1
|
Mazzeffi M, Tanaka KA, Gurbel PA, Tantry US, Levy JH. Platelet P2Y12 Receptor Inhibition and Perioperative Patient Management. Anesthesiology 2025; 142:202-216. [PMID: 39392789 DOI: 10.1097/aln.0000000000005148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Affiliation(s)
- Michael Mazzeffi
- University of Virginia School of Medicine, Department of Anesthesiology, Charlottesville, Virgina
| | - Kenichi A Tanaka
- Oklahoma University School of Medicine, Department of Anesthesiology, Oklahoma City, Oklahoma
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, Maryland
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research and Drug Development, Sinai Hospital of Baltimore, Baltimore, Maryland
| | - Jerrold H Levy
- Duke University School of Medicine, Department of Anesthesiology, Durham, North Carolina
| |
Collapse
|
2
|
Swieton J, Miklosz J, Bielicka N, Frackiewicz A, Depczynski K, Stolarek M, Bonarek P, Kaminski K, Rozga P, Yusa SI, Gromotowicz-Poplawska A, Szczubialka K, Pawlak D, Mogielnicki A, Kalaska B. Synthesis, Biological Evaluation and Reversal of Sulfonated Di- and Triblock Copolymers as Novel Parenteral Anticoagulants. Adv Healthc Mater 2024; 13:e2402191. [PMID: 39370656 DOI: 10.1002/adhm.202402191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Despite targeting different coagulation cascade sites, all Food and Drug Administration-approved anticoagulants present an elevated risk of bleeding, including potentially life-threatening intracranial hemorrhage. Existing studies have not thoroughly investigated the efficacy and safety of sulfonate polymers in animal models and fully elucidate the precise mechanisms by which these polymers act. The activity and safety of sulfonated di- and triblock copolymers containing poly(sodium styrenesulfonate) (PSSS), poly(sodium 2-acrylamido-2-methylpropanesulfonate) (PAMPS), poly(ethylene glycol) (PEG), poly(sodium methacrylate) (PMAAS), poly(acrylic acid) (PAA), and poly(sodium 11-acrylamidoundecanoate) (PAaU) blocks are synthesized and assessed. PSSS-based copolymers exhibit greater anticoagulant activity than PAMPS-based ones. Their activity is mainly affected by the total concentration of sulfonate groups and molecular weight. PEG-containing copolymers demonstrate a better safety profile than PAA-containing ones. The selected copolymer PEG47-PSSS32 exhibits potent anticoagulant activity in rodents after subcutaneous and intravenous administration. Heparin Binding Copolymer (HBC) completely reverses the anticoagulant activity of polymer in rat and human plasma. No interaction with platelets is observed. Selected copolymer targets mainly factor XII and fibrinogen, and to a lesser extent factors X, IX, VIII, and II, suggesting potential application in blood-contacting biomaterials for anticoagulation purposes. Further studies are needed to explore its therapeutic applications fully.
Collapse
Affiliation(s)
- Justyna Swieton
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Joanna Miklosz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Natalia Bielicka
- Department of Biopharmacy and Radiopharmacy, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Aleksandra Frackiewicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Karol Depczynski
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Marta Stolarek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, prof. S. Lojasiewicza 11 St., Krakow, 30-348, Poland
| | - Piotr Bonarek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Kamil Kaminski
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Piotr Rozga
- Drug Discovery and Early Development Department, Adamed Pharma S.A., Pienkow, Mariana Adamkiewicza 6A St., Czosnow, 05-152, Poland
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 167 Shosha, Himeji, 671-2280, Japan
| | - Anna Gromotowicz-Poplawska
- Department of Biopharmacy and Radiopharmacy, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Krzysztof Szczubialka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2 St., Krakow, 30-387, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Andrzej Mogielnicki
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| | - Bartlomiej Kalaska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C St., Bialystok, 15-089, Poland
| |
Collapse
|
3
|
Chan MV, Chen MH, Thibord F, Nkambule BB, Lachapelle AR, Grech J, Schneider ZE, Wallace de Melendez C, Huffman JE, Hayman MA, Allan HE, Armstrong PC, Warner TD, Johnson AD. Factors that modulate platelet reactivity as measured by 5 assay platforms in 3429 individuals. Res Pract Thromb Haemost 2024; 8:102406. [PMID: 38813256 PMCID: PMC11135030 DOI: 10.1016/j.rpth.2024.102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/05/2024] [Indexed: 05/31/2024] Open
Abstract
Background Assessment of platelet function is key in diagnosing bleeding disorders and evaluating antiplatelet drug efficacy. However, there is a prevailing "one-size-fits-all" approach in the interpretation of measures of platelet reactivity, with arbitrary cutoffs often derived from healthy volunteer responses. Objectives Our aim was to compare well-used platelet reactivity assays. Methods Blood and platelet-rich plasma obtained from the Framingham Heart Study (N = 3429) were assayed using a range of agonists in 5 platelet assays: light transmission aggregometry, Optimul aggregometry, Multiplate impedance aggregometry (Roche Diagnostics), Total Thrombus-Formation Analysis System, and flow cytometry. Using linear mixed-effect models, we determined the contribution of preanalytical and technical factors that modulated platelet reactivity traits. Results A strong intra-assay correlation of platelet traits was seen in all assays, particularly Multiplate velocity (r = 0.740; ristocetin vs arachidonic acid). In contrast, only moderate interassay correlations were observed (r = 0.375; adenosine diphosphate Optimul Emax vs light transmission aggregometry large area under the curve). As expected, antiplatelet drugs strongly reduced platelet responses, with aspirin use primarily targeting arachidonic acid-induced aggregation, and explained substantial variance (β = -1.735; P = 4.59 × 10-780; variance proportion = 46.2%) and P2Y12 antagonists blocking adenosine diphosphate responses (β = -1.612; P = 6.75 × 10-27; variance proportion = 2.1%). Notably, female sex and older age were associated with enhanced platelet reactivity. Fasting status and deviations from standard venipuncture practices did not alter platelet reactivity significantly. Finally, the agonist batch, phlebotomist, and assay technician (more so for assays that require additional sample manipulation) had a moderate to large effect on measured platelet reactivity. Conclusion Caution must be exercised when extrapolating findings between assays, and the use of standard ranges must be medication-specific and sex-specific at a minimum. Researchers should also consider preanalytical and technical variables when designing experiments and interpreting platelet reactivity measures.
Collapse
Affiliation(s)
- Melissa V. Chan
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Ming-Huei Chen
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Florian Thibord
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Bongani B. Nkambule
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Amber R. Lachapelle
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Joseph Grech
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Zoe E. Schneider
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | | | - Jennifer E. Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| | - Melissa A. Hayman
- Centre for Immunobiology, the Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Harriet E. Allan
- Centre for Immunobiology, the Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Paul C. Armstrong
- Centre for Immunobiology, the Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Timothy D. Warner
- Centre for Immunobiology, the Blizard Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, USA
| |
Collapse
|
4
|
Lyytinen G, Melnikov G, Brynedal A, Anesäter E, Antoniewicz L, Blomberg A, Wallén H, Bosson JA, Hedman L, Tehrani S, Lundbäck M. Use of heated tobacco products (IQOS) causes an acute increase in arterial stiffness and platelet thrombus formation. Atherosclerosis 2024; 390:117335. [PMID: 37872010 DOI: 10.1016/j.atherosclerosis.2023.117335] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/19/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND AND AIMS Heated tobacco products (HTPs) are novel alternative tobacco products being promoted as an alternative to cigarettes. To evaluate the impact of HTP use on vascular function, we investigated the effects of a brief HTP usage on arterial stiffness and platelet thrombus formation in healthy volunteers. METHODS In a randomised crossover study, twenty-four healthy young adults with occasional tobacco use smoked the HTP IQOS 3 Multi (Phillip Morris Int.) and "no-exposure" was used as a control, with a wash-out period of at least one week in-between. Arterial stiffness was assessed through pulse wave velocity and pulse wave analysis. Blood samples, collected at baseline and 5 min following exposure, were analysed with the Total-Thrombus-formation analysis system evaluating platelet and fibrin-rich thrombus formation tendency. RESULTS HTP exposure caused immediate heightened pulse wave velocity (+0.365 m/s, 95% CI: +0.188 to 0.543; p = 0.004) and enhanced augmentation index corrected to heart rate (+6.22%, 95% CI: +2.33 to 10.11; p = 0.003) compared to the no-exposure occasion. Similarly, blood pressure and heart rate transiently increased immediately following HTP inhalation. Platelet thrombus formation significantly increased following HTP exposure (area under the curve +59.5, 95% CI: +25.6 to 93.4; p < 0.001) compared to no-exposure. No effect was seen on fibrin-rich thrombus formation following HTP-exposure. CONCLUSIONS Brief HTP use in healthy young adults had immediate adverse effects on vascular function resulting in increased arterial stiffness and platelet thrombus formation, known risk factors for the development of atherosclerosis. Further research is needed to address long term health impacts.
Collapse
Affiliation(s)
- Gustaf Lyytinen
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Georgy Melnikov
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Amelie Brynedal
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Erik Anesäter
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Lukasz Antoniewicz
- Department of Medicine II, Division of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Anders Blomberg
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Håkan Wallén
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jenny A Bosson
- Dept. of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Linnea Hedman
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, The OLIN Unit, Umeå University, Umeå, Sweden
| | - Sara Tehrani
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Lundbäck
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Otsuka Y, Ishii M, Ikebe S, Nakamura T, Tsujita K, Kaikita K, Matoba T, Kohro T, Oba Y, Kabutoya T, Kario K, Imai Y, Kiyosue A, Mizuno Y, Nochioka K, Nakayama M, Iwai T, Miyamoto Y, Sato H, Akashi N, Fujita H, Nagai R. BNP level predicts bleeding event in patients with heart failure after percutaneous coronary intervention. Open Heart 2023; 10:e002489. [PMID: 38065584 PMCID: PMC10711837 DOI: 10.1136/openhrt-2023-002489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the association between heart failure (HF) severity measured based on brain natriuretic peptide (BNP) levels and future bleeding events after percutaneous coronary intervention (PCI). BACKGROUND The Academic Research Consortium for High Bleeding Risk presents a bleeding risk assessment for antithrombotic therapy in patients after PCI. HF is a risk factor for bleeding in Japanese patients. METHODS Using an electronic medical record-based database with seven tertiary hospitals in Japan, this retrospective study included 7160 patients who underwent PCI between April 2014 and March 2020 and who completed a 3-year follow-up and were divided into three groups: no HF, HF with high BNP level and HF with low BNP level. The primary outcome was bleeding events according to the Global Use of Streptokinase and t-PA for Occluded Coronary Arteries classification of moderate and severe bleeding. The secondary outcome was major adverse cardiovascular events (MACE). Furthermore, thrombogenicity was measured using the Total Thrombus-Formation Analysis System (T-TAS) in 536 consecutive patients undergoing PCI between August 2013 and March 2017 at Kumamoto University Hospital. RESULTS Multivariate Cox regression showed that HF with high BNP level was significantly associated with bleeding events, MACE and all-cause death. In the T-TAS measurement, the thrombogenicity was lower in patients with HF with high BNP levels than in those without HF and with HF with low BNP levels. CONCLUSIONS HF with high BNP level is associated with future bleeding events, suggesting that bleeding risk might differ depending on HF severity.
Collapse
Affiliation(s)
- Yasuhiro Otsuka
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - So Ikebe
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Nakamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takahide Kohro
- Department of Clinical Informatics, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yusuke Oba
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Tomoyuki Kabutoya
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yasushi Imai
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Arihiro Kiyosue
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshiko Mizuno
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Development Bank of Japan Inc, Tokyo, Japan
| | - Kotaro Nochioka
- Division of Cardiovascular Medicine, Tohoku University Hospital, Sendai, Japan
| | - Masaharu Nakayama
- Department of Medical Informatics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Takamasa Iwai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoshihiro Miyamoto
- Open Innovation Center, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Naoyuki Akashi
- Division of Cardiovascular Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Hideo Fujita
- Division of Cardiovascular Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Ryozo Nagai
- Jichi Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
6
|
Yamazaki M, Shirai Y, Ohnishi T, Hosokawa K, Dahlen JR, Kitagawa K. Differential inhibition of platelet function by cilostazol in combination with clopidogrel. Eur J Clin Pharmacol 2023; 79:1623-1630. [PMID: 37740121 DOI: 10.1007/s00228-023-03553-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/13/2023] [Indexed: 09/24/2023]
Abstract
PURPOSE To assess the antiplatelet effect of cilostazol clinically, we compared the effects of cilostazol in combination with clopidogrel on various platelet function tests. METHODS We recruited patients with ischemic stroke at high risk of recurrence who were treated with clopidogrel alone within 180 days after stroke onset. Subjects underwent baseline platelet function tests, and were then randomly assigned to receive dual antiplatelet therapy (DAPT) comprising clopidogrel and cilostazol or clopidogrel monotherapy (SAPT). After 6 months, platelet function was measured again and compared to that at baseline in each group, and the rate of change was compared between groups. RESULTS Thirty-four patients were enrolled, but 4 patients were excluded for various reasons. In total, 30 subjects (13 in DAPT and 17 in SAPT group) were analyzed. Adenosine diphosphate- and collagen-induced aggregation, VerifyNow P2Y12 reaction units, vasodilator-stimulated phosphoprotein (platelet reactivity index: PRI) and plasma p-selectin concentration were significantly lower (P = 0.004, 0.042, 0.049, 0.003 and 0.006 respectively), while VerifyNow % inhibition was significantly higher at 6 months compared to baseline (P = 0.003) in the DAPT group only. Comparison of the rate of change in each parameter from baseline to 6 months showed that while PRI decreased at a greater rate (P = 0.012), VerifyNow % inhibition increased at a greater rate (P = 0.003) in the DAPT group than the SAPT group. CONCLUSIONS The inhibitory effects of adjunctive cilostazol added to clopidogrel on platelet function differed by type of platelet function test. VerifyNow % inhibition and PRI were more inhibited than the other platelet function tests. TRIAL REGISTRATION CSPS.com substudy in TWMU (UMIN000026672), registered on April 1, 2017. This study was performed as a substudy of CSPS.com (UMIN000012180, registered on October 31, 2013) and was retrospectively registered.
Collapse
Affiliation(s)
- Masako Yamazaki
- Department of Neurology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
- Department of Artificial Intelligence Medicine Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| | - Yuka Shirai
- Department of Neurology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Tomoko Ohnishi
- Research Institute, Fujimori Kogyo Co., Ltd., 1-10-1 Sachiura, Kanazawa-ku, Yokohama, Kanagawa, 236-0003, Japan
| | - Kazuya Hosokawa
- Research Institute, Fujimori Kogyo Co., Ltd., 1-10-1 Sachiura, Kanazawa-ku, Yokohama, Kanagawa, 236-0003, Japan
| | | | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| |
Collapse
|
7
|
Omae T, Ishikawa T, Ogiwara K, Nogami K. Pediatric Mesangial Proliferative Glomerulonephritis Has Increased the Platelet Thrombus Formation Potentials under High-Shear Flow Condition. Nephron Clin Pract 2023; 148:415-425. [PMID: 37812914 DOI: 10.1159/000534494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
INTRODUCTION Blood coagulation is associated with glomerulonephritis (GN) pathophysiology. Using whole-blood-based rotational thromboelastometry, we recently reported that the degree of hypercoagulability in pediatric patients with immunoglobulin A nephropathy (IgAN), a GN, might be associated with pathological severity. To further clarify the coagulation status of mesangial proliferative GN (MesPGN), we assessed the platelet thrombus formation (PTF) under high-shear flow using a microchip-based flow chamber system (T-TAS®). METHODS Thirty-four pediatric patients definitively diagnosed with MesPGN by renal biopsy at Nara Medical University Hospital between 2015 and 2022 were enrolled, and 29 patients (case group; median age, 8.0 years) were assessed. Microchips coated with collagen (PL-chip) were used to assess PTF at high-shear in whole blood. The times to increase by 10 and 30 kPa (T10 and T30) from baseline were calculated and compared with those of the pediatric controls. Changes in the parameters during the treatment course and the relationship between pathological severity and the parameters were evaluated. RESULTS T10 and T30 parameters in the PL-chip were significantly shorter, and the area under the curves were greater in the case group than those in the control group (both p < 0.05). Each parameter was enhanced during the 3-week treatment but improved after the end of treatment. No significant relationship was observed between pathological severity and these parameters. Little PTF difference was observed between IgAN and Henoch-Schönlein purpura nephritis. CONCLUSIONS Pediatric MesPGN increased the potential for PTF under high-shear flow conditions.
Collapse
Affiliation(s)
- Takashi Omae
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
- Department of Community-Based Medicine, Nara Medical University, Kashihara, Japan
| | - Tomoaki Ishikawa
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| | - Kenichi Ogiwara
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Japan
| |
Collapse
|
8
|
Lyytinen G, Brynedal A, Anesäter E, Antoniewicz L, Blomberg A, Wallén H, Bosson JA, Hedman L, Mobarrez F, Tehrani S, Lundbäck M. Electronic Cigarette Vaping with Nicotine Causes Increased Thrombogenicity and Impaired Microvascular Function in Healthy Volunteers: A Randomised Clinical Trial. Cardiovasc Toxicol 2023; 23:255-264. [PMID: 37548804 PMCID: PMC10435650 DOI: 10.1007/s12012-023-09802-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/15/2023] [Indexed: 08/08/2023]
Abstract
Electronic cigarette (EC) vaping is increasingly popular, despite growing evidence of adverse health effects. To further evaluate the impact of EC use on vascular health, we investigated the effects of brief EC inhalation on flow-dependent thrombus formation and microcirculation in healthy volunteers. The study was performed with a randomised double-blind crossover design. Twenty-two healthy subjects aged between 18 and 45 years with occasional tobacco use were recruited. Subjects inhaled 30 puffs of EC aerosol with and without nicotine on two occasions separated by a wash-out period of at least 1 week. Blood samples were collected at baseline and at 15 and 60 min following exposure and analysed with the Total-Thrombus-formation analysis system evaluating fibrin-rich thrombus formation and platelet thrombus formation in whole blood under flow. Microvascular function was assessed at baseline and 30 min after exposure by laser speckle contrast imaging and iontophoresis of acetylcholine and sodium nitroprusside (SNP) to evaluate the endothelium-dependent and independent pathways of vasodilation. Compared with nicotine free EC aerosol, exposure to EC aerosol with nicotine significantly increased platelet thrombus formation and fibrin-rich thrombus formation at 15 min (p = 0.017 and p = 0.037, respectively) with normalisation after 60 min. Peak SNP-mediated microvascular perfusion, i.e. endothelium-independent vasodilation, was reduced following EC vaping with nicotine compared with baseline (p = 0.006). Thirty puffs of EC aerosol with nicotine increased platelet and fibrin-dependent thrombus formation and reduced microvascular dilatation capacity. No compelling effects of EC vaping without nicotine were observed, indicating nicotine as the main effector. Trial registration: ClinicalTrials.gov Identifier: NCT04175457 URL: https://clinicaltrials.gov/ct2/show/NCT04175457.
Collapse
Affiliation(s)
- Gustaf Lyytinen
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden.
| | - Amelie Brynedal
- Section of Medicine, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Erik Anesäter
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| | - Lukasz Antoniewicz
- Department of Medicine II, Division of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Anders Blomberg
- Section of Medicine, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Håkan Wallén
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| | - Jenny A Bosson
- Section of Medicine, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Linnea Hedman
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, The OLIN Unit, Umeå University, Umeå, Sweden
| | | | - Sara Tehrani
- Department of Clinical Sciences, Division of Internal Medicine, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Lundbäck
- Department of Clinical Sciences, Division of Cardiovascular Medicine, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Japanese high bleeding risk criteria status predicts low thrombogenicity and bleeding events in patients undergoing percutaneous coronary intervention. Cardiovasc Interv Ther 2023:10.1007/s12928-023-00920-3. [PMID: 36877333 DOI: 10.1007/s12928-023-00920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/13/2023] [Indexed: 03/07/2023]
Abstract
Although the Japanese high bleeding risk criteria (J-HBR) were established to predict bleeding risk in patients undergoing percutaneous coronary intervention (PCI), the thrombogenicity in the J-HBR status remains unknown. Here, we examined the relationships among J-HBR status, thrombogenicity and bleeding events. This study was a retrospective analysis of 300 consecutive patients who underwent PCI. Blood samples obtained on the day of PCI were used in the total thrombus-formation analysis system (T-TAS) to investigate the thrombus-formation area under the curve (AUC; PL18-AUC10 for platelet chip; AR10-AUC30 for atheroma chip). The J-HBR score was calculated by adding 1 point for any major criterion and 0.5 point for any minor criterion. We assigned patients to three groups based on J-HBR status: a J-HBR-negative group (n = 80), a low score J-HBR-positive group (positive/low, n = 109), and a high score J-HBR-positive group (positive/high, n = 111). The primary end point was the 1-year incidence of bleeding events defined by the Bleeding Academic Research Consortium types 2, 3, or 5. Both PL18-AUC10 and AR10-AUC30 levels were lower in the J-HBR-positive/high group than the negative group. Kaplan-Meier analysis showed worse 1-year bleeding event-free survival in the J-HBR-positive/high group compared with the negative group. In addition, both T-TAS levels in J-HBR positivity were lower in those with bleeding events than in those without bleeding events. In multivariate Cox regression analyses, the J-HBR-positive/high status was significantly associated with 1-year bleeding events. In conclusion, the J-HBR-positive/high status could reflect low thrombogenicity as measured by T-TAS and high bleeding risk in patients undergoing PCI.
Collapse
|
10
|
Fuchizaki A, Yasui K, Hayashi T, Tanaka M, Nagasato T, Ohnishi-Wada T, Hosokawa K, Fujimura Y, Shimogaki K, Hirayama F, Takihara Y, Kimura T. A novel quantitative method to evaluate the contribution of platelet products to white thrombus formation in reconstituted blood under flow conditions. Vox Sang 2023; 118:367-375. [PMID: 36862116 DOI: 10.1111/vox.13414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 03/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Currently, the quality of platelet (PLT) products is evaluated using a series of in vitro tests, which only analyse PLTs as an inspection material. However, it would be ideal to assess the physiological functions of PLTs under conditions similar to the sequential blood haemostatic process. In this study, we attempted to establish an in vitro system where the thrombogenicity of PLT products was evaluated in the presence of red blood cells (RBCs) and plasma using a microchamber under constant shear stress (600/s). MATERIALS AND METHODS Blood samples were reconstituted by mixing PLT products, standard human plasma (SHP) and standard RBCs. Each component was serially diluted keeping the other two components fixed. The samples were applied onto a flow chamber system (Total Thrombus-formation Analysis System [T-TAS]), and white thrombus formation (WTF) was assessed under large arterial shear conditions. RESULTS We observed a good correlation between the PLT numbers in the test samples and WTF. The WTF of samples containing ≦10% SHP was significantly lower than those containing ≧40% SHP, and no difference was observed in WTF among samples containing 40%-100% SHP. WTF significantly declined in the absence of RBCs, whereas no change in WTF was observed in the presence of RBCs, over haematocrit range of 12.5%-50%. CONCLUSION The WTF assessed on the T-TAS using reconstituted blood may serve as a new physiological blood thrombus test to quantitatively determine the quality of PLT products.
Collapse
Affiliation(s)
| | - Kazuta Yasui
- Japanese Red Cross Kinki Block Blood Center, Osaka, Japan
| | - Tomoya Hayashi
- Japanese Red Cross Kinki Block Blood Center, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Davidson S. Monitoring of Antiplatelet Therapy. Methods Mol Biol 2023; 2663:381-402. [PMID: 37204725 DOI: 10.1007/978-1-0716-3175-1_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In the late 1990s, the antithrombotic antiplatelet agent, clopidogrel, a P2Y12 inhibitor, was introduced. Around the same time, there was an increase in a number of new methods to measure platelet function (e.g., PFA-100 in 1995), and this has continued. It became evident that not all patients responded to clopidogrel in the same way and that some patients had a relative "resistance" to therapy, termed "high on-treatment platelet reactivity." This then led to some publications to advocate platelet function testing being used for patients on antiplatelet therapy. Platelet function testing was also suggested for use in patients awaiting cardiac surgery after stopping their antiplatelet therapy as a way of balancing thrombotic risk pre-surgery and bleeding risk perioperatively. This chapter will discuss some of the commonly used platelet function tests used in these settings, particularly those that are sometimes referred to as point-of-care tests or that require minimal laboratory sample manipulation. The latest guidance and recommendations for platelet function testing will be discussed following several clinical trials looking at the usefulness of platelet function testing in these clinical settings.
Collapse
Affiliation(s)
- Simon Davidson
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
12
|
Matsuo O, Ishii M, Kaikita K, Morinaga J, Miyamura F, Matsumoto S, Tsujita K, Nakamura K. Utility of the Total Thrombus-Formation Analysis System as a Tool for Evaluating Thrombogenicity and Monitoring Antithrombotic Therapy in Pediatric Fontan Patients. Pediatr Cardiol 2022; 44:1150-1159. [PMID: 36550317 DOI: 10.1007/s00246-022-03076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND There is no consensus regarding thromboprophylaxis after Fontan procedure, and novel tools to assess thrombogenicity are needed to establish optimal thromboprophylaxis. The Total Thrombus-formation Analysis System (T-TAS) was developed for the quantitative analysis of thrombus formation using microchips with thrombogenic surfaces. This prospective study evaluated the utility of T-TAS in the assessment of thrombogenicity in pediatric Fontan patients. METHODS AND RESULTS The participants included 20 consecutive Fontan patients who underwent cardiac catheterization and 30 healthy controls. Blood samples collected without and with antithrombotic therapy (aspirin or aspirin and warfarin) were used for T-TAS to compute the area under the curve (AUC) in the atheroma (AR10-AUC30) and platelet (PL18-AUC10) chips. A higher AUC indicates higher thrombogenicity. T-TAS values showed that patients in the Fontan group without antithrombotic therapy had lower thrombogenicity than those in the control group [PL18-AUC10, median (interquartile range) 356 (313-394) vs. 408 (392-424); AR10-AUC30, median (interquartile range) 1270 (1178-1351) vs. 1382 (1338-1421)]. Aspirin and warfarin therapies significantly decreased PL18-AUC10 and AR10-AUC30, respectively, compared with those of patients without antithrombotic therapy (P < 0.001 for each comparison). Subgroup analysis divided by low (< 9 mmHg) or high (≥ 9 mmHg) central venous pressure (CVP) showed that CVP affects the reduction in AR10-AUC30 with antithrombotic therapy. CONCLUSIONS T-TAS may be a useful tool for monitoring thrombogenicity and antithrombotic therapy in Fontan patients.
Collapse
Affiliation(s)
- Osamu Matsuo
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-Ku, Kumamoto City, Kumamoto, 860-8556, Japan.
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-Ku, Kumamoto City, Kumamoto, 860-8556, Japan.,Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Jun Morinaga
- Department of Clinical Investigation, Kumamoto University Hospital, Kumamoto, Japan
| | - Fumiya Miyamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shiro Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-Ku, Kumamoto City, Kumamoto, 860-8556, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
13
|
Liang B, Li R, Lu J, Tian XJ, Gu N. Tongue diagnostic parameters-based diagnostic signature in coronary artery disease patients with clopidogrel resistance after percutaneous coronary intervention. Explore (NY) 2022:S1550-8307(22)00202-6. [PMID: 36335058 DOI: 10.1016/j.explore.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/20/2022] [Accepted: 10/25/2022] [Indexed: 11/15/2022]
|
14
|
Nakanishi N, Kaikita K, Ishii M, Kuyama N, Tabata N, Ito M, Yamanaga K, Fujisue K, Hoshiyama T, Kanazawa H, Hanatani S, Sueta D, Takashio S, Arima Y, Araki S, Usuku H, Nakamura T, Suzuki S, Yamamoto E, Soejima H, Matsushita K, Tsujita K. Malnutrition-associated high bleeding risk with low thrombogenicity in patients undergoing percutaneous coronary intervention. Nutr Metab Cardiovasc Dis 2022; 32:1227-1235. [PMID: 35197212 DOI: 10.1016/j.numecd.2022.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/04/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Although antithrombotic treatments are established for coronary artery disease (CAD), they increase the bleeding risk, especially in malnourished patients. The total thrombus-formation analysis system (T-TAS) is useful for the assessment of thrombogenicity in CAD patients. Here, we examined the relationships among malnutrition, thrombogenicity and 1-year bleeding events in patients undergoing percutaneous coronary intervention (PCI). METHODS AND RESULTS This was a retrospective analysis of 300 consecutive CAD patients undergoing PCI. Blood samples obtained on the day of PCI were used in the T-TAS to compute the thrombus formation area under the curve. We assigned patients to two groups based on the geriatric nutritional risk index (GNRI): 102 patients to the lower GNRI group (≤98), 198 patients to the higher GNRI group (98<). The primary endpoint was the incidence of 1-year bleeding events defined by Bleeding Academic Research Consortium criteria types 2, 3, or 5. The T-TAS levels were lower in the lower GNRI group than in the higher GNRI group. Kaplan-Meier analysis showed worse 1-year bleeding event-free survival in the lower GNRI group compared with the higher GNRI group. The combined model of the GNRI and the Academic Research Consortium for High Bleeding Risk (ARC-HBR) had good calibration and discrimination for bleeding risk prediction. In addition, having a lower GNRI and ARC-HBR positivity was associated with 1-year bleeding events. CONCLUSION A lower GNRI could reflect low thrombogenicity evaluated by the T-TAS and determine bleeding risk in combination with ARC-HBR positivity.
Collapse
Affiliation(s)
- Nobuhiro Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Division of Cardiovascular Medicine and Nephrology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Japan.
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoto Kuyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenshi Yamanaga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Nakamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Soejima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Matsushita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
15
|
Oda Y, Ito T, Yamada Y, Koga T, Nagasato T, Ohnishi-Wada T, Hosokawa K, Fukase H, Hashiguchi T, Maruyama I. Cardiovascular risk factors are associated with augmented thrombogenicity in healthy individuals: analysis using the Total Thrombus-formation Analysis System. Thromb J 2021; 19:88. [PMID: 34789279 PMCID: PMC8597268 DOI: 10.1186/s12959-021-00341-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rupture of an atherosclerotic plaque and subsequent exposure of the subendothelial prothrombotic matrix to blood cause arterial thrombosis. Circulating platelets play an indispensable role in the growth of arterial thrombi partially owing to their unique ability to adhere to the subendothelial matrix and to aggregate to each other under flow conditions. Recently, the Total Thrombus-formation Analysis System (T-TAS) was developed for ex vivo analysis of the thrombogenic potential of whole blood samples under flow conditions. Despite the potential clinical utility of the T-TAS in assessing the risk for thrombosis and bleeding, reference intervals for T-TAS analysis in healthy individuals have not been determined. METHODS In total, 122 whole blood samples were collected from healthy volunteers ranging in age from 25 to 45 years. T-TAS analysis and hematological, physiological, and lifestyle assessments were conducted in these subjects. Whole blood samples anticoagulated with hirudin were perfused into a collagen-coated microchip (PL chip). The time to 10 kPa and the area under the flow pressure curve up to 10 min (AUC10) were analyzed as representative variables for thrombogenic potential. Reference intervals, which were defined as 2.5-97.5 percentiles, were determined. Additionally, univariate and multivariate analyses were performed to identify factors associated with the AUC10 in the T-TAS. RESULTS The time to 10 kPa and the AUC10 widely varied, even in healthy volunteers. The reference intervals were 1.50-4.02 min and 223.4-456.8, respectively, at a shear rate of 1500 s- 1. Univariate and multivariate analyses showed that platelet counts were most significantly associated with the AUC10 of the T-TAS. The presence of one or more cardiovascular risk factors of a high body mass index, a high pulse pressure, high fasting serum glucose levels, high low-density lipoprotein-cholesterol levels, a history of smoking, and no habitual exercise, had the second largest effect on the AUC10 of the T-TAS. CONCLUSIONS Healthy volunteers who had any cardiovascular risk factors showed augmented thrombogenicity, even in artificial uniform capillaries, compared with those without any risk factors in the T-TAS.
Collapse
Affiliation(s)
- Yuu Oda
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan. .,Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, 4-24-1 Kuhonji, Kumamoto, 862-0976, Japan.
| | | | | | - Tomoka Nagasato
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Research Institute, Fujimori Kogyo Co., Ltd., Yokohama, Japan
| | | | - Kazuya Hosokawa
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Research Institute, Fujimori Kogyo Co., Ltd., Yokohama, Japan
| | | | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
16
|
Melnichnikova OS, Nazarova IA, Sirotkina OV, Panov AV, Abesadze IT, Alugishvili MZ, Lokhovinina NL, Vavilova TV. [Integral tests of the hemostasis system in assessing the efficiency of acetylsalicylic acid in patients with ischemic heart disease]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:427-433. [PMID: 34730556 DOI: 10.18097/pbmc20216705427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Despite the fact that acetylsalicylic acid (ASA) is the "gold" standard for the prevention of cardiovascular complications in patients with coronary heart disease (CAD), a number of patients still have risks of atherothrombosis. In the present study, the antithrombotic effect of ASA in patients with CAD was assessed in platelet-rich plasma (PRP) using integral tests of the hemostasis study: the T-TAS system (Total Thrombus-formation Analysis System) and the thrombin generation test (TGT). The study involved 34 patients with stable CAD (11 women, 23 men) and people (15 women, 18 men) in the control group. As a result of assessing the activity of thrombus formation using the T-TAS system, a significant decrease in the area under the curve (AUC10) was found in the group with CAD patients compared with the control (135.6 [88.0-222.3] and 260.5 [217.3-301.9], respectively, p.
Collapse
Affiliation(s)
- O S Melnichnikova
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - I A Nazarova
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - O V Sirotkina
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia; Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre "Kurchatov Institute", St.Petersburg, Russia
| | - A V Panov
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - I T Abesadze
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - M Z Alugishvili
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - N L Lokhovinina
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| | - T V Vavilova
- V.A. Almazov National Medical Research Center, St.Petersburg, Russia
| |
Collapse
|
17
|
Sikora J, Karczmarska-Wódzka A, Bugieda J, Sobczak P. The Use of Total Thrombus Formation Analysis System as a Tool to Assess Platelet Function in Bleeding and Thrombosis Risk-A Systematic Review. Int J Mol Sci 2021; 22:8605. [PMID: 34445311 PMCID: PMC8395324 DOI: 10.3390/ijms22168605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Today there are many devices that can be used to study blood clotting disorders by identifying abnormalities in blood platelets. The Total Thrombus Formation Analysis System is an automated microchip flow chamber system that is used for the quantitative analysis of clot formation under blood flow conditions. For several years, researchers have been using a tool to analyse various clinical situations of patients to identify the properties and biochemical processes occurring within platelets and their microenvironment. METHODS An investigation of recent published literature was conducted based on PRISMA. This review includes 52 science papers directly related to the use of the Total Clot Formation Analysis System in relation to bleeding, surgery, platelet function assessment, anticoagulation monitoring, von Willebrand factor and others. CONCLUSION Most available studies indicate that The Total Thrombus Formation Analysis System may be useful in diagnostic issues, with devices used to monitor therapy or as a significant tool for predicting bleeding events. However, T-TAS not that has the potential for diagnostic indications, but allows the direct observation of the flow and the interactions between blood cells, including the intensity and dynamics of clot formation. The device is expected to be of significant value for basic research to observe the interactions and changes within platelets and their microenvironment.
Collapse
Affiliation(s)
- Joanna Sikora
- Research and Education Unit for Experimental Biotechnology, Department of Transplantology and General Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (A.K.-W.); (J.B.)
| | - Aleksandra Karczmarska-Wódzka
- Research and Education Unit for Experimental Biotechnology, Department of Transplantology and General Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (A.K.-W.); (J.B.)
| | - Joanna Bugieda
- Research and Education Unit for Experimental Biotechnology, Department of Transplantology and General Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (A.K.-W.); (J.B.)
| | - Przemysław Sobczak
- Department of Hematology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
18
|
Tyrosine Kinase Inhibitor Sunitinib Delays Platelet-Induced Coagulation: Additive Effects of Aspirin. Thromb Haemost 2021; 122:92-104. [PMID: 34130349 DOI: 10.1055/s-0041-1730312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sunitinib is a multitarget tyrosine kinase inhibitor (TKI) used for cancer treatment. In platelets, sunitinib affects collagen-induced activation under noncoagulating conditions. We investigated (1) the effects of sunitinib on thrombus formation induced by other TK-dependent receptors, and (2) the effects under coagulating conditions. Cardiovascular disease is a comorbidity in cancer patients, resulting in possible aspirin treatment. Sunitinib and aspirin are associated with increased bleeding risk, and therefore we also investigated (3) the synergistic effects of these compounds on thrombus and fibrin formation. METHODS Blood or isolated platelets from healthy volunteers or cancer patients were incubated with sunitinib and/or aspirin or vehicle. Platelet activation was determined by TK phosphorylation, flow cytometry, changes in [Ca2+]i, aggregometry, and whole blood perfusion over multiple surfaces, including collagen with(out) tissue factor (TF) was performed. RESULTS Sunitinib reduced thrombus formation and phosphatidylserine (PS) exposure under flow on collagen type I and III. Also, sunitinib inhibited glycoprotein VI-induced TK phosphorylation and Ca2+ elevation. Upon TF-triggered coagulation, sunitinib decreased PS exposure and fibrin formation. In blood from cancer patients more pronounced effects of sunitinib were observed in lung and pancreatic as compared to neuroglioblastoma and other cancer types. Compared to sunitinib alone, sunitinib plus aspirin further reduced platelet aggregation, thrombus formation, and PS exposure on collagen under flow with(out) coagulation. CONCLUSION Sunitinib suppresses collagen-induced procoagulant activity and delays fibrin formation, which was aggravated by aspirin. Therefore, we urge for awareness of the combined antiplatelet effects of TKIs with aspirin, as this may result in increased risk of bleeding.
Collapse
|
19
|
Herfs L, Swieringa F, Jooss N, Kozlowski M, Heubel-Moenen FCJ, van Oerle R, Machiels P, Henskens Y, Heemskerk JWM. Multiparameter microfluidics assay of thrombus formation reveals increased sensitivity to contraction and antiplatelet agents at physiological temperature. Thromb Res 2021; 203:46-56. [PMID: 33934017 DOI: 10.1016/j.thromres.2021.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/13/2021] [Accepted: 04/07/2021] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Current developments to assess qualitative and quantitative platelet traits in flowed whole-blood are based on microfluidic devices that mostly operate at room temperature. However, operation at physiological temperature (37 °C) may increase the assay's sensitivity, and facilitates the comparison to other platelet function tests of the diagnostic laboratory. MATERIALS AND METHODS We adapted the conventional microspot-based microfluidic device with a simple thermo-coupled pre-heating module. Automated analysis of microscopic images assisted in obtaining five time-dependent parameters of thrombus formation over collagen microspots (shear rate 1000 s-1). These modifications allowed rapid testing of control and patient blood samples at physiological temperature. RESULTS AND CONCLUSION The higher temperature enhanced platelet adhesion and aggregation as well as late thrombus characteristics such as size and contraction, when compared to room temperature. Moreover, assessment at 37 °C indicated a time-dependent impairment of the thrombus parameters in blood from patients taking common antiplatelet medication, i.e. aspirin and/or clopidogrel. This pointed to increased contribution of the autocrine platelet agonists thromboxane A2 and ADP in the buildup of contracted thrombi under flow. Overall, this study underlined the advantage of multiparameter assessment of microfluidic thrombus formation in detecting an acquired platelet dysfunction, when operating at physiological temperature. This work may bring microfluidics tests closer to the diagnostic laboratory.
Collapse
Affiliation(s)
- Linda Herfs
- FlowChamber B.V., Oxfordlaan 70, Maastricht, the Netherlands
| | - Frauke Swieringa
- Dept. of Biochemistry, CARIM, P.O. Box 616, 6200 MD, Maastricht University, Maastricht, the Netherlands
| | - Natalie Jooss
- Dept. of Biochemistry, CARIM, P.O. Box 616, 6200 MD, Maastricht University, Maastricht, the Netherlands; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TH, United Kingdom
| | - Mike Kozlowski
- YourRhythmics B.V., Oxfordlaan 70, Maastricht, the Netherlands
| | - Floor C J Heubel-Moenen
- Dept. of Hematology/Internal Medicine, Maastricht University Medical Centre(+), P. Debyelaan 25, Maastricht, the Netherlands
| | - René van Oerle
- Dept. of Biochemistry, CARIM, P.O. Box 616, 6200 MD, Maastricht University, Maastricht, the Netherlands; Central Diagnostic Laboratory, Maastricht University Medical Centre(+), P. Debyelaan 25, Maastricht, the Netherlands
| | - Patric Machiels
- FlowChamber B.V., Oxfordlaan 70, Maastricht, the Netherlands
| | - Yvonne Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre(+), P. Debyelaan 25, Maastricht, the Netherlands
| | - Johan W M Heemskerk
- FlowChamber B.V., Oxfordlaan 70, Maastricht, the Netherlands; Dept. of Biochemistry, CARIM, P.O. Box 616, 6200 MD, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
20
|
Zheng KL, Wallen H, Aradi D, Godschalk TC, Hackeng CM, Dahlen JR, Ten Berg JM. The Total Thrombus Formation (T-TAS) platelet (PL) assay, a novel test that evaluates whole blood platelet thrombus formation under physiological conditions. Platelets 2021; 33:273-277. [PMID: 33554695 DOI: 10.1080/09537104.2021.1882669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Dual antiplatelet therapy (DAPT, aspirin, and a P2Y12 inhibitor) reduces thrombotic events in patients with coronary artery disease (CAD). The T-TAS PL assay uses arterial shear flow over collagen surface, better mimicking in vivo conditions compared to conventional agonist-based platelet function assays, to evaluate platelet function. Here, the platelet function in patients taking DAPT is evaluated with the T-TAS PL assay. In 57 patients with CAD, taking DAPT ≥7 days (n = 22 for clopidogrel, n = 15 for prasugrel, n = 20 for ticagrelor), T-TAS PL assessments were performed in duplicate, and expressed as area under the flow pressure curve within a 10-minute period (AUC10). The duplicate measurements were strongly correlated (r = 0.90, p < .001), with an intra-assay coefficient of variation (CV) of 11,5%. For clopidogrel, the median AUC10 was 11.5 (IQR5.9-41.8), for prasugrel 28.8 (IQR10.3-37.6), and for ticagrelor 8.9 (IQR 6.4-10.9). All measurements were below the AUC10 cutoff of 260 measured in healthy volunteers, suggesting excellent discrimination of DAPT-treated and untreated persons. The new T-TAS PL assay demonstrated complete discrimination of platelet function in patients on DAPT based on an established cutoff. Ticagrelor showed lower levels of platelet function and a more uniform response compared to prasugrel and clopidogrel.
Collapse
Affiliation(s)
- K L Zheng
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - H Wallen
- Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - D Aradi
- Department of Cardiology, Heart Center Balatonfüred, and Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - T C Godschalk
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - C M Hackeng
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, Netherlands
| | | | - J M Ten Berg
- Department of Cardiology, St. Antonius Hospital, Nieuwegein, Netherlands
| |
Collapse
|
21
|
Nakanishi N, Kaikita K, Ishii M, Kuyama N, Tabata N, Ito M, Yamanaga K, Fujisue K, Hoshiyama T, Kanazawa H, Hanatani S, Sueta D, Takashio S, Arima Y, Araki S, Usuku H, Nakamura T, Suzuki S, Yamamoto E, Soejimaa H, Matsushita K, Tsujita K. Hemodialysis-related low thrombogenicity measured by total thrombus-formation analysis system in patients undergoing percutaneous coronary intervention. Thromb Res 2021; 200:141-148. [PMID: 33610886 DOI: 10.1016/j.thromres.2021.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Established antithrombotic therapies can increase bleeding risk, especially in hemodialysis (HD) patients. The Total Thrombus-formation Analysis System (T-TAS) is useful for evaluating thrombogenicity. The aim of this study was to examine the relationship between HD and thrombogenicity, or bleeding events in patients undergoing percutaneous coronary intervention (PCI). METHODS In this retrospective cohort study, 300 patients undergoing elective PCI were enrolled between April 2017 and March 2019. Blood samples obtained on the day of PCI were analyzed with T-TAS to compute the thrombus formation area under the curve (AUC; PL18-AUC10 for platelet chip; AR10-AUC30 for atheroma chip). The patients were divided into three groups according to estimated glomerular filtration rate (eGFR): 33 HD patients, 124 non-HD patients with eGFR <60 mL/min/1.73m2, and 143 non-HD patients with eGFR ≥60. We examined the thrombogenicity and spontaneous bleeding events within 1-year post-PCI. RESULTS HD was significantly associated with both low PL18-AUC10 and AR10-AUC30 levels determined by T-TAS. Bleeding events defined by the Bleeding Academic Research Consortium criteria types 2, 3, or 5 occurred during follow-up in 27 patients (9.0%): 7 in HD, 10 in non-HD with eGFR <60, and 10 in non-HD with eGFR ≥60. Both T-TAS parameters in the patients with bleeding were lower compared with those in the patients without bleeding, and HD was significantly associated with 1-year bleeding events. CONCLUSIONS The results suggested that HD patients undergoing PCI might be a predictor for low thrombogenicity measured by T-TAS and 1-year bleeding risk.
Collapse
Affiliation(s)
- Nobuhiro Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Miyazaki Prefectural Nobeoka Hospital, Miyazaki, Japan
| | - Naoto Kuyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenshi Yamanaga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Nakamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Soejimaa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Matsushita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
22
|
Tsujii N, Nogami K, Yoshizawa H, Sakai T, Fukuda K, Ishiguro A, Shima M. Assessment of Platelet Thrombus Formation under Flow Conditions in Patients with Acute Kawasaki Disease. J Pediatr 2020; 226:266-273. [PMID: 32553864 DOI: 10.1016/j.jpeds.2020.06.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To assess platelet thrombus formation (PTF) under flow conditions in patients with Kawasaki disease. Previously available platelet activation data were limited for nonphysiological shear stress condition. The total thrombus-formation analysis system (T-TAS) was developed for quantitative PTF analysis. STUDY DESIGN In total, 33 patients with acute Kawasaki disease were assessed. Whole blood samples, obtained immediately before treatment and 1 week and 1 month after treatment, were assessed using the T-TAS with a collagen-coated platelet chip under high shear values (1000 s-1 [PL12] and 2000 s-1 [PL24]). Measures, such as time to reach 5 kPa above the base pressure (T5+α) and area under the curve for flow pressure curve for 10 minutes (AUC10) were analyzed to quantify PTF. RESULTS Immediately before treatment, the median PL12-T5+α and PL24-T5+α were 3.3 minutes (IQR 2.0-4.5) and 1.3 minutes (0.9-1.9), respectively, and both values were significantly lower in adult controls (3.5 minutes [2.9-6.4] and 2.8 minutes [1.8-4.8]; P = .015 and P < .001, respectively). In addition, the PL12-AUC10 (151.7 U [94.5-279.9]) significantly decreased in adult controls (234.1 U [110.5-306.5], P = .007). By contrast, at 1 week and 1 month after the start of treatment, the T5+α was longer, and the PL12-AUC10 and PL24-AUC10 decreased. CONCLUSIONS In patients with acute Kawasaki disease, the PTF had an early onset and weak stability.
Collapse
Affiliation(s)
- Nobuyuki Tsujii
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan.
| | - Hiroyuki Yoshizawa
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Toshiyuki Sakai
- Department of Pediatrics, Kokuho Central Hospital, Shiki, Nara, Japan
| | - Kazuyoshi Fukuda
- Department of Pediatrics, Saiseikai Chuwa Hospital, Sakurai, Nara, Japan
| | - Akira Ishiguro
- Department of Postgraduate Education and Training, National Center for Child Health and Development, Tokyo, Japan
| | - Midori Shima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
23
|
Nakanishi N, Kaikita K, Ishii M, Kuyama N, Tabata N, Ito M, Yamanaga K, Fujisue K, Hoshiyama T, Kanazawa H, Hanatani S, Sueta D, Takashio S, Arima Y, Araki S, Usuku H, Nakamura T, Suzuki S, Yamamoto E, Soejima H, Matsushita K, Tsujita K. Development and assessment of total thrombus-formation analysis system-based bleeding risk model in patients undergoing percutaneous coronary intervention. Int J Cardiol 2020; 325:121-126. [PMID: 33053391 DOI: 10.1016/j.ijcard.2020.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/13/2020] [Accepted: 10/07/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Antithrombotic therapy is established for the treatment of various cardiovascular events. However, it has been shown to increase the bleeding risk. Total Thrombus-formation Analysis System (T-TAS) is reported to be useful for evaluating thrombogenicity. Here, we estimated whether T-TAS is useful for predicting bleeding events risk in patients undergoing percutaneous coronary intervention (PCI). METHODS This was a retrospective, observational study at Kumamoto University Hospital between April 2017 and March 2019. Blood samples obtained on the day of PCI were used in T-TAS to compute the thrombus formation area under the curve (AUC) (AR10-AUC30, AUC for AR chip). We divided the study population into 2 groups according to the Academic Research Consortium for High Bleeding Risk (ARC-HBR) (182 patients in ARC-HBR positive, 118 in ARC-HBR negative). The primary endpoint was 1-year bleeding events that were defined by Bleeding Academic Research Consortium type2, 3, or 5. RESULTS The AR10-AUC30levels were significantly lower in the ARC-HBR positive group than in the ARC-HBR negative group (median [interquartile range] 1571.4 [1277.0-1745.3] vs. 1726.2 [1567.7-1799.6], p < 0.001). The combination of ARC-HBR and AR10-AUC30 could discriminate the bleeding risk, and improved predictive capacity compared with ARC-HBR by c-statistics. Decision-curve analysis also revealed that combining AR10-AUC30 with ARC-HBR ameliorated bleeding risk-prediction. In multivariate Cox hazards analyses, combining ARC-HBR with lower AR10-AUC30 levels was significantly associated with 1-year bleeding events. CONCLUSIONS The results highlight that AR10-AUC30 evaluated by T-TAS could be a potentially useful marker for predicting high bleeding risk in patients undergoing PCI.
Collapse
Affiliation(s)
- Nobuhiro Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Miyazaki Prefectural Nobeoka Hospital, Miyazaki, Japan
| | - Naoto Kuyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenshi Yamanaga
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Nakamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Soejima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Matsushita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
24
|
Rossi JM, Diamond SL. Scalable manufacture of a disposable, storage-stable eight-channel microfluidic device for rapid testing of platelet, coagulation, and drug function under whole blood flow. BIOMICROFLUIDICS 2020; 14:054103. [PMID: 33014235 PMCID: PMC7527242 DOI: 10.1063/5.0023312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/17/2020] [Indexed: 05/04/2023]
Abstract
Custom polydimethylsiloxane (PDMS) microfluidic devices allow for small-volume human blood research under hemodynamic conditions of bleeding and clotting. However, issues of PDMS molding/assembly, bio-coating, and sample preparation often limit their point-of-care use. We aim to develop a microfluidic device that has the same utility as previously established PDMS devices but which is more usable in point-of-care operation. We designed an injection-molded 1 × 3 in.2 device with eight flow paths crossing a bio-printed surface of a collagen/tissue factor. The device is rapidly primed and compatible with multi-channel pipetting (<0.5 ml blood) and operates under venous or arterial shear rates using constant flow rate or constant pressure modes. Platelet and fibrin deposition were monitored dynamically by the imaging of immunofluorescence. For whole blood clotting at a wall shear rate of 200 s-1, the intrachip CV at 400 s for platelet and fibrin deposition was 10% and the interdonor CV at 400 s was 30% for platelet and 22% for fibrin deposition (across 10 healthy donors). No significant difference was detected for samples tested on a new chip vs a chip stored for 6 months at 4 °C. Using the fibrin signal, dose-response testing of whole blood revealed IC50's of 120 nM for rivaroxaban and apixaban, and 60 nM for dabigatran. A complete reversal of apixaban inhibition was observed for an equimolar addition of Xa DOAC reversal agent Andexanet Alfa. We demonstrate the ability to manufacture single-use, storage-stable eight-channel chips. In clinical settings, such chips may help evaluate patient bleeding risk, therapy choice, drug activity, or reversal.
Collapse
Affiliation(s)
| | - Scott L. Diamond
- Author to whom correspondence should be addressed:. Tel.: +1 215 573 5704. Fax: +1 215 573 6815
| |
Collapse
|
25
|
Bärnthaler T, Mahla E, Toth GG, Schuligoi R, Prüller F, Buschmann E, Heinemann A. Supplemental Fibrinogen Restores Platelet Inhibitor-Induced Reduction in Thrombus Formation without Altering Platelet Function: An In Vitro Study. Thromb Haemost 2020; 120:1548-1556. [PMID: 32772348 DOI: 10.1055/s-0040-1715445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND For patients treated with dual antiplatelet therapy, standardized drug-specific 3-to-7 day cessation is recommended prior to major surgery to reach sufficient platelet function recovery. Here we investigated the hypothesis that supplemental fibrinogen might mitigate the inhibitory effects of antiplatelet therapy. METHODS AND RESULTS To this end blood from healthy donors was treated in vitro with platelet inhibitors, and in vitro thrombus formation and platelet activation were assessed. Ticagrelor, acetylsalicylic acid, the combination of both, and tirofiban all markedly attenuated the formation of adherent thrombi, when whole blood was perfused through collagen-coated microchannels at physiological shear rates. Addition of fibrinogen restored in vitro thrombus formation in the presence of antiplatelet drugs and heparin. However, platelet activation, as investigated in assays of P-selectin expression and calcium flux, was not altered by fibrinogen supplementation. Most importantly, fibrinogen was able to restore in vitro thrombogenesis in patients on maintenance dual antiplatelet therapy after percutaneous coronary intervention. CONCLUSION Thus, our in vitro data support the notion that supplementation of fibrinogen influences the perioperative hemostasis in patients undergoing surgery during antiplatelet therapy by promoting thrombogenesis without significantly interfering with platelet activation.
Collapse
Affiliation(s)
- Thomas Bärnthaler
- Otto Loewi Research Center, Division of Pharmacology Medical University of Graz, Graz, Austria
| | - Elisabeth Mahla
- Division of Anaesthesiology for Cardiovascular and Thoracic Surgery and Intensive Care Medicine, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Gabor G Toth
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rufina Schuligoi
- Otto Loewi Research Center, Division of Pharmacology Medical University of Graz, Graz, Austria
| | - Florian Prüller
- Clinical Institute of Medical Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Eva Buschmann
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Otto Loewi Research Center, Division of Pharmacology Medical University of Graz, Graz, Austria
| |
Collapse
|
26
|
Iwanaga T, Miura N, Brainard BM, Brooks MB, Goggs R. A Novel Microchip Flow Chamber (Total Thrombus Analysis System) to Assess Canine Hemostasis. Front Vet Sci 2020; 7:307. [PMID: 32582782 PMCID: PMC7282356 DOI: 10.3389/fvets.2020.00307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/05/2020] [Indexed: 01/27/2023] Open
Abstract
Hemorrhagic diseases are common in dogs. Current coagulation assays do not model all aspects of in vivo hemostasis and may not predict bleeding risk. The Total-Thrombus Analysis System (T-TAS) is a novel hemostasis assay system in which whole blood flows through microfluidic channels at defined shear rates to provide qualitative and quantitative evaluation of platelet function (PL-chip) and coagulation function (AR-chip). The present study evaluated the T-TAS in dogs with hereditary bleeding disorders and with acquired hemorrhagic syndromes (Group 1), and healthy controls (Group 2). Hereditary defects included von Willebrand's disease (VWD; n = 4), hemophilia A (n = 2), and canine Scott syndrome (n = 2). Acquired hemorrhagic disorders included neoplastic hemoperitoneum (n = 2) and acute hemorrhagic diarrhea syndrome (n = 1). Citrate anticoagulated samples were collected from diseased dogs (Group 1, n = 11) and controls (Group 2, n = 11) for coagulation screening tests, fibrinogen analyses, D-dimer concentration, antithrombin activity, von Willebrand Factor antigen, PFA-100 closure time (PFA-CT), and thromboelastography (TEG). Citrate and hirudin anticoagulated samples were used for T-TAS analyses at two shear rates. Qualitative thrombus formation in each chip was recorded using the T-TAS video camera. Numeric parameters, derived from the instrument software, included occlusion start time (OST; time to 10 kPa), occlusion time (OT; time to 60 kPa (PL-chip) or 80 kPa (AR-chip)), and area under the pressure curve (AUC). Correlations between continuous variables were evaluated by Spearman's rank. Continuous variables were compared between groups by Student's t-test or the Mann-Whitney U-test. Alpha was set at 0.05. In combined analyses of all dogs, significant correlations were identified between T-TAS variables, between the PFA-CT and PL-chip parameters and between TEG variables and AR-chip parameters. The prothrombin time correlated with the AR-chip AUC at both shear rates. In Group 1 dogs, the AR-chip AUC at low shear was significantly reduced compared with Group 2 dogs. Aberrant thrombus formation was seen in video images recorded from dogs with VWD and hemophilia A. The T-TAS AR-chip analysis distinguished dogs with bleeding risk compared to healthy controls. Initial evaluations of the T-TAS suggest it may aid characterization of hemostasis in patients at-risk of bleeding and assist with delineating bleeding phenotypes.
Collapse
Affiliation(s)
- Tomoko Iwanaga
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Naoki Miura
- Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Benjamin M Brainard
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Marjory B Brooks
- Department of Population Medicine and Diagnostic Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| | - Robert Goggs
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, United States
| |
Collapse
|
27
|
Gorog DA, Lip GYH. Impaired Spontaneous/Endogenous Fibrinolytic Status as New Cardiovascular Risk Factor?: JACC Review Topic of the Week. J Am Coll Cardiol 2020; 74:1366-1375. [PMID: 31488274 DOI: 10.1016/j.jacc.2019.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/17/2019] [Accepted: 07/19/2019] [Indexed: 12/27/2022]
Abstract
Endogenous fibrinolysis is a powerful natural defense mechanism against lasting arterial thrombotic occlusion. Recent prospective studies have shown that impaired endogenous fibrinolysis (or hypofibrinolysis) can be detected in a significant number of patients with acute coronary syndrome (ACS) using global assays and is a strong marker of future cardiovascular risk. This novel risk biomarker is independent of traditional cardiovascular risk factors and unaffected by antiplatelet therapy. Most prospective prognostic data have been obtained using a global assay using native whole blood at high shear or plasma turbidimetric assays, which are described herein. Tests of endogenous fibrinolysis could be used to identify patients with ACS who, despite antiplatelet therapy, remain at high cardiovascular risk. This review discusses the impact of currently available medications and those in development that favorably modulate fibrinolytic status and may offer a potential new avenue to improve outcomes in ACS.
Collapse
Affiliation(s)
- Diana A Gorog
- National Heart and Lung Institute, Imperial College, London, United Kingdom; Postgraduate Medical School, University of Hertfordshire, Hertfordshire, United Kingdom.
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
28
|
Yamazaki M. Monitoring of Antithrombotic Therapy. J Atheroscler Thromb 2020; 27:201-203. [PMID: 31685715 PMCID: PMC7113145 DOI: 10.5551/jat.ed120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
29
|
Yamamoto K, Ito T, Nagasato T, Shinnakasu A, Kurano M, Arimura A, Arimura H, Hashiguchi H, Deguchi T, Maruyama I, Nishio Y. Effects of glycemic control and hypoglycemia on Thrombus formation assessed using automated microchip flow chamber system: an exploratory observational study. Thromb J 2019; 17:17. [PMID: 31496922 PMCID: PMC6717975 DOI: 10.1186/s12959-019-0206-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/26/2019] [Indexed: 01/13/2023] Open
Abstract
Background Thrombus formation is an important factor affecting cardiovascular events and venous thromboembolism in type 2 diabetes. However, it is unclear whether glycemic control reduces thrombogenicity. We investigated the effect of short-term glycemic control (STUDY 1) and hypoglycemia (STUDY 2) on thrombus formation using an automated microchip flow chamber system. Methods For STUDY 1, we recruited 10 patients with type 2 diabetes. Before and after 2 weeks of treatment, blood glucose was analyzed with a continuous glucose monitoring system, and thrombogenicity was analyzed with an automated microchip flow chamber system. For STUDY 2, we recruited 10 subjects without diabetes who underwent an insulin tolerance test. We evaluated the change in thrombogenic potential with hypoglycemia. Results STUDY1: The mean blood glucose level reduced from 10.1 ± 2.6 to 6.9 ± 0.97 mM (P < 0.01). T10, an indicator of thrombogenicity, significantly attenuated after glycemic control (338 ± 65 vs. 425 ± 117 s, P < 0.05). The attenuation in T10 was significantly correlated with changes in mean blood glucose level after treatment (r = - 0.718, P < 0.05). STUDY 2: Platelet function was enhanced with decreasing blood glucose; increased platelet function was strongly correlated with an increase in epinephrine. Conclusions We demonstrated attenuation in thrombogenicity with short-term comprehensive diabetes care and enhancement in thrombogenicity with hypoglycemia, using a new flow chamber system. Trial registration UMIN-CTR UMIN 000019899, registered 26-Jan-2015 (STUDY 2).
Collapse
Affiliation(s)
- Kiyoaki Yamamoto
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Takashi Ito
- 2Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Tomoka Nagasato
- 2Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan.,3Fujimori Kogyo Co., Research Institute, 1-10-1, Yokohama, Kanagawa, 236-0003 Japan
| | - Atsushi Shinnakasu
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Mihoko Kurano
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Aiko Arimura
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Hiroshi Arimura
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Hiroshi Hashiguchi
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Takahisa Deguchi
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Ikuro Maruyama
- 2Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| | - Yoshihiko Nishio
- 1Department of Diabetes and Endocrine Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8544 Japan
| |
Collapse
|
30
|
Mitsuse T, Kaikita K, Ishii M, Oimatsu Y, Nakanishi N, Ito M, Arima Y, Sueta D, Iwashita S, Fujisue K, Kanazawa H, Takashio S, Araki S, Usuku H, Suzuki S, Sakamoto K, Yamamoto E, Soejima H, Tsujita K. Total Thrombus-Formation Analysis System can Predict 1-Year Bleeding Events in Patients with Coronary Artery Disease. J Atheroscler Thromb 2019; 27:215-225. [PMID: 31391352 PMCID: PMC7113144 DOI: 10.5551/jat.49700] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aims: The assessment of bleeding risk in patients with coronary artery disease (CAD) is clinically important. We recently developed the Total Thrombus-Formation Analysis System (T-TAS) for the quantitative analysis of thrombus formation using microchips with thrombogenic surfaces. Here, we assessed the utility of T-TAS parameters in predicting 1-year bleeding events in patients with CAD. Methods: The study subjects were 561 consecutive patients who underwent coronary angiography (CAG) between August 2013 and September 2016 for suspected CAD. Blood samples collected at the time of CAG were used for T-TAS to compute the area under the curve (AUC) (AR10-AUC30) in the AR chip. Patients were divided into three groups according to AR10-AUC30 (low: ≤ 1603, intermediate, and high: > 1765, n = 187 each). One-year bleeding events were defined by the Platelet Inhibition and Patient Outcomes criteria. Results: Bleeding occurred in 21 (3.7%) patients and was classified as major (8 [1.4%]) and minor (13 [2.3%]). The AR10-AUC30 levels were significantly lower in the bleeding group than the non-bleeding group (median [interquartile range] 1590 [1442–1734] vs. 1687 [1546–1797], p = 0.04). Univariate Cox regression analysis demonstrated that low AR10-AUC30, high prothrombin time-international normalized ratio levels, and diabetes correlated with bleeding events. Multivariate Cox regression analysis identified low AR10-AUC30 levels as a significant determinant of bleeding events. Kaplan-Meier survival curves showed a higher rate of bleeding events in the low than the high AR10-AUC30 group (p = 0.007). Conclusions: The results highlight the potential usefulness of the AR10-AUC30 levels in the prediction of 1-year bleeding events in patients with CAD treated with various antithrombotic therapies.
Collapse
Affiliation(s)
- Tatsuro Mitsuse
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Yu Oimatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Nobuhiro Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Satomi Iwashita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Koichiro Fujisue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Seiji Takashio
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Satoshi Araki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Hiroki Usuku
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Satoru Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Hirofumi Soejima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, and Center for Metabolic Regulation of Healthy Aging Kumamoto University
| |
Collapse
|
31
|
Mazzeffi M, Hasan S, Abuelkasem E, Meyer M, Deatrick K, Taylor B, Kon Z, Herr D, Tanaka K. Von Willebrand Factor-GP1bα Interactions in Venoarterial Extracorporeal Membrane Oxygenation Patients. J Cardiothorac Vasc Anesth 2019; 33:2125-2132. [DOI: 10.1053/j.jvca.2018.11.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Indexed: 02/06/2023]
|
32
|
Lee H, Na W, Lee BK, Lim CS, Shin S. Recent advances in microfluidic platelet function assays: Moving microfluidics into clinical applications. Clin Hemorheol Microcirc 2019; 71:249-266. [PMID: 30584134 DOI: 10.3233/ch-189416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The analysis of platelet aggregation and thrombosis kinetics has significantly advanced with progress in microfluidic technology. However, the results of platelet aggregation tests do not fully reflect the observed clinical outcomes. To address the present unmet clinical needs, the basic but essential biology of platelets should be reconsidered in relation to the characteristics of microfluidic systems employed for platelet tests. To this end, the present article provides an overview of commercially available point of care devices and focuses on recent microfluidic studies, describing their measurement principles. We critically discuss the characteristics of the microfluidics systems used to evaluate the complex processes underlying platelet aggregation, and that are specifically designed to mimic the pathophysiological environment of blood vessels, including hemodynamic factors as well as blood vessel injury. To this end, we summarize unsolved issues related to the application of platelet function tests based on microfluidics. Overall, we confirm that platelet function tests based on microfluidics provide a versatile platform that encompasses a variety of basic research, as well as clinical diagnostic applications.
Collapse
Affiliation(s)
- Hoyoon Lee
- Department of Mechanical Engineering, Korea University, Seoul, Korea
| | - Wonwhi Na
- Engineering Research Center for Biofluid Biopsy, Korea University, Seoul, Korea
| | - Byoung-Kwon Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University Medical College, Seoul, Korea
| | - Chae-Seung Lim
- Department of Laboratory Medicine, Guro Hospital, Korea University, Seoul, Korea
| | - Sehyun Shin
- Department of Mechanical Engineering, Korea University, Seoul, Korea.,Engineering Research Center for Biofluid Biopsy, Korea University, Seoul, Korea
| |
Collapse
|
33
|
Ichikawa S, Tsukahara K, Kikuchi S, Minamimoto Y, Kimura Y, Okada K, Matsuzawa Y, Konishi M, Maejima N, Iwahashi N, Hibi K, Kosuge M, Ebina T, Tamura K, Kimura K. Impact of Total Antithrombotic Effect on Bleeding Complications in Patients Receiving Multiple Antithrombotic Agents. Circ J 2019; 83:1309-1316. [PMID: 30971637 DOI: 10.1253/circj.cj-18-1236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Few reports have evaluated the total antithrombotic effect of multiple antithrombotic agents. METHODS AND RESULTS Thrombus formation was evaluated with the Total Thrombus-formation Analysis System (T-TAS®) using 2 types of microchips in 145 patients with stable coronary artery disease receiving oral anticoagulants plus single- or dual-antiplatelet therapy. The PL-chip coated with collagen is designed for analysis of the platelet thrombus formation process under shear stress condition (18 µL/min). The AR-chip coated with collagen and tissue thromboplastin is designed for analysis of the fibrin-rich platelet thrombus formation process under shear stress condition (4 µL/min). The results were expressed as an area under the flow pressure curve (PL18-AUC10and AR4-AUC30, respectively). Bleeding events occurred in 43 patients during a 22-month follow-up. AR4-AUC30was significantly lower in patients with bleeding events than in those without (584 [96-993] vs. 1,028 [756-1,252], P=0.0003). Multivariate logistic regression analysis identified AR4-AUC30(odds ratio 3.18) as a significant predictor of bleeding events, in addition to baseline anemia and usage of the standard dose of direct oral anticoagulants. However, PL18-AUC10was not significantly related to bleeding events. CONCLUSIONS A lower AR4-AUC30level was associated with increasing risk of subsequent bleeding complications in patients with stable coronary artery disease who received multiple antithrombotic agents.
Collapse
Affiliation(s)
- Shinya Ichikawa
- Division of Cardiology, Yokohama City University Medical Center
| | - Kengo Tsukahara
- Division of Cardiology, Yokohama City University Medical Center
| | | | - Yugo Minamimoto
- Division of Cardiology, Yokohama City University Medical Center
| | - Yuichiro Kimura
- Division of Cardiology, Yokohama City University Medical Center
| | - Kozo Okada
- Division of Cardiology, Yokohama City University Medical Center
| | | | - Masaaki Konishi
- Division of Cardiology, Yokohama City University Medical Center
| | | | | | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center
| | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center
| | - Toshiaki Ebina
- Division of Cardiology, Yokohama City University Medical Center
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center
| |
Collapse
|
34
|
Frelinger AL. Platelet Function Testing in Clinical Research Trials. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
35
|
Baaten CCFMJ, Meacham S, de Witt SM, Feijge MAH, Adams DJ, Akkerman JWN, Cosemans JMEM, Grassi L, Jupe S, Kostadima M, Mattheij NJA, Prins MH, Ramirez-Solis R, Soehnlein O, Swieringa F, Weber C, White JK, Ouwehand WH, Heemskerk JWM. A synthesis approach of mouse studies to identify genes and proteins in arterial thrombosis and bleeding. Blood 2018; 132:e35-e46. [PMID: 30275110 PMCID: PMC6293874 DOI: 10.1182/blood-2018-02-831982] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/19/2018] [Indexed: 12/25/2022] Open
Abstract
Antithrombotic therapies reduce cardiovascular diseases by preventing arterial thrombosis and thromboembolism, but at expense of increased bleeding risks. Arterial thrombosis studies using genetically modified mice have been invaluable for identification of new molecular targets. Because of low sample sizes and heterogeneity in approaches or methodologies, a formal meta-analysis to compare studies of mice with single-gene defects encountered major limitations. To overcome these, we developed a novel synthesis approach to quantitatively scale 1514 published studies of arterial thrombus formation (in vivo and in vitro), thromboembolism, and tail-bleeding of genetically modified mice. Using a newly defined consistency parameter (CP), indicating the strength of published data, comparisons were made of 431 mouse genes, of which 17 consistently contributed to thrombus formation without affecting hemostasis. Ranking analysis indicated high correlations between collagen-dependent thrombosis models in vivo (FeCl3 injury or ligation/compression) and in vitro. Integration of scores and CP values resulted in a network of protein interactions in thrombosis and hemostasis (PITH), which was combined with databases of genetically linked human bleeding and thrombotic disorders. The network contained 2946 nodes linked to modifying genes of thrombus formation, mostly with expression in megakaryocytes. Reactome pathway analysis and network characteristics revealed multiple novel genes with potential contribution to thrombosis/hemostasis. Studies with additional knockout mice revealed that 4 of 8 (Apoe, Fpr2, Ifnar1, Vps13a) new genes were modifying in thrombus formation. The PITH network further: (i) revealed a high similarity of murine and human hemostatic and thrombotic processes and (ii) identified multiple new candidate proteins regulating these processes.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stuart Meacham
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Susanne M de Witt
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marion A H Feijge
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - David J Adams
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Jan-Willem N Akkerman
- Laboratory of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Luigi Grassi
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Steve Jupe
- EMBL-European Bioinformatics Institute, Cambridge, United Kingdom
| | - Myrto Kostadima
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Nadine J A Mattheij
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Martin H Prins
- Department of Clinical Epidemiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany; and
- Department of Pathology, AMC, Amsterdam, The Netherlands
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany; and
| | | | - Willem H Ouwehand
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
36
|
Al Ghaithi R, Mori J, Nagy Z, Maclachlan A, Hardy L, Philippou H, Hethershaw E, Morgan NV, Senis YA, Harrison P. Evaluation of the Total Thrombus-Formation System (T-TAS): application to human and mouse blood analysis. Platelets 2018; 30:893-900. [PMID: 30365350 DOI: 10.1080/09537104.2018.1535704] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/26/2022]
Abstract
The Total Thrombus-formation Analyser System (T-TAS) is a whole blood flow chamber system for the measurement of in vitro thrombus formation under variable shear stress conditions. Our current study sought to evaluate the potential utility of the T-TAS for the measurement of thrombus formation within human and mouse whole blood. T-TAS microchips (collagen, PL chip; collagen/tissue thromboplastin, AR chip) were used to analyze platelet (PL) or fibrin-rich thrombus formation, respectively. Blood samples from humans (healthy and patients with mild bleeding disorders) and wild-type (WT), mice were tested. Light transmission lumi-aggregometer (lumi-LTA) was performed in PRP using several concentrations of ADP, adrenaline, arachidonic acid, collagen, PAR-1 peptide and ristocetin. Thrombus growth (N = 22) increased with shear within PL (4:40 ± 1.11, 3:25 ± 0.43 and 3:12 ± 0.48 mins [1000, 1500 and 2000s-1]) and AR chips (3:55 ± 0.42 and 1:49 ± 0.19 [240s-1 and 600s-1]). The area under the curve (AUC) on the PL chip was also reduced at 1000s-1 compared to 1500/2000s-1 (260 ± 51.7, 317 ± 55.4 and 301 ± 66.2, respectively). In contrast, no differences in the AUC between 240s-1 and 600s-1 were observed in the AR chip (1593 ± 122 and 1591 ± 158). The intra-assay coefficient of variation (CV) (n = 10) in the PL chip (1000s-1) and AR chip (240s-1) were T1014.1%, T6016.7%, T10-6022.8% and AUC1024.4% or T10 9.03%, T808.64%, T10-8023.8% and AUC305.1%. AR chip thrombus formation was inhibited by rivaroxaban (1 µM), but not with ticagrelor (10 µM). In contrast, PL chip thrombus formation was totally inhibited by ticagrelor. T-TAS shows an overall agreement with lumi-LTA in 87% of patients (n = 30) with normal PL counts recruited into the genotyping and phenotyping of platelet (GAPP) study and suspected to have a PL function defect. The onset (T10) of thrombus formation in WT mice (N = 4) was shorter when compared to humans e.g. PL chip (1000s-1) T10 were 02:02 ± 00:23 and 03:30 ± 0:45, respectively). T-TAS measures in vitro thrombus formation and can be used for monitoring antithrombotic therapy, investigating patients with suspected PL function defects and monitoring PL function within mice.
Collapse
Affiliation(s)
- Rashid Al Ghaithi
- Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
- Haematology and Blood Transfusion Department, The Royal Hospital, Ministry of Health , Muscat , Sultanate of Oman
| | - Jun Mori
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Zoltan Nagy
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Annabel Maclachlan
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Lewis Hardy
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Helen Philippou
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Emma Hethershaw
- Leeds Institute of Cardiovascular and Metabolic Medicine LIGHT Laboratories, University of Leeds , Leeds , West Yorkshire , UK
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Yotis A Senis
- Institute of Cardiovascular Sciences, Institute for Biomedical Research, University of Birmingham , Birmingham , UK
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| |
Collapse
|
37
|
Wu CH, Lin YJ, Chang SL, Lo LW, Tuan TC, Chao TF, Chung FP, Liao JN, Tzeng CH, Hu YF, Lu TM, Chen SA, Tsao HM. Differences in high on-treatment platelet reactivity between intracoronary and peripheral blood after dual anti-platelet agents in patients with coronary artery disease. Thromb Haemost 2017; 110:124-30. [DOI: 10.1160/th13-01-0034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/20/2013] [Indexed: 12/31/2022]
Abstract
SummaryThe differences of high on-treatment platelet reactivity (HPR) between the coronary arteries and peripheral veins might be associated with poor prediction of adverse cardiovascular events in patients with coronary artery diseases. HPR from the peripheral blood might not adequately reflect the platelet responses in the coronary artery. A total of 21 patients were recruited, and regional differences in HPR were compared between blood samples from the intra-coronary artery (IC), femoral artery (FA), and femoral vein (FV) by light aggregometry (agonists: arachidonic acid, LTA-AA; ADP, LTA-ADP), VerifyNow P2Y12 assays, and a platelet function analyser (PFA-100, collagen and epinephrine cartridge, PFA-CEPI). There were regional differences in the platelet reactivities observed by LTA-AA, VerifyNow P2Y12 assays, and PFACEPI. Platelets from the IC had higher platelet reactivities than those from the FV and FA by the VerifyNow P2Y12 assays but lower reactivities by LTA-AA and PFA-CEPI. HPR values from the blood in the FA were more similar to those from the IC than those from the FV by any test. The monocyte percentages were the only factors associated with differences in HPR between the FV and IC by the VerifyNow P2Y12 assays. Triglyceride levels were associated with the differences in HPR between the FV and IC by LTA-ADP. During the six-month follow-up period, two patients developed cardiovascular events and exhibited differences in HPR between different sites by VerifyNow P2Y12 assays. In conclusions, there were regional differences in HPR in patients with coronary artery diseases, which might prevent the adequate prediction of cardiovascular events.
Collapse
|
38
|
Holmström M, Schmidt DE, Hosokawa K, Blombäck M, Hjemdahl P, Ågren A. Monitoring of coagulation factor therapy in patients with von Willebrand disease type 3 using a microchip flow chamber system. Thromb Haemost 2017; 117:75-85. [DOI: 10.1160/th16-06-0430] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/23/2016] [Indexed: 01/22/2023]
Abstract
SummaryPatients with type 3 von Willebrand disease (VWD-3) have no measurable levels of VW factor (VWF) and usually require treatment with VWF-FVIII concentrate to prevent and/or stop bleeding. Even though the patients are treated prophylactically, they may experience bleeding symptoms. The aim of this study was to evaluate the effect of VWF-FVIII concentrate treatment in VWD-3 patients with the Total Thrombus Analysis System (T-TAS®), which measures thrombus formation under flow conditions. Coagulation profiles of 10 VWD-3 patients were analysed using T-TAS before and 30 minutes after VWF-FVIII concentrate (Haemate®) injection. Results were compared to VWF- and FVIII activity in plasma, and results with thromboelastometry and ris-tocetin-activated platelet impedance aggregometry (Multiplate®) in whole blood. For comparison, 10 healthy controls were also analysed with T-TAS. A median dose of 27 (range 15–35) IU/kg of VWF-FVIII concentrate increased VWF- and FVIII activity as expected. T-TAS thrombus formation was enhanced when a tissue factor/collagen-coated flow chamber was used at low shear, but treatment effects at high shear using a collagen-coated flow chamber were minimal. Whole blood coagulation assessed by thromboelastometry was normal and did not change (p > 0.05) but ristocetin-induced platelet aggregation improved (p < 0.001). In conclusion, T-TAS detects effects of VWF-FVIII concentrate treatment on coagulation-dependent thrombus formation at low shear, but minor effects are observed on platelet-dependent thrombus formation at high shear. The poor prediction of bleeding by conventional laboratory monitoring in VWD-3 patients might be related to insufficient restoration of platelet-dependent thrombus formation.
Collapse
|
39
|
Mazzeffi MA, Lee K, Taylor B, Tanaka KA. Perioperative management and monitoring of antiplatelet agents: a focused review on aspirin and P2Y 12 inhibitors. Korean J Anesthesiol 2017; 70:379-389. [PMID: 28794832 PMCID: PMC5548939 DOI: 10.4097/kjae.2017.70.4.379] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/26/2017] [Indexed: 01/02/2023] Open
Abstract
Platelets play pivotal roles in hemostasis as well as pathological arterial thrombosis. The combination of aspirin and a P2Y12 inhibitor has become the mainstay therapy in the ageing population with cardiovascular conditions, particularly during and after percutaneous coronary intervention. A number of novel P2Y12 inhibitors has become available in the recent years, and they markedly vary in pharmacokinetic and pharmacodynamic properties. Perioperative physicians today face a challenge of preventing hemorrhage due to platelet inhibitors, while minimizing thrombotic risks. There are several point-of-care platelet function tests available in the peri-procedural assessment of residual platelet aggregation. However, these platelet function tests are not standardized in terms of sample processing, agonist type and potency as well as methods of detecting platelet activity. Understanding the differences in pharmacological properties of antiplatelet agents, principles of platelet function tests, and pertinent hemostatic strategies may be useful to anesthesiologists and intensivists who manage perioperative issues associated with antiplatelet agents. The objectives of this review are: 1) to discuss clinical data on aspirin and P2Y12 inhibitors relating to perioperative bleeding, 2) to outline different features of point-of-care platelet function tests, and 3) to discuss therapeutic options for the prevention and treatment of bleeding associated with antiplatelet agents.
Collapse
Affiliation(s)
- Michael A Mazzeffi
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Khang Lee
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bradley Taylor
- Department of Surgery, Division of Cardiac Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kenichi A Tanaka
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Oimatsu Y, Kaikita K, Ishii M, Mitsuse T, Ito M, Arima Y, Sueta D, Takahashi A, Iwashita S, Yamamoto E, Kojima S, Hokimoto S, Tsujita K. Total Thrombus-formation Analysis System Predicts Periprocedural Bleeding Events in Patients With Coronary Artery Disease Undergoing Percutaneous Coronary Intervention. J Am Heart Assoc 2017; 6:JAHA.116.005263. [PMID: 28438734 PMCID: PMC5533019 DOI: 10.1161/jaha.116.005263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Periprocedural bleeding events are common after percutaneous coronary intervention. We evaluated the association of periprocedural bleeding events with thrombogenicity, which was measured quantitatively by the Total Thrombus-formation Analysis System equipped with microchips and thrombogenic surfaces (collagen, platelet chip [PL]; collagen plus tissue factor, atheroma chip [AR]). METHODS AND RESULTS Between August 2013 and March 2016, 313 consecutive patients with coronary artery disease undergoing elective percutaneous coronary intervention were enrolled. They were divided into those with or without periprocedural bleeding events. We determined the bleeding events as composites of major bleeding events defined by the International Society on Thrombosis and Hemostasis and minor bleeding events (eg, minor hematoma, arteriovenous shunt and pseudoaneurysm). Blood samples obtained at percutaneous coronary intervention were analyzed for thrombus formation area under the curve (PL24-AUC10 for PL chip; AR10-AUC30 for AR chip) by the Total Thrombus-formation Analysis System and P2Y12 reaction unit by the VerifyNow system. Periprocedural bleeding events occurred in 37 patients. PL24-AUC10 levels were significantly lower in patients with such events than those without (P=0.002). Multiple logistic regression analyses showed association between low PL24-AUC10 levels and periprocedural bleeding events (odds ratio, 2.71 [1.22-5.99]; P=0.01) and association between PL24-AUC10 and periprocedural bleeding events in 176 patients of the femoral approach group (odds ratio, 2.88 [1.11-7.49]; P=0.03). However, PL24-AUC10 levels in 127 patients of the radial approach group were not significantly different in patients with or without periprocedural bleeding events. CONCLUSIONS PL24-AUC10 measured by the Total Thrombus-formation Analysis System is a potentially useful predictor of periprocedural bleeding events in coronary artery disease patients undergoing elective percutaneous coronary intervention.
Collapse
Affiliation(s)
- Yu Oimatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsuro Mitsuse
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Aya Takahashi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satomi Iwashita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Sunao Kojima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
41
|
Yamazaki M, Ohnishi T, Hosokawa K, Yamaguchi K, Yoneyama T, Kawashima A, Okada Y, Kitagawa K, Uchiyama S. Measurement of residual platelet thrombogenicity under arterial shear conditions in cerebrovascular disease patients receiving antiplatelet therapy. J Thromb Haemost 2016; 14:1788-97. [PMID: 27328457 DOI: 10.1111/jth.13391] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 05/22/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Essentials A consensus methodology for assessing the effects of antiplatelet agents has not been established. Measuring platelet thrombus formation (PTF) for evaluating antiplatelet effects was assessed. PTF differentially reflected antiplatelet effects compared to other tests. PTF may be associated with the severity of carotid or intracranial arterial stenosis. Click to hear a presentation on platelet function testing in the clinic by Gresele and colleagues SUMMARY Background A consensus methodology for assessing the effects of antiplatelet agents has not been established. Objective We investigated the usefulness of directly measuring platelet thrombus formation (PTF) using a microchip-based flow chamber system for evaluating antiplatelet therapy. Patients/Methods Platelet thrombus formation in the whole blood of 94 patients with ischemic cerebrovascular disease treated with clopidogrel and/or aspirin was measured in a flow chamber system at a shear rate of 1500 s(-1) and was compared with the results of assays for agonist-induced platelet aggregability, phosphorylation of vasodilator-stimulated phosphoprotein, platelet p-selectin expression (PS), and platelet-monocyte complexes. Results In all patients tested, area under the flow pressure curve (AUC10), which represents platelet thrombogenicity, showed weak correlation with platelet aggregation induced by either adenosine diphosphate or collagen. In addition, AUC10 was lower in patients treated with dual antiplatelet therapy (median 79.4) compared with patients treated with aspirin or clopidogrel alone (217.7 and 301.0, respectively), whereas the parameters evaluated by the other assays did not reflect the combined treatment efficacy. In clopidogrel monotherapy patients, AUC10 was associated with the severity of arterial stenosis (R(2) = 0.127, β = 1.25), and AUC10 and PS were higher in patients with severe carotid or intracranial arterial stenosis than in those with mild stenosis. Conclusions Platelet thrombus formation measurement using a flow-chamber system was useful for evaluating the efficacy of treatment with aspirin and clopidogrel, both alone and in combination. The present findings indicate that high residual platelet thrombogenicity in patients treated with clopidogrel may be associated with the severity of carotid or intracranial arterial stenosis.
Collapse
Affiliation(s)
- M Yamazaki
- Department of Neurology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan.
| | - T Ohnishi
- Research Institute, Fujimori Kogyo Co., Ltd, Yokohama, Kanagawa, Japan
| | - K Hosokawa
- Research Institute, Fujimori Kogyo Co., Ltd, Yokohama, Kanagawa, Japan
| | - K Yamaguchi
- Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - T Yoneyama
- Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - A Kawashima
- Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Y Okada
- Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - K Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - S Uchiyama
- Department of Neurology, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
- Clinical Research Center for Medicine, International University of Health and Welfare, Center for Brain and Cerebral Vessels, Sanno Hospital and Sanno Medical Center, Tokyo, Minato-ku, Japan
| |
Collapse
|
42
|
Goto M, Miura SI, Suematsu Y, Idemoto Y, Takata K, Imaizumi S, Uehara Y, Saku K. Rivaroxaban, a factor Xa inhibitor, induces the secondary prevention of cardiovascular events after myocardial ischemia reperfusion injury in mice. Int J Cardiol 2016; 220:602-7. [PMID: 27390997 DOI: 10.1016/j.ijcard.2016.06.212] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/25/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Rivaroxaban has been shown to reduce overall death from cardiovascular causes in patients with recent acute coronary syndrome. Therefore, we evaluated the secondary prevention of cardiovascular events after myocardial ischemia reperfusion injury and its mechanisms in mice. METHODS After myocardial reperfusion injury, C57BL/6J mice were randomized to receive either no treatment or treatment for 14days with low and high doses of rivaroxaban. After 7days, mice were administered tissue factor as a secondary event. RESULTS Based on a Kaplan-Meier curve analysis, the high-dose rivaroxaban group showed a significantly higher % survival than the no-treatment group from day 7 (after the administration of tissue factor) to day 14 (at the end of the experimental period). Left ventricular (LV) ejection fraction in both the low- and high-dose rivaroxaban groups improved compared to that in the no-treatment group. Moreover, mRNA levels of interleukin-6 and collagens 1α2 and 3α1 in the LV in the high-dose group were significantly suppressed compared to those in the no-treatment group. CONCLUSIONS Rivaroxaban improved the survival rate, probably by improving cardiac function through the reduction of inflammatory and fibrotic factors in the LV. This effect may be due to the pleiotropic effects of rivaroxaban beyond its main effect as an anti-coagulant.
Collapse
Affiliation(s)
- Masaki Goto
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan; Department of Molecular Cardiovascular Therapeutics, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan.
| | - Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Yoshiaki Idemoto
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Kouhei Takata
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Satoshi Imaizumi
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Yoshinari Uehara
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan; Department of Molecular Cardiovascular Therapeutics, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| | - Keijiro Saku
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan; Department of Molecular Cardiovascular Therapeutics, Fukuoka University School of Medicine, Fukuoka 814-1080, Japan
| |
Collapse
|
43
|
Idemoto Y, Miura SI, Norimatsu K, Suematsu Y, Hitaka Y, Shiga Y, Morii J, Imaizumi S, Kuwano T, Iwata A, Zhang B, Ogawa M, Saku K. Evaluation of the antithrombotic abilities of non-vitamin K antagonist oral anticoagulants using the Total Thrombus-formation Analysis System ®. Heart Vessels 2016; 32:309-316. [PMID: 27325226 DOI: 10.1007/s00380-016-0864-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/10/2016] [Indexed: 12/28/2022]
Abstract
The Total Thrombus-formation Analysis System (T-TAS®) is a novel automated microchip flow-chamber system for the quantitative evaluation of thrombus formation under blood flow conditions. T-TAS® uses two types of microchip to evaluate thrombus formation: the AR-chip quantifies white thrombus formation and the PL-chip quantifies platelet thrombus formation. We assessed the antithrombotic abilities of various non-vitamin K antagonist oral anticoagulants (NOACs) using T-TAS®. One hundred and three consecutive patients who were hospitalized with cardiovascular diseases were enrolled. We divided the patients into 2 groups; a control group that did not receive an anticoagulant (non-AC group) and an anticoagulant group (AC group). The AC group was further divided into warfarin, dabigatran, rivaroxaban and apixaban groups. We performed common coagulation tests and evaluated the area under the flow pressure curve (AR-AUC and PL-AUC) to quantify antithrombotic ability using T-TAS® at the trough. There were no significant differences in patient characteristics between the non-AC and AC groups. Only 55.1 % of patients in the AC group achieved the target blood pressure (BP) of less than 130/80 mmHg. Compared with the non-AC group, AR-AUC was significantly decreased in the AC, warfarin, dabigatran and apixaban groups. Only the rivaroxaban group did not show a significant decrease in AR-AUC. NOACs showed a significant decrease in PL-AUC compared with the non-AC group. In conclusion, T-TAS® was a useful tool for evaluating anticoagulation activity. NOACs was significantly effective as an antiplatelet agent. BP control should be a higher priority than the selection of an anticoagulant drug, especially NOACs.
Collapse
Affiliation(s)
- Yoshiaki Idemoto
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan. .,Department of Molecular Cardiovascular Therapeutics, Fukuoka University School of Medicine, Fukuoka, Japan.
| | - Kenji Norimatsu
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Yuka Hitaka
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Yuhei Shiga
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Joji Morii
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Satoshi Imaizumi
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Takashi Kuwano
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Atsushi Iwata
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Bo Zhang
- Department of Biochemistry, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Masahiro Ogawa
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan
| | - Keijiro Saku
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814 0180, Japan.,Department of Molecular Cardiovascular Therapeutics, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
44
|
Arima Y, Kaikita K, Ishii M, Ito M, Sueta D, Oimatsu Y, Sakamoto K, Tsujita K, Kojima S, Nakagawa K, Hokimoto S, Ogawa H. Assessment of platelet-derived thrombogenicity with the total thrombus-formation analysis system in coronary artery disease patients receiving antiplatelet therapy. J Thromb Haemost 2016; 14:850-9. [PMID: 26773298 DOI: 10.1111/jth.13256] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/18/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND Accurate evaluation of thrombogenicity helps to prevent thrombosis and excessive bleeding. The total thrombus-formation analysis system (T-TAS) was developed for quantitative analysis of platelet thrombus formation by the use of microchips with thrombogenic surfaces (collagen, platelet chip [PL-chip]; collagen plus tissue factor, atherome chip [AR-chip]). We examined the utility of the T-TAS in the assessment of the efficacy of antiplatelet therapy in patients with coronary artery disease (CAD). METHODS AND RESULTS In this cross-sectional study, 372 consecutive patients admitted to the cardiovascular department were divided into three groups: patients not receiving any antiplatelet therapy (control, n = 56), patients receiving aspirin only (n = 69), and patients receiving aspirin and clopidogrel (n = 149). Blood samples were used for the T-TAS to measure the platelet thrombus-formation area under the curve (AUC) at various shear rates (1500 s(-1) [PL18 -AUC10 ] and 2000 s(-1) [PL24 -AUC10 ] for the PL-chip; 300 s(-1) [AR10 -AUC30 ] for the AR-chip). The on-clopidogrel platelet aggregation was measured by the use of P2Y12 reaction units (PRUs) with the VerifyNow system. The mean PL24 -AUC10 levels were 358 ± 111 (± standard deviation) (95% confidence interval [CI] 328.9-387.1) in the control group, 256 ± 108 (95% CI 230.5-281.5) in the aspirin group, and 113 ± 91 (95% CI 98.4-127.6) in the aspirin/clopidogrel group. In the aspirin/clopidogrel group, the PL24 -AUC10 was higher in poor metabolizers (PMs) with cytochrome P450 2C19(CYP2C19) polymorphisms (152 ± 112, 95% CI 103.4-200.6) than in the non-PM group (87 ± 74, 95% CI 73.8-100.2). CONCLUSIONS Our findings suggest that the PL24 -AUC10 level measured by the T-TAS is a potentially suitable index for the assessment of antiplatelet therapy in CAD patients.
Collapse
Affiliation(s)
- Y Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - K Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - M Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - M Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - D Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Y Oimatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - K Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - K Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - S Kojima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - K Nakagawa
- Division of Pharmacology and Therapeutics, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - S Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - H Ogawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
45
|
Nogami K, Ogiwara K, Yada K, Shida Y, Takeyama M, Yaoi H, Minami H, Furukawa S, Hosokawa K, Shima M. Assessing the clinical severity of type 1 von Willebrand disease patients with a microchip flow-chamber system. J Thromb Haemost 2016; 14:667-74. [PMID: 27061057 DOI: 10.1111/jth.13273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND The clinical phenotype of von Willebrand disease (VWD) is heterogeneous, and von Willebrand factor ristocetin cofactor activity (VWF:RCo) does not always reflect clinical severity, especially in VWD type 1. We have reported the potential of a microchip flow-chamber system (Total-Thrombus Formation Analysis System [T-TAS®]) for assessing physiologic hemostasis in VWD. Aim To evaluate the relationship between T-TAS, bleeding score (BS) and laboratory test results in type 1 VWD patients. METHODS Microchips coated with collagen (platelet chip [PL-chip]) or collagen/thromboplastin (AR-chip) were used to assess platelet thrombus formation (PTF) at high shear rates or fibrin-rich PTF at low shear rates, respectively, in whole blood from 50 patients. The times needed for the flow pressure to increase by 10 kPa and 30 kPa (T10 and T30 ) from baseline were calculated from flow pressure curves. BS was determined by the use of a standardized questionnaire. RESULTS PL-T10 values correlated with BS (R(2) ~ 0.45) better than VWF:RCo (R(2) ~ 0.36), irrespective of the flow rate, whereas AR-T10 showed only a weak correlation with BS (R(2) ~ 0.18). Patients with PL-T10 > 10 min or AR-T10 > 30 min had lower VWF levels and higher BS than those with PL-T10 ≤ 10 min or AR-T10 ≤ 30 min, and the greatest differences were observed with PL-T10. Clinical severity appeared to correlate best with PL-T10 > 8 min. BS was significantly higher in patients with VWF:RCo of < 10 IU dL(-1) than in those with VWF:RCo of 10 IU dL(-1) to < 25 IU dL(-1) and 25-40 IU dL(-1). In patients with VWF:RCo of < 10 IU dL(-1) , BS was significantly higher in those with PL-T10 > 8 min than in those with PL-T10 ≤ 8 min. CONCLUSION T-TAS could be a useful technique for discriminating and predicting BS in VWD type 1 patients.
Collapse
Affiliation(s)
- K Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - K Ogiwara
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - K Yada
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Y Shida
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - M Takeyama
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - H Yaoi
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - H Minami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - S Furukawa
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - K Hosokawa
- Research Institute, Fujimori Kogyo Co., Ltd, Yokohama, Kanagawa, Japan
| | - M Shima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
46
|
Microfluidic approaches for the assessment of blood cell trauma: a focus on thrombotic risk in mechanical circulatory support devices. Int J Artif Organs 2016; 39:184-93. [PMID: 27034318 DOI: 10.5301/ijao.5000485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2016] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Mechanical circulatory support devices (MCSDs) are emerging as a valuable therapeutic option for the management of end-stage heart failure. However, although recipients are routinely administered with anti-thrombotic (AT) drugs, thrombosis persists as a severe post-implant complication. Conventional clinical assays and coagulation markers demonstrate partial ability in preventing the onset of thrombosis. Through years, different laboratory techniques have been proposed as potential tools for the evaluation of platelets' hemostatic response in MCSD recipients. Most rely on platelet aggregation tests; they are performed in static or low shear conditions, neglecting the prominent contribution of MCSD shear-induced mechanical load in enhancing platelet activation (PA). On the other hand, those tests able to account for shear-induced PA have limited possibility of effective clinical translation. AIMS AND METHODS Advances on this side have been addressed by microfluidic technology. Microfluidic devices have been developed for AT drug monitoring under flow, able to replicate physiological and/or constant shear flow conditions in vitro. In this paper, we present a newly developed microfluidic platform able to expose platelets to MCSD-specific dynamic shear stress patterns. We performed in vitro tests circulating human platelets in the microfluidic platform and quantifying the dynamics of PA by means of the Platelet Activity State (PAS) assay. RESULTS Our results prove the feasibility of using microfluidics for the diagnosis of MCSD-related thrombotic risk. This study paves the way for the development of a miniaturized point-of-care device for monitoring AT drug regimen. Such a system may have significant impact on limiting the incidence of thrombosis in MCSD recipients.
Collapse
|
47
|
Ito M, Kaikita K, Sueta D, Ishii M, Oimatsu Y, Arima Y, Iwashita S, Takahashi A, Hoshiyama T, Kanazawa H, Sakamoto K, Yamamoto E, Tsujita K, Yamamuro M, Kojima S, Hokimoto S, Yamabe H, Ogawa H. Total Thrombus-Formation Analysis System (T-TAS) Can Predict Periprocedural Bleeding Events in Patients Undergoing Catheter Ablation for Atrial Fibrillation. J Am Heart Assoc 2016; 5:JAHA.115.002744. [PMID: 26811167 PMCID: PMC4859393 DOI: 10.1161/jaha.115.002744] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Non–vitamin K antagonist oral anticoagulants are used to prevent thromboembolism in patients with atrial fibrillation. The T‐TAS “Total Thrombus‐formation Analysis System” (Fujimori Kogyo Co Ltd) was developed for quantitative analysis of thrombus formation using microchips with thrombogenic surfaces (collagen, platelet chip [PL] ; collagen plus tissue factor, atheroma chip [AR]). We evaluated the utility of T‐TAS in predicting periprocedural bleeding in atrial fibrillation patients undergoing catheter ablation (CA). Methods and Results After exclusion of 20 from 148 consecutive patients undergoing CA, the remaining 128 patients were divided into 2 treatment groups: the warfarin group (n=30) and the non–vitamin K antagonist oral anticoagulants group (n=98). Blood samples obtained on the day of CA (anticoagulant‐free point) and at 3 and 30 days after CA were used in T‐TAS to compute the thrombus formation area under the curve (AUC; AUC for the first 10 minutes for PL tested at flow rate of 24 μL/min [PL24‐AUC10]; AUC for the first 30 minutes for AR tested at flow rate of 10 μL/min [AR10‐AUC30]). AR10‐AUC30 and PL24‐AUC10 levels were similar in the 2 groups on the day of CA. Levels of AR10‐AUC30, but not PL24‐AUC10, were significantly lower in the 2 groups at days 3 and 30 after CA. Multiple logistic regression analyses identified the AR10‐AUC30 level on the day of CA as a significant predictor of periprocedural bleeding events (odds ratio 5.7; 95% CI 1.54–21.1; P=0.009). Receiver operating characteristic analysis showed that the AR10‐AUC30 level on the day of CA significantly predicted periprocedural bleeding events (AUC 0.859, 95% CI 0.766–0.951; P<0.001). The cutoff AR10‐AUC30 level was 1648 for identification of periprocedural bleeding events. Conclusions These results suggested that the AR10‐AUC30 level determined by T‐TAS is a potentially useful marker for prediction of bleeding events in atrial fibrillation patients undergoing CA.
Collapse
Affiliation(s)
- Miwa Ito
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Daisuke Sueta
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Yu Oimatsu
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Satomi Iwashita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Aya Takahashi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Tadashi Hoshiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Hisanori Kanazawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Megumi Yamamuro
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Sunao Kojima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Hiroshige Yamabe
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan (M.I., K.K., D.S., M.I., Y.O., Y.A., S.I., A.T., T.H., H.K., K.S., E.Y., K.T., M.Y., S.K., S.H., H.Y., H.O.)
| |
Collapse
|
48
|
Sueta D, Kaikita K, Okamoto N, Arima Y, Ishii M, Ito M, Oimatsu Y, Iwashita S, Takahashi A, Nakamura E, Hokimoto S, Mizuta H, Ogawa H. A novel quantitative assessment of whole blood thrombogenicity in patients treated with a non-vitamin K oral anticoagulant. Int J Cardiol 2015; 197:98-100. [DOI: 10.1016/j.ijcard.2015.06.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/18/2015] [Indexed: 10/23/2022]
|
49
|
Minami H, Nogami K, Ogiwara K, Furukawa S, Hosokawa K, Shima M. Use of a microchip flow-chamber system as a screening test for platelet storage pool disease. Int J Hematol 2015; 102:157-62. [DOI: 10.1007/s12185-015-1819-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/19/2015] [Accepted: 06/02/2015] [Indexed: 11/30/2022]
|
50
|
Branchford BR, Ng CJ, Neeves KB, Di Paola J. Microfluidic technology as an emerging clinical tool to evaluate thrombosis and hemostasis. Thromb Res 2015; 136:13-9. [PMID: 26014643 DOI: 10.1016/j.thromres.2015.05.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 10/23/2022]
Abstract
Assessment of platelet function and coagulation under flow conditions can augment traditional static assays used to evaluate patients with suspected hemostatic or thrombotic disorders. Among the available flow-based assays, microfluidic devices require the smallest blood volume and provide multiple output options. These assays are based on the presence of wall shear stress that mimics in vivo interactions between blood components and vessel walls. Microfluidic devices can generate essential information regarding homeostatic regulation of platelet activation and subsequent engagement of the coagulation cascade leading to fibrin deposition and clot formation. Emerging data suggest that microfluidic assays may also reveal consistent patterns of hemostatic or thrombotic pathology, and could aid in assessing and monitoring patient-specific effects of coagulation-modifying therapies.
Collapse
Affiliation(s)
- Brian R Branchford
- Dept. of Pediatrics - Hematology/Oncology, University of Colorado School of Medicine, Aurora, CO, USA; University of Colorado Hemophilia and Thrombosis Center, Aurora, CO, USA
| | - Christopher J Ng
- Dept. of Pediatrics - Hematology/Oncology, University of Colorado School of Medicine, Aurora, CO, USA; University of Colorado Hemophilia and Thrombosis Center, Aurora, CO, USA
| | - Keith B Neeves
- Dept. of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Jorge Di Paola
- Dept. of Pediatrics - Hematology/Oncology, University of Colorado School of Medicine, Aurora, CO, USA; University of Colorado Hemophilia and Thrombosis Center, Aurora, CO, USA; Graduate Program- Human Medical Genetics and Genomics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|