The Effect of Combination of CDK Inhibitor (Roscovitine) and Derivatives of Tetracycline on Human Leukemic Cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4562-4562
Author(s):  
Hairong Song ◽  
Zuzana Hassan

Abstract Roscovitine is a 2,6,9- trisubstituted aminopurine analogue that compete with ATP for binding to the active site of Cyclin-dependent kinases (CDKs). It inhibits CDK2/cyclinE, CDK7/cyclinH and CDK9/cyclinT. The cytotoxic effect of roscovitine and its analogues has been reported in several cancer cell lines in vitro and in animal models of cancer xenografts in vivo. The analogues of tetracycline (Doxycycline and Minocycline) possess anti-tumor, anti-metastatic and anti-angiogenesis properties with low toxicity besides acting as antibiotics. The effect of single agent is usually limited in cancer therapy and the use of single agent may mediate drug resistance. We have studied a cytotoxic effect of the CDK inhibitor roscovitine and tetracycline derivatives either alone or in combinations in human leukemia cells lines HL-60 (myeloid), Jurkat (lymphoblastic) and K562 (CML). HL-60, Jurkat and K562 cells were cultured in RPMI 1640 supplemented with 10% FBS. Cells were treated with roscovitine (0.5 – 100 μM), doxycyline (0.5 – 100 μg/ml) or minocycline (0.5 – 100 μg/ml) up to 48 hours alone or in combinations. The cells were examined for viability using AlarmaBlue assay, proliferation using 3H-thymidine incorporation assay, apoptosis using morphological criteria in Giemsa staining, cell cycle using propidium iodide and flow cytometry. Mitochondrial membrane permeabilization was assessed using tetramethylrhodamine ethyl ester. Specific proteins were detected by Western blotting. Incubation with roscovitine, doxycycline or minocycline decreased the cell viability in concentration-dependent manner in all three cell lines. Pretreatment of cells with doxycycline and minocycline for 24 hours followed by roscovitine for 24 hours have shown the additional decrease in cell viability. No such effect was observed when the drugs were used concomitantly. Roscovitine alone decreased proliferation in concentration- and time-dependent way in all three cell lines. Apoptotic morphology was firstly observed at 3 hrs after the treatment with roscovitine and markedly increased at 6 hrs in HL60 and Jurkat cells, but not in K562 cells. In HL60 and Jurkat cells, the cell cycle analysis has shown an increase in sub-G1 cells at 6 hours with maximum at 24h without preceding cell cycle arrest. In K562 cells sub-G1 peak increased subsequently to G2/M arrest. A marked loss of ΔΨm was observed at 2 hrs of exposure to 25uM of roscovitine in Jurkat and HL-60 cell lines, respectively. In HL60 and Jurkat cells, release of cytochrome c, AIF and Smac from mitochondria into cytosol were detected along with activation of caspases and PARP. However in K562, no caspase activation was detected at studied time points. Thus, all three agents have a cytotoxic effect in different types of leukemic cells, however, the additional cytotoxic effect of combination of Roscovitine and derivatives of tetracycline might be a potent therapeutic approach in treatment of leukemia.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2478-2478
Author(s):  
Andrea Ghelli Luserna Di Rora ◽  
Ilaria Iacobucci ◽  
Enrica Imbrogno ◽  
Enrico Derenzini ◽  
Anna Ferrari ◽  
...  

Abstract Nowadays the effectiveness of the treatments for adult Acute Lymphoblastic Leukemia (ALL) patients is still inadequate and frequently many patients after years of response to treatments develop relapses. Thus there is a need to find novel targets for specific therapies and to maximize the effect of the actual treatments. Recently different Checkpoint Kinase (Chk)1/Chk2 inhibitors has been assessed for the treatment of different type of cancers but only few studies have been performed on hematological diseases. We evaluated the effectiveness of the Chk1 inhibitor, LY2606368, as single agent and in combination with tyrosine kinase inhibitors (imatinib and dasatinib) or with the purine nucleoside antimetabolite clofarabine in B-/T- acute lymphoblastic leukemia (ALL) cell lines and in primary blasts. Human B (BV-173, SUPB-15, NALM-6, NALM-19 and REH) and T (MOLT-4, RPMI-8402 and CEM) ALL cell lines were incubated with increasing concentrations of drug (1-100 nM) for 24 and 48 hours and the reduction of the cell viability was evaluated using WST-1 reagent. LY2606368 deeply reduced the cell viability in a dose and time dependent manner in all the cell lines, with the BV-173 (6.33 nM IC50 24hrs) and RPMI-8402 (8.07 nM IC50 24hrs) being the most sensitive while SUP-B15 (61.4 nM IC50 24hrs) and REH (96.7 nM IC50 24hrs) being the less sensitive cell lines. Moreover the sensitivity to the compound was no correlated with the different sub-type of ALL or with the mutational status of p53, which is a marker of the functionality of the G1/S checkpoint. The cytotoxic activity was confirmed by the significant increment of apoptosis cells (Annexin V/Propidium Iodide), by the increment of gH2AX foci and by the activation of different apoptotic markers (Parp-1 and pro-Caspase3 cleavage). To understand the relationship between the activation of apoptosis and the effect on cell cycle and to identify hypothetical mechanisms of death, different cell cycle analyses were performed (Propidium Iodide staining). The inhibition of Chk1, deeply changed the cell cycle profile. Indeed in all the cell lines the percentage of cells in S phase and in G2/M phase were reduced by the treatment while the numbers of cells in sub-G1 and G1 phase were increased. The hypothetical function of LY2606368 as a chemosensitizer agent was evaluated combining the compound with different drugs normally used in clinical trials. For each drugs the combination strongly reduced the cell viability when compared to the cytotoxic effect of the single drugs. Moreover the combination showed an additive efficacy in term of induction of DNA damages as showed by the increase number of gH2AX foci and the activation of pChk1 (ser 317). The results found on the cell lines were confirmed also using primary leukemic blast isolated from adult Philadelphia-positive ALL patients. Indeed LY2606368 as single agent or in combination with the Tki, imatinib, was able to deeply reduce the cell viability and to induce DNA damages (gH2AX foci). In conclusion LY2606368 showed a strong cytotoxic activity on B-/T-All cell lines and primary blasts as single agent and in combination with other drugs. In our opinion this data are the basis for a future clinical evaluation of this compound in the treatment of leukemia. Supported by ELN, AIL, AIRC, progetto Regione-Università 2010-12 (L. Bolondi), FP7 NGS-PTL project. Disclosures Soverini: Novartis, Briston-Myers Squibb, ARIAD: Consultancy. Cavo:JANSSEN, CELGENE, AMGEN: Consultancy. Martinelli:ROCHE: Consultancy; Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau; Pfizer: Consultancy; Ariad: Consultancy; AMGEN: Consultancy; MSD: Consultancy.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5201-5201
Author(s):  
Shaowei Qiu ◽  
Jing Yu ◽  
Tengteng Yu ◽  
Haiyan Xing ◽  
Na An ◽  
...  

Abstract Introduction: As the important suprressor of P53, iASPP was found to be overexpressed in leukemia, and functioned as oncogene that inhibited apoptosis of leukemia cells. Sertad1 is identified as one of the proteins that can bind with iASPP in our previous study by two-hybrid screen. Sertad1 is highly expressed in carcinomas from pancreatic, lung and ovarian tissues, which considered Sertad1 as an oncoprotein. In this study, our findings revealed that Sertad1 could interact with iASPP in the cytoplasm near nuclear membrane, which could block iASPP to enter into nucleus to interact with P53, and inhibited the function of iASPP eventually. Methods: Co-immunoprecipitation and fluorescence confocal microscopic imaging were used to confirm the interaction between iASPP and Sertad1, the exact binding domains and the subcellular colocalization.The plasmids of iASPP and Sertad1 were transfected alone or co-transfected into K562 cells, the stable subclones that highly expressed iASPP, Sertad1 or both of them were then established by limiting dilution and named as K562-iASPPhi, K562-Sertad1hi, and K562-Douhi, respectively. The cell proliferation, cell cycle and apoptosis of above subclones were investigated by flow cytometry. Further, silence of the above two proteins was performed to confirm their functions. Immunoblotting analysis and immunofluorescence were performed to explore the possible mechanisms of difference between the biological functions of the above subclones. Results: Sertad1 expression level varied in leukemic cell lines and AML patients irrespectively of iASPP and P53. Interaction between iASPP and Sertad1 did exist in 293 cell and leukemic cells, both iASPP and Sertad1 scattered in the cytoplasm and nucleus, and their colocalizations were mainly in the cytoplasm, which encircled the nucleus. iASPP binds directly to Sertad1 through its PHD-bromo domain, C-terminal domain and Cyclin-A domain in a reduced order, and Serta domain failed to bind to iASPP. Overexpression of iASPP in K562 cells (iASPPhi) could result in the increased cell proliferation, cell cycle arrest in G2/M phase and resistance to apoptosis induced by chemotherapy drugs. While overexpression of iASPP and Sertad1 at the same time (Douhi) could slow down the cell proliferation, lead the cells more vulnerable to the chemotherapy drugs. As figure showed, in K562-Douhi cells, both iASPP and Sertad1 were obviously located in the cytoplasm, which encircled the nuclei, the subcellular colocalization was nearly outside the nuclei. The immunoblotting analysis further supported the conclusions. The resistance of iASPP to chemotherapeutic drug was accompanied by Puma protein expression in a p53-independent manner. By knocking down the expersssion of iASPP and Sertad separately, we found that iASPP is dispensable for maintenance of anti-apoptotic function and Sertad1 is indispensable for cell cycle in leukemic cells. Conclusions: In normal situation, the protein iASPP and Sertad1 scatter in the nucleus and cytoplasm, mainly in the cytoplasm. As convinced by our study, iASPP was overexpressed in the leukemia cell lines and primary AML patients, it could function as oncogene through its binding with P53 protein in the nucleus, inhibit the function of P53. When iASPPhi cells were exposed to apoptosis stimuli, Puma protein could play an important role in this process, irrespective of the expression level of P53. But when iASPP and Sertad1 were both overexpressed in the leukemic cells, Sertad1 could tether iASPP outside the nucleus mainly through its PHD-bromo domain, prevent it from inhibiting P53 function, suppress the leukemic cell growth and stimulate cell apoptosis by rescuing the P53 eventually. Our data provided a new insight to overcome iASPP protein, namely through its binding partners, when the similar proteins or drugs that can tether iASPP outside the nucleus such as Sertad1 are transfected into the leukemic cells, it may restore p53 function to eliminate the leukemic cells. Figure 1 Figure 1. Disclosures Wang: Novartis: Consultancy; Bristol Myers Squibb: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3210-3210
Author(s):  
Maria Todaro ◽  
Valentina Griggio ◽  
Candida Vitale ◽  
Chiara Salvetti ◽  
Chiara Riganti ◽  
...  

Abstract BACKGROUND: Despite the therapeutic efficacy of new target drugs in chronic lymphocytic leukemia (CLL), treatment of high-risk patients remains an unmet clinical need. Disease aggressiveness can be ascribed to intrinsic features of the tumor cells such as immunoglobulin heavy chain (IGHV) mutational status and TP53 disruption, which are strong determinant of drug response. Many of the tumor suppressor and growth regulatory proteins with a known pathogenic role in CLL (i.e. p53, NF-kB, Akt, IkB) bind the nuclear export protein XPO1 (Chromosome Region Maintenance 1; CRM1) and are carried through the nuclear pore complex into the cell cytoplasm. Elevated protein levels of XPO1 and specific XPO1 mutations have been reported in various hematologic and solid tumors. In particular, XPO1 is overexpressed and recurrently mutated in CLL cells (Puente XS et al, Nature 2011; Lapalombella R et al, Blood 2012). Selinexor (KPT-330) an oral inhibitor of XPO1, is active as single agent in different hematologic malignancies including acute myeloid leukemia, non-Hodgkin lymphomas, CLL and multiple myeloma. The combination of selinexor and ibrutinib elicits a synergistic cytotoxic effect in primary CLL cells and increases overall survival of a CLL mouse model compared with ibrutinib alone (Hing ZA et al, Blood 2015). AIM: The aim of this study is to evaluate the additive or synergistic in vitro cytotoxic effects of selinexor, used in combination with chemotherapeutic drugs or the new PI3k inhibitor idelalisib against primary CLL cells. Specifically, this study aims at identifying combination regimens that might overcome single agent resistance. METHODS: 15 patients with CLL were included in the study, among these 9 with mutated (M) and 3 patients with unmutated (UM) IGHV; for 3 patients the mutational status was not available at the moment of data analyses. Purified CLL cells were exposed, alone or in presence of the murine stromal cell line M2-10B4, to selinexor (10 nM, 100 nM, 1 uM and 10 uM) in combination with fludarabine (F-ara-A, 10 nM, 100 nM, 1 uM and 10 uM), bendamustine (Ben, 3 mM, 10 mM, 30 mM and 50 mM) or idelalisib (Ide, 10 nM, 100 nM, 1 uM and 10 uM) for 24, 72 and 120 hours. Cell viability was analysed by Annexin-V/propidium Iodide (AnnV/PI) immunostaining and flow cytometry. Samples were considered resistant when the relative viability of F-ara-A treated CLL cells compared to untreated control was >0.5. Combination analysis was performed using Calcusyn software; combinations were considered synergistic when CI was <1. RESULTS: Leukemic cells were cultured in the presence of increasing concentrations of selinexor, used alone or in combination with F-ara-A and Ben, and with the PI3Kδ inhibitor, Ide. After 72 hours of culture, the mean percentage of viable AnnV-/PI- CLL cells significantly decreased by 0,62-ratio following KPT-330 (100 nM), 0,42-ratio following F-ara-A (1 uM) and 0,24-ratio after dual treatment, compared to untreated controls. Combination analysis showed that selinexor and F-ara-A strongly synergize in inducing CLL cells apoptosis with a CI < 1. Similarly, we observed a synergistic interaction between selinexor (100 nM) and Ben (30 mM) that significantly enhanced the cytotoxic effect of the individual drugs (0,62-ratio for selinexor, 0,68-ratio for Ben and 0,41-ratio for selinexor + Ben) with a CI <1, at the same time point. The combination between selinexor (100 nM) and Ide (10 nM) at 72 hours resulted in a weaker, although significant, viability reduction (0,62-ratio for KPT-330, 0,5 for Ide and 0,38 for selinexor + Ide). We observed that IGHV UM CLL cells showed higher fold reduction values in cell viability when exposed to synergistic combinations, compared to IGHV M cells. Selinexor (100 nM) was also effective in impairing the viability of CLL cells that showed intrinsic resistance to F-ara-A (n=6, 0,61-ratio for selinexor, 0,74-ratio for F-ara-A and 0,4-ratio for selinexor + F-ara-A). Lastly, we exposed CLL-stromal cells co-coltures to the identified synergistic combinations and found that selinexor + F-ara-A or selinexor + Ide significantly reduced the viability of leukemic cells, effectively counteracting the protective effect exerted by stromal cells toward drug-induced apoptosis. CONCLUSIONS: Our data demonstrate that the combination of selinexor with chemotherapy or Ide has synergistic cytotoxic effects, also counteracting intrinsic or stromal cells-mediated drug resistance. Disclosures Boccadoro: SANOFI: Honoraria, Research Funding; CELGENE: Honoraria, Research Funding; Abbivie: Honoraria; Novartis: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; BMS: Honoraria, Research Funding; Mundipharma: Research Funding. Landesman:Karyopharm Therapeutics Inc: Employment, Other: stockholder. Coscia:ROCHE: Honoraria, Other: Advisory board; Karyopharm: Research Funding; Mundipharma: Honoraria; Janssen: Honoraria; Gilead: Honoraria.


2021 ◽  
Vol 22 (19) ◽  
pp. 10445
Author(s):  
Haseeb A. Khan ◽  
Uday Kishore ◽  
Hamed M. Alsulami ◽  
Salman H. Alrokayan

Nanoparticles are efficient drug delivery vehicles for targeting specific organs as well as systemic therapy for a range of diseases, including cancer. However, their interaction with the immune system offers an intriguing challenge. Due to the unique physico-chemical properties, carbon nanotubes (CNTs) are considered as nanocarriers of considerable interest in cancer diagnosis and therapy. CNTs, as a promising nanomaterial, are capable of both detecting as well as delivering drugs or small therapeutic molecules to tumour cells. In this study, we coupled a recombinant fragment of human surfactant protein D (rfhSP-D) with carboxymethyl-cellulose (CMC) CNTs (CMC-CNT, 10–20 nm diameter) for augmenting their apoptotic and immunotherapeutic properties using two leukemic cell lines. The cell viability of AML14.3D10 or K562 cancer cell lines was reduced when cultured with CMC-mwCNT-coupled-rfhSP-D (CNT + rfhSP-D) at 24 h. Increased levels of caspase 3, 7 and cleaved caspase 9 in CNT + rfhSP-D treated AML14.3D10 and K562 cells suggested an involvement of an intrinsic pathway of apoptosis. CNT + rfhSP-D treated leukemic cells also showed higher mRNA expression of p53 and cell cycle inhibitors (p21 and p27). This suggested a likely reduction in cdc2-cyclin B1, causing G2/M cell cycle arrest and p53-dependent apoptosis in AML14.3D10 cells, while p53-independent mechanisms appeared to be in operation in K562 cells. We suggest that CNT + rfhSP-D has therapeutic potential in targeting leukemic cells, irrespective of their p53 status, and thus, it is worth setting up pre-clinical trials in animal models.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2808-2808
Author(s):  
Andrea Ghelli Luserna di Rorà ◽  
Ilaria Iacobucci ◽  
Neil Beeharry ◽  
Maria Vittoria Falzacappa ◽  
Chiara Ronchini ◽  
...  

Abstract Although impressive developments have been made in the treatment of Acute Lymphoblastic Leukemia (ALL) patients, the overall survival is still very poor. With the exception of novel therapeutic strategies based on monoclonal antibodies (Bi-specific T-cell engagers, BiTEs) or immunogenic cells (CART cells), the therapeutic approaches for adult ALL patients are still base on non-selective chemotherapy or on tyrosine kinase inhibitors (TKIs) for the patients harboring the BCR-ABL1 fusion transcript. In addition a large percentage of initial successfully treated patients frequently develop relapses. Thus there is a need to improve the efficacy of conventional therapies, in particular those related to TKIs and to DNA damaging agents, in order to reduce the off-target toxicity and avoid relapses. In the present study we evaluated the in vitro, ex vivo and in vivo efficacy of MK-1775, a specific Wee1 inhibitor, in single agent and in combination with different therapeutic agents normally used for the treatment of B-/T-ALL. We firstly started by evaluated the efficacy of the compound in single agent on a panel of human B and T ALL cell lines (n=8) and on primary cells isolated from the bone marrow of adult B-ALL patients (n=8). The inhibition of Wee1 deeply reduced the cell viability and the proliferation rate, induced the apoptosis and increased the DNA damages of both leukemic cell lines and primary cells. Further cell-cycle analysis showed that in leukemic cell lines the treatment increased the number of cell in late S and G2/M phase. Light microscopy analyses, looking for nuclei morphology, confirmed that MK-1775 increased the number of mitotic cells but it interfered with normal mitotic division (induction of aberrant mitosis as showed by the increment of DNA bridges and micro-nuclei). The effects of the compound on the cell cycle profile and on the G2/M checkpoint were confirmed also in immunoblotting analyses, by the increment of phospho-HH3(ser10) and of Myt1 (mitotic isoform), and by gene expression analysis looking to specific genes involved in the G2/M checkpoints (PrimePcr DNA damage assay, Biorad). In particular genes like GADD45A and CCNB1/CCNB2 were significantly up-regulated between treated and untreated samples. Finally using a T-ALL mouse model we evaluated the effect of MK-1775 in single agent. Although no significative differences were seen between treated and un-treated samples, due to a very aggressive phenotype of the disease (all animal died after only 18 days from the engraftment), molecular analyses confirmed that the treatment induced DNA damages (increase of H2A.X and p-Chk1 ser317) and inhibited Wee1 functionality (reduction of pCDC2) on leukemic blasts isolated from both spleens and bone marrows. To evaluate if the inhibition of the G2/M checkpoint could sensitize leukemic cells to the toxicity of antineoplastic drugs, Philadelphia-negative ALL cell lines and primary leukemic cells (n=9) where treated with increasing concentration of MK-1775 and increasing concentration of the nucleotide analogue, clofarabine. Statistical analyses (Combination index value) confirmed the synergy of the combination in the reduction of the cell viability, in the inhibition of the proliferation and in the induction of the apoptosis. Similar results were seen on Philadelphia-positive ALL cell lines and primary cells (n=3) combining the MK-1775 with the TKI, bosutinib. The simultaneously inhibition of the Wee1 and the BCR-ABL downstream pathway resulted in a synergic inhibition of the cell viability, reduction of the proliferation and induction of apoptosis. In our opinion the pre-clinical results of this study are the basis for a future clinical evaluation of MK-1775 for the treatment of ALL patients. Acknowledgments: ELN, AIL, AIRC, progetto Regione-Università 2010-12 (L. Bolondi), Fondazione del Monte di Bologna e Ravenna, FP7 NGS-PTL project. Disclosures Martinelli: Novartis: Speakers Bureau; BMS: Speakers Bureau; Amgen: Consultancy, Speakers Bureau; Roche: Consultancy, Speakers Bureau; MSD: Consultancy; Pfizer: Consultancy, Speakers Bureau; Ariad: Consultancy, Speakers Bureau; Genentech: Consultancy; Celgene: Consultancy, Speakers Bureau.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2481-2481 ◽  
Author(s):  
Rakesh Popat ◽  
Lyndsey Goff ◽  
Heather E. Oaekervee ◽  
Jamie D. Cavenagh ◽  
Simon P. Joel

Abstract Background: Proteasome inhibition has been shown to be effective against a variety of tumours. In multiple myeloma the response rates to bortezomib (B) in relapsed patients is 46% (APEX study ≥ MR), but are likely to be up to 75% when combined with melphalan (M). Mechanisms underlying this effect are yet to be fully determined. Aims: To investigate the effect of B, M and the combination of the two on myeloma cell lines and primary patient cells, focusing on the anti-apoptotic molecule Mcl-1 and the pro-apoptotic molecule Bim in the mediation of drug activity. Cell cycle analysis using propidium iodide and flow cytometry was also performed in parallel. Methods: The human multiple myeloma cell lines (HMCLs) RPMI 8226/S, U266 and purified primary patient malignant plasma cells were used for cell culture and viability assays using an ATP bioluminescence method. Cells were incubated for 24 or 48 hours with differing concentrations of B, M and combinations in varying schedules. EC50 values were calculated using a sigmoidal Emax model and the observed cell viability of the combination of the two drugs was compared with the additive effect expected. Mcl-1, Bim, caspase-3, and PARP were probed for by Western Blotting of HMCLs. Results: Both U266 and RPMI 8226/S cells showed dramatic reductions in cell viability to B, with EC50 values of 4.7nM and 5.3nM respectively, and responded to high concentrations of M with EC50 values of 95.6 uM and 91.5 uM respectively after 48 hour incubations. Synergistic responses were seen when M was added 24 hours prior to B, but not with B pre-treatment. This was also observed with primary patient cells. Mcl-1 levels increased after 6 hours of B exposure, likely due to proteasome inhibition, but decreased by 24 hours with associated cleavage. This effect was concentration-dependent with partial cleavage observed at 4nM (approximately EC30) and full cleavage at 50nM. Bim was present in untreated cells, unchanged after 6 hours of B exposure, but decreased at 24 hours at both concentrations. All of these changes were associated with cleavage of caspase-3 and the appearance of cleaved PARP, and persisted out to 48 hours exposure. Six hours following M exposure, there was an increase in Mcl-1 at the sub-toxic 10uM concentration (possibly a cell survival response), but a reduction at 100uM. Following 24/48 hour exposures changes were no different to control cells with 10uM M, however at 100uM (EC50 concentration) cleavage of Mcl-1 and a decrease in Bim were observed, similar to changes seen with 50nM B. On combining the two drugs simultaneously in a 48 hour exposure, B 4nM and M 10uM failed to induce any changes in U266 cells, but resulted in partial cleavage of Mcl-1 in RPMI 8226 cells. When the M concentration was increased to 100uM there was a decrease of both Mcl-1 and Bim and the associated cleavage of caspase-3 and PARP. There were no differences whether B preceded or followed M. Cell cycle analysis demonstrated G2 arrest following B therapy at 24 hours and in combination with M. Conclusions: This work demonstrates that in multiple myeloma, both Mcl-1 and Bim are closely involved in proteasome mediated cellular apoptosis and in M mediated cytotoxicity. In keeping with work in Jurkat cells (Nencioni et al., Blood 2005), Mcl-1 was found to transiently increase following proteasome inhibition, but then decreased at 24 hours as apoptosis occurred. An early rise in anti-apoptotic proteins such as Mcl-1 may explain why synergistic responses with B and M were seen only with M pretreatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4106-4106
Author(s):  
Ming-Yang Lee ◽  
Jing-Jing Chuang ◽  
Yi-Wen Liu

Abstract Abstract 4106 Introduction Indirubin-3-monoxime (IO) is the active ingredient of Danggui Longhui Wan, a mixture of plants that is used in traditional Chinese medicine. It is a potent inhibitor of cyclin-dependent kinases (CDKs), especially CDK2. In clinical studies demonstrated that indirubin did not cause major side effects in patients with chronic myelogenous leukemia. However, the functional action of indirubin on acute lymphoblastic leukemia (ALL) is still unclear. In the present study, we investigated that cytotoxic effect and mechanism study of IO in human acute lymphocytic leukemia cell line JM1 and human chronic myeloid leukemia cell line K562. We also analyzed the viability influence of IO in normal human granulocytes and lymphocytes. Materials & Methods After the treatment of IO, cell viability of JM1 and K562 cells was determined by WST-1 assay. The influence of IO on cell-cycle distribution was analyzed by FACScan. To identify which cell death types were induced during IO treatment we measured the caspase-3 activity for analysis the induction of apoptosis; evaluated the LDH release in the necrotic analysis and the expression level of LC3-II was determined by Western blotting analysis for autophagic cell death. Furthermore, we also analyzed the toxicity of IO in normal human granulocytes and lymphocytes by the treatment dose of IO with value of IC50. Results IO significantly affected the cell viability on JM1 but also on K562 cells in a dose dependent manner. The G2/M phase of cell cycle was arrested and sub-G1 proportion was relative increased. In addition, the finding showed that IO initiated caspase-3-dependent apoptosis in JM1 and K562 cells. The expression of autophagosome-incorporated LC3-II protein was also clearly increased once cells treated with IO. However, the necrotic phenomenon through measuring LDH release from K562 and JM1 cells could not be observed. Excitingly, by the treatment dose of IO with value of IC50, the cell viability of lymphocytes was marginally affected, whereas the cell cytotoxitity of granulocytes was not induced. Conclusions IO has the ability to induce potent cytotoxic effect on K562 and JM1 cells. The cell death after IO treatment is mainly caused by apoptosis and autophagy, not by necrosis. This effect is also associated with interrupted cell cycle. Importantly, IO has few or little cytotoxic effect on human lymphocytes and granulocytes. The results suggest that IO might be useful for clinical anti- ALL treatment. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e16056-e16056
Author(s):  
Roham Salman Roghani ◽  
Ali Sanjari moghaddam ◽  
Gabrielle Rupprecht ◽  
Erdem Altunel ◽  
So Young Kim ◽  
...  

e16056 Background: Colorectal cancer (CRC) is the 3rdmost common form of cancer in the US, responsible for over 50,000 death each year. Therapeutic options for advanced colorectal cancer are limited, and there remains an unmet clinical need to identify new therapies to treat this deadly disease. To address this need, we have developed a precision medicine pipeline that integrates high throughput chemical screens with matched patient-derived cell lines and patient-derived xenografts (PDXs) to identify new treatments for CRC. Methods: We used high-throughput chemical screens of 2,100 compounds across five low-passage, patient-derived CRC cell lines. These results were validated using dose-response IC50curves for CDK1, CDK2, CDK9 or CDK1/2/9 inhibitors and by siRNA-mediated knockdown of CDK9 with or without CDK2 inhibition. Cell cycle arrest analysis was performed by flow cytometry and anaphase catastrophe was analyzed by immunofluorescence staining. For in vivo studies, matched PDXs were treated with either CDK2, CDK9 or dual CDK2/9 inhibitors. Results: We identified the CDK inhibitor drug class as among the most effective cytotoxic compounds across all five CRC lines. Further analysis of the CDK inhibitor class revealed that combined targeting of CDK1, 2, and 9 was the most effective, with IC50 in the range of 110 nM to 1.2 μM. We further validated the efficacy of combined CDK2/9 inhibition using siRNA-mediated knockdown of CDK9 in the presence of a CDK2 inhibitor(CVT-313), and showed that CDK9 knockdown acted synergistically with CDK2 inhibition. Dual CDK2/9 inhibition led to significant G2/M cell cycle arrest and anaphase catastrophe. Finally, combined CDK2/9 inhibition in vivo synergistically inhibited PDX tumor growth as compared to single-agent CDK inhibitors. Conclusions: Our precision medicine pipeline revealed CDK2/9 dual inhibition as a combinatorial therapy to treat CRC and can also be used to identify new and novel therapies


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.


2021 ◽  
Vol 14 (7) ◽  
pp. 682
Author(s):  
Jianling Bi ◽  
Garima Dixit ◽  
Yuping Zhang ◽  
Eric J. Devor ◽  
Haley A. Losh ◽  
...  

Angiogenesis plays a crucial role in tumor development and metastasis. Both bevacizumab and cediranib have demonstrated activity as single anti-angiogenic agents in endometrial cancer, though subsequent studies of bevacizumab combined with chemotherapy failed to improve outcomes compared to chemotherapy alone. Our objective was to compare the efficacy of cediranib and bevacizumab in endometrial cancer models. The cellular effects of bevacizumab and cediranib were examined in endometrial cancer cell lines using extracellular signal-related kinase (ERK) phosphorylation, ligand shedding, cell viability, and cell cycle progression as readouts. Cellular viability was also tested in eight patient-derived organoid models of endometrial cancer. Finally, we performed a phosphoproteomic array of 875 phosphoproteins to define the signaling changes related to bevacizumab versus cediranib. Cediranib but not bevacizumab blocked ligand-mediated ERK activation in endometrial cancer cells. In both cell lines and patient-derived organoids, neither bevacizumab nor cediranib alone had a notable effect on cell viability. Cediranib but not bevacizumab promoted marked cell death when combined with chemotherapy. Cell cycle analysis demonstrated an accumulation in mitosis after treatment with cediranib + chemotherapy, consistent with the abrogation of the G2/M checkpoint and subsequent mitotic catastrophe. Molecular analysis of key controllers of the G2/M cell cycle checkpoint confirmed its abrogation. Phosphoproteomic analysis revealed that bevacizumab and cediranib had both similar and unique effects on cell signaling that underlie their shared versus individual actions as anti-angiogenic agents. An anti-angiogenic tyrosine kinase inhibitor such as cediranib has the potential to be superior to bevacizumab in combination with chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document