Consumption Of Viiirrag During Delayed Coagulation Process

1981 ◽  
Author(s):  
M Hada ◽  
S Ikematsu ◽  
M Fujimaki ◽  
K Fukutake

Coagulational functions of VIIIR:AG are still unknown, while its serological significance in laboratory medicine has been established. In this study it will be analyzed the mechanism of the consumption of VIIIR:AG during the process of prolonged blood coagulation.Quantitative assay of VIIIR:AG in plasma and serum is measured by Laurell’s method using 1% agarose with anti-F.VIII rabbit serum and qualitative assay of VIIIR:AG is performed by crossed-immunoelectrophoresis with anti F.VIII rabbit serum.In 28 patients with Hemophilia A a negative relationship between serum VIIIR:AG / plasma VIIIR:AG ratio and prolongation of PTT is estimated and also a positive relationship between serum VIIIR:AG / plasma VIIIR:AG ratio and serum VIII:vW / plasma VIII:vW ratio is obtained. However, serum VIIIR:AG shows the similar quantity to plasma VIIIR:AG in the cases within normal clotting time, which have been treated by the in vitro addition of thrombin or by transfusion of AHG in vivo. When heparin or synthetic antithrombin ( MCI-9038 ) is added into normal whole blood or F.XIII deficient whole blood, the case of normal whole blood shows delayed clotting time and decrease of serum VIIIR:AG, but the case of F.XIII deficient whole blood indicates no decrease of serum VIIIR:AG. Furthermore, in those conditions change of the concentration of CIg in serum, which has been pointed out with fibrin crosslinking, indicate the similar behavior as serum VIIIR:AG.The results obtained above might suggest that serum VIIIR:AG tend to decrease in the cases with prolonged clotting time, and the F.XIII activity might be involved in the consumption mechanism of serum VIIIR:AG in such an abnormal condition, as fibrin crosslinkage could not carry out properly.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 5129-5129
Author(s):  
Jawed Fareed ◽  
Debra Hoppensteadt ◽  
Omer Iqbal ◽  
Jeanine M. Walenga ◽  
Bruce E Lewis

Abstract Abstract 5129 Several generic versions of argatroban) (Mitsubishi; Tokyo, Japan) have been introduced in Japan (Argaron, Gartban, Slovastan). In addition, other generic versions of argatroban are being considered by the European and North American regulatory bodies. While the generic versions of argatroban exhibit similar antithrombin potency (Ki values), because of the differential compositional variations their anticoagulant effects in whole blood systems may differ due to their cellular and plasmatic protein interactions. Branded and generic versions of argatroban may exhibit differential anticoagulant actions in the whole blood and plasma based assays due to their differential interactions with blood cells, platelets and plasma proteins. Three generic versions of argatroban that are commercially available in Japan namely Argaron, Gartban and Slovastan and a powdered version of generic argatroban (Lundbeck) were compared with the branded argatroban. Native whole blood thrombelastographic (TEG) analysis was carried out at 0.1 ug/mL, the Activated Clotting Time (ACT) assay was carried out in a concentration range of 0–10 ug/mL, and such coagulation tests as the PT/INR, aPTT, Heptest, and calcium thrombin time were performed. Plasma retrieved from the supplemented whole blood was also assayed. Ratios of the clotting time test values from whole blood and plasma were calculated. Retrieved plasma samples were also assayed in the thrombin generation assays (TGA). All of the different versions of argatroban produced a concentration dependent anticoagulant effect in the native whole blood TEG and ACT. In the TEG, while argatroban and Slovastan showed a similar effect, Gartban, Argaron and a powdered generic showed weaker effects. Argatroban was also different in the ACT assay. At a concentration of 5 ug/ml the ACTs were, Arg 340+15.2 secs, S 297+10.5 secs, G 292.0+19.1 secs and A 285.2+21.7 secs. In the citrated whole blood systems, all agents produced a concentration dependent anticoagulant effect; however, the generic versions produced a stronger anticoagulant effect in comparison to branded argatroban (p<0.001). In the PT assay at 5 ug/mL, argatroban showed 32 ± 3 sec vs 40–50 sec for the generic products. Similarly in the aPTT, Heptest and thrombin time tests argatroban was weaker than the generic products. Differences among generic versions were also evident. Similar results were obtained in the retrieved plasma, however the ratio of whole blood over plasma varied from product to product. The IC50 of the generic and branded argatrobans in the TGA were also different. These results show that while in the thrombin inhibition assays generic and branded argatroban may show similar effects, these agents exhibit assay dependent differences in the whole blood and plasma based assays. Such differences may be more evident in the in vivo studirs where endothelial cells and other interactions may contribute to product individuality. Therefore, based on the in vitro antiprotease assays, generic argatrobans may not be considered equivalent and require a multi-parametric study. Currently available generic argatrobans may not be equivalent in the in vivo anticoagulant effects. Therefore, clinical validation of the clinical equivalence for these drugs is warranted. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3382-3382
Author(s):  
Junka Haku ◽  
Kenichi Ogiwara ◽  
Tomoko Matsumoto ◽  
Koji Yada ◽  
Midori Shima ◽  
...  

Abstract Abstract 3382 There is no optimal assay to monitor the hemostatic effect of bypassing therapy for hemophilia A patients (HA) with inhibitor. Clot waveform analysis (CWA) is a convenient method of assessing global clotting function based on the continuous monitoring of light transmittance or absorbance during routine coagulation tests such as aPTT. We attempted to optimize the aPTT based CWA for hemostatic monitoring during the bypassing therapy in patients of HA with inhibitor. An automated laboratory system, MDA-II® (Trinity Biotech) was used for CWA. Clot waveform was plotted from transmittance change during the clot formation. By the first and second derivation of the waveform, parameters such as clotting time, the maximum velocity (|min1|) and acceleration (|min2|) were calculated. Three trigger reagents were used; [R1] ellagic acid (E) and phospholipid (PL) that is used as an aPTT reagent, [R2] tissue factor (TF, Innovin®) and PL as a 17,000-diluted PT reagent, [R3] the mixed condition of E, TF and PL, based on our recent report on thrombin generation test optimized for HA (Matsumoto, IJH, 2009). Results were expressed as a percentage (% of control) relative to control pooled plasma. The great difference in all parameters between factor (F)VIII-deficient and control plasma resulted in R1>R3>>R2 in order (|min2| in R1, R2, R3 were 13, 119, 21% of control, respectively). Since significant difference between both plasma was not observed, we excluded R2. By the addition of clinically therapeutic concentration of recombinant FVIIa (rFVIIa, NovoSeven®) (25 nM), parameters in R3 were more improved than R1 (|min2| in R1, R3 were 18, 47% of control, respectively). Similar results were observed by the addition of activated prothrombin complex concentrates (APCC, FEIBA®) (1 U/ml) (|min2| in R1, R3 was 32, 71% of control, respectively). According to these in vitro experiments, R3 (E/TF/PL) was regarded as most optimal reagent. Next, in order to confirm the usefulness of the R3-CWA system in vivo, six patients with HA with inhibitor to whom rFVIIa (n=6, 37 (12–59) BU/ml, 105 (91–175) ƒÊg/kg dosage) or APCC (n=3, 25 (12–49) BU/ml, 91 (91–100) U/kg dosage) was administrated, were evaluated. All patients showed the clinical hemostatic efficacy by each bypassing agent. As shown in Table 2 and Fig. 1, the hemostatic effects by bypassing agents were confirmed by improvement in all R3-CWA parameters. APTT based CWA system should be promising method for quantitative monitoring during the bypassing therapy with routine automated clotting machine and only with the modified reagents such as well-balanced mixtures of E, TF and PL. Table 1. Parameters of CWA using three reagents in FVIII-deficient plasma with bypassing agents in vitro Parameter Reagent R1 R2 R3 Clotting time % of control     FVIII-def plasma 372 86 147     +rFVIIa (25 nM) 271 – 100     +APCC (1 U/ml) 122 – 69 |min1| % of control     FVIII-def plasma 16 90 33     +rFVIIa (25 nM) 20 – 54     +APCC (1 U/ml) 27 – 69 |min2| % of control     FVIII-def plasma 13 119 21     +rFVIIa (25 nM) 18 – 47     +APCC (1 U/ml) 32 – 71 Values are shown as median of triplicate. Table 2. Effect of bypassing agents in vivo on parameters of R3-triggered CWA Parameter rFVIIa (n=6) APCC (n=3) Pre Post Pre Post % of control % of control Clotting time 202 103 135 99 (129–268) (91–122) (127–159) (89–104) |min1| 37 56 41 60 (19–48) (40–78) (30–42) (46–75) |min2| 19 53 23 41 (13–32) (44–75) (16–24) (34–60) Values are shown as median (range). Fig. 1 Representative data of R3-triggered CWA in a case of HA with inhibitor on bypassing therapy using rFVIIa (dotted line) or APCC (solid line). Fig. 1. Representative data of R3-triggered CWA in a case of HA with inhibitor on bypassing therapy using rFVIIa (dotted line) or APCC (solid line). Disclosures: Shima: Chugai Pharmaceutical Co., Ltd.: Consultancy, Honoraria, Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 544-544 ◽  
Author(s):  
Emily K. Waters ◽  
Jeffrey C. Kurz ◽  
Ryan Genga ◽  
Jennifer A Nelson ◽  
Kathleen E. McGinness ◽  
...  

Abstract Abstract 544 Hemophilia A is a bleeding disorder characterized by a deficiency in coagulation factor VIII (FVIII) rendering the body incapable of maintaining hemostasis. Abnormally prolonged bleeding can occur either spontaneously or after an injury or surgery. The most effective treatment for hemophilia A is FVIII replacement therapy; however, for patients with FVIII inhibitors, this therapy is not possible. In these patients, expensive bypassing agents such as recombinant factor VIIa (rFVIIa) are the only currently availabel treatment. We have developed an aptamer that binds to and inhibits tissue factor pathway inhibitor (TFPI) as a novel therapeutic strategy to treat hemophilia A patients. TFPI inhibits factor Xa and is the primary regulator of the FVIIa:tissue factor complex, the initiator of blood coagulation. By blocking TFPI, sufficient thrombin could be generated through the extrinsic pathway to bypass the defect in clot propagation caused by the deficiency of FVIII. We compared our anti-TFPI aptamer to rFVIIa in a number of in vitro and in vivo assays, including a plasma-based thrombin generation assay–the calibrated automated thrombogram (CAT) assay–initiated with 1 pM tissue factor. These experiments were carried out with normal plasma from healthy volunteers, plasma from severe hemophilia A patients, and plasma from hemophilia A patients containing high titers of inhibitory antibodies. We also measured activity using tissue factor-initiated thromboelastography (TEG), in whole blood from healthy volunteers depleted of FVIII by preincubation with a polyclonal anti-FVIII antibody. In the CAT assay, the aptamer demonstrated a dose-dependent improvement in both endogenous thrombin potential (ETP) and peak thrombin similar to that achieved with rFVIIa in hemophilia A plasma, either with or without inhibitors. In the TEG assay, R-values–a measure of clotting time–were prolonged in the whole blood upon FVIII antibody treatment, and then restored in a dose-dependent manner with both the aptamer and rFVIIa, to levels similar to untreated whole blood. The rate of clot development, measured by the TEG angle, was also improved in a dose-dependent manner in response to both aptamer and rFVIIa. We also tested the effectiveness of our aptamer in a non-human primate model of hemophilia A that mimics the inhibitor patient. In this model, cynomolgus monkeys are treated with a single intravenous (IV) bolus of a purified polyclonal antibody against human FVIII. The antibody treatment was followed by an IV bolus of aptamer, rFVIIa, or saline. The effects of these agents on hemostasis were analyzed by TEG in plasma obtained from blood samples drawn at various times prior to or following treatment. Other measures included prothrombin time (PT), activated partial thromboplastin time (aPTT) and FVIII activity. Upon treatment with the FVIII antibody, FVIII activity in the monkey plasma was reduced to less than 1%. The administration of the polyclonal antibody had no effect on the PT of the monkey plasma, but did prolong the aPTT. In the TEG, 2.5 hours after administration of the antibody, there was a prolongation in clotting time (R-value) from a baseline R-value of 5-10 minutes to an R-value of ≥ 25 minutes. Within 15 minutes after IV bolus administration of either aptamer or rFVIIa, there was an improvement in the TEG R-value to levels similar to baseline, pre-study measurements. Infusion of rFVIIa subsequently shortened the PT by 2 seconds and shortened the aPTT by 20 seconds. These changes were not observed with aptamer infusion. These in vitro and in vivo studies suggest we have developed a TFPI inhibitory aptamer that could be a novel procoagulant therapeutic in the treatment of inhibitor and non-inhibitor hemophilia A patients. Disclosures: Waters: Archemix Corporation: Employment. Kurz:Archemix Corporation: Employment. Genga:Archemix Corporation: Employment. Nelson:Archemix Corporation: Employment. McGinness:Archemix Corporation: Employment. Schaub:Archemix Corporation: Employment.


2018 ◽  
Vol 20 (1) ◽  
pp. 79-86 ◽  
Author(s):  
Sheng Zhang ◽  
Amanda R Guido ◽  
Richard G Jones ◽  
Benjamin J Curry ◽  
Angela S Burke ◽  
...  

Purpose: The use of cyanoacrylate for intravenous catheter securement is of interest to clinicians and patients, because of the superior adhesive strength and hemostatic effect of cyanoacrylate compared to current securement devices. The purpose of this study is to use novel in vitro and in vivo testing methods to analyze the hemostatic effect of a catheter securement cyanoacrylate (cyanoacrylate). Methods: An unprecedented in vitro method was performed to determine the effects of a cyanoacrylate on a customized modified activated clotting time assay and blood flow inhibition assay by exposing blood or plasma to either one or three drops of cyanoacrylate. For the in vivo testing, full-thickness incisions were made on swine, and the bleeding was scored prior to treatment and at 3, 6, 9, and 12 min after treatment. Results: The cyanoacrylate rapidly achieved hemostasis in the presence of anticoagulated whole blood, platelet-poor plasma, and non-anticoagulated whole blood, in vitro. The cyanoacrylate achieved hemostasis 12-fold faster than thromboplastin in the modified activated clotting time assay. The cyanoacrylate does not alter normal blood clotting, as measured by prothrombin time. In vivo, the bleeding score of cyanoacrylate prior to treatment and at 3, 6, 9, and 12 min after treatment were 2.3 ± 1.0, 0.3 ± 0.5, 0.2 ± 0.5, 0.2 ± 0.4, and 0.2 ± 0.4, respectively. Conclusion: This study indicates that cyanoacrylate demonstrates a potent mechanical hemostatic effect and cyanoacrylate in the presence of anticoagulated whole blood has an activated clotting time that is 12 times quicker than thromboplastin. The cyanoacrylate was found to be significantly equivalent to two known hemostatic agents, in vivo.


1979 ◽  
Vol 42 (04) ◽  
pp. 1248-1260 ◽  
Author(s):  
Lyle F Mockros ◽  
Samuel D Hirsch ◽  
Leon Zuckerman ◽  
Joseph A Caprini ◽  
William P Robinson ◽  
...  

SummaryBolus injections of beef-lung heparin at doses of 50, 100 and 200 u/kg body weight were administered to mongrel dogs. Neutralization of the anticoagulant effect was evaluated using sequential samples withdrawn from the animals (in vivo samples) and aliquots from a 100 ml sample withdrawn from the dog at 30 minutes post-injection (in vitro samples). Tests of the activated partial thromboplastin time (APTT) and prothrombin time (PT) did not indicate the degree of anticoagulation. Tests of the whole blood clotting time (WBCT), celite- activated whole blood clotting time (ACT), and celite-activated thromboelastography (ATEG) indicated pronounced hypocoagulability immediately after the injection, followed by a fairly rapid decay in anticoagulability, and a slight Ziype/coagulability at three to four hours post injection. The results from the in vitro ATEG samples were essentially identical to those on the in vivo samples, whereas the in vitro WBCT and ACT generally indicated higher degrees of anticoagulation. Calculated half-lives of the anticoagulant effect are significantly shorter than previously reported, being 18 to 36 minutes, and slightly dose dependent. The decay of the effects, however, does not appear to follow a single exponential curve, dropping very rapidly immediately post-injection and at a somewhat slower rate 60 or more minutes post-injection.


1990 ◽  
Vol 63 (02) ◽  
pp. 220-223 ◽  
Author(s):  
J Hauptmann ◽  
B Kaiser ◽  
G Nowak ◽  
J Stürzebecher ◽  
F Markwardt

SummaryThe anticoagulant effect of selected synthetic inhibitors of thrombin and factor Xa was studied in vitro in commonly used clotting assays. The concentrations of the compounds doubling the clotting time in the various assays were mainly dependent on their thrombin inhibitory activity. Factor Xa inhibitors were somewhat more effective in prolonging the prothrombin time compared to the activated partial thromboplastin time, whereas the opposite was true of thrombin inhibitors.In vivo, in a venous stasis thrombosis model and a thromboplastin-induced microthrombosis model in rats the thrombin inhibitors were effective antithrombotically whereas factor Xa inhibitors of numerically similar IQ value for the respective enzyme were not effective at equimolar dosageThe results are discussed in the light of the different prelequisiles and conditions for inhibition of thrombin and factor Xa in the course of blood clotting.


1997 ◽  
Vol 77 (05) ◽  
pp. 0920-0925 ◽  
Author(s):  
Bernd Pötzsch ◽  
Katharina Madlener ◽  
Christoph Seelig ◽  
Christian F Riess ◽  
Andreas Greinacher ◽  
...  

SummaryThe use of recombinant ® hirudin as an anticoagulant in performing extracorporeal circulation systems including cardiopulmonary bypass (CPB) devices requires a specific and easy to handle monitoring system. The usefulness of the celite-induced activated clotting time (ACT) and the activated partial thromboplastin time (APTT) for r-hirudin monitoring has been tested on ex vivo blood samples obtained from eight patients treated with r-hirudin during open heart surgery. The very poor relationship between the prolongation of the ACT and APTT values and the concentration of r-hirudin as measured using a chromogenic factor Ila assay indicates that both assays are not suitable to monitor r-hirudin anticoagulation. As an alternative approach a whole blood clotting assay based on the prothrombin-activating snake venom ecarin has been tested. In vitro experiments using r-hirudin- spiked whole blood samples showed a linear relationship between the concentration of hirudin added and the prolongation of the clotting times up to a concentration of r-hirudin of 4.0 µg/ml. Interassay coefficients (CV) of variation between 2.1% and 5.4% demonstrate the accuracy of the ecarin clotting time (ECT) assay. Differences in the interindividual responsiveness to r-hirudin were analyzed on r-hirudin- spiked blood samples obtained from 50 healthy blood donors. CV- values between 1.8% and 6% measured at r-hirudin concentrations between 0.5 and 4 µg/ml indicate remarkably slight differences in r-hirudin responsiveness. ECT assay results of the ex vivo blood samples linearily correlate (r = 0.79) to the concentration of r-hirudin. Moreover, assay results were not influenced by treatment with aprotinin or heparin. These findings together with the short measuring time with less than 120 seconds warrant the whole blood ECT to be a suitable assay for monitoring of r-hirudin anticoagulation in cardiac surgery.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Michele Dei Cas ◽  
Jessica Rizzo ◽  
Mariangela Scavone ◽  
Eti Femia ◽  
Gian Marco Podda ◽  
...  

AbstractLow-dose aspirin (ASA) is used to prevent cardiovascular events. The most commonly used formulation is enteric-coated ASA (EC-ASA) that may be absorbed more slowly and less efficiently in some patients. To uncover these “non-responders” patients, the availability of proper analytical methods is pivotal in order to study the pharmacodynamics, the pharmacokinetics and the metabolic fate of ASA. We validated a high-throughput, isocratic reversed-phase, negative MRM, LC–MS/MS method useful for measuring circulating ASA and salicylic acid (SA) in blood and plasma. ASA-d4 and SA-d4 were used as internal standards. The method was applied to evaluate: (a) the "in vitro" ASA degradation by esterases in whole blood and plasma, as a function of time and concentration; (b) the "in vivo" kinetics of ASA and SA after 7 days of oral administration of EC-ASA or plain-ASA (100 mg) in healthy volunteers (three men and three women, 37–63 years). Parameters of esterases activity were Vmax 6.5 ± 1.9 and Km 147.5 ± 64.4 in plasma, and Vmax 108.1 ± 20.8 and Km 803.2 ± 170.7 in whole blood. After oral administration of the two formulations, tmax varied between 3 and 6 h for EC-ASA and between 0.5 and 1.0 h for plain-ASA. Higher between-subjects variability was seen after EC-ASA, and one subject had a delayed absorption over eight hours. Plasma AUC was 725.5 (89.8–1222) for EC-ASA, and 823.1(624–1196) ng h/mL (median, 25–75% CI) for plain ASA. After the weekly treatment, serum levels of TxB2 were very low (< 10 ng/mL at 24 h from the drug intake) in all the studied subjects, regardless of the formulation or the tmax. This method proved to be suitable for studies on aspirin responsiveness.


Transfusion ◽  
2021 ◽  
Vol 61 (S1) ◽  
Author(s):  
Turid Helen Felli Lunde ◽  
Lindsay Hartson ◽  
Shawn Lawrence Bailey ◽  
Tor Audun Hervig
Keyword(s):  

2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 1347.2-1347
Author(s):  
S. Y. Ki ◽  
H. Shin ◽  
Y. Lee ◽  
H. R. Bak ◽  
H. Yu ◽  
...  

Background:Janus kinases (JAK1, JAK2, JAK3, and TYK2) play critical roles in mediating various cytokine signaling, and has been developed as a target for autoimmune diseases such as RA. Tofacitinib, oral Pan-JAK inhibitor, demonstrated efficacy in RA patients, but its widespread use is limited by safety issues. Baricitinib, JAK1/2 inhibitor, is also known to interfere with the hematopoiesis system, such as anemia and thrombocytopenia associated with suppression of JAK2 signals. Therefore, it is necessary to develop a new potent compound that selectively inhibits JAK1 over JAK2, 3Objectives:To identify the pharmacological characteristic based on efficacy of CJ-15314 as potent and selective JAK1 inhibitor for treatment of autoimmune disease.Methods:In vitro, cell-based, kinase panel, Kd value and human whole blood assay were performed to determine the inhibition potency and selectivity for JAK subfamily kinases. In vivo therapeutic potential was evaluated by RA model including rat Adjuvant-Induced Arthritis (AIA) and collagen-induced arthritic (CIA). To confirm the possibility of further expansion into the autoimmune disease, BioMAP® Diversity PLUS® Panel was performed by discoverX.Results:In vitro assay, CJ-15314 inhibited JAK kinase family in a concentration-dependent manner with IC50 values of 3.8 nM against JAK1, Selectivity for JAK1 over JAK2, 3 was approximately 18, 83 fold greater for CJ-15314. In 1mM ATP condition, CJ-15314 has been confirmed to have the highest JAK1 selectivity over competing drugs, under 1 mM ATP condition that reflects the physiological environment in the body. Similarly, Kd values has also confirmed the selectivity of JAK1, which is 10 fold higher than JAK2, 3. Accordingly, in human whole blood assays, CJ-15314 is 11 fold more potent against IL-6 induced pSTAT1 inhibition through JAK1 (IC50 value: 70 nM) than GM-CSF-induced pSTAT5 inhibition (JAK2) whereas baricitinib and filgotinib exhibited only 2 fold and 7 fold respectively.In vivo efficacy model, CJ-15314 inhibited disease severity scores in a dose dependent manner. In the rat AIA model, CJ-15314 at 30 mg/kg dose showed 95.3% decrease in arthritis activity score, 51.2% in figotinib at 30 mg/kg, 97.7% showed baricitinib at 10 mg/kg. CJ-15314 showed superior anti-arthritic efficacy than filgotinib. CJ-15314 also minimally affected anemia-related parameters but not bricitinib end of the 2-week treatment. In the rat CIA model, like 10 mg/kg of bricitinib, 30 mg/kg of CJ-15314 also has a similar effect, with a significant reduction in histopathological scores.In biomap diversity panel, CJ-15314 inhibited the expression of genes such as MCP-1, VCAM-1, IP-10, IL-8, IL-1, sTNF-α and HLA-DR confirming the possibility of expansion into other diseases beyond arthritis.Conclusion:CJ-15314 is a highly selective JAK1 inhibitor, demonstrates robust efficacy in RA animal model and is good candidate for further development for inflammatory diseases.* CJ-15314 is currently conducting a phase I trial in south Korea.References:[1]Clark JD et al. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014; 57(12):5023-38.[2]Burmester GR et al. Emerging cell and cytokine targets in rheumatoid arthritis. Nat Rev Rheumatol. 2014; 10(2):77-88[3]Jean-Baptiste Telliez et al. Discovery of a JAK3-selective inhibitor: functional differentiation of JAK3-selective inhibition over pan-JAK or JAK1-selective inhibition. ACS Chem. Biol., 2016; 11 (12):3442-3451Disclosure of Interests:so young Ki Employee of: CJ healthcare, hyunwoo shin Employee of: CJ healthcare, yelim lee Employee of: CJ healthcare, Hyoung rok Bak Employee of: CJ healthcare, hana yu Employee of: CJ healthcare, Seung Chan Kim Employee of: CJ healthcare, juhyun lee Employee of: CJ healthcare, donghyun kim Employee of: CJ healthcare, Dong-hyun Ko Employee of: CJ Healthcare, dongkyu kim Employee of: CJ healthcare


Sign in / Sign up

Export Citation Format

Share Document