Antitumor effect of the combination of proteasome inhibitor (Pi) and histone deacytelase inhibitor (HDACi) in breast cancer (BC).

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e23156-e23156
Author(s):  
Joseph Kannarkatt ◽  
Omar Abed Alkharabsheh ◽  
Burra V Madhukar ◽  
Cheryl Leece ◽  
Deimante Tamkus

e23156 Background: Previous studies demonstrated the efficacy of HDACi and Pi as single agents in BC. The synergistic combination of HDACi and Pi has been shown to be effective in clinical studies in other tumor types, including multiple myeloma, non-small cell lung cancer, and pancreatic cancer. In this in vitro study, we explored the hypothesis that combination of these two agents may improve overall antitumor effect in BC. Methods: In the experiment, we used 3 BC cell lines: MCF7 (ER+), MDA-MB-231 (ER-), and BCSV (isolated from metastatic ER+ tumor which lost ER expression with disease progression). The cells were treated with carfilzomib (Pi) and scriptaid (HDACi) as single agents and in combination. Cell viability was determined using MTT assay, and combination indexes were determined using CalcuSyn software. Additionally, cells were cultured in 3D tumor spheroid model, and response to treatment was assessed on day 5, 10, 15 and 20. Results: In comparison with control, single agent HDACi, single agent Pi and combination HDACi/Pi increased cell viability by 6%, decreased cell viability by 60% and by 69% respectively (p < 0.0001) in MCF7 cells; and decreased cell viability by 25%, 59% and 66% respectively (p = 0.0061) in BCSV cells. The combination indexes for most HDACi and Pi combinations were lower than 1.0, indicating synergistic effects in MCF-7 and BCSV cells but not in MDA-MB-231 cells. In 3D cultures, the combination of carfilzomib 1µM and scriptaid 10µM led to significant reduction of tumor spheroid size as compared to single agent. By day 20, BCSV tumor spheroid size increased by 258% in control group and by 50% in Pi, decreased by 17% in HDACi and by 30% in HDACi/Pi combination group (p < 0.0001). Conclusions: Combination of HDACi and Pi exhibited synergistic activity in both MCF7 and BCSV monolayer cultures. Additionally, combination HDACi and Pi significantly decreased 3D tumor spheroid size as compared to either single agent alone. Further study is needed to evaluate the mechanisms of Pi/HDACi combination in estrogen-mediated BC tumorigenesis.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4991-4991
Author(s):  
T. Elter ◽  
J. Kilp ◽  
T. Piironen ◽  
P. Borchmann ◽  
H. Schulz ◽  
...  

Abstract Combination chemoimmunotherapy regimens have shown substantial efficacy in the treatment of lymphoproliferative disorders, particularly in comparison to the efficacy of single-agent therapies. Fludarabine has become an established treatment regimen in CLL, and although overall response rates (ORR) in previously untreated patients range between 60% to 80%, patients who are refractory to fludarabine have poor outcomes. Alemtuzumab, the anti-CD52 monoclonal antibody, is the most effective single-agent therapy in CLL, and is capable of inducing minimal residual disease (MRD)-negative responses even among patients with fludarabine-refractory disease. Our previous clinical experience with the combination of alemtuzumab and fludarabine (FluCam) resulted in 83% ORR in 36 patients with relapsed/refractory CLL, with 30% achieving a complete response (CR; Elter et al J Clin Oncol2005;23:7024–7031). In addition, among 12 patients with fludarabine-refractory disease, 8 achieved responses (4 CRs), and median time-to-progression (TTP) for all patients was 13 months. In order to optimize the dose and schedule of the FluCam combination, we performed pharmacokinetic (PK) analysis of the previously reported 6-cycle regimen. PK data were collected for a 14-patient cohort that participated in the phase 2 FluCam trial. Median patient age was 60 years (range, 49–73), 9 patients had Binet C disease (5 were Binet B), and patients received a mean 2.5 prior therapies. Alemtuzumab 30 mg (after initial dose escalation) and fludarabine 30 mg/m2 were administered on days 1–3 of a 28-day cycle for up to 6 cycles. PK parameters were measured from samples collected before each subsequent cycle, and at days 1, 4, 7, 14, 21, 28, and 42, for a total of 158 patient samples, of which 120 were tested. Plasma concentration of alemtuzumab increased steadily from day 1 to day 4 of therapy to a median Cmax 1.55 mg/mL, but decreased to a median 0.145 mg/mL by 7 days after initiation of treatment. By day 21 of therapy, alemtuzumab plasma concentration decreased to undetectable levels. Because efficacy of alemtuzumab has been shown to correlate with serum levels of this antibody, significant improvement in progression free survival (PFS) may require a elevated plasma levels of alemtuzumab for the duration of the treatment cycle. Therefore, the significant responses seen in this trial can be attributed to documented synergistic activity between alemtuzumab and fludarabine, which has been demonstrated both in vitro and in vivo. Despite low CD4 counts through the duration of therapy, favorable safety results seen in the trial could be attributed to opportunity for hematologic recovery between treatment cycles. Detailed PK analysis is currently being completed and will be presented at the conference. Conclusions: Treatment with the FluCam immunotherapy combination yielded positive results among patients with fludarabine resistant/refractory CLL, a difficult-to-treat population. As shown previously, response rates correlate with higher alemtuzumab plasma concentrations. Therefore, longer PFS durations may require longer, more sustained alemtuzumab plasma levels, which may be achieved with either consolidation or maintenance.


PEDIATRICS ◽  
1993 ◽  
Vol 92 (1) ◽  
pp. 86-89
Author(s):  
Suat Caglayan ◽  
Halil Candemir ◽  
Sadik Aksit ◽  
Savas Kansoy ◽  
Sezin Asik ◽  
...  

Objective. To determine the value of oral agar in the treatment of neonatal hyperbilirubinemia and to compare it with two other treatment modalities: phototherapy alone and phototherapy plus oral agar. Methods. Two hundred eight jaundiced full-term newborns were divided into four groups. They were given either phototherapy alone, phototherapy plus oral agar, oral agar alone, or no treatment (control group). The changes in the serum bilirubin values were determined and the results were compared statistically, mainly using analysis of variance. Results. In all three therapy groups, the time required to reduce the bilirubin level to either 15 mg/dL or to 10 mg/dL was significantly shorter than that required by the control group. Although oral agar was found to be as effective as phototherapy, the most significant decrease in bilirubin level was in the combination group. Conclusions. The efficacy of phototherapy in decreasing the serum bilirubin level in neonatal hyperbilirubinemia can be augmented with the use of oral agar. Oral agar can also be used as a single agent for the treatment of neonatal hyperbilirubinemia, since it is as effective as phototherapy.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1384-1384 ◽  
Author(s):  
Russell R. Hoover ◽  
Matthew W. Harding

Abstract MK-0457 (VX-680) is a reversible small molecule kinase inhibitor that targets Aurora A, B, and C with Ki values of 0.7, 18, and 4.6 nM, respectively. MK-0457 also inhibits Flt3 (Ki = 30 nM), and both the wild type and the T315I mutant of BCR-ABL (Ki = 30 and 40 nM, respectively). Clinical trials are ongoing in patients with solid tumors and hematological malignancies. Recent data show that MK-0457 is active in patients against subtypes of AML, BCR-ABL T315I mutant CML, and Philadelphia positive (Ph+) ALL. To support multi-agent clinical trials, the activity of MK-0457 in combination with idarubicin, Ara-C, and BCR-ABL inhibitors was investigated. The viability of a panel of AML, ALL, and CML cell lines was assessed following single agent and either simultaneous or sequential combinations of agents. Combination effects were evaluated using the Bliss Independence Model. MK-0457 as a single agent markedly inhibited leukemia cell viability (at 72 hrs) with an IC50 range of 20–300 nM for MV4-11, Molt-4, Molm-13, K562, LAMA-84, MEG-01, and KU812F cells. Additionally, MK-0457 inhibited the viability of BaF3 cells transformed by wild type, T315I, or Y253F mutants of BCR-ABL with similar IC50s (approximately 300 nM). The sequential combination of MK-0457 followed by either idarubicin or Ara-C showed greater synergy than simultaneous combinations in a cell line dependent manner. MK-0457 displayed strong synergy in simultaneous combination with Gleevec (imatinib mesylate) in a panel of human CML-derived cell lines and BaF3 cells expressing wild type BCR-ABL. MK-0457 enhanced the Gleevec-mediated cell death of K562 leukemia cells as evidenced by increased caspase activity, PARP cleavage, and induction of the sub-G1 population. At concentrations where synergy was observed by cell viability analysis, the MK-0457/Gleevec combination resulted predominantly in aneuploidy and G2/M arrest, consistent with inhibition of Aurora kinases by MK-0457. These results support the clinical evaluation of MK-0457 combined with idarubicin and Ara-C in AML and with BCR-ABL inhibitors in CML and Ph+ ALL.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1514-1514
Author(s):  
Enrique M. Ocio ◽  
David Vilanova ◽  
Laura San-Segundo ◽  
Patricia Maiso ◽  
Mercedes Garayoa ◽  
...  

Abstract Introduction Panobinostat (LBH589) is a novel histone deacetylase (HDAC) inhibitor being evaluated in clinical trials in hematological and solid malignancies. In multiple myeloma (MM), investigators have demonstrated its in vitro antimyeloma effect in cell lines and patients cells. Cancer treatment is typically based on the concept of combining agents with different mechanisms of action to overcome drug resistance. This was the rationale of the present study in which the in vitro and in vivo benefit of combinations of pabinostat with conventional antimyeloma agents has been explored. Material and Methods The potential in vitro synergism of pabinostat with 6 antimyeloma agents (melphalan, doxorubicin, dexamethasone, thalidomide, lenalidomide, bortezomib) was analyzed in MM1S cell line. The two most favorable combinations were tested in 120 NOD/SCID mice implanted with a human subcutaneous plasmocytoma. Mice were randomized into 12 treatment groups. Drugs were given ip, 5 days/week × 7 weeks. Doses were: pabinostat: 10 mg/Kg × 3 weeks and 5 mg/Kg afterwards; dexamethasone (D): 1 mg/Kg; bortezomib (B): 0.1 mg/Kg; and lenalidomide (L): 15 mg/Kg. Tumor volumes clinical features and weight were monitored three times a week. Mice were sacrificed when their tumors reached 2 cm. Immunohistochemistry was performed in selected tumors. Results Three agents potentiated the effect of pabinostat in vitro: bortezomib, dexamethasone and, to a lesser extent, lenalidomide. Moreover, the triple combination of pabinostat+L+D and pabinostat+B+D resulted in high synergistic activity. These studies provided the rationale for testing these combinations in vivo: Single agent pabinostat at a dose of 10 mg/Kg completely abrogated the growth of plasmocytomas without significant toxicity. In fact, after three weeks of treatment, the median volume of tumors in the pabinostat group was 163±75 mm3 as compared to 1891±1182 mm3 in the control group (p=0.001). Immunohistochemistry of pabinostat treated tumors revealed a decrease in BrdU uptake, an increase in histone acetylation and phosphorylation of H2AX suggesting DNA damage. This antiproliferative action was associated with survival advantage: median survival 70±1.8 vs 30±2.1 days (p&lt;0.001) for the pabinostat and vehicle treated groups respectively. Subsequently the dose of pabinostat was decreased by 50% in order to gain further insights into the potential advantage of the combinations. Interestingly, the addition of D and suboptimal doses of either B or L significantly improved the antimyeloma effect of pabinostat. In this sense, median survival increased up to 86±2.6 days in pabinostat+D+B (p&lt;0.001) and 88±1.2 days for pabinostat+D+L (p&lt;0.001). The efficacy of these triple combinations was significantly higher than any of the respective double combinations (pabinostat+D; pabinostat+B; pabinostat+L; B+D; L+D). Some of these combinations (including or not pabinostat) initially induced a slight toxicity (5%–15% body weight loss) which spontaneously recovered after the third week of treatment. Conclusion Combinations of pabinostat + dexamethasone with either bortezomib or lenalidomide are safe and display promising antimyeloma efficacy. This study provides the rationale for the clinical development of triple combinations of these drugs to improve the outcome of MM patients.


2011 ◽  
Vol 365 ◽  
pp. 222-227
Author(s):  
Li Tang ◽  
Li Yu

Recent studies have shown that Genistein can obviously suppress growth of gynecologic carcinoma. In this study we examined whether Genistein and Cisplatin, alone and in combination, exhibited a inhibitory effect on the growth of ovarian carcinoma. SKOV3 cells were treated with 20µM Genistein, 20µM Cisplatin and combination group (G+C) for 24 to 72 hours, antiproliferative effect was tested by MTT method. Apoptosis was evaluated using flow cytometry and transmission electron microscopy. Then the transplanted xenografts were treated with Genistein and Cisplatin, the tumor volume and tumor weight were measured, HE staining were performed for morphologic observation. A time and dose-dependent growth inhibition was observed. When treated the cells with 20µM Genistein, 20µMCisplatin and combination group (G+C), compared with the control group, the growth of cells in different treatment group was inhibited, while the combination of Genistein and Cisplatin showed significant inhibition effect than single agent. When Genistein (0.4mg/kg, s.c.) treated the transplanted xenograft in vivo, the tumor volume and tumor weight decreased, T/C ratio (mean volume of treated group/mean volume of control group) also was reduced compared to untreated group, and extensive necrotic areas in the tumor treated with Genistein and Cisplatin appeared in HE section. A weight loss of nude mice after treatment with Genistein was not significant as compared with the control group. And when treated with Genistein (0.4mg/kg, s.c.) and Cisplatin (4mg/kg, i.v.), it showed a synergistic effect. Genistein and Cisplatin, alone and in combination, suppress cell growth. The combination of Genistein and Cisplatin, as compared to single treated group, caused a synergistic increase in antiproliferative effect on SKOV3 cells. The implication for treatment of ovarian cancer may be combination of Genistein and Cisplatin. Further studies are needed to supply more evidence.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3832-3832 ◽  
Author(s):  
Jeffrey A Steinberg ◽  
Jing Shen ◽  
Eric Sanchez ◽  
Haiming Chen ◽  
Zhi-Wei Li ◽  
...  

Abstract Abstract 3832 Poster Board III-768 Introduction ALA is an antioxidant often used in the management of peripheral neuropathy (PN) for patients with multiple myeloma (MM). A clinical trial evaluating ALA in diabetic neuropathy showed this drug to be effective for patients with both somatic and autonomic neuropathies. It also normalized the endoneural blood flow, reduced oxidative stress and improved vascular dysfunction. Bortezomib (Velcade®), the first-in-class proteasome inhibitor (PI), which is approved for the treatment of patients with MM, may cause PN. As a result, patients are often treated empirically with ALA. In this study, we investigated whether ALA has any impact on the anti-MM effects of bortezomib. Methods First, cells from the MM cell lines RPMI8226 and MM1S (1×105 cells per 100μl) were treated with ALA alone to determine whether ALA had any effects on their growth as determined with an MTS assay. MM cells were plated in a 96-well plate using serum-free media. The cells were treated with either media alone or ALA at concentrations ranging from 1 to 1000 μM for 48 hours. The acidity of ALA at these doses was determined and if the pH was less than 7, we neutralized it using NaOH. Second, we measured the proliferation of cells exposed to bortezomib alone and combinations of a fixed concentration of bortezomib and escalating concentrations of ALA. Results The exposure of cells to ALA alone had a stimulatory effect on the growth of both MM cell lines in vitro. ALA alone at 1000 μM resulted in an increase in cell viability of MM1S cells by approximately 10% when compared to the control group. ALA alone also stimulated the growth of RPMI8226 cells but at much lower concentrations than observed for MM1S. Compared to untreated cells, there was an increase in cell viability with ALA at concentrations as low as 1 μM and a concentration dependent increase at concentrations of 1, 10, 100, and 1000 μM in RPMI8226 cells. At the highest concentration (1000 μM) of ALA, cell viability increased 150% when compared to RPMI8226 cells incubated with media alone. Next, we evaluated the effect of ALA on bortezomib's anti-MM activity. As a single agent, bortezomib reduced MM1S (20 nM) and RPMI8226 (5 nM) cell viability by 93% and 70% respectively. When ALA was added at a clinically achievable concentration (100 μM) to bortezomib (RPMI8226, 5 nM; MM1S, 20 nM), a reduction in the anti-MM effects of bortezomib on these cell lines was observed when compared to bortezomib treatment alone. Compared to bortezomib alone, the combination of ALA plus bortezomib doubled cell viability (increased RPMI8226 and MM1S cell viability from 32.5% to 65% and 7.5% to 15%, respectively). These negative effects of ALA on bortezomib's anti-MM activity were consistently observed in multiple experiments involving both of these cell lines evaluating concentrations of ALA ranging from 100 to 1000 μM and bortezomib ranging from 5 to 20 nM. Conclusions Our data suggest that ALA has the potential to antagonize the anti-MM effects of bortezomib based on our in vitro results using MM cell lines. Thus, it is possible that ALA could negatively impact the therapeutic benefit of bortezomib for MM patients and this requires further study especially if ALA is accepted as an intervention in bortezomib-related neuropathy. We are currently completing studies evaluating primary MM patients' tumor cells in vitro and our human MM xenograft models in vivo to further validate this impact of ALA on bortezomib's anti-MM activity and whether changes in treatment schedule of these drugs may prevent this inhibitory effect from occurring. In addition, because part of bortezomib's anti-tumor effects are related to reactive oxygen species (ROS) levels, we are evaluating whether the inhibitory effects of ALA on this PI may be overcome by increasing intracellular ROS levels. Disclosures: Hilger: Millennium Pharmaceutcals: Employment. Berenson:Millennium Pharmaceuticals, Inc.: Consultancy, Honoraria, Research Funding, Speakers Bureau.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2226-2226 ◽  
Author(s):  
Lianne E Rotin ◽  
Marcela Gronda ◽  
Neil Maclean ◽  
Feng-Hsu Lin ◽  
Jeff Wrana ◽  
...  

Abstract Ibrutinib is a small-molecule Bruton’s tyrosine kinase (BTK) inhibitor yielding impressive clinical responses in B-cell malignancies, where BTK contributes to disease development and progression. We noted that BTK is expressed and constitutively active in acute myeloid leukemia (AML) cell lines and in a subset of AML patients. We therefore sought to determine BTK’s potential role in AML. Using BTK-pY223 expression on immunoblot as a marker of BTK activity, we demonstrated that ibrutinib doses as low as 1μM were sufficient to inhibit BTK activity in the AML cells TEX and OCI-AML2. Yet, these cell lines were insensitive to ibrutinib compared to the B-cell lymphoma Daudi cell line (IC50 10.4μM and 23.7μM versus 1.1μM, respectively). Although inactive as a single agent, we sought to identify drug combinations that sensitized AML cells to pharmacologically relevant concentrations of ibrutinib by conducting a combination chemical screen with our in-house known drug library in TEX and OCI-AML2 cells. According to excess-over-Bliss additivism criteria, we determined that in both cell lines, the poly(ADP-ribose) glycohydrolase inhibitor ethacridine lactate was the most synergistically cytotoxic, producing an 8-fold reduction in the IC50 of ibrutinib. Synergistic cytotoxicity was also observed in a subset of primary AML cells, but not normal hematopoietic cells. Mechanistically, the combination’s synergistic cytotoxicity resulted from excessive ROS production (>10-fold increase, versus a <2-fold increase with either drug alone in TEX and OCI-AML2), since α-tocopherol addition to combination-treated cells rescued cell viability from 24% to 85% and from 4% to 84% in TEX and OCI-AML2, respectively. Combining ibrutinib with other ROS-inducing agents such as parthenolide and the first-line AML therapy daunorubicin produced similar synergistic effects in AML cells,with α-tocopherol also rescuing cell viability. However, the synergistic effects were likely not related to inhibition of BTK, as knockdown of BTK with shRNA did not sensitize TEX cells to ethacridine or daunorubicin and thus did not recapitulate the effects of ibrutinib. We therefore conclude that the BTK inhibitor ibrutinib lacks single agent anti-AML activity, but synergizes with ROS-inducing agents including daunorubicin by inhibiting targets beyond BTK. Thus, clinical trials of ibrutinib in combination with standard chemotherapy could be warranted in AML. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 67 (7) ◽  
pp. 1067-1075 ◽  
Author(s):  
Lei Huang ◽  
Zhaoyun Liu ◽  
Huijuan Jiang ◽  
Lijuan Li ◽  
Rong Fu

Most of the International Prognostic Scoring System (IPSS) high-risk patients with myelodysplastic syndrome partly responded to hypomethylating therapy even with transient remission, while arsenic trioxide (ATO) had partial effect in patients with MDS. Therefore, we sought to investigate the effects and possible mechanisms of the combination of ATO and decitabine (DAC) in MDS cells. In our study, the MUTZ-1 and SKM-1 cells were treated with ATO, DAC or both. Cell viability, cell apoptosis, levels of reactive oxygen species (ROS) and expressions of the endoplasmicreticulum (ER) stress-associated genes and proteins were examined. Results showed the combination of ATO and DAC synergistically inhibited the proliferation and induced apoptosis of MDS cells. Through the RNA-sequence and GSEA gene function analysis, ER stress-related pathway played an important role in apoptosis of MDS cells induced by the combination of ATO and DAC. ER stress-related genes DNA damage inducible transcript 3, GRP78, and activating transcription factor-6 were significantly highly expressed in combination group than those in single agent groups; proteins were confirmed by western blot. The levels of ROS significantly increased in the combination group. Furthermore, the apoptosis of (ATO+DAC) group MDS cells could be partially reversed by antioxidant agent N-acetylcysteine, accompanied by decreased expression of intracellular ROS and ER stress-related genes. These results suggested that the combination of ATO and DAC synergistically induced the apoptosis of MDS cells by increased ROS-related ER stress in MDS cells.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 22-23
Author(s):  
Sophia Adamia ◽  
Shruti Bhatt ◽  
Yu-Tzu Tai ◽  
Kenneth Wen ◽  
Catherine A Nicholas ◽  
...  

Whole-genome sequencing analysis of newly diagnosed and relapsed multiple myeloma (MM) samples identified recurrent mutations in genes involved in the MAPK pathway, highlighting the potential of RAS/RAF/MEK/ERK signaling as a therapeutic target. Genomic studies identified translocations that involve IGH and set of partner genes MMSET, FGFR3, and CCND1 as primary events in MM. CDK4/CDK6 is overexpressed in MM, and CDK6 overexpression correlates with poor OS, suggesting that CDK4/6 are promising targets for MM therapy. Recent studies demonstrated synergistic activity of combined novel ERK1/2i inhibitor LY3214996 and CDK4/6i LY2835219 in solid tumors, but analogous studies have not been done in MM. Here we used preclinical models of MM to investigate inhibiting Erk1/2, CDK4/6, or both using ERK1/2i, CDK4/6i, or combination therapy. MM cell lines, RAS mutated or wild type (WT), were sensitive to ERK1/2i at IC50&lt;0.5uM, and CDK4/6i at IC50&lt;3uM. Synergistic effects of the Erk1/2i and CDK4/6i were noted in both RAS mutated and WT MM cell lines when ERK1/2i combined with CDK4/6i. Combination of ERK1/2i+CDK4/6i resulted in dose-dependent G0/G1 arrest in RAS mutated and WT MM cells. Similar effects were seen in RAS mutated cells treated with ERK1/2i or CDK4/6i as a single agent. ERK1/2i + CDK4/6i treatment triggered modest early apoptosis in RAS mutated MM cells, while in RAS WT MM cells this effect was more evident. Using dynamic BH3 profiling assay, we found that short-term treatment of MM cell with ERK1/2i and CDK4/6i led to increased overall mitochondrial priming in response to promiscuous BIM peptide in all MM cell lines. Even single agent treatment with ERK1/2i and CDK4/6i was able to enhance priming of RAS mutated or WT cells. Thus, ERK1/2i and CDK4/6i may activate mitochondrial apoptotic signaling in MM cells alone or in combination, consistent with observed synergistic cytotoxicity. HD PBMC and ARH77 cells were tested as controls. These cells were resistant to ERK1/2i and CDK4/6i at a broad range of concentrations, suggesting a favorable therapeutic index. The clinical potential of CDK4/6i+ERK1/2i was supported by an in vivo study demonstrating a significant (P=0.0004) decrease of the MM burden in CDK4/6i+ERK1/2i treated mice, without adverse effects. Proliferation and apoptosis studies of PCs from MM patient BM samples in the presence and absence of autologous BMSC/BMSCI-CM suggest potent and strong synergistic effects of ERK1/2i+CDK4/6i in MM and may allow successful use in clinic. To address the underlying mechanism of the synergism between Erk1/2i and CDK4/6i, we evaluated their cellular and transcriptional activity in MM cells. Gene expression profiling showed significant downregulation of RAS and CDK4/6 signaling pathway genes in MM cells as a result of ERK1/2i and CDK4/6i treatment at specific concentration ratios (3:1/1:3). Further evaluation of functional effects of ERK1/2i and CDK4/6i, alone or in combination, demonstrated that the synergistic effect of these inhibitors in MM cells is achieved through inhibition of p-S6, downregulation of c-myc, and correlate with ERK1/2i+CDK4/6i induced cell arrest in the G1 cell cycle phase. We noted increased ERK1/2 phosphorylation, which generally results in compensatory activation of parallel signaling pathways or in the loss of negative feedback. Regardless, ERK1/2i+CDK4/6i retained the inhibitory activity of the downstream signaling network, as demonstrated by the inhibition of cytoplasmic (p-RSK1) and nuclear (c-myc) targets of ERK at protein and mRNA levels. Treatment with ERK1/2i+CDK4/6i significantly decreased the levels of p-Rb and E2F1, downstream targets of CDK4/6. Recent studies shown that, in addition to cell cycle regulation, CDK4 and CDK6 induce tumorigenesis through regulation of inflammatory cytokines that are induced via NFκB pathway activation. CDK4/6i functional effects on MM cells cannot be limited to cell cycle arrest, CDK4/6i might also inhibit cytokines, which are produced in MM cells by NFκB activation. Overall, we shown that ERK1/2i+CDK4/6i induced cell proliferation and led to the key target molecule (p-c-myc, p-RSK, p-S6, p-RB, and E2F1) downregulations suggesting on-target activity of these inhibitors in MM cells. Importantly, our studies demonstrate strong synergistic anti-MM activity with ERK1/2+CDK4/6 therapy, providing a preclinical framework for clinical trials to improve patient outcome in MM. Disclosures Letai: Novartis: Research Funding; AbbVie: Consultancy; AstraZeneca: Consultancy; Zentalis: Membership on an entity's Board of Directors or advisory committees; Flash Therapeutics: Membership on an entity's Board of Directors or advisory committees; Dialectic: Membership on an entity's Board of Directors or advisory committees; Chugai: Other: Lecture Fees. Anderson:Sanofi-Aventis: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Millenium-Takeda: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Oncopep and C4 Therapeutics.: Other: Scientific Founder of Oncopep and C4 Therapeutics..


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3020-3020
Author(s):  
Frank C. Passero ◽  
Ravi Dashnamoorthy ◽  
Afshin Beheshti ◽  
J. Tyson McDonald ◽  
Andrew M Evens

Abstract Background: Activation of NF-kB pathways are a hallmark feature of TCL and HL, making proteasome inhibition an attractive therapeutic target. Previous studies have demonstrated prominent in vivo efficacy of ixazomib, an oral proteasome inhibitor for both TCL and HL. Among the common significantly regulated genes identified via systems biology approach include upregulation of genes encoding for ubiquitin proteasome subunits (Ravi et al. Cancer Res. 2016). Further combination studies with HDACi belinostat were synergistic in Jurkat, HH and L428 cell lines, and NRF2 was discovered as mediator of proteasome gene expression. We hypothesized that HDACi abrogates NRF2 mediated proteasome recovery leading to synergistic effects on cell viability in combination with ixazomib. Methods: Global transcriptome analysis was performed on RNA isolated from multiple cell lines include TCL (Jurkat) and HL (L540, L428) treated with ixazomib and control, as well as Jurkat cell lines treated with single agent ixazomib, belinostat and combination. Significant genes were determined by applying a one-way ANOVA with an adjusted Bonferroni correction for a false discovery rate (FDR) < 0.05. Further pathway analysis from significant genes was performed by using a fold change greater than ±1.2 comparing all samples to each other and observing pathway relationships using Ingenuity Pathway Analysis. Gene Set Enrichment Analysis was performed with FDR <0.05 for functional analysis. Proteasome-Glo cell based assay was used to evaluate caspase-, chymotrypsin, and trypsin-like activity. Proteasome activity was measured at 24 and 72 hours after Jurkat cell lines were treated with control, ixazomib or belinostat as single agents, and in combination. SiRNA knockdown experiments were performed in Jurkat cell line with NRF2 and non-targeting (NT) SiRNA transfection. Real-time quantitative PCR (qPCR) for proteasome subunit and NRF2 genes was performed on RNA isolated from treated cells. Results: Transcriptome analysis revealed upregulation of proteasome genes in ixazomib treated cell lines Jurkat, L540 and L428 at 24 hours. In Jurkat TCL, ixazomib caused decreased caspase-like and chymotrypsin-like proteasome activity at 24 hours that was followed by recovery of these activities at 72 hours. The combination of ixazomib and belinostat significantly decreased proteasome activity for chymotrypsin-like, caspase-like and trypsin-like activity at 72 hours compared to single agent ixazomib or belinostat. In Jurkat cells, NRF2 was identified as a transcriptional regulator involved in proteasome gene regulation, showing upregulation of proteasomal genes and NRF2 with ixazomib single agent, downregulation with belinostat single agent and in combination with ixazomib. These results were confirmed with qPCR for NRF2 and proteasome genes in Jurkat and L428. SiRNA knockdown for NRF2 in Jurkat cells resulted in decreased cell viability, NRF2 and proteasome gene expression compared with NT SiRNA following ixazomib treatment. Conclusions: Treatment with single agent ixazomib induced prominent proteasome gene expression in all TCL and HL cell lines. In Jurkat, recovery of chymotrypsin and caspase-like proteasome activity occurred by 72 hours suggesting that transcriptional changes induced by proteasome inhibition contributed to proteasome function recovery. Combination therapy with belinostat resulted in downregulation of proteasome genes in Jurkat and L428 and prevented functional recovery of the proteasome observed in Jurkat. Our results suggest that targeting the proteasome itself with ixazomib and preventing the induced recovery of proteasome genes with belinostat contributes to synergistic effects observed on proteasome function and cell viability in TCL and HL. Further studies with CRISPR/Cas to confirm the effect of NRF2 on proteasome gene and functional recovery in the context of proteasome inhibition are ongoing and will be reported. Disclosures Evens: Takeda: Other: Advisory board.


Sign in / Sign up

Export Citation Format

Share Document