The IMPDH Inhibitor VX-944 Demonstrates In Vivo Efficacy in an Aggressive Leukemia Model.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2530-2530 ◽  
Author(s):  
Jugnu Jain ◽  
Jianguo Ma ◽  
Brinley Furey ◽  
Christian Recher ◽  
Cecile Demur ◽  
...  

Abstract VX-944 is a small molecule, orally bioavailable, specific inhibitor of inosine monophosphate dehydrogenase (IMPDH), an essential rate-limiting enzyme in de novo guanine nucleotide synthesis. We have previously shown that VX-944 has broad anti-cancer properties in vitro and that its potency is not affected by MDR pumps (Jain et al, Blood 2002, 100:572a; ibid 2003, 102: 622a). Here, we describe studies that demonstrate its oncolytic activity in blast cells derived from patients with acute myeloid leukemia (AML). We also report the efficacy of VX-944 in an aggressive murine model of myeloproliferative disease. The anti-leukemic activity of VX-944 was established both in colony formation assays and in viability assays using primary cells from AML patients. VX-944 inhibited the clonogenic proliferation of acute myeloid progenitor cells in a dose-dependent manner. The mean and median IC50 values were 218±92 and 199 nM respectively (n=8), indicating that no samples were resistant to VX-944. In another study, VX-944 reduced the viability of AML blasts from 4 additional patient samples, with IC50 values ranging from 20–200 nM. VX-944 was observed to be 3–40-fold more potent than mycophenolic acid (MPA, MMF), another IMPDH inhibitor. Importantly, genotyping of these samples revealed that VX-944 is active against cancer cells with wild type and point- or ITD-mutations in Flt3 that are implicated in approx 30% of AML patients. The anti-proliferative activity of VX-944 was at least additive, and in some patient samples, synergistic, when tested in combination with Daunorubicin, a standard chemotherapy drug for AML. To determine the therapeutic potential of VX-944 in vivo, VX-944 was tested in a leukemia model using Ba/F3 cells transduced with an activating human Flt-3 mutation injected into Balb/c mice. The anti-proliferative activity of VX-944 was first established in vitro in the cell lines used in the model. VX-944 inhibited the proliferation of the human MV-4-11 and murine Ba/F3-Flt3-ITD-dependent cell lines with IC50 values of 26 and 30 nM, respectively. In PK studies, a dose-dependent increase in Cmax and AUC values were observed in Balb/c mice. In the leukemia model, VX-944 was administered orally at 75 or 150 mg/kg BID. Doxorubicin (3 mg/kg, weekly), a standard AML therapy, and MLN518 (60 mg/kg BID), a selective Flt3 inhibitor, were administered as reference compounds. All treatments began 5 days following cell implantation. Both dose groups of VX-944 provided a significant increase in median survival time as compared with the vehicle treated group (p <0.001). Three of the 12 mice treated with 150 mg/kg VX-944 were still alive on Day 35 when the study was terminated. Further studies are underway to optimize the dosing regimen of VX-944 in xenograft models. In conclusion, we have demonstrated both potency and efficacy of VX-944 in in vitro and in vivo leukemia models. These preclinical results support further clinical development of VX-944 for the treatment of patients with leukemias and other rapidly proliferating hematological malignancies. VX-944 may provide significant therapeutic benefit when used alone, or in combination with approved chemotherapy agents such as Daunorubicin.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3357-3357
Author(s):  
Renate Burger ◽  
Steven Legouill ◽  
Yu-Tzu Tai ◽  
Reshma Shringarpure ◽  
Klaus Podar ◽  
...  

Abstract In multiple myeloma (MM), IL-6 plays an important role for tumor cell growth, survival, and drug resistance. Janus kinases (JAKs) are protein tyrosine kinases and constitutively associated with the gp130 chain of the IL-6 receptor complex. Their activation is one of the first steps in cytokine receptor-mediated signaling and critical for virtually all subsequent downstream signaling cascades. INCB20 is a small-molecule synthetic compound which, in biochemical assays, potently inhibited all four JAKs with IC50 values between 0.3 nM and 1.2 nM (for comparison, IC50 of AG490, another JAK inhibitor, was >50 μM). Consistent with the central role of JAKs in gp130-mediated signaling, INCB20 inhibited IL-6 induced phosphorylation of SHP-2, STAT1, STAT3, ERK1/2, and AKT in MM1.S cells. In contrast, AKT phosphorylation induced by IGF-1 remained unchanged. Evaluation of the cellular efficacy of INCB20 was performed using the IL-6 dependent INA -6 cell line. Growth of INA-6 cells was inhibited in a dose-dependent manner with an IC50 of approx. 0.5 μM, as measured by [3H]-thymidine uptake and an MTS-based assay (for comparison, the cellular IC50 of AG490 was 15–20 μM). This correlated with an increase in the percentage of apoptotic cells, as evaluated by Apo2.7 staining after 48 hours. Importantly, INA-6 growth was inhibited in the presence of bone marrow stromal cells accompanied by a decrease in phospho-STAT3 levels. Furthermore, in a subcutaneous INA-6-SCID model, INCB20 inhibited tumor growth (and phosphorylated STAT3) in a dose-dependent manner. Our studies provide the conceptual basis for the use of JAK inhibitors as a therapeutic approach in MM.


PeerJ ◽  
2020 ◽  
Vol 8 ◽  
pp. e9909
Author(s):  
Carol Haddoub ◽  
Mohamad Rima ◽  
Sandrine Heurtebise ◽  
Myriam Lawand ◽  
Dania Jundi ◽  
...  

Background Montivipera bornmuelleri’s venom has shown immunomodulation of cytokines release in mice and selective cytotoxicity on cancer cells in a dose-dependent manner, highlighting an anticancer potential. Here, we extend these findings by elucidating the sensitivity of murine B16 skin melanoma and 3-MCA-induced murine fibrosarcoma cell lines to M. bornmuelleri’s venom and its effect on tumor growth in vivo. Methods The toxicity of the venom on B16 and MCA cells was assessed using flow cytometry and xCELLigence assays. For in vivo testing, tumor growth was followed in mice after intratumoral venom injection. Results The venom toxicity showed a dose-dependent cell death on both B16 and MCA cells. Interestingly, overexpression of ovalbumin increased the sensitivity of the cells to the venom. However, the venom was not able to eradicate induced-tumor growth when injected at 100 µg/kg. Our study demonstrates a cytotoxic effect of M. bornmuelleri’s venom in vitro which, however, does not translate to an anticancer action in vivo.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3302-3302
Author(s):  
Timothy Pardee ◽  
Evan Gomes ◽  
Jamie Jennings-Gee ◽  
David L. Caudell ◽  
William Gmeiner

Abstract Abstract 3302 Acute Myeloid Leukemia (AML) is an aggressive myeloid malignancy that leads to marrow failure and death. This disease affects approximately 12,000 people per year in the United States, causing 9,000 deaths. Despite decades of research, therapy remains essentially unchanged and outcomes are poor. In patients over the age of 60 less then 10% of patients survive 5 years from diagnosis. There is a desperate need for the identification of new active agents with favorable toxicity profiles. The novel polymeric fluoropyrimidine (FP) FdUMP[10] is an oligodeoxynucleotide pro-drug of the thymidylate synthase (TS)-inhibitory FP metabolite 5-fluoro-2'-deoxyuridine-5`-O-monophosphate (FdUMP). The observation that this compound was highly active against several leukemia lines in the NCI 60 cell line screen prompted us to evaluate its activity in several preclinical models of AML. In vitro, FdUMP[10] exhibited remarkable activity against 3 human acute leukemia cell lines, HL60, Jurkat and THP-1, with IC50 values of 3.378 nM (95% CI 2.984 to 3.825), 5.438 nM (4.609 to 6.417) and 4.093 nM (3.413 to 4.907) respectively. We next tested its efficacy against a more genetically defined murine model of AML driven by expression of MLL-ENL. FdUMP[10] exhibited even greater activity against all murine lines tested. The IC50 values of FdUMP[10] against two MLL-ENL driven murine AML cell lines were 214 pM (95%CI 178.9 to 255.9) and 292.3 pM (251.8 to 339.4). The IC50 values observed for FdUMP[10] for all the murine lines tested were lower than both Ara-C (30-40 nM) and doxorubicin (2-4 nM). We then determined the cytotoxic mechanism for FdUMP[10] in vitro. Upon treatment with FdUMP[10] both the human and murine cell lines undergo extensive apoptosis as indicated by Annexin V and propidium iodide staining. Treated cells developed γH2AX foci, rapid and complete TS inhibition and display trapped Topoisomerase I (Topo I) cleavage complexes. FdUMP[10]-mediated induction of apoptosis was p53 independent as murine AML cells that had p53 knocked down by RNAi demonstrated resistance to both Ara-C and doxorubicin, but not to FdUMP[10]. We next tested the efficacy of FdUMP[10] in vivo. The MLL-ENL driven murine AML model results in blasts that can be transplanted into sublethally irradiated, immunocompetent, syngeneic recipients. The recipients develop a fatal and therapy-resistant AML. Lines were generated that expressed a luciferase reporter. Animals were imaged 6–7 days after injection of the leukemias to ensure engraftment and then began treatment with either the combination of Ara-C plus doxorubicin, single-agent FdUMP[10], or observation. Studies were performed using 2 doses of FdUMP[10] at 150 or 300 mg/kg injected on days 1 and 3 and compared to animals treated with 100 mg/kg Ara-C and 3mg/kg doxorubicin injected on days 1 through 5. Both treatments resulted in a statistically significant survival advantage over observation. A preliminary toxicology study compared FdUMP[10], 150 mg/kg daily, to 5-fluorouracil (5 FU), 150 mg/kg daily, or the combination of Ara-C at 100 mg/kg plus doxorubicin at 3 mg/kg daily. All groups were treated for 3, 4 or 5 days. On day 6 animals were sacrificed and organs harvested, sectioned, and stained. Slides were then reviewed by a veterinary pathologist. Tissues most affected were the small intestine, colon, and the bone marrow. The 5FU-treated animals had severe villous blunting and fusion with crypt necrosis in both large and small intestine. In contrast, FdUMP[10]-treated animals had only mild crypt epithelial apoptosis with mitoses. The 5 FU and Ara-C plus doxorubicin groups had a severe pan-cytopenia in the marrow compared to FdUMP[10] treated animals that showed only minimal to mild apoptosis. These data support the assertion that FdUMP[10] has lower toxicity then either Ara-C plus doxorubicin or identically dosed 5 FU. In summary FdUMP[10] exhibited remarkable activity against AML cells in vitro and in vivo. Additionally, FdUMP[10] had decreased toxicity compared to treatment with either single agent 5 FU or combination treatment with Ara-C plus doxorubicin. Disclosures: Gmeiner: Salzburg Therapeutics: Equity Ownership.


2019 ◽  
Vol 63 (1) ◽  
pp. 39-49 ◽  
Author(s):  
Soojin Kim ◽  
Daksh Thaper ◽  
Samir Bidnur ◽  
Paul Toren ◽  
Shusuke Akamatsu ◽  
...  

Neuroendocrine (NE) differentiation of advanced prostate adenocarcinoma following androgen receptor (AR) axis-directed therapy is becoming increasingly recognized. Several models of this transdifferentiation provide insight into its molecular pathogenesis and have highlighted the placental gene PEG10 for further study. Using our unique model of enzalutamide resistance (ENZR) and NE differentiation, we studied PEG10/AR interplay in enzalutamide treatment-resistant cell lines 42DENZR and 42FENZR compared to LNCaP and castration-resistant 16DCRPC cells. ENZR cell lines with positive terminal NE marker status also displayed higher baseline expression of PEG10 compared to LNCaP and 16DCRPC. Antagonism of AR activity increased PEG10 expression followed by an increase in terminal NE markers. Conversely, stimulating AR activity via androgen supplementation reversed PEG10 and NE marker expression in a time and dose-dependent manner. These results were supported by human data showing that PEG10 expression is highest in NEPC and that AR-dependent gene, PSA, is negatively correlated with PEG10 in adenocarcinoma. Further, ChIP assay confirmed binding of activated AR to the PEG10 enhancer, decreasing PEG10 expression. While PEG10 did not drive NEPC, its knockdown reduced NE markers in our cell lines. Moreover, PEG10 knockdown in vitro- and in vivo-attenuated tumor growth. Overall, these observations indicate that PEG10 is an AR-repressed gene which modulates NE markers in ENZR cells and targeting PEG10 in advanced prostate cancer with NE features is a rational and viable option.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2555-2555 ◽  
Author(s):  
Sha Liu ◽  
Lin Chen ◽  
Xiaojiao Wang ◽  
Ruihua Mi ◽  
Qingsong Yin ◽  
...  

Background Acute myeloid leukaemia (AML) is a malignancy derived from haematopoietic stem cells in which maturation and apoptosis are blocked and proliferation is uncontrolled during differentiation. With the optimization of chemotherapy regimens, the emergence of new drugs and the development of haematopoietic stem cell transplantation technology, the complete remission (CR) rate and long-term survival of AML patients have significantly improved, but 20-40% of patients still have difficulty achieving a CR. Furthermore, approximately 60% of patients eventually relapse after a CR. Patients who are unable to obtain a CR and who relapse after remission are more likely to develop refractory AML. For patients with refractory/relapsed AML, there is no standard rescue therapy, and general chemotherapy drugs have limited efficacy. It is of great significance to explore new salvage treatment options. Venetoclax (ABT-199) is an effective oral inhibitor of Bcl-2. ABT-199 has been used to treat various in vitro and in vivo tumour models, including AML models, and these experiments have shown its effectiveness as a single agent. However, its efficacy is very limited in refractory/relapsed AML, Mcl-1 play important roles in ABT-199 resistance. As an ancient pro-apoptotic drug, arsenic trioxide (ATO) has been shown to induce apoptosis in vivo and in vitro. ATO can downregulate the expression of Mcl-1, thereby inducing apoptosis. objective To explore not only the synergistic pro-apoptotic effects of the Bcl-2 inhibitor ABT-199 and ATO on AML cell lines and AML primary cells, but also the clinical effects on refractory/relapsing AML patients. Methods 1. Bcl-2 and Mcl-1 mRNA and protein levels in AML cell lines (MOLM13, MV4-11, THP-1, OCI-AML3, U937) were detected by real-time PCR and Western blot, respectively; 2.Apoptosis rates of AML cell lines after treatment with different concentrations of ABT-199 or ATO alone or in combination were measured by flow cytometry; 3. The transcriptomes of THP-1 and OCI-AML3 cells before and after treatment with ABT-199 or ATO alone or in combination were analysed; 4. Western blot, co-immunoprecipitation, cell cycle assay ABT-199, ATO effect on oci-AML3 cells; 5. ABT-199 combined with ATO in the treatment of patients with relapsed and refractory AML, the specific program is: ABT-199 100mg d1, 200mg d2, 400mg d3-28, oral 30 minutes after breakfast, ATO 6mg/m2, d1-28, daily intravenous infusion for 6 hours, review the effect of bone marrow aspiration on the 28th day. Results 1.MOLM13, MV4-11, THP-1, OCI-AML3 and U937 cells expressed both Bcl-2 and Mcl-1. The sensitivity of AML cell lines to ABT-199 was positively correlated with the Bcl-2 protein expression level; 2.In AML cell lines whether ABT-sensitive or ABT-resistant and primary AML cells, ABT-199 and ATO synergistically promoted apoptosis in a time- and concentration-dependent manner; 3. ABT-199 up-regulated the expression of Mcl-1 in ABT-resistant OCI-AML3 cells, ATO down-regulated the expression of p-AKT, p-GSK3β, and Mcl-1, and the expression of Mcl-1 decreased after the combination of the two drugs. Bim and Mcl-1 protein binding decreased after treatment OCI-AML3 cells with the combination of the two drugs for 24h; 4. ATO promotes DNA damage, and the effect is further enhanced when combinated with ABT-199; 5. A total of 7 patients with relapse were treated with ABT-ATO combined regimen. Four of the 7 patients had multiple relapses and 3 patients had short-term recurrence after remission, with an average age of 49 years. On the 28th day, the bone marrow was reviewed. Two of them achieved CRi, one case with Leukemia-free state (MLFS), two cases with PR, one case with SD, and one case with PD. Conclusion We demenstrated ATO could reverse AML resistance to ABT-199 and the synergistic effects of the combination of ATO and ABT-199, which may provide a new and probably more effective treatment strategy for refractory/relapsed AML . Key Words Bcl-2 inhibitor; ABT-199; arsenic trioxide; Mcl-1; AKT; apoptosis Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (9) ◽  
pp. 1765-1773 ◽  
Author(s):  
Parvathi Ranganathan ◽  
Xueyan Yu ◽  
Caroline Na ◽  
Ramasamy Santhanam ◽  
Sharon Shacham ◽  
...  

AbstractChromosome maintenance protein 1 (CRM1) is a nuclear export receptor involved in the active transport of tumor suppressors (eg, p53 and nucleophosmin) whose function is altered in cancer because of increased expression and overactive transport. Blocking CRM1-mediated nuclear export of such proteins is a novel therapeutic strategy to restore tumor suppressor function. Orally bioavailable selective inhibitors of nuclear export (SINE) that irreversibly bind to CRM1 and block the function of this protein have been recently developed. Here we investigated the antileukemic activity of KPT-SINE (KPT-185 and KPT-276) in vitro and in vivo in acute myeloid leukemia (AML). KPT-185 displayed potent antiproliferative properties at submicromolar concentrations (IC50 values; 100-500nM), induced apoptosis (average 5-fold increase), cell-cycle arrest, and myeloid differentiation in AML cell lines and patient blasts. A strong down-regulation of the oncogene FLT3 after KPT treatment in both FLT3-ITD and wild-type cell lines was observed. Finally, using the FLT3-ITD–positive MV4-11 xenograft murine model, we show that treatment of mice with oral KPT-276 (analog of KPT-185 for in vivo studies) significantly prolongs survival of leukemic mice (P < .01). In summary, KPT-SINE are highly potent in vitro and in vivo in AML. The preclinical results reported here support clinical trials of KPT-SINE in AML.


2020 ◽  
Vol 17 (4) ◽  
pp. 502-511
Author(s):  
Mingxia Song ◽  
Bing Liu ◽  
Shengwang Yu ◽  
Shihui He ◽  
Yuqiu Liang ◽  
...  

Background: Several series of hydrazone derivatives of pyrazole-4-carboxaldehydes (4- 11) were designed and synthesized to screen their inflammatory activity. Methods: The products were characterized by 1H NMR, 13C NMR and HRMS. In vitro LPS-induced TNF-α model and in vivo xylene-induced ear-edema model were used to evaluate their antiinflammatory activity. Results and Conclusion: Bioassays indicated that most of the compounds markedly inhibited the expression of TNF-α at the concentration of 10 µg/mL. Compounds 7b and 11c, two of the most potent compounds, exhibited TNF-α inhibitory ability in a dose-dependent manner with IC50 values of 5.56 and 3.69 µM, respectively. In vivo, intraperitoneal administration with 7b and 11c markedly inhibited the ear edema at the doses of 20 and 50 mg/kg. Compound 11c, inhibited edema by 49.59 % at the dose of 20 mg/kg, was comparable to the reference drug dexamethasone at the same dose (with an inhibition of 50.49 %). To understand the binding pattern, molecular docking of representative 7b and 11c was performed, which demonstrated that both compounds have a forceful binding with the TNF-α, and that the phenyl and hydrazide moieties of them play a significant role in binding with the target site.


2014 ◽  
Vol 121 (6) ◽  
pp. 1483-1491 ◽  
Author(s):  
Ho-Shin Gwak ◽  
Myung-Jin Park ◽  
In-Chul Park ◽  
Sang Hyeok Woo ◽  
Hyeon-Ok Jin ◽  
...  

Object Local invasiveness of malignant glioma is a major reason for the failure of current treatments including surgery and radiation therapy. Tetraarsenic oxide (As4O6 [TAO]) is a trivalent arsenic compound that has potential anticancer and antiangiogenic effects in selected cancer cell lines at a lower concentration than arsenic trioxide (As2O3 [ATO]), which has been more widely tested in vitro and in vivo. The authors tried to determine the cytotoxic concentration of TAO in malignant glioma cell lines and whether TAO would show anti-invasive effects under conditions independent of cell death or apoptosis. Methods The human phosphatase and tensin homolog (PTEN)-deficient malignant glioma cell lines U87MG, U251MG, and U373MG together with PTEN-functional LN428 were cultured with a range of micromolar concentrations of TAO. The invasiveness of the glioma cell lines was analyzed. The effect of TAO on matrix metalloproteinase (MMP) secretion and membrane type 1 (MT1)-MMP expression was measured using gelatin zymography and Western blot, respectively. Akt, or protein kinase B, activity, which is a downstream effector of PTEN, was assessed with a kinase assay using glycogen synthesis kinase-3β (GSK-3β) as a substrate and Western blotting of phosphorylated Akt. Results Tetraarsenic oxide inhibited 50% of glioma cell proliferation at 6.3–12.2 μM. Subsequent experiments were performed under the same TAO concentrations and exposure times, avoiding the direct tumoricidal effect of TAO, which was confirmed with apoptosis markers. An invasion assay revealed a dose-dependent decrease in invasiveness under the influence of TAO. Both the gelatinolytic activity of MMP-2 and MT1-MMP expression decreased in a dose-dependent manner in all cell lines, which was in accordance with the invasion assay results. The TAO decreased kinase activity of Akt on GSK-3β assay and inhibited Akt phosphorylation in a dose-dependent manner in all cell lines regardless of their PTEN status. Conclusions These results showed that TAO effectively inhibits proliferation of glioblastoma cell lines and also exerts an anti-invasive effect via decreased MMP-2 secretion, decreased MT1-MMP expression, and the inhibition of Akt phosphorylation under conditions devoid of cytotoxicity. Further investigations using an in vivo model are needed to evaluate the potential role of TAO as an anti-invasive agent.


1994 ◽  
Vol 72 (01) ◽  
pp. 119-124 ◽  
Author(s):  
Juerg F Tschopp ◽  
Curt Mazur ◽  
Kenneth Gould ◽  
Raymond Connolly ◽  
Michael D Pierschbacher

SummaryMembrane glycoprotein αIIbβ3 on platelets plays a pivotal role in hemostasis by mediating RGD-(arginine-glycine-aspartic acid)-dependent platelet adhesion and aggregation. Antagonists of αIIbβ3 ligand binding function, such as antibodies, snake venom peptides, or synthetic RGD-containing peptides can completely inhibit platelet aggregation in vitro and cause significant prolongation of bleeding times when injected into experimental animals. The in vitro and in vivo properties of an αIIbβ3 specific RGD-containing peptide 2G (G(Ten)GHRGDLRCA) were compared to two non-specific RGD-containing peptides IN (G(Pen)GRGDTPCA) and 2H (GRGDSPDG). All three peptides have similar IC50 values in human patelet aggregation (14-22 μM) and ELISA-based μIIbβ3 receptor assays (0.2–0.3 αM) but show different inhibitory activity (IC50 values) in the αv㯂5 (2G = 10 μM; IN = 0.06 μM; 2H = 0.05 μM) and receptor assays (2G = 8.3 μM; IN = 0.06 μM; 2H = 0.04). The αIIbβ3 specific peptide 2G had no effect on monolayers of human saphenous vein endothelial cells while IN and 2H caused many cells to detach and contract. Peptides 2G and IN inhibited ADP-stimulated ex vivo platelet aggregation in dogs in a dose dependent manner. When complete inhibition (>90%) of ex vivo platelet aggregation was achieved with either a 10 mg/kg bolus followed by a 16mg/kg/h infusion of 2G or with a 15 mg/kg bolus and 24 mg/kg/h infusion of IN, peptide IN caused a dose-dependent increase of the template bleeding time, while peptide 2G had no effect, even at doses up to 15 mg/kg bolus followed by 24 mg/kg/h infusion. The in vivo properties of peptides 2G and 2H were also examined in a baboon ex vivo shunt model for their ability to block platelet uptake and fibrinogen deposition on small caliber GORE-TEX® grafts and for their effect on the hemostatic system. Systemic administration of peptide 2G at 10 mg/kg bolus followed by 10 mg/kg/h infusion (or at a 2-fold lower dose) abolished platelet uptake and fibrinogen deposition on the graft surface without affecting the hemostasis and template bleeding time of the animal. By contrast, peptide 2H caused a 3-4-fold increase in bleeding time at a dose of 10 mg/kg. The results suggest that efficacy and the effect of specific aIIbp3antagonists on bleeding time can be separated and that selective aIIbP3 receptor blockade may be an efficient and safe approach to improve the patency and the success rate pf small caliber vascular grafts and to treat unwanted platelet-dependent thromboses. While peptide 2G may represent a unique class of antithrombotic agent, the clinical use of this type of molecule would require a significant enhancement in potency.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4183-4183 ◽  
Author(s):  
Jordan N Boyle ◽  
Caroline R Kim ◽  
Hui Guo ◽  
Taylor Bell ◽  
Shengjian Huang ◽  
...  

Abstract Introduction Mantle cell lymphoma (MCL) is an aggressive lymphoma with elevated B-cell receptor activity. Ibrutinib (IBN), a Bruton's tyrosine kinase (BTK) inhibitor, has been shown to have a response rate of 68% in relapsed or refractory MCL patients. However, with the emergence of IBN-resistant lymphomas, new therapies are needed. It is suspected that upregulation of the PI3K-Akt-mTOR pathway allows survival in the presence of IBN. CUDC-907 is a next generation PI3K and HDAC dual-inhibitor currently in phase II clinical trials. Our objective is to investigate the effects of CUDC-907 in IBN-resistant MCL cells in vitro and in PDX model. Methods MCL cells were seeded at 10,000 cells per well in 96-well plate and were treated with various doses of compounds at the following concentrations: CUDC-907/Ibrutinib 0.015, 0.05, 0.15, 0.5, 1.5, 5, and 15 uM. Cell viability was tested by CellTiter-Glo luminescent cell viability assay (Promega) after a 72-hour incubation. Next, MCL cells were incubated with IBN at varying doses (0.39 uM, 1.56 uM, 6.25 uM), CUDC-907 (0.39 uM, 1.56 uM, 6.25 uM), and IBN+CUDC-907 (0.39 uM, 1.56 M, 6.25 uM) for 24 hours. Apoptosis was detected by Annexin V-binding assay. In patient derived xenograft (PDX) model: CUDC-907 was administered at a dose of 50 mg/kg in ibrutinib-resistant MCL-bearing PDX mice daily. Tumor volumes were measured as the length X width2 X 0.5 weekly. Toxicity was also observed every week. Mice were sacrificed once diameter of tumor mass reached 15 mm size. Results CUDC-907 inhibited the growth of both ibrutinib-sensitive and resistant MCL cells in vitro. Sensitive cell lines include: Rec-1, Mino, and JVM-13 with IC50 values of 1.1 nM, 1.0 nM, and 5 nM respectively. Resistant cell lines include: Granta-519, Maver-1, and Z-138 with IC50 values of 2 nM, 3 nM, and 1.5 nM respectively. All tested cell lines were more sensitive to CUDC-907 than to ibrutinib. In addition, combination treatments of CUDC-907 and IBN increased cell death in comparison to single agent treatments. Next, one pair of MCL cell lines, Jeko-1 (sensitive to IBN) and Jeko-R (resistant to IBN) were treated for 24 hours with varying doses of CUDC-907 or ibrutinib either as single agent inhibitors or in combination therapies. The results demonstrated that CUDC-907 induced apoptosis in both Jeko-1 and Jeko-R cell lines in a dose-dependent manner. Combination therapies increased cell death in a dose-dependent manner as well. In PDX model, tumor volume in treated mice of ibrutinib-resistant PDX decreased significantly compared with vehicle control (pvalue = 0.032). Control mice also weighed considerably more than treated mice (p value = 0.073). Common toxicities included a decrease in body mass for first 28 days of treatment. Conclusion CUDC-907, a dual inhibitor of PI3K-Akt-mTOR and HDAC, inhibits tumor growth of ibrutinib-resistant MCL in vitro and in PDX model. It would be a potential drug for the patients with ibrutinib-resistant/relapsed MCL. Disclosures Wang: Celgene: Research Funding; Onyx: Research Funding; Kite Pharma: Research Funding; Acerta Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pharmacyclics: Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BeiGene: Research Funding; Juno Therapeutics: Research Funding; Asana BioSciences: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document