Depsipeptide in the Treatment of Relapsed and Refractory Multiple Myeloma (MM): A Prospective Evaluation of the Cell Cycle.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1497-1497
Author(s):  
Zoe Goldberg* ◽  
Scott Ely ◽  
Selina Chen-Kiang ◽  
Martha Chesi ◽  
Peter L. Bergsagel ◽  
...  

Abstract Background: Dysregulation of the cell cycle and apoptosis are two critical events in the pathophysiology of MM. This notion is supported by: 1)A high tumor burden is often present despite a low rate of tumor cell proliferation. 2)G1 arrest is common in MM cells while normal plasma cells are permanently withdrawn from cell cycle. 3) Cyclin D1 is often overexpressed without a defined genetic substrate. Herein, we show that cell cycle evaluation in vivo is feasible and that the histone-deacetylase inhibitor depsipeptide might be effective in selected patients with MM. Patients and Methods: In vitro studies were performed in 12 human MM cell lines with defined cytogenetic abnormalities. The IC50 for depsipeptide was determined by evaluation of apoptosis by standard methods. In vivo studies where done as correlates in a phase II protocol. These include: Immunohistochemistry (IHC) for co-expression of CD138/Ki-67 as a proliferation index (PCPI), cyclin D1, D3, caspase 3 cleavege, CD31 and bcl-2 before treatment and at 24 hrs and 30 days after treatment. Gene array studies are being performed on selected patients at those timepoints. To date, four stage III patients (PTS) with relapsed MM with four or fewer prior lines of therapy have been treated with one to three cycles of depsipeptide at a dose of 13mg/m2,as a 4-hour infusion on days 1, 8, and 15, repeated every 28 days. Mean age was 63 years (range, 56 to 72). KPS of >80%. Mean albumin was 3.5, (range, 3.2 to 4), mean LDH was 243 (range, 179 to 315). Results: 1)Depsipeptide induces apoptosis in several MM cell lines. All lines were susceptible to depsipeptide, however, differential sensitivities were noted. Three cell lines (ie U266) that contained 11q13 translocation (cyclin D1 overexpression) were the most sensitive with IC50s at least 2 fold lower than other lines. 2) Cell cycle changes are induced by depsipeptide: In 2/4 PTS, a significant increase of the PCPI was seen, whereas a marked reduction in the PCPI in a patient with cyclin D3 overexpression (27% to 16%) was also noted. One patient had an increase of cyclin D1 post treatment. No changes where seen in bcl-2, CD-31, or cleaved caspase-3 expression. 3) Depsipeptide is safe in a limited cohort of MM PTS: Grade 2 fatigue and anorexia were the most common toxicities. Mild thrombocytopenia (mean of 67) did not require transfusions. One patient had stable disease after 3 cycles of treatment, one patient had progression of disease after 3 cycles, one patient progressed after the 1st cycle, and one patient is too early for evaluation. Conclusions: 1)Patients with 11q13 translocation should be a target for treatment with depsipeptide. 2)Depsipeptide given on this schedule is safe and can stabilize tumor-mass in PTS with otherwise progressive relapsed and refractory disease.3) Evidence of cell cycle modulation can be seen during treatment with depsipeptide. No profound changes in apoptosis is evident.4)Further studies may help to understand the mechanism of transcriptional regulation by depsipeptide and will help design rational therapy and combinations. This study continues to accrue patients as part of New York Phase II Consortium. Supported by NCI grant (SAIC1N01-CO-12400-02) and a SCOR for Myeloma grant from the Leukemia and Lymphoma Society of America.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4304-4304
Author(s):  
Dirk Winkler ◽  
Daniela Nitsch ◽  
Christof Schneider ◽  
Annett Habermann ◽  
Hartmut Döhner ◽  
...  

Abstract Apoptosis can be induced by various stimuli including DNA-damaging anticancer drugs and chemical agents such as the protein kinase inhibitor staurosporine. To investigate the expression of apoptosis and cell cycle regulating proteins the lymphoma cell lines JURKAT (non-EBV transformed T-ALL) and EHEB (EBV-transformed B-CLL) were incubated with staurosporine. FACS analyses were performed with double staining of Annexin PE-V/7AAD to determine the rates of staurosporine induced apoptosis and for detection of active Caspase-3 after 24 hours and 48 hours. Similar rates of apoptosis which were achieved with lower concentrations of staurosporine in JURKAT (0.125μM-0.5μM vs. 0.5μM-2μM in EHEB). Expression changes after staurosporine treatment were examined for the following proteins: procaspase-8, procaspase-9, Apaf-1, active caspase-3, PARP, CDK4, CDK2, Survivin, p21, p27, BCL-2, BAX, Cyclin-D1/D2/D3, Rb, cIAP2, XIAP, and Akt1 by Western blotting. Cleavage of procaspase-8 and procaspase-9 was observed in both cell lines upon treatment. In JURKAT, subsequent activation of caspase-3 could be detected by Western Blotting as well as by FACS. In contrast, no active caspase-3 was detected in treated EHEB cells by Western blotting and only moderate activation was observed by FACS, although PARP-cleavage was clearly detected in both cell lines by Western blotting. Apoptotic regulators were differentially regulated when comparing treated JURKAT and EHEB cells. Treatment of JURKAT cells led to an up-regulation of BCL-2 and down-regulation of Akt1 and BAX, but not to expression changes of XIAP and Apaf-1. In contrast, XIAP and Apaf-1 were down-regulated in EHEB upon treatment, whereas no change in protein levels was observed for BCL-2 and BAX. Furthermore, differences between the two cell lines in response to staurosporine treatment were observed for the cell cycle proteins p27, p21, CDK4, Cyclin-D1/D2/D3 and Rb. Down-regulation of p27, p21 and Cyclin-D1 and up-regulation of Cyclin-D3 was only seen in treated EHEB cells. In the opposite, JURKAT showed up-regulation of Cyclin-D1 and down-regulation of Cyclin-D3 and CDK4 upon treatment. Interestingly, in EHEB Cyclin-D2 was initially down-regulated (after 24 hours) followed by an up-regulation later on. Both cell lines responded with cleavage of Rb upon treatment. Levels of cIAP2, Survivin and CDK2 were not altered in either cell line. In summary, characteristic responses to staurosporine treatment were detected in EHEB and JURKAT. In both cell lines apoptosis induction resulted in a cleavage of Rb despite opposite effects on Cyclin-D1 and Cyclin-D3 expression. The most striking difference in response to staurosporine incubation was a PARP cleavage in EHEB cells without significant activation of caspase-3 or alteration in BCL-2 expression in combination with a higher resistance to apoptosis induction by staurosporine when compared to JURKAT. A previous study indicated that apoptosis resistance in EBV-infected B cell lymphomas is promoted by an inactive caspase-3 pathway and an elevated expression of BCL-2 that is not altered by etoposide treatment. Therefore, the distinct protein expression response of EHEB to staurosporine treatment might be in part a result of its immortalization by EBV transformation. Further analyses are in progress to elucidate the response of lymphoma cell lines to fludarabine and etoposide.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 780-780
Author(s):  
Andrew G. Muntean ◽  
Liyan Pang ◽  
Mortimer Poncz ◽  
Steve Dowdy ◽  
Gerd Blobel ◽  
...  

Abstract Megakaryocytes, which fragment to give rise to platelets, undergo a unique form of cell cycle, termed endomitosis, to become polyploid and terminally differentiate. During this process, cells transverse the cell cycle but the late stages of mitosis are bypassed to lead to accumulation of DNA up to 128N. While the mechanisms of polyploidization in megakaryocytes are poorly understood, a few cell cycle regulators, such as cyclin D3, have been implicated in this process. Hematopoietic transcription factors, including GATA-1 and RUNX1 are also essential for polyploidization, as both GATA1-deficient and RUNX1-null megakaryocytes undergo fewer rounds of endomitosis. Interestingly, GATA-1 deficient megakaryocytes are also smaller than their wild-type counterparts. However, the link between transcription factors and the growth and polyploidization of megakaryocytes has not been established. In our studies to identify key downstream targets of GATA-1 in the megakaryocyte lineage, we discovered that the cell cycle regulators cyclin D1 and p16 were aberrantly expressed in the absence of GATA-1: cyclin D1 expression was reduced nearly 10-fold, while that of p16ink4a was increased 10-fold. Luciferase reporter assays revealed that GATA-1, but not the leukemic isoform GATA-1s, promotes cyclinD1 expression. Consistent with these observations, megakaryocytes that express GATA-1s in place of full-length GATA-1 are smaller than their wild-type counterparts. Chromatin immunoprecipitation studies revealed that GATA-1 is bound to the cyclin D1 promoter in vivo, in primary fetal liver derived megakaryocytes. In contrast, GATA-1 is not associated with the cyclin D1 promoter in erythroid cells, which do not become polyploid. Thus, cyclin D1 is a bona fide GATA-1 target gene in megakaryocytes. To investigate whether restoration of cyclin D1 expression could rescue the polyploidization defect in GATA-1 deficient cells, we infected fetal liver progenitors isolated from GATA-1 knock-down mice with retroviruses harboring the cyclin D1 cDNA (and GFP via an IRES element) or GFP alone. Surprisingly, expression of cyclin D1 did not increase the extent of polyploidization of the GATA-1 deficient megakaryocytes. However, co-overexpression of cyclin D1 and Cdk4 resulted in a dramatic increase in polyploidization. Consistent with the model that cyclinD:Cdk4/6 also regulates cellular metabolism, we observed that the size of the doubly infected cells was also significantly increased. Finally, in support of our model that cyclin D:Cdk4/6 kinase activity is essential for endomitosis, we discovered that introduction of wild-type p16 TAT fusion protein, but not a mutant that fails to interact with Cdk4/6, significantly blocked polyploidization of primary fetal liver derived megakaryocytes. Taken together, our data reveal that the process of endomitosis and cell growth relies heavily on cyclinD:Cdk4/6 kinase activity and that the maturation defects in GATA-1 deficient megakaryocytes are due, in part, to reduced Cyclin D1 and increase p16 expression.


2001 ◽  
Vol 15 (8) ◽  
pp. 1370-1380 ◽  
Author(s):  
Simon Skjøde Jensen ◽  
Mogens Winkel Madsen ◽  
Jiri Lukas ◽  
Lise Binderup ◽  
Jiri Bartek

Abstract The nuclear hormone 1α,25-dihydroxyvitamin D3 induces cell cycle arrest, differentiation, or apoptosis depending on target cell type and state. Although the antiproliferative effect of 1α,25-dihydroxyvitamin D3 has been known for years, the molecular basis of the cell cycle blockade by 1α,25-dihydroxyvitamin D3 remains largely unknown. Here we have investigated the mechanisms underlying the G1 arrest induced upon 1α,25-dihydroxyvitamin D3 treatment of the human breast cancer cell line MCF-7. Twenty-four-hour exposure of exponentially growing MCF-7 cells to 1α,25-dihydroxyvitamin D3 impeded proliferation by preventing S phase entry, an effect that correlated with appearance of the growth-suppressing, hypophosphorylated form of the retinoblastoma protein (pRb), and modulation of cyclin-dependent kinase (cdk) activities of cdk-4, -6, and -2. Time course immunochemical and biochemical analyses of the cellular and molecular effects of 1α,25-dihydroxyvitamin D3 treatment for up to 6 d revealed a dynamic chain of events, preventing activation of cyclin D1/cdk4, and loss of cyclin D3, which collectively lead to repression of the E2F transcription factors and thus negatively affected cyclin A protein expression. While the observed 10-fold inhibition of cyclin D1/cdk 4-associated kinase activity appeared independent of cdk inhibitors, the activity of cdk 2 decreased about 20-fold, reflecting joint effects of the lower abundance of its cyclin partners and a significant increase of the cdk inhibitor p21CIP1/WAF1, which blocked the remaining cyclin A(E)/cdk 2 complexes. Together with a rapid down-modulation of the c-Myc oncoprotein in response to 1α,25-dihydroxyvitamin D3, these results demonstrate that 1α,25-dihydroxyvitamin D3 inhibits cell proliferation by targeting several key regulators governing the G1/S transition.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e20047-e20047 ◽  
Author(s):  
Robert W Georgantas ◽  
Katie Streicher ◽  
Xiaobing Luo ◽  
Wei Zhu ◽  
Zheng Liu ◽  
...  

e20047 Background: MiR-206 has been implicated in a large number of cancers. However, its role in tumor biology is unknown and its biological function has yet to be fully characterized. To examine the role of miR-206 in cancer, we examined the expression of miR-206 in melanoma and identified potential target transcriptss that could be important for the progression of this disease. Methods: Using quantitative RT-PCR we compared expression of 364 microRNAs in melanoma skin biopsies skin from normal donors, melanoma cell lines, and normal melanocytes. The effects of miR-206 on cell growth, apoptosis, and cellular migration/invasion were determined using in vitro assays comparing melanoma cell lines to normal melanocytes. Putative mRNA targets of miR-206 were bioinfomatically identified, and empirically tested by luciferase-3’UTR reporter assays. The effect of miR-206 on the cell cycle of melanoma cells was assayed by flow cytometry. Results: Expression profiling of microRNAs in melanoma lesional skin biopsies compared to normal donor skin biopsies revealed numerous differentially regulated miRs. One such microRNA, miR-206, was significantly highly down-regulated in melanoma biopsies (-75.4-fold, p=1.7x10-4) compared to normal skin and normal melanocytes. Functional analysis showed that miR-206 substantially reduced growth and migration/invasion of multiple melanoma cell lines. Bioinformatic analysis identified the cell cycle genes CDK2, CDK4, Cyclin C, and Cyclin D1 as strong candidate mRNA targets of miR-206. Luciferase reporter gene assays revealed that miR-206 inhibits translation of CDK4, Cyclin D1, and Cyclin C. Consistent with this inhibition of CDK4 and Cyclin D1, miR-206 transfection induced robust G1 arrest in multiple melanoma cell lines. Conclusions: MiR-206 expression was decreased in melanoma tissue and cell lines compared to normal skin and melanocytes, respectively. Inhibition of Cyclin C, Cyclin D1 and CDK4 by miR-206 highlights its role in regulating cell cycle progression, a key aspect of melanoma progression. These observations support miR-206 as a potential tumor suppressor in melanoma, and possibly other cancers.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3771-3771
Author(s):  
Mark G. Frattini ◽  
David Shum ◽  
Kristen M O'Dwyer ◽  
Renier J. Brentjens ◽  
Peter Maslak ◽  
...  

Abstract Abstract 3771 Poster Board III-707 High throughput screening of compounds comprising the Memorial Sloan Kettering chemical library resulted in several confirmed hits against the recombinant Cdc7:Dbf4 heterodimeric kinase, a key regulator in the initiation of DNA replication and the G1 to S phase transition. Chemoinformatic analysis of the hits revealed an enrichment in one chemical cluster made up of several naturally occurring compounds, of which the most potent compound, CKI-7, was selected for further investigation. First, CKI-7 was found to be a non competitive inhibitor for ATP and prompted us to prolife it against a panel of 200 known kinases in order to assess its selectivity profile. The results were as predicted and very few kinases were specifically affected. Second, CKI-7 cytotoxic activity was assessed against a panel of well established cancer cell lines representing both hematopoietic and solid tumor malignancies as well as against a panel of primary hematopoietic cells derived from leukemia patients (both chemotherapy naïve and relapsed/refractory samples) and was found to be a very effective agent with potencies in the low nanomolar range. Subsequent studies using an isogenic pair of cell lines with one over expressing the Bcl_xL anti-apoptotic protein further confirmed the induction of the intrinsic apoptotic pathway via caspase-3 activation in the absence and attenuation of the activity in the presence of Bcl_xL. This was further demonstrated through standard cell cycle synchronization studies revealing that exposure to the Cdc7 inhibitor results in an S phase arrest, cell cycle dependent caspase-3 activation, and apoptotic cell death. This cell death is the direct result of Cdc7 kinase inhibition by CKI-7 as demonstrated using a Cdc7 substrate biomarker assay. Third, the physicochemical properties of this class of naturally occurring compounds also prompted us to investigate their effect on several multidrug resistence (MDR) over-expressing cell lines. We found that CKI-7 was not a substrate for the efflux pumps demonstrating that this novel compound can overcome a major mechanism of chemotherapy resistence in human tumor cells. Based of the above observations, in vivo dose-dependent anti-tumor activity of CKI-7 was subsequently demonstrated in a SCID-Beige mouse systemic tumor model utilizing a recently isolated Philadelphia chromosome positive acute lymphoblastic leukemia cell line (PhALL3.1). Taken together, our data confirm that Cdc7 is a new promising target for cancer therapy, and that the newly discovered inhibitor CKI-7, a naturally occurring selective small molecule inhibitor of this enzyme, is an equally promising novel cancer therapeutic agent. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1624-1624 ◽  
Author(s):  
Chandrima Sinha ◽  
John Bowling ◽  
Aman Seth ◽  
Bensheng Ju ◽  
Bhaskar Kahali ◽  
...  

Abstract The marine environment has been shown to be a rich source of pharmacologically-active secondary metabolites. Three marine- compounds have FDA approval for cancer indications. Aaptamine is a sponge-derived alkaloid that exhibits multiple pharmacological activities including proapoptotic/antiproliferative effects on leukemia cell lines. The effect of the aaptamine class has not been previously studied for high risk leukemias with mixed-lineage leukemia (MLL) gene rearrangements. Using the CellTiter-Glo cell viability assay we evaluated the cytotoxic effect of aaptamine against a panel of leukemia cell lines. We observed that cell lines containing t(4;11) are the most sensitive to aaptamine. Translocation (4;11) is associated with mixed-lineage leukemia and responsible for a very aggressive and refractory pediatric leukemia. Specifically, infants less than one year with t(4;11) have poor survival rates (≈ 19%) and new therapies are urgently needed. Interestingly other MLL cell lines that contain t(9;11) are comparatively less susceptible to aaptamine-mediated cytotoxicity. Jurkat cells overexpressing MLL-AF4 fusion protein are also more sensitive to aaptamine-induced cytotoxicity than wild type or MLL-AF9 overexpressing Jurkat cells indicating the specificity of aaptamine for t(4;11). To further confirm the specificity we conducted a flow based apoptosis assay and observed that aaptamine induces significant apoptosis and necrosis in RS4;11 and MV4;11 cell lines starting at 10µM but not in the t(9;11) containing THP1 cell line. We also found that aaptamine treatment induced G0/G1 arrest specifically in t(4;11) containing cell lines but not in THP1. Additionally we observed that aaptamine did not induce any resistance to the sensitive cell lines after 27 days of chronic exposure. Importantly the compound was well tolerated by healthy activated PBMCs and mice at high concentrations. In order to decipher the mechanism of specificity, we conducted a global proteomic study with treated and untreated RS4;11 and THP1 cell lines. Our proteomic data revealed a significant upregulation of p21 and p27 in aaptamine treated RS4;11 cells but not in THP1. In agreement with the proteomic data, we observed a dose-dependent upregulation of p21 and p27 in both protein and mRNA levels in RS4;11 and MV4;11 cells but not in resistant THP1 cells. Using p21 and p27 promoter-driven luciferase reporter constructs, we observed a significant upregulation of luminescence signal in the RS4;11 cell line at much lower concentration of aaptamine (1µM) whereas the THP1 cell line required 50µM of aaptamine for significant increase in luminescence signal. Cyclin-dependent kinase regulates the G1/S cell cycle transition by phosphorylating retinoblastoma protein (RB). Upregulation of cyclin-dependent kinase inhibitors, such as p27 and p21, promote RB hypophosphorylation and induce G0/G1 arrest. To confirm that this molecular mechanism is responsible for aaptamine induced G0/G1 arrest, we investigated the effect of aaptamine on Rb phosphorylation. We observed a dose dependent downregulation of Rb phosphorylation by aaptamine in sensitive cell lines and predicted it as a major cause of cell cycle arrest. Previous studies have shown that translocation (4;11) is associated with p27 upregulation; thus we hypothesize by further upregulating p27, aaptamine may induce G0/G1 arrest specifically in t(4;11) containing cell lines. To validate the efficacy of aaptamine in vivo, we xenografted 10 NSG mice with 1 million luciferase expressing RS4;11 cells. Four days after leukemia induction we treated half of the mice with subcutaneous injection of aaptamine (100mg/kg, daily) and the other half received vehicle treatment. Bioluminescence imaging (BLI) data revealed a significantly lower disease (p< 0.03) burden in the aaptamine treated group compared to vehicle treated group after 2 weeks. These findings are being confirmed in patient samples. Additional aaptamine analogs are being designed and will be evaluated for improved therapeutic efficacy. Together our in vitro and in vivo findings suggest that by inducing p21 and p27 aaptamine can induce cell cycle arrest and eventually apoptosis specifically in leukemia cells that contain t(4;11) with relatively low toxicity . Therefore the aaptamine class of drug may provide additional therapeutic options for t(4;11) containing high-risk MLL leukemia patients. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (24) ◽  
pp. 6176
Author(s):  
Andrew J. Robles ◽  
Wentao Dai ◽  
Saikat Haldar ◽  
Hongyan Ma ◽  
Victoria M. Anderson ◽  
...  

A screening program designed to identify natural products with selective cytotoxic effects against cell lines representing different types of pediatric solid tumors led to the identification of altertoxin II as a highly potent and selective cytotoxin against Ewing sarcoma cell lines. Altertoxin II, but not the related compounds altertoxin I and alteichin, was highly effective against every Ewing sarcoma cell line tested, with an average 25-fold selectivity for these cells as compared to cells representing other pediatric and adult cancers. Mechanism of action studies revealed that altertoxin II causes DNA double-strand breaks, a rapid DNA damage response, and cell cycle accumulation in the S phase. Our studies also demonstrate that the potent effects of altertoxin II are partially dependent on the progression through the cell cycle, because the G1 arrest initiated by a CDK4/6 inhibitor decreased antiproliferative potency more than 10 times. Importantly, the cell-type-selective DNA-damaging effects of altertoxin II in Ewing sarcoma cells occur independently of its ability to bind directly to DNA. Ultimately, we found that altertoxin II has a dose-dependent in vivo antitumor efficacy against a Ewing sarcoma xenograft, suggesting that it has potential as a therapeutic drug lead and will be useful to identify novel targets for Ewing-sarcoma-specific therapies.


2020 ◽  
Author(s):  
HAIBIAO XIE ◽  
Kaifang Ma ◽  
Kenan Zhang ◽  
Jingcheng Zhou ◽  
Lei Li ◽  
...  

Abstract Background P53 is a classic tumor suppressor, but its role in kidney cancer remains unclear. In our study, we tried to explain the role of p53 in kidney cancer through p53 enhancer RNA-related pathway. Methods qPCR and luciferase reporters were used to detect the expression of p53-related enhancer RNA. Nutlin3 and artificial "microRNA" were used to induce and inhibit the expression of p53 enhancer RNA, respectively. Cycle analysis and β-galactosidase assay were used to explore whether P53-bound enhancer regions 2(p53BER2) plays a role in the cell cycle and senescence response of p53-wild type (WT) renal cancer cells. The function of p53BER2 was further analyzed in vivo by nude mice. RNA sequencing was used to identify the potential target of p53BER2. Results The results showed that P53BER2 expression was down-regulated in renal cancer tissues and cell lines and could specifically express in p53-WT renal cancer cell lines. Knockdown p53BER2 could reverse nutlin-3-induced cytotoxic effect in p53-WT cell lines. Further, downregulation of p53BER2 could reverse nutlin-3-induced G1 arrest and senescence in p53-WT cell lines. What is more, knockdown of p53BER2 showed a resistance to nutlin-3 treatment in Vivo. Additionally, we found BRCA2 could be regulated by p53BER2 in vitro and vivo, which suggested BRCA2 might mediate the function of p53BER2 in RCC. Conclusions The p53-associated enhancer RNA-p53BER2 mediates the cell cycle and senescence of p53 in p53-WT renal cancer cells. This further provides a novel approach and insight for the RCC and p53 research in renal cancer.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2574-2574 ◽  
Author(s):  
Ruben Niesvizky ◽  
Scott Ely ◽  
Maurizio DiLiberto ◽  
Hearn J. Cho ◽  
Udi Y. Gelbshtein ◽  
...  

Abstract Background: Depsipeptide is a cyclic compound with histone deacetylase inhibitory activity as well as antiproliferative and apoptotic effects. Dysregulation of the cell cycle and apoptosis control have been implicated as critical events behind the genesis of MM. Previously, we demonstrated that depsipeptide induces apoptosis in vitro in MM cell lines; those containing t (11q13-cyclin D1) were most sensitive. In vivo studies of primary MM cells treated with depsipeptide show evidence of cell cycle modulation. Here, we report the preliminary results of a Phase II trial which evaluates the safety and efficacy of depsipeptide in MM patients (pts) with documented progressive disease. Methods: To date, 12 pts with relapsed or refractory MM Stage IIIa with progressive disease documented after 4 (mean 3) lines of therapy (Rx) were treated. Mean age 63 years (range 54–74). Baseline mean levels: WBC 4.6 K/uL (range 3.2–5.9); platelets 193 K/uL (63–272); hemoglobin 11g/dL (8–14); albumin 3.8 g/dL (range 2.7–4.6); creatinine 1.0 mg/dL (range 0.6–1.5); LDH 197 u/L (range 156–245); calcium 9.1 mg/dL (range 8.2–10.2). Pretreatment FISH in 9 pts identified 2 with del 13q14 including one with t(11;14), one patient with tetrasomy 11, one with trisomy 11 and 6 normals. Conventional cytogenetics identified one with inversion 9. Depsipeptide was administered IV at 13 mg/m2 as a 4-hr infusion on days 1, 8 and 15 of a 28 day cycle. Cardiac monitoring included pre and post Rx EKG as well as serum troponin-I levels. Results: 11 of 12 pts had stable disease (SD) measured by SPEP or free light chain measurement after receiving one to 6 cycles of depsipeptide; 2 of these pts continue with SD with ongoing Rx (including one patient in cycle 7), 5 discontinued the trial with SD, and 4 progressed after completion of 1 to 2 cycles. Preliminary results indicate that the drug has been well tolerated apart from thrombocytopenia (grade 3) reported in 2 pts necessitating dose reductions; one patient had ST-segment depression (grade 1) noted by EKG. Toxicities commonly associated with depsipeptide administration including fatigue and nausea (grade 2) were reported in 4 and 5 pts, respectively. Conclusions: Depsipeptide treatment was generally well tolerated at the dose and regimen used. Results suggest that the drug is active in pts with advanced MM producing stabilization of disease in those who had progressive relapsed and refractory disease after numerous cycles of prior chemotherapy. Clinical beneffit was noted with improvement of hypercalcemia and pain in 2 pts. Correlative IHC (CD138/Ki-67, BCL-2, MCL-1, CD56, cleaved caspase 3, nuclear cyclin D1 and D3, nuclear p18, p21, p27) and gene array studies are being conducted in these pts and will be reported. Future studies will evaluate depsipeptide at other dosing schedules and combinations with other agents such as bortezomib. Patient accrual continues by the NY Phase II Trial Consortium. Supported by The LLS SCOR grant, RFP S03-058 SAIC-Frederick, NCI K24 CA100287-02 and NCI K23 CA109260-01


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2790-2790 ◽  
Author(s):  
Elodie Lainey ◽  
Marie Sebert ◽  
Cyrielle Bouteloup ◽  
Carole Leroy ◽  
Sylvain Thepot ◽  
...  

Abstract Abstract 2790 Background: TKIs were initially developed as targeted therapies that would solely interfere with a “specific” aberrant signaling pathway in malignant cells. However, we and others showed that the EGFR TKI erlotinib (Erlo) has in vivo and in vitro efficacy in MDS and AML (Boehrer et al., Blood, 2008). We also previously observed, in a preliminary study, the potentiation of apoptosis upon combination of Erlo with azacitidine (Aza), but not with decitabine, in HL-60 cells and in cells from a few (five) MDS/AML patients (ASH 2010, 974). We decided to expand this pre-clinical study to define the potential interest of combining different TKI (Dasatinib (Dasa), Sorafenib (Sora) or Erlo) with Aza, now a reference first line treatment in higher risk MDS (Lancet Oncol, 2009). Methods: Erlo (10μM), Dasa (500nM), or Sora (5μM) were combined to Aza (1μM) and apoptosis over-time (24, 48 and 72h) quantified by FACS analysis following DioC3(6)/PI staining in different MDS/AML-derived cell lines (MOLM-13, SKM-1, MV4-11, Kasumi-1). Quantification of apoptosis at 48h or 72h was recapitulated ex vivo in CD34+ cells from patients with MDS (n=12), AML (n=14) or AML post MDS (n=5). For each single drug, as well as the respective combinationsErlo, Aza and Erlo+Aza, the capacity to induce cell cycle arrest (PI staining), cytotoxicity (MTT assay) and decrease of proliferation (Click-it EdU Assay) was assessed concomitantly. Differentiation was assessed by staining with CD11b on day 3 (FACS), evaluation of ROS production at 24/48h by FACS staining with CM-DFCDA (oxidative stress indicator) and HE (Hydroethidine). Functional relevance of apoptosis-related signaling pathways was determined by co-incubation of Aza (and drugs combinations with biochemical inhibitors against MAPK: JNK (SP600125, 10μM), p-38MAPK (SB203580, 10μM) and MEK (U0126, 5μM). Immunoblot analyses of caspase-3, PARP, Mcl-1 and Bcl-xl were performed at 24h and 48h. Results: Whereas co-incubation of Dasa or Sora with Aza did not increase the degree of apoptosis observed with Aza alone, combination of Erlo with Aza had synergistic effects already observed at 24h, an effect that increased over-time (SKM-1 72h (mean PI+ cells, n=3), Erlo: 15%, Aza: 40%, Erlo+Aza: 81% - MOLM-13 72h, Erlo: 21%, Aza: 25%, Erlo+Aza: 64%). The % of cells with intact metabolic activity at 72h (determined by the MTT assay using 2.5μM Erlo and 0.5μM Aza) decreased from 83% (Erlo) and 79% (Aza) to 23% (Erlo+Aza) in SKM-1 (similar results for MOLM-13 cells). To determine if Erlo also impacts on apoptosis in CD34+ cells from patients with MDS or AML, we screened 31 samples and observed a synergistic effect in 5/31 samples (2/12 MDS, 2/14 LAM, 1/5 AML post MDS) and an additive effect in 8/31 samples. Induction of apoptosis was not preceded by differentiation (no increase in CD11b) or production of ROS. On the other hand, analysis of cell cycle distribution at 24h showed that apoptosis was accompanied by an increase in G0/G1 arrest and a decrease in the % of cells in S phase upon incubation with the combination of Erlo+Aza (G0/G1 (mean, n=2) in SKM-1, DMSO: 51%, Erlo: 52%, Aza: 61%, Erlo+Aza: 78%, G0/G1 in MOLM-13, DMSO: 40%, Erlo: 51%, Aza: 38%, Erlo+Aza: 65%) confirmed by the study of newly synthesized DNA before induction of apoptosis (15h) with an alternative BrdU test (Click-EdU assay). Indeed, in SKM-1 cells, proliferation was decreased by 20% upon Erlo or Aza and by 80% with the association. Moreover, cell cycle arrest was associated with an increase in the % of apoptotic cells with a sub-diploid DNA content (about 10% for Erlo or Aza and 32% for Erlo+Aza in SKM-1. Immunoblot analyses confirmed an increase in cleaved PARP and caspase-3 upon incubation with Aza +Erlo as compared to the single agents, as well as a decrease of the anti-apoptotic protein Mcl-1 and Bcl-xl. Incubation of Aza with SP600125 induced the same extent of apoptosis observed with Erlo+Aza, whereas no effect was observed with the p-38MAPK inhibitor SB203580 or the MEK inhibitor U0126. In addition, Erlo decreased phosphorylation of JNK on Thr183/Tyr185, suggesting a potential implication of the JNK pathway in the mechanism of apoptosis. Conclusions: In this study, Erlo was the only TKI tested able to increase sensitivity towards Aza in MDS/AML cell lines and in patient-derived cells. These results suggest a potential clinical interest of combining Aza to Erlo in MDS/AML. Disclosures: Fenaux: Celgene: Honoraria, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document