WP-1034, a Novel Jak-Stat Inhibitor, Has Proapoptotic and Antileukemic Activity in Acute Myeloid Leukemia (AML) Cell Lines and AML Patient Samples.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2528-2528
Author(s):  
Stefan Faderl ◽  
Alessandra Ferrajoli ◽  
David Harris ◽  
Quin Van ◽  
Waldemar Priebe ◽  
...  

Abstract Proliferation and growth of AML cells result from stimulation by cytokines and high levels of cytokines are associated with poor prognosis in AML. Cytokines act through cellular receptors that are associated with members of the Jak family of protein tyrosine kinases. Upon phosphorylation and activation of Jak, proteins bound to Jak initiate signaling pathways including those regulated by Stat proteins. Since constitutive activation of Jak-Stat has been associated with leukemogenesis, we hypothesize that inhibition of Jak-Stat inhibits proliferation of AML cells. To do this, we studied the effects of WP-1034, a novel and potent inhibitor of Jak-Stat, in the OCIM2 AML cell line and fresh samples from AML patients. OCIM2 cells were deprived of serum for 2 hours and then incubated with 1 to 5 μM WP-1034 to investigate its effect on OCIM2 cell proliferation. After incubation of the cells without and with 1, 2.5, 5, 7.5, and 10 μM WP-1034 for 1 hour, and at 5 μM for 0, 20 min, 40 min, and 1, 2, 3, and 4 hours, we determined expression of Stat 1, 3, and 5, as well as Phospho-stat 1, 3, and 5 in the cells by Western Immunoblotting. In addition, we analyzed cell cycle status by PI staining and flow cytometry. We further evaluated induction of apoptosis of OCIM2 cells following incubation with WP-1034 at 3, 5, and 6 μM using the annexin V-CY5 assay and analyzed caspase 3 and PARP cleavage using Western Immunoblotting. To demonstrate the effect of WP-1034 on marrow cells from AML patients and healthy volunteers we incubated marrow cells with WP-1034 at increasing concentrations and used the blast colony assay to measure inhibition of proliferation. Our results show that: i) WP-1034 effectively inhibits proliferation of OCIM2 cells and AML blast proliferation from patient samples; ii) WP-1034 blocks activation of Stat 3 and 5 by decreasing the amount of Phospho-stat 3 and 5 in OCIM2 cells; iii) WP-1034 causes cell cycle arrest in sub-G0 phase and is able to induce apoptosis in OCIM2 cells; and iv) WP-1034 induces apoptosis involving cleavage of caspase 3 and PARP. Our data suggest that WP-1034, a potent inhibitor of Jak-Stat, inhibits proliferation of AML cells by inhibition of Stat 3 and 5 and induction of caspase-dependent apoptosis.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2462-2462
Author(s):  
Stefan Faderl ◽  
Alessandra Ferrajoli ◽  
David Harris ◽  
Quin Van ◽  
Hagop M. Kantarjian ◽  
...  

Abstract Cytokines and growth factors stimulate AML cell proliferation by activating various signaling pathways and high levels of cytokines have been associated with poor prognosis. Blocking signal transduction of cytokine-stimulated pathways may therefore inhibit AML growth and proliferation. Atiprimod is a cationic amphiphilic azaspirane. Although their mechanisms of action are not completely understood, azaspiranes were demonstrated among others to downregulate various cytokine receptors such as for interleukin (IL)-1, IL-2, IL-6, interferon-γ, and tumor necrosis factor-α. Owing to this spectrum of activities, we hypothesized that atiprimod inhibits activation of intracellular signaling pathways in AML cells resulting in apoptosis and growth inhibition. We first studied the antiproliferative effect of atiprimod on 5 AML cell lines (K562, HL-60, KG-1, OCIM2, OCI/AML3) using the MTT assay. Cells were incubated for 72 hours without and with increasing concentrations of atiprimod (1, 2, 3, and 4 μM), then harvested and their metabolic activity and viability determined as optical density measurements. To corroborate the results, we also studied the effect of atiprimod on OCIM2 cells using a clonogenic cell line assay. Next we determined expression of Stat3 and Stat5, as well as Phospho-stat3 and phospho-stat5 in the K562 cells by Western Immunoblotting where cells were incubated in the absence and presence of increasing atiprimod concentrations (0.5, 1, 2, 3, 4 μM). We further evaluated induction of apoptosis of OCIM2 and K562 cells following incubation with atiprimod at 1 and 4 μM using the annexin V-FITC assay and finally analyzed caspase 3 and PARP cleavage in K562 cells at atiprimod concentrations of 0.5, 1, 2, 3, and 4 μM using Western Immunoblotting. To demonstrate the effect of atiprimod on marrow cells from AML patients and healthy volunteers we incubated marrow cells with atprimod at increasing concentrations and used the blast colony assay to measure inhibition of proliferation. Our results demonstrate that: 1) atiprimod inhibits proliferation of AML cell lines and AML blast proliferation from patient samples, but not significantly normal hematopoietic progenitors from samples of healthy controls; 2) atiprimod inhibits phosphorylation of Stat 3 and 5; and 3) atiprimod induces apoptosis in OCIM2 and K562 cells by cleavage of caspase 3 and PARP. In summary, our data suggest that atiprimod inhibits phopshorylation of Stat 3 and 5, induces caspase-dependent apoptosis, and blocks AML cell proliferation. Further evaluation of atiprimod in clinical trials of AML and MDS should be considered.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 153-153 ◽  
Author(s):  
Stefan Faderl ◽  
William Bornmann ◽  
David Maxwell ◽  
Ashutosh Pal ◽  
Zheng-Hong Peng ◽  
...  

Abstract Tight control of protein tyrosine kinase (TK) activity is crucial for the regulation and maintenance of vital cellular functions such as proliferation, differentiation, and apoptosis. c-KIT is a TK and transmembrane receptor for stem cell factor (SCF). Binding of SCF to c-KIT results in activation of marrow precursors and other blood cells. Activating mutations of c-KIT associated with amino acid Asp-816 (D816) have been identified in leukemic cells of patients with AML and are thought to play an important pathophysiologic role in leukemogenesis. Identification of activating c-KIT mutations and development of novel compounds targeting these mutations may therefore be of therapeutic benefit in AML. Based on the 3-dimensional structure of c-KIT we have generated a number of compounds with activity against c-KIT mutated cells. Here we present initial results of the activity and mechanism of action of the novel c-KIT inhibitor APCK110 in AML cell lines and primary samples from patients with AML. Using an MTT assay, we first studied the antiproliferative effect of APCK110 in the AML cell lines OCI/AML3 and the SCF-responsive cell line OCIM2. Cells were incubated for 72 hours without or with APCK110 at concentrations of 50, 100, 250, and 500 nM, then harvested and their metabolic activity and viability determined as optical density. Next we determined expression of phospho-AKT and -STAT3 in the mastocytosis cell line HMC1.1 and phospho-c-KIT in the AML cell line OCI/AML3 by Western Immunoblotting. We further analyzed induction of caspase 3 and PARP cleavage in OCI/AML3 cells at APCK110 concentrations of 50, 100, 250, and 500 nM using Western Immunoblotting. To demonstrate the effect of APCK110 on primary AML cells, we incubated diagnostic marrow cells from 3 patients with AML with increasing concentrations of APCK110 and used the blast colony assay to measure inhibition of proliferation. We then compared the antiproliferative effect of APCK110 with that of dasatinib and imatinib in OCI/AML3 cells. We show that 1) APCK110 strongly inhibits proliferation of AML cells with 80% inhibition at 500 nM; 2) similar to cell lines, APCK110 also inhibits AML colony growth of primary samples in a dose-dependent manner of up to 80% at 500 nM concentration; 3) APCK110 blocks activation of phospho-AKT, phospho-STAT3, and phospho-c-KIT; 4) APCK110 induces apoptosis by cleavage of caspase 3 and PARP; and 5) APCK110 demonstrates more potent inhibition (up to 100% at 500 nM) of AML cell proliferation than dasatinib (60% at 500 nM) and dasatinib (none at 500 nM). In summary, APCK110 is a novel and potent inhibitor of mutated c-KIT that inhibits AML cell proliferation, blocks activation of intracellular signaling molecules, and induces caspase-dependent apoptosis. Further development of APCK110 for clinical trials of patients with AML should be pursued.


2020 ◽  
Vol 16 (3) ◽  
pp. 340-349
Author(s):  
Ebrahim S. Moghadam ◽  
Farhad Saravani ◽  
Ernest Hamel ◽  
Zahra Shahsavari ◽  
Mohsen Alipour ◽  
...  

Objective: Several anti-tubulin agents were introduced for the cancer treatment so far. Despite successes in the treatment of cancer, these agents cause toxic side effects, including peripheral neuropathy. Comparing anti-tubulin agents, indibulin seemed to cause minimal peripheral neuropathy, but its poor aqueous solubility and other potential clinical problems have led to its remaining in a preclinical stage. Methods: Herein, indibulin analogues were synthesized and evaluated for their in vitro anti-cancer activity using MTT assay (on the MCF-7, T47-D, MDA-MB231 and NIH-3T3 cell lines), annexin V/PI staining assay, cell cycle analysis, anti-tubulin assay and caspase 3/7 activation assay. Results: One of the compounds, 4a, showed good anti-proliferative activity against MCF-7 cells (IC50: 7.5 μM) and low toxicity on a normal cell line (IC50 > 100 μM). All of the tested compounds showed lower cytotoxicity on normal cell line in comparison to reference compound, indibulin. In the annexin V/PI staining assay, induction of apoptosis in the MCF-7 cell line was observed. Cell cycle analysis illustrated an increasing proportion of cells in the sub-G-1 phase, consistent with an increasing proportion of apoptotic cells. No increase in G2/M cells was observed, consistent with the absence of anti-tubulin activity. A caspase 3/7 assay protocol showed that apoptosis induction by more potent compounds was due to activation of caspase 3. Conclusion: Newly synthesized compounds exerted acceptable anticancer activity and further investigation of current scaffold would be beneficial.


2019 ◽  
Vol 19 (6) ◽  
pp. 826-837 ◽  
Author(s):  
Pratibha Pandey ◽  
Preeti Bajpai ◽  
Mohammad H. Siddiqui ◽  
Uzma Sayyed ◽  
Rohit Tiwari ◽  
...  

Background:Plant sterols have proven a potent anti-proliferative and apoptosis inducing agent against several carcinomas including breast and prostate cancers. Jab1 has been reported to be involved in the progression of numerous carcinomas. However, antiproliferative effects of sterols against Jab1 in gall bladder cancer have not been explored yet.Objective:In the current study, we elucidated the mechanism of action of stigmasterol regarding apoptosis induction mediated via downregulation of Jab1 protein in human gall bladder cancer cells.Methods:In our study, we performed MTT and Trypan blue assay to assess the effect of stigmasterol on cell proliferation. In addition, RT-PCR and western blotting were performed to identify the effect of stigmasterol on Jab1 and p27 expression in human gall bladder cancer cells. We further performed cell cycle, Caspase-3, Hoechst and FITC-Annexin V analysis, to confirm the apoptosis induction in stigmasterol treated human gall bladder cancer cells.Results:Our results clearly indicated that stigmasterol has up-regulated the p27 expression and down-regulated Jab1 gene. These modulations of genes might occur via mitochondrial apoptosis signaling pathway. Caspase-3 gets activated with the apoptotic induction. Increase in apoptotic cells and DNA were confirmed through annexin V staining, Hoechst staining, and cell cycle analysis.Conclusion:Thus, these results strongly suggest that stigmasterol has the potential to be considered as an anticancerous therapeutic agent against Jab1 in gall bladder cancer.


Cells ◽  
2018 ◽  
Vol 7 (10) ◽  
pp. 171 ◽  
Author(s):  
Rohit Gundamaraju ◽  
Ravichandra Vemuri ◽  
Wai Chin Chong ◽  
Stephen Myers ◽  
Shaghayegh Norouzi ◽  
...  

Sustained endoplasmic reticular stress (ERS) is implicated in aggressive metastasis of cancer cells and increased tumor cell proliferation. Cancer cells activate the unfolded protein response (UPR), which aids in cellular survival and adaptation to harsh conditions. Inhibition of apoptosis, in contrast, is a mechanism adopted by cancer cells with the help of the inhibitor of an apoptosis (IAP) class of proteins such as Survivin to evade cell death and gain a proliferative advantage. In this study, we aimed to reveal the interrelation between ERS and Survivin. We initially verified the expression of Survivin in Winnie (a mouse model of chronic ERS) colon tissues by using immunohistochemistry (IHC) and immunofluorescence (IF) in comparison with wild type Blk6 mice. Additionally, we isolated the goblet cells and determined the expression of Survivin by IF and protein validation. Tunicamycin was utilized at a concentration of 10 µg/mL to induce ERS in the LS174T cell line and the gene expression of the ERS markers was measured. This was followed by determination of inflammatory cytokines. Inhibition of ERS was carried out by 4Phenyl Butyric acid (4PBA) at a concentration of 10 mM to assess whether there was a reciprocation effect. The downstream cell death assays including caspase 3/7, Annexin V, and poly(ADP-ribose) polymerase (PARP) cleavage were evaluated in the presence of ERS and absence of ERS, which was followed by a proliferative assay (EdU click) with and without ERS. Correspondingly, we inhibited Survivin by YM155 at a concentration of 100 nM and observed the succeeding ERS markers and inflammatory markers. We also verified the caspase 3/7 assay. Our results demonstrate that ERS inhibition not only significantly reduced the UPR genes (Grp78, ATF6, PERKandXBP1) along with Survivin but also downregulated the inflammatory markers such as IL8, IL4, and IL6, which suggests a positive correlation between ERS and the inhibition of apoptosis. Furthermore, we provided evidence that ERS inhibition promoted apoptosis in LS174T cells and shortened the proliferation rate. Moreover, Survivin inhibition by YM155 led to a comparable effect as that of ERS inhibition, which includes attenuation of ERS genes and inflammatory markers as well as the promotion of programmed cell death via the caspase 3/7 pathway. Together, our results propose the interrelation between ERS and inhibition of apoptosis assigning a molecular and therapeutic target for cancer treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1689-1689 ◽  
Author(s):  
Deshpande S. Deshpande ◽  
Mary Jo Lechowicz ◽  
Rajni Sinha ◽  
Jonathan L. Kaufman ◽  
Lawrence H. Boise ◽  
...  

Abstract Abstract 1689 Poster Board I-715 Introduction The use of the proteasome inhibitor bortezomib has demonstrated activity in multiple myeloma and lymphomas. The HDAC inhibitor romidepsin is being evaluated in CTCL and PTCL, though its activity in B-cell lymphomas is less clear. We hypothesized that the combination of bortezomib and romidepsin would result in synergistic apoptosis in different B-cell NHL cell lines based upon the observed activity of this combination in more mature B-cell malignancies such as myeloma. Experimental Design Daudi, HT, Ramos and SUDHL-4 cell lines were exposed to different concentrations of bortezomib and romidepsin, separately, concurrently, and sequentially. Cell viability was assessed using MTT-assay, induced apoptosis was evaluated using Annexin V and PI staining from 24-48 hours. Apoptosis was also evaluated using western blot analysis of caspases and PARP cleavage. LC3 and HDAC6 level expressions were performed to determine if the effect of the combination was a result of the aggresome or autophagy pathway. Cell cycle studies were also performed to study if there were any changes after treating cells with the combination. Results The combination of bortezomib and romidepsin resulted in synergistic B-cell apoptosis as measured by MTT-assay with combination indices of < 0.5. This was associated with increased caspases and PARP cleavage as early as 24 hours after exposure. Order of addition experiments demonstrated definite sequence specificity. When romidepsin was added first, and 6 hours later followed by bortezomib, apoptosis was enhanced, compared to both agents being given concurrently or when bortezomib was administered first. Cell cycle analysis studies demonstrated that pretreatment of cells with romidepsin for 6 hours followed by the addition of bortezomib arrested the cells in G2M phase. HDAC6 expression was significantly reduced following combination therapy, and LC3-I was cleaved to LC3-II in treated cells suggesting that the combination affected aggresome formation and autophagy. Conclusion The combination of romidepsin and bortezomib at low nanomolar concentrations suggests that this may be an important clinical combination to test in patients with relapsed or refractory B-cell malignancies. Sequence of administration data is currently being tested to determine if the effect is a result of autophagy inhibition as is seen in myeloma cell lines. Additional mechanistic studies will be presented with the goals of identifying predictors of response that can then be validated in prospective clinical trials. Disclosures Lechowicz: Gloucester: Consultancy. Kaufman:Millennium: Consultancy; Genzyme: Consultancy; Celgene: Consultancy; Merck: Research Funding; Celgene: Research Funding. Lonial:Gloucester: Research Funding; Novartis: Consultancy; BMS: Consultancy; Millennium: Consultancy, Research Funding; Celgene: Consultancy. Flowers:Millennium: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1847-1847
Author(s):  
Chirag Acharya ◽  
Mike Y Zhong ◽  
Daniel Tannenbaum ◽  
Michelle Chen ◽  
Matt Ma ◽  
...  

Abstract Abstract 1847 Aminopeptidases (AP) are necessary for the growth and development of malignant cells and have a selectively important role in the maintenance of intracellular amino acid (AA) levels in neoplastic cells. CHR2797 is a novel, low nanomolar inhibitor of the M1 family of AP, a group of metalloenzymes containing a central Zn2+ ion. CHR2797 has antiproliferative and apoptotic effects against MM in vitro by inducing the AA deprivation response (AADR). TST, an oral, chronically administered agent with a good safety profile has demonstrated activity in patients with relapsed/refractory AML and is currently under study as part of combination therapy for untreated elderly patients with AML. At the epigenetic regulatory level, Zn-dependent histone deacetylase (HDAC) cause the deacetylation of histone and non-histone cellular proteins which are critical for gene expression, inducing apoptosis and cell cycle arrest in cancer cells. LBH589 (Panobinostat) is an established pan-HDAC inhibitor with potent in vitro anti-cancer activity in many hematological malignancies. The clinical efficacy of Panobinostat is currently being studied in several Phase II/III clinical trials with particular promise seen in the treatment of MM. Here we examined the potential therapeutic effect of CHR2797, alone and with LBH589, against MM cells. Using MTS and CTG assays, CHR2797, at clinically achievable concentrations, decreased survival and proliferation in MM1S and IL-6-dependent ANBL6 cells, in the presence or absence of bone marrow stromal cells following 72 hours incubation. CHR2797 induces apoptosis in MM cells via activation of Caspase 3/7 and 9 but not Caspase 8. Significantly, CHR2797 (10 μM) induced apoptosis in patient MM cells, as seen by % of annexin V and PI from 22 + 1.5% to 39 + 2.3% after 48h incubation. Combined treatment with CHR2797 and LBH589 in MM cells (MM1S, ANBL6, and INA6) further reduced cell viability following 72 hour incubation when compared with CHR2797 treatment alone, as determined by CTG viability luminescent assay. Both drugs together also augmented growth inhibitory effects when compared with single agent alone, after 72 hours incubation followed by MTS assay. Importantly, the combination of both drugs increased caspase 3/7- & 9-mediated apoptosis than CHR2797 alone in these MM cells following 24h-treatment. Cell cycle analysis (CHR2797 at 1μM; LBH589 at 1 nM) showed an increased growth arrest in G0/G1 cells in MM1R cells treated with both drugs versus CHR2797 alone after 24 hours: 68.5±3.3% versus 36±2.5%. Furthermore, CHR2797 inhibited anti-apoptotic protein Mcl-1 in MM1R and U266 MM cells by immunoblottings. Combined treatment with CHR2797 and LBH589 further blocked Mcl-1 when compared with either treatment alone after 24 hours incubation. Together, these results show that the combination of CHR2797 and LBH589 enhanced anti-myeloma effects when compared with either drug alone. This combination, which also has the potential of being without overlapping clinical toxicities, provides a promising novel approach to anti-myeloma therapy. Disclosures: Singer: Cell Therapeutics, Inc: Employment, Equity Ownership. Richardson:Novartis: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4478-4478 ◽  
Author(s):  
Noriyoshi Iriyama ◽  
Hirotsugu Hino ◽  
Shota Moriya ◽  
Masaki Hiramoto ◽  
Yoshihiro Hatta ◽  
...  

Abstract Background:Multiple myeloma (MM) is a hematologic malignancy characterized by the accumulation of abnormal plasma cells in the bone marrow. D-type cyclins (CCNDs), an important family of cell cycle regulators, are thought to be implicated in multiple myeloma (MM) development because CCNDs are commonly expressed in myeloma cells. CCND is known to positively regulate the cell cycle from G1 to S-phase initiation by binding to cyclin-dependent kinase (CDK) 4/6, resulting in potentiation of myeloma cell growth. These findings suggest a possible role for CDK4/6-targeting therapy in MM, yet the details remain incompletely understood. In this regard, we investigated the biological activity of abemaciclib, a potent, highly selective CDK4/6 inhibitor, in myeloma cell lines, to elucidate the mechanisms underlying the involvement of the CCND-CDK4/6 complex in cell cycle regulation and survival. Methods:The effects of abemaciclib on myeloma cells were investigated using three myeloma cell lines, KMS12-PE (CCND1-positive and CCND2-negative), RPMI8226 (CCND1-negative and CCND2-positive), and IM-9 (both CCND1- and CCND2-positive). Cell growth was assessed by trypan blue exclusion assay. Cell cycle analysis was performed using propidium iodide (PI) and apoptosis was measured using annexin V/PI staining via flow cytometry. Cell cycle regulated proteins, including p21 and p27, and phosphorylated proteins, including STAT1, STAT3, ERK, JNK, p38, and AKT, were evaluated using a phospho-flow method. Autophagy was assessed using CYTO-ID via flow cytometry. PARP cleavage was investigated via western blotting. Clarithromycin, an antibiotic agent belonging to the macrolide class, was used as an autophagy inhibitor. Results:Abemaciclib inhibited myeloma cell growth in a dose-dependent manner in all the cell lines evaluated, with significant differences seen at a concentration of 320 nM. Annexin V/PI staining revealed that 1 μM abemaciclib showed little or no effect on apoptosis, but 3.2 μM abemaciclib induced apparent myeloma cell apoptosis, with an increase in both the early and late apoptotic fractions. Therefore, 1 and 3.2 μM of abemaciclib were used in subsequent experiments for the assessment of cell growth and apoptosis, respectively. Cell cycle analyses revealed that 1 μM abemaciclib increased the fraction of cells in G0/G1 phase and decreased the fraction in S-G2/M phase. Furthermore, this effect was associated with the upregulation of p21 and p27 in the evaluated myeloma cells. PARP cleavage was observed in KMS12-PE cells treated with 3.2 μM abemaciclib, but not 1 μM, suggesting a close connection between the degree of PARP cleavage and apoptosis in myeloma cells. Importantly, abemaciclib induced autophagy in a dose-dependent manner. However, no apparent inhibitory effect on the autophagy-related phosphorylated proteins STAT1 (Y701), STAT3 (Y705), ERK (T202/Y204), JNK (T183/Y185), p38 (T180/Y182), or AKT (Y315) was observed in myeloma cells treated with 3.2 μM abemaciclib. To investigate the role of abemaciclib-induced autophagy on myeloma cell apoptosis, we further assessed the apoptotic effect of 3.2 μM abemaciclib or 50 μg/mL clarithromycin, alone or in combination. Clarithromycin did not induce apoptosis of myeloma cells. Importantly, clarithromycin treatment in combination with abemaciclib attenuated the apoptotic effect of abemaciclib. Discussion & Conclusions: Although the underlying mechanisms conferring the level of CCND expression are known to differ greatly (e.g., CCND translocation, hyperdiploidy, or activation of upstream pathways of CCND transcription), the results of the current study indicate that the CCND-CDK4/6 complex is closely involved in myeloma cell growth and survival regardless of the CCND family member present. In addition, we demonstrate that abemaciclib exerts multiple effects, such as myeloma cell apoptosis, via the PARP pathway or autophagy, as well as cell cycle regulation. Because abemaciclib in combination with clarithromycin inhibits myeloma cell apoptosis, the autophagy induced by abemaciclib is considered to have a critical role in the induction of apoptosis, so-called "autophagic cell death." These results provide novel insights into a possible therapeutic approach using abemaciclib to target CDK4/6 in patients with MM, and offer new possibilities for combination therapy with CDK4/6 inhibitors and autophagy regulators. Disclosures Iriyama: Novartis: Honoraria, Speakers Bureau; Bristol-Myers Squibb: Honoraria, Speakers Bureau. Hatta:Novartis Pharma: Honoraria.


2019 ◽  
Vol 9 (2) ◽  
pp. 319-323 ◽  
Author(s):  
Li Ping ◽  
Li Mingzhu ◽  
Lü Yuchun

Objective: To explore on the antitumor effect of gefitinib and rapamycin and possible mechanism in normal glucose and high lactic acid microenvironment. Methods: Hela cells are cultured in six conditions: the normal glucose group (NG, glucose 3 mmol/L); the normal glucose + gefitinib group (NGG, glucose 3 mmol/L, gefitinib 2.67 μmol/L); the normal glucose + rapamycin group (NGR, glucose 3 mmol/L, rapamycin 2.67 μmol/L); the high lactate group (NGHL, glucose 3 mmol/L, lactic acid 2.5 mmol/L); the normal glucose + high lactate + gefitinib group (NGHLG, glucose 3 mmol/L, lactic acid 2.5 mmol/L, gefitinib 2.67μmol/L); the normal glucose + high lactate + rapamycin group (NGHLG, glucose 10 mmol/L, lactic acid 2.5 mmol/L, rapamycin 2.67μmol/L). Growth inhibitory rate of Hela cell is determined by CCK-8; Flow cytometry (FCM) is performed to evaluate the cell cycle; The annexin V-phycoerythrin/Propidium Iodide (annexin V-PE/PI) staining combined with flow cytometry is used to examine the cell cycle and apoptosis of Hela cells. Results: Under normal glucose with gefitinib or rapamycin environment, the apoptosis rate of Hela cells is higher than that of the normal glucose group. But the cell apoptosis rate of the gefitinib or rapamycin group decreases in high lactic acid and normal glucose, which is lower than that of the normal glucose and high lactate. Combined with the results of cell cycle, compared with the normal glucose group, percentage of Hela cells in G1/G0 phase increases significantly, the proportion of S phase cells decreases significantly in high lactic acid environment. In the normal glucose and gefitinib environment, Hela cells in G1/G0 phase and S phase are slightly higher than the proportion of normal glucose group, and G2/M phase cells are mild lower than the proportion of normal glucose group. Under the environment of high lactate and normal glucose, the percentage of G1/G0 and S phase cells in the gefitinib increase. As for rapamycin, normal glucose and high lactic acid environment makes cells stay in G1/G0 phase. The presence of rapamycin in the environment of normal sugar and high lactate makes more cells stay in G1/G0 or G2/M phase. Conclusion: Normal glucose and high lactic acid environment is conducive to Hela cell survival, and can promote the expression of EGFR and mTOR. Gefitinib is an antagonist of EGFR and rapamycin is an inhibitor of mTOR.


2004 ◽  
Vol 286 (3) ◽  
pp. H902-H908 ◽  
Author(s):  
Antonio Curcio ◽  
Daniele Torella ◽  
Giovanni Cuda ◽  
Carmela Coppola ◽  
Maria Concetta Faniello ◽  
...  

Synthetic polymers, like methacrylate (MA) compounds, have been clinically introduced as inert coatings to locally deliver drugs that inhibit restenosis after stent. The aim of the present study was to evaluate the effects of MA coating alone on vascular smooth muscle cell (VSMC) growth in vitro. Stainless steel stents were coated with MA at the following doses: 0.3, 1.5, and 3 ml. Uncoated/bare metal stents were used as controls. VSMCs were cultured in dishes, and a MA-coated stent or an uncoated bare metal stent was gently added to each well. VSMC proliferation was assessed by bromodeoxyuridine (BrdU) incorporation. Apoptosis was analyzed by three distinct approaches: 1) annexin V/propidium iodide fluorescence detection; 2) DNA laddering; and 3) caspase-3 activation and PARP cleavage. MA-coated stents induced a significant decrease of BrdU incorporation compared with uncoated stents at both the low and high concentrations. In VSMCs incubated with MA-coated stents, annexin V/propidium iodide fluorescence detection showed a significant increase in apoptotic cells, which was corroborated by the typical DNA laddering. Apoptosis of VSMCs after incubation with MA-coated stents was characterized by caspase-3 activation and PARP cleavage. The MA-coated stent induced VSMC growth arrest by inducing apoptosis in a dose-dependent manner. Thus MA is not an inert platform for eluting drugs because it is biologically active per se. This effect should be taken in account when evaluating an association of this coating with antiproliferative agents for in-stent restenosis prevention.


Sign in / Sign up

Export Citation Format

Share Document