99%. Cells were then treated with 30 nM silvestrol. Viability was 94% at 48 hr post-treatment and returned to 99% within a week, while parental 697 cells with the same treatment were completely dead. Baseline Mcl-1 levels remained elevated in 697-R even with prolonged silvestrol-free incubation. These results indicate that the resistance phenotype is not rapidly reversible, as is seen with transient upregulation of multi-drug resistance or stress-response proteins. Additionally, silvestrol moderately induces the transcription of several pro-apoptotic Bcl-2 family members and results in elevated levels of these proteins despite its translation inhibitory activity. Interestingly, no such activity is detected in silvestrol-treated normal T-cells. Together, these results support the hypothesis that in B-cells, silvestrol induces cell death by altering the balance of pro- and anti-apoptotic factors, and that increased Mcl-1 protein can force the balance back toward survival. This work further underscores the importance of Mcl-1 in silvestrol-mediated cytotoxicity. We are now investigating the mechanism of Mcl-1 upregulation in 697-R cells to identify a factor or pathway that can be targeted therapeutically to circumvent resistance. Silvestrol is currently undergoing preclinical pharmacology and toxicology investigation by the U.S. National Cancer Institute Drug Development Group at the Stage IIA level to facilitate its progression to Phase I clinical testing. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document