Novel Small Molecule Inhibitors Of The Gas6/TAM Signaling Pathway Mediate Synergistic Inhibition Of Platelet Aggregation In Combination With ADP/P2Y Antagonists

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3507-3507
Author(s):  
Gilbert Acevedo ◽  
Brian R. Branchford ◽  
Luke Law ◽  
Christine Brzezinski ◽  
Susan Sather ◽  
...  

Abstract Background ADP activates platelets through P2Y1 and P2Y12 receptors. Various ADP/P2Y inhibitors are used clinically for arterial thrombosis prophylaxis. However, these agents exhibit inter-individual response variability and bleeding complications. A more recently described platelet activation signaling pathway is activated by Growth Arrest Specific gene 6 (Gas6), a ligand for the Tyro3/Axl/Mer (TAM) family of platelet surface receptor tyrosine kinases. Previous have shown that inhibiting this pathway decreases platelet activation responses and β3 integrin-mediated thrombus stabilization, and protects mice from arterial and venous thrombosis, without evidence of significant bleeding side effects. Here, we investigate the effects of a novel Mer-selective small molecule inhibitor (SMI) on platelet aggregation and murine models of induced thrombosis relative to ADP/P2Y pathway antagonists. Objectives We hypothesized that inhibition of the Gas6/TAM pathway with a novel SMI would decrease platelet aggregation and thrombosis, comparable to that seen with known ADP inhibitors. Additionally, we examined the effect of the combination of these 2 inhibitor types on platelet aggregation. Methods We compared the inhibitory effect of 2 known ADP inhibitors (1uM MRS2179 administered concurrently with 1uM 2-MeSAMP) to 1uM of a novel Mer-selective SMI (UNC Mer TKI), using standard light-transmission aggregometry with washed human platelets (30-minute incubation at 37 ¢ªC) and two murine thrombosis models (collagen/epinephrine-induced systemic venous thrombosis and FeCl3-induced carotid artery injury). Thrombosis studies were performed using WT C57BL/6 mice treated with either UNC Mer TKI or ADP/P2Y inhibitors compared to mice treated with vehicle only. Mean values +/- SEM are shown and statistical significance (p<0.05) was determined using the student’s paired t-test. Results ADP/P2Y antagonists and Gas6/TAM inhibitor both mediate protection from thrombosis in mice relative to vehicle-treated controls. Following FeCl3 –induced carotid artery injury, control mice (n=11) experienced stable vessel occlusion at a mean time of 6.77 +/- 0.25 minutes. In contrast, stable occlusion occurred at 46.6 +/- 7.72 minutes (n=9, p=0.001) in mice treated with UNC Mer TKI, and 18.74 +/- 4.35 minutes (n=3, p<0.001) for mice treated with the two ADP/P2Y inhibitors. Survival times following venous injection of collagen and epinephrine significantly differed between mice treated with ADP/P2Y antagonists or UNC Mer TKI, compared to vehicle control. Mice pre-treated with UNC Mer TKI (n=9, p=0.04) or ADP/P2Y inhibitors (n=5, p<0.001) survived for 19.84 +/- 4.4 and 19.9 +/- 4.92 minutes, respectively. In contrast, mice treated with vehicle control (n=21), only survived for 3.21 +/- 2.4 minutes. Both the ADP/P2Y antagonists and UNC Mer TKI also inhibit platelet aggregation. At 1uM doses, the maximum percent aggregation in UNC Mer TKI-treated samples (n=7) differed significantly from samples treated with vehicle alone, with mean values of 69 +/- 2.2% (p=0.04), 77 +/- 1.8% (n=7), and 76.9 +/- 2.1% (n=7), respectively. 1uM ADP/P2Y inhibitor-treated samples (n=7) exhibited a mean maximum aggregation of 62 +/- 5.2% (p=0.008), and the combination of ADP/P2Y inhibitors and UNC TAM TKI had a maximum percent aggregation of 31.3 +/- 7.7% (n=7, p=0.001). The Chou-Talalay Combination Index (a quantitative estimation of the effect of combined inhibitors) was 0.78, indicating a synergistic, rather than additive, effect. Similarly, using the Bliss additivity equation, 38.1 +/- 7.9% inhibition of aggregation was predicted for an additive interaction, but the actual % observed in samples treated with UNC Mer TKI combined with ADP/P2Y inhibitors was 64.3 +/- 8.9%, a statistically significant difference (p = 0.02), suggesting a synergistic effect of the combination therapy. Conclusion A novel Mer-selective SMI mediated inhibition of platelet aggregation and protection from arterial and venous thrombosis in a manner comparable to that seen with known ADP/P2Y inhibitors. Additionally, the two drugs mediate synergistic inhibition of platelet aggregation when used in combination. This observation suggests that combination therapies consisting of a Mer-inhibitor and an ADP/P2Y inhibitor could allow for dose reduction of one or both agents, thereby decreasing off-target effects and/or bleeding complications. Disclosures: Branchford: University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Sather:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. DeRyckere:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Zhang:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Earp:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Frye:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Graham:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Di Paola:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2296-2296
Author(s):  
Gilbert Acevedo ◽  
Brian R. Branchford ◽  
Christine Brzezinski ◽  
Susan Sather ◽  
Gary Brodsky ◽  
...  

Abstract Background Growth Arrest Specific gene 6 (Gas6) is a ligand for the Tyro3/Axl/Mer (TAM) family of receptor tyrosine kinases found on the surface of platelets. Previous studies have shown that stimulation of these receptors results in amplification of platelet activation and thrombus stabilization via activation of phosphatidylinositol-3-kinase (PI3K) and Akt, leading to phosphorylation of the β3 integrin. Previous work (from our lab and others) demonstrated that inhibition of the Gas6/TAM pathway results in impaired platelet aggregation, reduced aggregate stability, and decreased platelet spreading. Additionally, knockout mice deficient in the receptor or ligand are protected from venous and arterial thrombosis, but retain normal tail bleeding times. Here, we describe development and characterization of novel Mer-selective small molecule inhibitors (SMIs) for thrombosis applications. Objectives To determine if Mer-selective SMIs can inhibit platelet aggregation and protect mice from thrombosis using in vitro and in vivo models Methods We used aggregometry and in vivo murine models of arterial and venous thrombosis to compare two Mer-selective SMIs (UNC Mer TKI1 and UNC Mer TKI2) and determine the most effective inhibitor of platelet aggregation and thrombus formation. The inhibitory effect of two doses (1µM and 5 µM) of the compounds were determined using standard light-transmission aggregometry after a 30 minute incubation with washed human platelets at 37 ¢ªC and compared to platelets treated with vehicle control or with a TKI control (UNC TKI Null), a SMI with similar structure but minimal anti-TAM activity. Both collagen/epinephrine-induced systemic venous thrombosis and FeCl3-induced carotid artery injury models were used to determine effects on thrombosis mediated by UNC TKIs. Wild type C57Bl/6 mice were treated with one of the two inhibitors and compared to mice treated with vehicle control. Mean values +/- SEM are shown and statistical significance (p<0.05) was determined using the student’s paired t-test. Results UNC Mer TKI1 exhibited more potent inhibition of platelet aggregation in vitro relative to UNC Mer TKI2, although both compounds mediated dose-dependent effects. At a concentration of 1uM, the maximum percent aggregation in UNC Mer TKI1-treated samples (n=7) was significantly greater than samples treated with UNC TKI Null (n=7), 20% DMSO vehicle (n=7), or UNC TKI2 (n=7), with mean values of 69 +/- 2.2%, 76.7 +/-1.8% (p<0.01), 76.9 +/- 2.1% (p=0.001), and 77 +/- 1.8% (p<0.001), respectively. At a concentration of 5 µM, UNC Mer TKI1-treated samples (n=7) exhibited a mean maximum percent aggregation of 23.7 +/- 2.4% compared to 50.4 +/- 4.8% for samples treated with UNC Mer TKI2 (n=7, p<0.001). UNC Mer TKIs also mediated protection from thrombus formation in mice. Following FeCl3 injury to the carotid artery, vehicle-treated mice (n=11) developed stable vessel occlusions with a mean time of 6.77 +/- 0.25 min. In contrast, stable occlusion occurred at a mean time of 46.6 +/- 7.72 min (n=9, p=0.001) for UNC Mer TKI1-treated mice. Survival times following venous injection of collagen and epinephrine were also significantly increased in mice treated with either UNC Mer TKI relative to the UNC TKI Null or vehicle controls. Mice pre-treated with UNC Mer TKI1 (n=9, p=0.04 compared to vehicle alone) or UNC Mer TKI2 (n=9, p=0.03 compared to vehicle alone) survived for 19.84 +/- 4.4 and 21.25 +/- 4.65 minutes, respectively. In contrast, mice given UNC TKI Null (n=3) or vehicle (n=21), only survived for 3.21 +/- 2.4 min and 3.09 +/- 0.22 minutes, respectively. Conclusion UNC Mer TKIs mediate dose-dependent inhibition of platelet aggregation and protect mice from arterial and venous thrombosis. Their pronounced activity compared to an inactive scaffold protein with minimal anti-TAM activity suggest that Gas6/TAM pathway inhibition is the mechanism of action for these novel compounds. UNC Mer TKI1 has more potent anti-thrombotic properties than UNC Mer TKI2. Disclosures: Branchford: University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Sather:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. DeRyckere:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Zhang:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Liu:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Earp:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Wang:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Frye:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Graham:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties. Di Paola:University of Colorado: inventor on a patent application relevant to this work , inventor on a patent application relevant to this work Patents & Royalties.


Author(s):  
Layla Van Doren ◽  
Nga Nguyen ◽  
Christopher Garzia ◽  
Elizabeth Fletcher ◽  
Ryan Stevenson ◽  
...  

Objective: 12-LOX (12-lipoxygenase) produces a number of bioactive lipids including 12(S)-HETE that are involved in inflammation and platelet reactivity. The GPR31 (G-protein–coupled receptor 31) is the proposed receptor of 12(S)-HETE; however, it is not known whether the 12(S)-HETE-GPR31 signaling axis serves to enhance or inhibit platelet activity. Approach and Results: Using pepducin technology and biochemical approaches, we provide evidence that 12(S)-HETE-GPR31 signals through Gi to enhance PAR (protease-activated receptor)-4–mediated platelet activation and arterial thrombosis using both human platelets and mouse carotid artery injury models. 12(S)-HETE suppressed AC (adenylyl cyclase) activity through GPR31 and resulted in Rap1 and p38 activation and low but detectable calcium flux but did not induce platelet aggregation. A GPR31 third intracellular (i3) loop–derived pepducin, GPR310 (G-protein–coupled receptor 310), significantly inhibited platelet aggregation in response to thrombin, collagen, and PAR4 agonist, AYPGKF, in human and mouse platelets but relative sparing of PAR1 agonist SFLLRN in human platelets. GPR310 treatment gave a highly significant 80% protection ( P =0.0018) against ferric chloride–induced carotid artery injury in mice by extending occlusion time, without any effect on tail bleeding. PAR4-mediated dense granule secretion and calcium flux were both attenuated by GPR310. Consistent with these results, GPR310 inhibited 12(S)-HETE–mediated and PAR4-mediated Rap1-GTP and RASA3 translocation to the plasma membrane and attenuated PAR4-Akt and ERK activation. GPR310 caused a right shift in thrombin-mediated human platelet aggregation, comparable to the effects of inhibition of the Gi-coupled P2Y 12 receptor. Co-immunoprecipitation studies revealed that GPR31 and PAR4 form a heterodimeric complex in recombinant systems. Conclusions: The 12-LOX product 12(S)-HETE stimulates GPR31-Gi–signaling pathways, which enhance thrombin-PAR4 platelet activation and arterial thrombosis in human platelets and mouse models. Suppression of this bioactive lipid pathway, as exemplified by a GPR31 pepducin antagonist, may provide beneficial protective effects against platelet aggregation and arterial thrombosis with minimal effect on hemostasis.


1987 ◽  
Vol 32 (6) ◽  
pp. 167-168 ◽  
Author(s):  
B.J.Z. Danesh ◽  
A.R. Saniabadi ◽  
R.I. Russell ◽  
G.D.O. Lowe

We have compared the effects of acetyl salicylic acid (ASA, aspirin) and choline magnesium trisalicylate (CMT), a non-acetylated salicylate product, on platelet aggregation in human whole blood ex-vivo. Using a whole blood platelet counter, platelet aggregation was quantified by measuring the fall in the number of single platelets at peak aggregation in response to collagen, arachidonic acid (AA), as well as spontaneous aggregation. In double blind and random order, 12 healthy volunteers received, on two separate occasions 10 days apart, a single oral dose of 652 mg ASA or 655 mg CMT. Despite a comparable absorption of salicylic acid from the two drugs, ingestion of ASA resulted in a marked inhibition of platelet aggregation induced by collagen (p<0.005), AA (p<0.01) and spontaneous aggregation (p<0.01), whereas such effects were not observed after CMT ingestion. We suggest that CMT may have therapeutic potential as an alternative to aspirin when inhibition of platelet aggregation can induce bleeding complications.


1987 ◽  
Author(s):  
B J Z Danesh ◽  
A R Saniabadi ◽  
R I Russell ◽  
G D O Lowe ◽  
C D Forbes

Suppression of platelet aggregation by ASA limits the therapeutic use of this drug as an analgesic in patients with bleeding tendencies. CMT is a non-acetylated salicylate derivative with analgesic and anti-inflammatory effect similar to that of ASA. We compared platelet aggregation in human whole blood ex-vivo, three hours after ingestion of ASA and. CMT. Using a whole blood platelet counter, platelet aggregation was quantified by measuring the fall in the number of single platelets at peak aggregation in response to collagen (lμg/ml) arachidonic acid (AA, 0.5 mM) as well as spontaneous aggregation. In double blind and random order, 12 healthy volunteers received a single oral dose of ASA and CMT containing 500 mg equivalent salicylate, on two separate occasions, 10 days apart. Despite a comparable absorption of salicylic acid from the two drugs, ingestion of ASA resulted in a marked inhibition of platelet aggregation induced by collagen, AA and spontaneous aggregation, whereas such effects were not observed after CMT ingestion.We suggest that CMT may have therapeutic potential as an alternative to aspirin when inhibition of platelet aggregation can induce bleeding complications.


1990 ◽  
Vol 67 (4) ◽  
pp. 941-947 ◽  
Author(s):  
B Adelman ◽  
C Gennings ◽  
J Strony ◽  
E Hanners

1991 ◽  
Vol 66 (03) ◽  
pp. 355-360 ◽  
Author(s):  
Harve C Wilson ◽  
William Coffman ◽  
Anne L Killam ◽  
Marlene L Cohen

SummaryThe present study was designed to evaluate the effectiveness of the ergoline 5HT2 receptor antagonist, LY53857 in a rabbit model of vascular arterial occlusion. LY53857 (1 and 10 εM) inhibited serotonin amplified platelet aggregation responses to threshold concentrations of ADP in rabbit platelets in vitro. LY53857 (1 εM) not only inhibited the serotonin component of rabbit platelet aggregation, but also inhibited in vitro aggregation induced by ADP (48.7 ± 16.7% inhibition), collagen (76.1 ± 15.9% inhibition) and U46619 (65.2 ± 12.3% inhibition). The effectiveness of this ergoline 5HT2 receptor antagonist in blocking aggregation to ADP, collagen and U46619 may be related to its ability to inhibit a serotonin component of platelet aggregation since rabbit platelets possess high concentrations of serotonin that may be released during aggregation produced by other agents. Based on the effectiveness of LY53857 to inhibit rabbit platelet aggregation, we explored the ability of LY53857 to extend the time to carotid artery occlusion in rabbits following electrical stimulation of the artery. Reproducible carotid artery occlusion was induced in rabbits by moderate stenosis coupled to arterial cross clamping, followed by electrical stimulation. With this procedure, occlusion occurred at 47.0 ± 7 min (n = 30) after initiation of the electrical stimulation. Animals pretreated with LY53857 (50 to 500 εg/kg i.v.) showed a delay in the time to carotid artery occlusion (at 100 εg/kg i.v. occlusion time extended to 164 ± 16 min). Furthermore, ex vivo platelet aggregation from animals treated with LY53857 (300 εg/kg i.v.) resulted in 40.5% inhibition of platelet aggregation in response to the combination of ADP (1 εM) and serotonin (1 εM). These studies document the ability to obtain reproducible arterial occlusion in the rabbit and showed that intravenously administered LY53857 prolonged the time to carotid artery occlusion. Prolongation of carotid artery occlusion time was accompanied by inhibition of serotonin-amplified ADP-induced aggregation in rabbit platelets, an effect observed both in vitro and ex vivo. Thus, the rabbit is a useful model for studying the effectiveness of 5HT2 receptor antagonists in prolonging vascular occlusion induced by insult of the carotid artery.


Sign in / Sign up

Export Citation Format

Share Document