ArticlePDF Available

The Reevaluation of Thrombin Time Using a Clot Waveform Analysis

MDPI
Journal of Clinical Medicine
Authors:
  • Mie University Hospital, Tsu- city, Mie, Japan

Abstract and Figures

Object: Although thrombin burst has attracted attention as a physiological coagulation mechanism, clinical evidence from a routine assay for it is scarce. This mechanism was therefore evaluated by a clot waveform analysis (CWA) to assess the thrombin time (TT). Material and methods: The TT with a low concentration of thrombin was evaluated using a CWA. We evaluated the CWA-TT of plasma deficient in various clotting factors, calibration plasma, platelet-poor plasma (PPP), and platelet-rich plasma (PRP) obtained from healthy volunteers, patients with thrombocytopenia, and patients with malignant disease. Results: Although the TT-CWA of calibration plasma was able to be evaluated with 0.01 IU/mL of thrombin, that of FVIII-deficient plasma could not be evaluated. The peak time of CWA-TT was significantly longer, and the peak height significantly lower, in various deficient plasma, especially in FVIII-deficient plasma compared to calibration plasma. The second peak of the first derivative (1st DP-2) was detected in PPP from healthy volunteers, and was shorter and higher in PRP than in PPP. The 1st DP-2 was not detected in PPP from patients with thrombocytopenia, and the 1st DP-2 in PRP was significantly lower in patients with thrombocytopenia and significantly higher in patients with malignant disease than in healthy volunteers. Conclusion: The CWA-TT became abnormal in plasma deficient in various clotting factors, and was significantly affected by platelets, suggesting that the CWA-TT may be a useful test for hemostatic abnormalities.
Content may be subject to copyright.
Journal of
Clinical Medicine
Article
The Reevaluation of Thrombin Time Using a Clot
Waveform Analysis
Hideo Wada 1,* , Yuhuko Ichikawa 2, Minoru Ezaki 2, Takeshi Matsumoto 3, Yoshiki Yamashita 4,
Katsuya Shiraki 1, Motomu Shimaoka 5and Hideto Shimpo 6


Citation: Wada, H.; Ichikawa, Y.;
Ezaki, M.; Matsumoto, T.; Yamashita,
Y.; Shiraki, K.; Shimaoka, M.; Shimpo,
H. The Reevaluation of Thrombin
Time Using a Clot Waveform
Analysis. J. Clin. Med. 2021,10, 4840.
https://doi.org/10.3390/jcm10214840
Academic Editor:
Giancarlo Castaman
Received: 30 August 2021
Accepted: 18 October 2021
Published: 21 October 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Department of General and Laboratory Medicine, Mie Prefectural General Medical Center,
Yokkaichi 510-0885, Japan; katsuya-shiraki@mie-gmc.jp
2Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan;
ichi911239@yahoo.co.jp (Y.I.); ajbyd06188@yahoo.co.jp (M.E.)
3Department of Transfusion Medicine and Cell Therapy, Mie University Hospital, Tsu 514-8507, Japan;
matsutak@clin.medic.mie-u.ac.jp
4
Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan;
yamayamafan4989@yahoo.co.jp
5Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of
Medicine, Tsu 514-8507, Japan; motomushimaoka@gmail.com
6Mie Prefectural General Medical Center, Yokkaichi 510-0885, Japan; hideto-shimpo@mie-gmc.jp
*Correspondence: wadahide@clin.medic.mie-u.ac.jp; Tel.: +81-59-345-2321
Abstract:
Object: Although thrombin burst has attracted attention as a physiological coagulation
mechanism, clinical evidence from a routine assay for it is scarce. This mechanism was therefore
evaluated by a clot waveform analysis (CWA) to assess the thrombin time (TT). Material and Methods:
The TT with a low concentration of thrombin was evaluated using a CWA. We evaluated the CWA-TT
of plasma deficient in various clotting factors, calibration plasma, platelet-poor plasma (PPP), and
platelet-rich plasma (PRP) obtained from healthy volunteers, patients with thrombocytopenia, and
patients with malignant disease. Results: Although the TT-CWA of calibration plasma was able to
be evaluated with 0.01 IU/mL of thrombin, that of FVIII-deficient plasma could not be evaluated.
The peak time of CWA-TT was significantly longer, and the peak height significantly lower, in
various deficient plasma, especially in FVIII-deficient plasma compared to calibration plasma. The
second peak of the first derivative (1st DP-2) was detected in PPP from healthy volunteers, and was
shorter and higher in PRP than in PPP. The 1st DP-2 was not detected in PPP from patients with
thrombocytopenia, and the 1st DP-2 in PRP was significantly lower in patients with thrombocytopenia
and significantly higher in patients with malignant disease than in healthy volunteers. Conclusion:
The CWA-TT became abnormal in plasma deficient in various clotting factors, and was significantly
affected by platelets, suggesting that the CWA-TT may be a useful test for hemostatic abnormalities.
Keywords: CWA; thrombin time; platelet; thrombin burst
1. Introduction
It is well known that thrombin directly activates fibrinogen to generate fibrin forma-
tion [
1
]. Furthermore, thrombin activates many coagulation factors in the upper stream,
such as clotting factor XI (FXI), FVIII, Factor X, and Factor V, resulting in thrombin genera-
tion to enhance coagulation reaction, a process known as thrombin burst [
2
,
3
]. The thrombin
time (TT) is generally used to detect abnormalities of fibrinogen, such as dysfibrinogen-
emia [
4
,
5
], disseminated intravascular coagulation (DIC) [
6
], and liver dysfunction [
7
].
Therefore, the TT is used to measure fibrinogen concentrations [
8
]. In addition, it is also
used to monitor anti-thrombin reagents [
9
]. Thrombin burst has generally been evaluated
by thromboelastography (TEG) [10] and the thrombin generation test (TGT) [11,12].
The activated partial thromboplastin time (APTT) and prothrombin time (PT),
which are inexpensive to conduct and allow for the easy performance of multiple
J. Clin. Med. 2021,10, 4840. https://doi.org/10.3390/jcm10214840 https://www.mdpi.com/journal/jcm
J. Clin. Med. 2021,10, 4840 2 of 8
assays, are established as routine assays for the coagulation system. However, where
the APTT and PT reflect only a single dimension, such as the clotting time, the TEG
and TGT are able to present results in two dimensions, including the time, width,
height, or area. Unfortunately, these tests are expensive and time-consuming to perform
compared to routine assays, at present.
A clot waveform analysis (CWA)-APTT [
13
,
14
] and small-amount tissue factor (TF)-
induced FIX activation (sTF/FIXa) assay [
14
,
15
] can show the peak time and peak height,
allowing the evaluation of thrombin burst to be performed as easily as a routine assay.
The CWA-APTT has been reported to detect very low levels of FVIII activity in patients
with hemophilia A [
16
], and has proven useful for the differential diagnosis of hemophilia,
acquired hemophilia A, lupus anticoagulant (LA), and DIC, as well as monitoring the
results of anticoagulant therapy or bypass therapy in patients with FVIII inhibitors [
17
21
].
In the present study, a CWA using TT (CWA-TT) for physiological coagulation was
used to investigate the mechanism underlying thrombin burst in calibration plasma and
plasma deficient of various clotting factors. We also demonstrate the role of platelets in the
coagulation system.
2. Materials and Methods
Platelet-rich plasma (PRP) and platelet-poor plasma (PPP) were collected from
12 patients with thrombocytopenia, 16 patients with malignant diseases, and 18 healthy
volunteers (8 males and 12 females; 21 to 56 years old). The TT was measured using 0.5 IU
thrombin (Thrombin 500 units, Mochida Pharmaceutical CO., LTD, Tokyo, Japan) with an
ACL-TOP
®
system (Instrumentation Laboratory, Bedford, MA, USA). Three types of curves
are shown on this system monitor [
19
]. One shows the changes in the absorbance observed
while measuring the TT, corresponding to the fibrin formation curve (FFC). The second
is the first derivative peak of the absorbance (1st DP), corresponding to the coagulation
velocity. The third is the second derivative peak of the absorbance (2nd DP), corresponding
to the coagulation acceleration. FII-deficient plasma, FV-deficient plasma, FVII-deficient
plasma, FVIII-deficient plasma, FIX-deficient plasma, FX-deficient plasma, FXI-deficient
plasma, FXII-deficient plasma (Instrumentation Laboratory), and FXIII-deficient plasma
(George King Bio-Medical Inc, Overland Park, KS, USA) were used as clotting factor-
deficient plasma, and calibration plasma (Instrumentation Laboratory) was used as normal
plasma. The fibrinogen concentrations in various deficient and calibration plasma were
measured using a Thrombocheck Fib (L) (Sysmex, Kobe, Japan) and CS-5100 (Sysmex).
PRP was prepared by centrifugation at 900 rpm for 15 min (platelet count, 40
×
10
10
/L
in healthy volunteers), and PPP was prepared by centrifugation at 3000 rpm for 15 min
(platelet count, <0.5 ×1010/L in healthy volunteers) [15].
Statistical Analyses
The data are expressed as the median (25th to 75th percentile). Differences between
PRP and PPP were examined for significance using Student’s t-test, and differences between
independent groups were examined using the Mann–Whitney U-test. p-values of
0.05
were considered to indicate statistical significance. All statistical analyses were performed
using the Stat flex software program (version 6. Artec Co Ltd., Osaka, Japan).
3. Results
The 2nd DP and 1st DP of calibration plasma using the CWA-TT with 0.01 IU/mL
of thrombin were detected at 270 s, and the peak times of CWA-TT gradually shortened,
whereas the peak heights of CWA-TT gradually increased, as the concentrations of thrombin
increased (Figure 1). The 2nd DP and 1st DP of FVIII-deficient plasma using the CWA-TT
with 0.01 to 0.1 IU/m of thrombin were not detected within 500 s, and the heights of the
2nd DP, 1st DP, and FFC of FVIII-deficient plasma using the CWA-TT with 0.5 to 1.0 IU/mL
of thrombin were significantly lower than those of calibration plasma using the CWA-TT
J. Clin. Med. 2021,10, 4840 3 of 8
with the same concentration of thrombin. A total of 5 IU/mL of thrombin showed similar
CWA-TT between calibration and FVIII-deficient plasma samples.
Figure 1.
A clot waveform analysis for thrombin time. (
I
) Calibration plasma; (
II
) FVIII-deficient plasma; (
a
) thrombin
0.01 IU/mL; (
b
) thrombin 0.05 IU/mL; (
c
) thrombin 0.1 IU/mL; (
d
) thrombin 0.5 IU/mL; (
e
) thrombin 1.0 IU/mL;
(
f
) thrombin 5.0 IU/mL. Navy line, fibrin formation curve; red line, 1st derivative curve (velocity); light blue, 2nd derivative
curve (acceleration).
The heights of the 2nd DP, 1st DP, and FFC in FII-, FV-, FVII-, FVIII-, FIX-, FX-, FXI-,
and FXII-deficient plasma using the CWA-TT with 0.5 IU thrombin were significantly
lower than those in calibration plasma and FXIII-deficient plasma using the CWA-TT with
0.5 IU thrombin (Figure 2). In particular, the heights of the 2nd DP, 1st DP, and FFC in FVIII-
deficient plasma were extremely low. The second peak of the 1st DP using the CWA-TT
was observed only in calibration plasma, and FVII-, FXI-, FXII-, and FXIII-deficient plasma.
The mean concentration of fibrinogen was 280 mg/dL in calibration plasma, 298 mg/dL
in FII-, 313 mg/dL in FV-, 311 mg/dL in FVII-, 276 mg/dL in FVIII-, 310 mg/dL in FIX-,
295 mg/dL in FX-, 306 mg/dL in FXI-, and 334 mg/dL in FXII-deficient plasma.
The absorbances of FFC at 100 s in the mixing tests (n= 3) between calibration plasma
and factor-deficient plasma showed dose dependence (Figure 3). The steep slope of the
dose-dependent curve was inclined toward larger values in FVIII-deficient plasma, and
toward smaller values in FXII-deficient plasma. The second peak of the 1st DP in normal
plasma from healthy volunteers was significantly shorter and higher in PRP than in PPP
(Figure 4).
J. Clin. Med. 2021,10, 4840 4 of 8
Figure 2.
A clot waveform analysis for thrombin time. (
a
) Calibration plasma; (
b
) FII-deficient plasma; (
c
) FV-deficient
plasma; (
d
) FVII-deficient plasma; (
e
) FVIII-deficient plasma; (
f
) FIX-deficient plasma; (
g
) FX-deficient plasma; (
h
) FXI-
deficient plasma; (
i
) FXII-deficient plasma; (
j
) FXIII-deficient plasma; thrombin 0.5 IU/mL. Navy line, fibrin formation
curve; red line, 1st derivative curve (velocity); light blue, 2nd derivative curve (acceleration).
Figure 3.
Mixing test between calibration plasma and plasma deficient of each factor using a clot waveform analysis for
thrombin time (0.5 IU/mL of thrombin). The height of fibrin formation at 100 s was plotted. The mean values of three
assays are shown. The standard deviation in each assay was less than 45 mm absorbance. DP, deficient plasma.
J. Clin. Med. 2021,10, 4840 5 of 8
Figure 4.
A clot waveform analysis of thrombin time (0.5 IU/mL) in platelet-poor plasma (
I
) and platelet-rich plasma
(
II
) from healthy volunteers (
a
), patients with thrombocytopenia (
b
), and patients with malignant disease (
c
). Navy line,
fibrin formation curve; red line, 1st derivative curve (velocity); light blue, 2nd derivative curve (acceleration); red arrow
shows 1st DPT-2.
Regarding the analysis of the CWA-TT between PPP and PRP from healthy volunteers,
the 2nd DPT and 1st DPT were significantly longer in PRP than in PPP, but the 1st DPT-2
and FFCT were significantly shorter in PRP than in PPP (Table 1). Although the 1st DPH-1
was significantly lower in PRP than in PPP, the 1st DPH-2 and FFCH were significantly
higher in PRP than in PPP. The CWA-TT using PPP did not show a second peak of the
1st-derivative in patients with thrombocytopenia, although the CWA-TT using PRP did
show a second peak of the 1st derivative. Using PPP for the CWA-TT, the 2nd DPT and
1st DPT-1 were significantly longer, and the 1st DPH-1 was significantly lower in patients
with thrombocytopenia than in healthy volunteers. The 2nd DPH, 1st DPH-1, 1st DPH-2,
and FFCH were significantly higher in patients with malignant disease than in healthy
volunteers (Table 2). Using PRP for the CWA-TT, the 1st DPT-2 and FFCT were significantly
longer in patients with thrombocytopenia than in healthy volunteers, and the FFCT was
significantly shorter in patients with malignant disease than in healthy volunteers. The
1st DPH-2 was significantly lower in patients with thrombocytopenia than in healthy
volunteers, and the 2nd DPH, 1st DPH-1, and 1st DPH-2 were significantly higher in
patients with malignant diseases than in healthy volunteers (Table 3).
Table 1. Parameters of CWA-TT in PPP and PRP from healthy volunteers.
2nd DPT
(seconds)
2nd DPH
(mm ABS)
1st DPT-1
(seconds)
1st DPH-1
(mm ABS)
1st DPT-2
(seconds)
1st DPH-2
(mm ABS)
FFCT
(seconds)
FFCH
(mm ABS)
PPP
(n= 18)
30.1 ***
(28.9–30.9)
164
(137–219)
36.3 **
(35.0–37.5)
113 *
(96.5–132)
234 ***
(220–278)
18.1 ***
(15.4–22.9)
194 ***
(185–204)
615 **
(560–664)
PRP
(n= 18)
37.4 ***
(33.1–43.3)
124
(110–180)
45.4 **
(39.2–50.2)
101 *
(82.5–120)
134 ***
(112–137)
45.3 ***
(40.0–62.1)
127 ***
(115–139)
730 **
(700–843)
Data are shown as the median (25–75 percentile). ***, p< 0.001, **, p< 0.01; *, p< 0.05 on comparing PPP and PRP. PPP, platelet-poor
plasma; PRP (n= 18), platelet-rich plasma (n= 18); 2nd DPT, second derivative peak time; 2nd DPH, second derivative peak height;
1st DPT, first derivative peak time; 1st DPH, first derivative peak height, FFCT, fibrin formation curve time; FFCH, fibrin formation
curve height; ABS, absorbance.
J. Clin. Med. 2021,10, 4840 6 of 8
Table 2.
Parameters of CWA-TT in PRP from healthy volunteers, patients with solid cancer, and patients with malignancy.
2nd DPT
(seconds)
2nd DPH
(mm ABS)
1st DPT-1
(seconds)
1st DPH-1
(mm ABS)
1st DPT-2
(seconds)
1st DPH-2
(mm ABS)
FFCT
(seconds)
FFCH
(mm ABS)
Healthy volunteers
(n= 18)
37.4
(33.1–43.3)
124
(110–180)
45.4
(39.2–50.2)
101
(82.5–120)
134
(112–137)
45.3
(40.0–62.1)
127
(115–139)
730
(700–842)
Thrombocytopenia
(n= 12)
35.6
(32.3–38.7)
174
(98.6–219)
43.9
(39.9–44.7)
115
(73.6–140)
188 ***
(146–216)
29.2 ***
(26.6–34.4)
153 *
(131–198)
763
(682–865)
Malignant diseases
(n= 16)
33.7
(30.1–38.5)
268 **
(185–339)
39.3
(34.7–43.0)
154 ***
(126–176)
111
(104–129)
82.2 ***
(76.4–108)
102 *
(98.2–118)
953
(813–1067)
Data are shown as the median (25–75 percentile). ***, p< 0.001, **, p< 0.01; *, p< 0.05 compared with healthy volunteers. PRP, platelet-rich
plasma; 2nd DPT, second derivative peak time; 2nd DPH, second derivative peak height; 1st DPT, first derivative peak time; 1st DPH, first
derivative peak height, FFCT, fibrin formation curve time; FFCH, fibrin formation curve height; ABS, absorbance.
Table 3.
Parameters of CWA-TT in PPP from healthy volunteers, patients with solid cancer, and patients with malignancy.
2nd DPT
(seconds)
2nd DPH
(mm ABS)
1st DPT-1
(seconds)
1st DPH-1
(mm ABS)
1st DPT-2
(seconds)
1st DPH-2
(mm ABS)
FFCT
(seconds)
FFCH
(mm ABS)
Healthy volunteers
(n= 18)
30.5
(28.9–30.9)
164
(137–219)
36.3
(35.0–37.5)
113
(96.5–132)
234
(220–278)
18.1
(15.4–22.9)
194
(185–204)
615
(560–664)
Thrombocytopenia
(n= 12)
34.9 *
(31.0–39.2)
143
(78.5–255)
43.2 *
(37.6–47.5)
106 *
(70.2–157)
Not
detectable
Not
detectable
202
(152–209)
550
(492–637)
Malignant diseases
(n= 16)
30.3
(28.6–46.1)
321 *
(137–420)
35.0
(33.7–52.7)
166 ***
(103–207)
228
(173–273)
28.5 **
(21.6–43.3)
187
(157–236)
738 **
(656–874)
Data are shown as the median (25–75 percentile). ***, p< 0.001, **, p< 0.01; *, p< 0.05 compared with healthy volunteers. PPP, platelet-poor
plasma; 2nd DPT, second derivative peak time; 2nd DPH, second derivative peak height; 1st DPT, first derivative peak time; 1st DPH, first
derivative peak height, FFCT, fibrin formation curve time; FFCH, fibrin formation curve height; ABS, absorbance.
4. Discussion
The CWA-TT with FVIII-deficient plasma showed that a small amount of thrombin
(
0.1 IU/mL) failed to induce clot formation, suggesting that FVIII is required for phys-
iological coagulation. As FVIII is reported to be markedly catalyzed and activated by
thrombin [
22
], FVIII may play an important role in thrombin burst. Furthermore, the
CWA-TT in plasma deficient of various clotting factors showed prolonged peak time,
and decreased peak height. These findings suggest that various clotting factors are also
required for the coagulation system involving thrombin burst induced by a small amount
of thrombin [
4
,
14
]. However, CWA-TT reflects thrombin burst at thrombin concentrations
1.0 IU/mL, whereas at thrombin concentrations
5.0 IU/mL, CWA-TT strongly reflects
the fibrinogen concentration [
23
]. In mixing texts to evaluate thrombin burst, a test with
calibration plasma and FII-deficient plasma is useful as a control without thrombin burst,
as FII-deficient plasma cannot cause a cycle of thrombin burst resulting in fibrin clot forma-
tion without thrombin burst. Therefore, a mixing test using CWA-TT proves that many
coagulation factors, except for FXII, in which the millimeter absorbance was lower than
that with FII-deficient plasma, may play an important role in thrombin burst.
In addition, the CWA-TT may be useful for evaluating the physiological and patho-
logical coagulation induced by a small amount of thrombin. Physiological coagulation
starts after small amounts of TF and FVIIa activate FIX, resulting in a small amount of
thrombin. This thrombin activates not only fibrinogen, but also FV, FVIII, FIX, FX, and FXI,
with the activation cycle from thrombin to FXI continuing for a short time thereafter [
14
].
The CWA-sTF/FIXa with a 2000-fold diluted PT reagent (recombinant TF) [24,25] was de-
veloped to evaluate physiological coagulation, and was shown to be capable of measuring
the FVIII concentration. As a cross-mixing test of the CWA-TT between calibration plasma
and FVIII-deficient plasma showed a good dose-response curve, the CWA-TT may be able
to measure the FVIII concentration.
J. Clin. Med. 2021,10, 4840 7 of 8
The physiological coagulation system includes enhancement of clotting activation
by phospholipids of platelets. However, most APTT reagents have some contact acti-
vation substance, and cannot demonstrate physiological coagulation [
15
]. Therefore, an
sTF/FIXa assay [
15
,
24
] uses PRP as a physiological phospholipid instead of commercial
APTT reagents. The CWA-TT also showed that the peak height, especially the 1st DPH-2,
was higher in PRP than in PPP. The 1st DPH-2 was absent in PPP, and low in PRP. Activated
FVII has been reported to generate thrombin in hemophilia via both platelet-dependent and
platelet-independent mechanisms [
26
]. These findings therefore suggest that the thrombin
burst mechanism may depend, at least partially, on platelets. However, the peak heights of
the CWA-TT were extremely high, suggesting that patients with malignant diseases are in
a hypercoagulable state. Further studies of the CWA-TT will be required to investigate its
utility for the differential diagnosis of thrombocytopenia and hypercoagulability.
5. Conclusions
The peak time and height of CWA-TT became abnormal in plasma deficient of various
clotting factors, and the CWA-TT was markedly affected by platelet counts, suggesting that
the CWA-TT may be useful in testing for hemostatic abnormalities, such as thrombocytope-
nia or hypercoagulability.
Author Contributions:
Conceptualization, H.W.; methodology, Y.I., M.E.; validation, K.S., formal
analysis, H.W.; investigation, T.M., Y.Y.; resources, Y.I.; data curation, M.E.; writing—original draft
preparation, H.W.; writing—review and editing, M.S.; visualization, H.W.; supervision, H.S.; project
administration, K.S.; funding acquisition, H.W. All authors have read and agreed to the published
version of the manuscript.
Funding:
This research was funded by a Grant-in-Aid from the Ministry of Health, Labour and
Welfare of Japan (H30-015), and the Rare/Intractable Disease Project of Japan from Japan Agency for
Medical Research and Development, AMED.
Institutional Review Board Statement:
The study protocol (O-0057) was approved by the Human
Ethics Review committees of Mie Prefectural General Medical Center, and informed consent was
obtained from each patient.
Informed Consent Statement:
Informed consent was obtained from all subjects involved in the study.
Data Availability Statement:
The data presented in this study are available on request to the corre-
sponding author. The data are not publicly available due to privacy restrictions.
Acknowledgments:
The authors thank Nisii H and Sakano Y for their kind support in performing
the assay for the CWA.
Conflicts of Interest:
The measurements of CWA were partially supported by Instrumentation
Laboratory Japan. In the other points, the authors declare no conflict of interest.
References
1.
Winter, W.E.; Greene, D.N.; Beal, S.G.; Isom, J.A.; Manning, H.; Wilkerson, G.; Harris, N. Clotting factors: Clinical biochemistry
and their roles as plasma enzymes. Int. Rev. Cytol. 2020,94, 31–84. [CrossRef]
2.
Berntorp, E.; Salvagno, G.L. Standardization and clinical utility of thrombin-generation assays. Semin. Thromb. Hemost.
2008
,34,
670–682. [CrossRef] [PubMed]
3.
Salvagno, G.L.; Berntorp, E. thrombin generation testing for monitoring hemophilia treatment: A clinical perspective. Semin.
Thromb. Hemost. 2010,36, 780–790. [CrossRef]
4.
Li, S.; Wang, M.; Li, X.; Xu, Q.; Liu, S.; Luo, S.; Chen, Y. Analysis of an Inherited dysfibrinogenemia pedigree associated with a
heterozygous mutation in the FGA Gene. Hämostaseologie 2020,40, 642–648. [CrossRef] [PubMed]
5.
Marchi, R.; Neerman-Arbez, M.; Gay, V.; Mourey, G.; Fiore, M.; Mouton, C.; Gautier, P.; De Moerloose, P.; Casini, A. Comparison
of different activators of coagulation by turbidity analysis of hereditary dysfibrinogenemia and controls. Blood Coagul. Fibrinolysis
2021,32, 108–114. [CrossRef]
6.
Haupt, L.; Vieira, M.; Brits, H.; de Beer, J.; Erasmus, E.; Esterhuyse, W.; Fraser, R.; Joubert, G. An audit of disseminated
intravascular coagulation screen requests at an academic hospital laboratory in central South Africa. Int. J. Lab. Hematol.
2021
,43,
1174–1180. [CrossRef]
J. Clin. Med. 2021,10, 4840 8 of 8
7.
Martinez, J.; Macdonald, K.A.; Palascak, J.E. The role of sialic acid in the dysfibrinogenemia associated with liver disease:
Distribution of sialic acid on the constituent chains. Blood 1983,61, 1196–1202. [CrossRef]
8. Winter, W.E.; Flax, S.D.; Harris, N.S. Coagulation Testing in the Core Laboratory. Lab. Med. 2017,48, 295–313. [CrossRef]
9.
Van Cott, E.M.; Roberts, A.J.; Dager, W.E. Laboratory monitoring of parenteral direct thrombin inhibitors. Semin. Thromb. Hemost.
2017,43, 270–276.
10.
Konstantinidi, A.; Sokou, R.; Parastatidou, S.; Lampropoulou, K.; Katsaras, G.; Boutsikou, T.; Gounaris, A.K.; Tsantes, A.E.;
Iacovidou, N. Clinical Application of Thromboelastography/Thromboelastometry (TEG/TEM) in the Neonatal Population: A
Narrative Review. Semin. Thromb. Hemost. 2019,45, 449–457. [CrossRef]
11.
Tripodi, A. Thrombin Generation Assay and Its Application in the Clinical Laboratory. Clin. Chem.
2016
,62, 699–707. [CrossRef]
[PubMed]
12.
Bendetowicz, A.V.; Kai, H.; Knebel, R.; Caplain, H.; Hemker, H.C.; Lindhout, T.; Béguin, S. The effect of subcutaneous injection of
unfractionated and low molecular weight heparin on thrombin generation in platelet rich plasma—A study in human volunteers.
Thromb. Haemost. 1994,72, 705–712. [CrossRef]
13.
Maeda, K.; Wada, H.; Shinkai, T.; Tanemura, A.; Matsumoto, T.; Mizuno, S. Evaluation of hemostatic abnormalities in patients
who underwent major hepatobiliary pancreatic surgery using activated partial thromboplastin time-clot waveform analysis.
Thromb. Res. 2021,201, 154–160. [CrossRef] [PubMed]
14.
Wada, H.; Matsumoto, T.; Ohishi, K.; Shiraki, K.; Shimaoka, M. Update on the clot waveform analysis. Clin. Appl. Thromb.
2020
,26.
[CrossRef] [PubMed]
15.
Wada, H.; Shiraki, K.; Matsumoto, T.; Ohishi, K.; Shimpo, H.; Shimaoka, M. Effects of platelet and phospholipids on clot formation
activated by a small amount of tissue factor. Thromb. Res. 2020,193, 146–153. [CrossRef]
16.
Matsumoto, T.; Fukuda, K.; Kubota, Y.; Tanaka, I.; Nishiya, K.; Giles, A.R.; Yoshioka, A.; Shima, M. The Utility of activated Partial
Thromboplastin Time (aPTT) Clot Waveform Analysis in the Investigation of Hemophilia A Patients with very Low Levels of
Factor VIII Activity (FVIII:C). Thromb. Haemost. 2002,87, 436–441. [CrossRef]
17.
Matsumoto, T.; Nogami, K.; Shima, M. A combined approach using global coagulation assays quickly differentiates coagulation
disorders with prolonged aPTT and low levels of FVIII activity. Int. J. Hematol. 2016,105, 174–183. [CrossRef]
18.
Tokutake, T.; Baba, H.; Shimada, Y.; Takeda, W.; Sato, K.; Hiroshima, Y.; Kirihara, T.; Shimizu, I.; Nakazawa, H.; Kobayashi,
H.; et al. Exogenous magnesium chloride reduces the activated partial thromboplastin times of lupus anticoagulant-positive
patients. PLoS ONE 2016,11, e0157835. [CrossRef]
19.
Matsumoto, T.; Wada, H.; Fujimoto, N.; Toyoda, J.; Abe, Y.; Ohishi, K.; Yamashita, Y.; Ikejiri, M.; Hasegawa, K.; Suzuki, K.; et al.
An evaluation of the activated partial thromboplastin time waveform. Clin. Appl. Thromb. 2017,24, 764–770. [CrossRef]
20.
Hasegawa, M.; Wada, H.; Tone, S.; Yamaguchi, T.; Wakabayashi, H.; Ikejiri, M.; Watanabe, M.; Fujimoto, N.; Matsumoto, T.;
Ohishi, K.; et al. Monitoring of hemostatic abnormalities in major orthopedic surgery patients treated with edoxaban by APTT
waveform. Int. J. Lab. Hematol. 2017,40, 49–55. [CrossRef] [PubMed]
21.
Suzuki, K.; Wada, H.; Matsumoto, T.; Ikejiri, M.; Ohishi, K.; Yamashita, Y.; Imai, H.; Iba, T. Katayama N: Usefulness of the APTT
waveform for the diagnosis of DIC and prediction of the outcome or bleeding risk. Thromb. J.
2019
,17, 12. [CrossRef] [PubMed]
22.
Nakajima, Y.; Nogami, K. The C-terminal acidic region in the A1 domain of factor VIII facilitates thrombin-catalyzed activation
and cleavage at Arg 372. J. Thromb. Haemost. 2021,19, 677–688. [CrossRef]
23.
Suzuki, A.; Suzuki, N.; Kanematsu, T.; Shinohara, S.; Arai, N.; Kikuchi, R.; Matsushita, T. Clot waveform analysis in Clauss
fibrinogen assay contributes to classification of fibrinogen disorders. Thromb. Res. 2019,174, 98–103. [CrossRef] [PubMed]
24.
Hasegawa, M.; Tone, S.; Wada, H.; Naito, Y.; Matsumoto, T.; Yamashita, Y.; Shimaoka, M.; Sudo, A. The evaluation of hemostatic
abnormalities using a CWA-Small amount tissue factor induced fix activation assay in major orthopedic surgery patients. Clin.
Appl. Thromb. 2021,27. [CrossRef] [PubMed]
25.
Matsumoto, T.; Wada, H.; Toyoda, H.; Hirayama, M.; Yamashita, Y.; Katayama, N. Modified clot waveform analysis to measure
plasma coagulation potential in the presence of the anti-factor IXa/factor X bispecific antibody emicizumab: Comment. J. Thromb.
Haemost. 2018,16, 1665–1666. [CrossRef]
26.
Keshava, S.; Pendurthi, U.R.; Esmon, C.T.; Rao, L.V.M. Vijaya Mohan Rao: Therapeutic doses of recombinant factor VIIa in
hemophilia generates thrombin in platelet-dependent and -independent mechanisms. J. Thromb. Haemost.
2020
,18, 1911–1921.
[CrossRef]
... Furthermore, the use of a small scale of TF-induced FIX activation (sTF/FIXa) (CWA-sTF/ FIXa) assay can evaluate hemostatic abnormalities including platelet abnormalities [17]. Hypercoagulability is considered to be caused by a thrombin burst [20,21] which is evaluated by thrombin time using a small amount of thrombin with a CWA (CWA-TT) [22]. ...
... The CWA-TT was measured using 0.5 IU thrombin (Thrombin 500 units Mochida Pharmaceutical CO., LTD, Tokyo, Japan) with an ACL-TOP ® system (Instrumentation Laboratory) [17]. Three types of curves are shown on this system monitor [22]. The 1 st DP showed two peaks and the second peak height of the 1 ST DP (1 st DPH-2) reflected the thrombin burst. ...
... Markedly high peak heights of the sTF/FIXa using small amount of TF suggest that intrinsic TF released from cancer cells exists in plasma. Thrombin causes an elevation in FVIII activity and increases the activation cycle from thrombin to FXIa, called the thrombin burst [22]. Elevated peak heights, especially the second peak of the 1 st DPH (1 st DPH-2) of CWA-TT suggest that the thrombin burst worsens hypercoagulability in cancer patients including those with pancreatic cancer. ...
Article
Background: Cancer, especially pancreatic cancer, is frequently associated with thrombosis which is one of the causes of poor outcomes; moreover hypercoagulability can be present in cancer patients. Hypercoagulability is considered to be caused by a thrombin burst. Methods: Activated Partial Tthromboplastin Time (APTT), small amount of tissue factor induced FIX activation assay (sTF/FIXa) and Thrombin Time (TT) assessment using Clot Waveform Analysis (CWA) were performed in 138 patients with malignant neoplasms, including pancreatic cancer. Results: The first derivative peak (1st DP) time (1st DPT), 1st DP height (1st DPH) and 1st DPH/1st DPT ratio were increased in a clotting-factor-FVIII-dependent manner. Thrombosis was frequently associated with pancreatic cancer and was observed in the early stage. CWA-APTT and CWA-sTF/FIXa indicated that the peak times and heights were markedly longer and higher, respectively, in cancer patients, especially pancreas cancer patients, than in patients without cancer. The 1st DPH/1st DPT ratios of CWA-sTF/FIXa were significantly high in patients with pancreas cancer (median value 1.5). CWA-TT showed that the peak times were significantly shorter in cancer patients than in healthy volunteers and that the peak heights were significantly higher in cancer than in benign pancreas diseases. The cutoff value of the 1st DPH/ 1st DPT of sTF/FIXa for cancer patients with thrombosis vs. all patients without cancer was 1.3. Conclusions: Cancer patients, including those with pancreatic cancer were frequently associated with thrombosis due to hypercoagulability caused by thrombin burst detected by CWA. A high 1st DPH/1st DPT ratio of sTF/FIXa may suggest an association with cancer or thrombosis.
... However, few reports have described the relationship between FVIII activity, which is assessed using the peak time and height of CWA-APTT, including a small amount of tissue factor-induced activated FIX (sTF/FIXa) assay [15]. A small amount of thrombin reflects the thrombin burst, including the activation of FXI, FVIII, and FV instead of fibrinogen, and thrombin time (TT) using only a small amount of thrombin can evaluate the intrinsic pathway and FVIII activity [16]. Recently, it was reported that CWA-TT can measure FVIII activity independent of the presence of emicizumab [17]. ...
... This study was conducted in accordance with the principles of the Declaration of Helsinki. The CWA-TT ( Figure 1a) using 0.5 IU thrombin (Thrombin 500 units; Mochida Pharmaceutical Co., Ltd., Tokyo, Japan) was measured using an ACL-TOP ® system (Instrumentation Laboratory, Bedford, MA, USA) [16,17]. This system shows three types of curves [16,17]. ...
... The CWA-TT ( Figure 1a) using 0.5 IU thrombin (Thrombin 500 units; Mochida Pharmaceutical Co., Ltd., Tokyo, Japan) was measured using an ACL-TOP ® system (Instrumentation Laboratory, Bedford, MA, USA) [16,17]. This system shows three types of curves [16,17]. One illustrates the changes in the absorbance observed while measuring CWA-TT, corresponding to the fibrin formation curve (FFC). ...
Article
Full-text available
Background: Regular prophylactic therapy has become an increasingly common treatment for severe hemophilia. Therefore, hypercoagulability-a potential risk factor of thrombosis-is a cause for concern in hemophilic patients treated with a high dose of FVIII concentrate. In clot waveform analysis (CWA)-thrombin time (TT), a small amount of thrombin activates clotting factor VIII (FVIII) instead of fibrinogen, resulting in FVIII measurements using CWA-TT with a small amount of thrombin. Methods: The coagulation ability of patients treated with FVIII concentrate or emicizumab was evaluated using activated partial thromboplastin time (APTT), TT and a small amount of tissue factor-induced FIX activation assay (sTF/FIXa) using CWA. Results: The FVIII activity based on CWA-TT was significantly greater than that based on the CWA-APTT or chromogenic assay. FVIII or FVIII-like activities based on the three assays in plasma without emicizumab were closely correlated; those in plasma with emicizumab based on CWA-TT and chromogenic assays were also closely correlated. CWA-APTT and CWA-TT showed different patterns in patients treated with FVIII concentrates compared to those treated with emicizumab. In particular, CWA-TT in patients treated with FVIII concentrate showed markedly higher peaks in platelet-rich plasma than in platelet-poor plasma. CWA-APTT showed lower coagulability in hemophilic patients treated with FVIII concentrate than in healthy volunteers, whereas CWA-sTF/FIXa did not. In contrast, CWA-TT showed hypercoagulability in hemophilic patients treated with FVIII concentrate. Conclusions: CWA-TT can be used to evaluate the thrombin bursts that cause hypercoagulability in patients treated with emicizumab. Although routine APTT evaluations demonstrated low coagulation ability in patients treated with FVIII concentrate, CWA-TT showed hypercoagulability in these patients, suggesting that the evaluation of coagulation ability may be useful when using multiple assays.
... However, few reports have described this relationship between the FVIII activity assessed using the peak time and height of CWA-APTT, including a small amount of tissue factor-induced activated FIX (sTF/FIXa) assay [14]. A CWA-small amount of thrombin time (CWA-TT) also reflects thrombin burst and FVIII activity [15] and can be used to measure the FVIII activity independent of the presence of emicizumab [16]. ...
... This study was carried out in accordance with the principles of the Declaration of Helsinki. The CWA-TT was measured using 0.5 IU thrombin (Thrombin 500 units; Mochida Pharmaceutical Co., Ltd., Tokyo, Japan) with an ACL-TOP ® system (Instrumentation Laboratory, Bedford, MA, USA) [15,16]. Three types of curves are shown on this system monitor [15,16]. ...
... The CWA-TT was measured using 0.5 IU thrombin (Thrombin 500 units; Mochida Pharmaceutical Co., Ltd., Tokyo, Japan) with an ACL-TOP ® system (Instrumentation Laboratory, Bedford, MA, USA) [15,16]. Three types of curves are shown on this system monitor [15,16]. One shows the changes in the absorbance observed while measuring the TT, corresponding to the fibrin formation curve (FFC). ...
Preprint
Full-text available
Background: Although the use of regular replacement therapy including emicizumab for severe hemophilia has been spread, the assessment of the hemostatic ability using routine activated partial thromboplastin time (APTT) is still difficult in patients being treated with emicizumab. Methods: The hemostatic ability in patients treated with FVIII concentrate or emicizumab was evaluated by APTT, thrombin time (TT) and a small amount of tissue factor induced FIX activation assay (sTF/FIXa) using a clot waveform analysis (CWA). Results: FVIII activities based on a CWA-TT were significantly higher than those based on a CWA-APTT or chromogenic assay. FVIII activities based on the three assays in plasma without emicizumab were closely correlated, and those in plasma with emicizumab based on a CWA-TT and chromogenic assays were also closely correlated. The CWA-APTT and CWA-TT showed different patterns in patients treated with FVIII concentrates from those treated with emicizumab. In particular, the CWA-TT in patients treated with FVIII concentrate showed that the peak heights were significantly higher in platelet-rich plasma than in platelet-poor plasma. In plasma with approximately 16% of FVIII activity based on APTT assay from patients treated with FVIII concentrate, the peak height on the CWA-sTF/FIXa showed a higher hemostatic abilitythan normal plasma. Conclusions: The CWA-TT can measure the FVIII activity in patients treated with emicizumab. Although routine APTT evaluations demonstrate a low hemostatic ability in patients treated with FVIII concentrate, the CWA-TT and CWA-sTF/FIXa show hypercoagulability in those patients.
... Clot waveform analysis (CWA) [1][2][3][4][5] is based on the activated partial thromboplastin time (APTT) [1][2][3], prothrombin time (PT) [4], or thrombin time (TT) [5] (CWA-APTT, CWA-PT, and CWA-TT, respectively) ( Table 1). Although conventional clotting assays such as the APTT, PT, and TT are inexpensive, easy, and automated, enabling the measurement of multiple samples, they cannot visualize the clotting process. ...
... Clot waveform analysis (CWA) [1][2][3][4][5] is based on the activated partial thromboplastin time (APTT) [1][2][3], prothrombin time (PT) [4], or thrombin time (TT) [5] (CWA-APTT, CWA-PT, and CWA-TT, respectively) ( Table 1). Although conventional clotting assays such as the APTT, PT, and TT are inexpensive, easy, and automated, enabling the measurement of multiple samples, they cannot visualize the clotting process. ...
... Modified CWA is based on CWA-dilute PT [32,35], dilute TT [5], and clot-fibrinolysis waveform analysis (CFWA) [36,37]. CWAdilute PT shows both the extrinsic and intrinsic pathways. ...
Article
Clot waveform analysis (CWA) observes changes in transparency in a plasma sample based on clotting tests such as activated partial thromboplastin time (APTT), prothrombin time (PT), and thrombin time (TT). Evidence indicates that not only an abnormal waveform but also peak times and heights in derivative curves of CWA are useful for the evaluation of hemostatic abnormalities. Modified CWA, including the PT with APTT reagent, dilute PT (small amount of tissue factor [TF]-induced clotting factor IX [FIX] activation; sTF/FIXa), and dilute TT, has been proposed to evaluate physiological or pathological hemostasis. We review routine and modified CWA and their clinical applications. In CWA-sTF/FIXa, elevated peak heights indicate hypercoagulability in patients with cancer or thrombosis, whereas prolonged peak times indicate hypocoagulability in several conditions, including clotting factor deficiency and thrombocytopenia. CWA-dilute TT reflects the thrombin burst, whereas clot-fibrinolysis waveform analysis reflects both hemostasis and fibrinolysis. The relevance and usefulness of CWA-APTT and modified CWA should be further investigated in various diseases.
... reported to be caused by a thrombin burst. [53][54][55][56] Although the mechanism underlying the prolonged clotting time in patients with HCC may involve hepatic dysfunction, the concomitance of both prolonged peak times and increased peak heights cannot be sufficiently understood. These findings in the CWA of prolonged peak times and increased peak heights indicate that a strong coagulation process might continue for a long time, suggesting that a thrombotic risk may exist in patients with HCC. ...
Article
Full-text available
Background: Although hepatocellular carcinoma (HCC) is frequently associated with thrombosis, it is also associated with liver cirrhosis (LC) which causes hemostatic abnormalities. Therefore, hemostatic abnormalities in patients with HCC were examined using a clot waveform analysis (CWA). Methods: Hemostatic abnormalities in 88 samples from HCC patients, 48 samples from LC patients and 153 samples from patients with chronic liver diseases (CH) were examined using a CWA-activated partial thromboplastin time (APTT) and small amount of tissue factor induced FIX activation (sTF/FIXa) assay. Results: There were no significant differences in the peak time on CWA-APTT among HCC, LC, and CH, and the peak heights of CWA-APTT were significantly higher in HCC and CH than in HVs and LC. The peak heights of the CWA-sTF/FIXa were significantly higher in HCC than in LC. The peak times of the CWA-APTT were significantly longer in stages B, C, and D than in stage A or cases of response. In the receiver operating characteristic (ROC) curve, the fibrin formation height (FFH) of the CWA-APTT and CWA-sTF/FIXa showed the highest diagnostic ability for HCC and LC, respectively. Thrombosis was observed in 13 HCC patients, and arterial thrombosis and portal vein thrombosis were frequently associated with HCC without LC and HCC with LC, respectively. In ROC, the peak time×peak height of the first derivative on the CWA-sTF/FIXa showed the highest diagnostic ability for thrombosis. Conclusion: The CWA-APTT and CWA-sTF/FIXa can increase the evaluability of HCC including the association with LC and thrombotic complications.
... Elevated PT and APTT values signify prolonged clotting time, impaired coagulation function, and increased bleeding risk [17,18]. TT evaluates coagulation function by examining plasma fibrinogen's capacity to form fibrin polymers [19]. Prolonged TT indicates potential dysfunction in fibrinogen-to-fibrin conversion [20]. ...
... FIB is the substrate of the process, so its level affects the length of TT. 30,31 Based on the activation of FVIII, TT can reflect a thrombin burst, which was synergistic with the generation of FIXa caused by the process that APTT activated the intrinsic pathway, thus contributing to the conversion of FIB to fibrin. 31,32 In our study, the results suggested the decrease of TT with accompanying by the increase of FIB. The shorter TT in the adenomyosis patients than in the patients with uterine leiomyoma or the control group may be due to the higher level of FIB in the adenomyosis. ...
Article
Full-text available
Objective Adenomyosis patients are in a hypercoagulable state, and studies have shown that carbohydrate antigen125 (CA125) may relate to the hypercoagulability and thrombosis of patients with adenomyosis, but there is still a lack of clarity regarding the changes in CA125-related coagulation indicators. This study was to explore the changes and influencing factors of CA125-related coagulation parameters in patients with adenomyosis. Methods Retrospective observational study conducted on 200 patients with adenomyosis (AM group), 240 patients with uterine leiomyoma (LM group) and 81 patients with cervical intraepithelial neoplasia (CIN)-III (control group), of which the coagulation parameters were detected by clinical blood sample collection and statistical method analysis and informed consent was obtained. Results The level of CA125 in the AM group was significantly higher than that in the LM group and control group. However, thrombin time (TT) shortened in the AM group when compared with the LM and control group. Activated partial thromboplastin time (APTT) in the AM group was shorter than in the control group. Multivariate logistic regression analysis found that adenomyosis was associated with CA125 level (OR=323.860, 95% CI 90.424–1159.924, P<0.001), APTT (OR=1.295, 95% CI 1.050–1.598, P=0.016), TT (OR=0.642, 95% CI 0.439–0.938, P=0.022), menorrhagia (OR=7.363, 95% CI 2.544–21.315, P<0.001), dysmenorrhea (OR=22.590, 95% CI 8.185–62.347, P<0.001). Correlation analysis revealed that APTT (r= −0.207) and TT (r = −0.174) were negatively correlated with the level of CA125. Conclusion The shortening of CA125-related APTT and TT indicates that it is meaningful to detect coagulation parameters of patients with elevated CA125 levels early, dysmenorrhea and menorrhagia, and maybe further discover the hypercoagulability and prevent the occurrence of thrombus in adenomyosis.
... Although PT was the most important indicator for detecting the coagulation status of patients, it was influenced by various factors such as liver synthesis function and inflammatory factors [42]. TT was the time at which fibrinogen was converted into fibrin after the addition of thrombin, and the prolongation of TT to a certain extent reflected the level and state of fibrinogen [43]. The decrease in fibrinogen levels also indicated liver synthesis dysfunction, reflecting long-term damage to liver function [44]. ...
Article
Full-text available
Objective The predictive biomarkers of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC) still need to be further explored. This study aims to establish a new immune prognosis biomarker to predict the clinical outcomes of hepatocellular carcinoma patients receiving immune checkpoint inhibitors. Methods The subjects of this study were 151 HCC patients receiving ICIs at Harbin Medical University Cancer Hospital from January 2018 to December 2021. This study collected a wide range of blood parameters from patients before treatment and used Cox’s regression analysis to identify independent prognostic factors in blood parameters, as well as their β coefficient. The hepatocellular carcinoma immune prognosis score (HCIPS) was established through Lasso regression analysis and COX multivariate analysis. The cut-off value of HCIPS was calculated from the receiver operating characteristic (ROC) curve. Finally, the prognostic value of HCIPS was validated through survival analysis, stratified analyses, and nomograms. Results HCIPS was composed of albumin (ALB) and thrombin time (TT), with a cut-off value of 0.64. There were 56 patients with HCIPS < 0.64 and 95 patients with HCIPS ≥ 0.64, patients with low HCIPS were significantly related to shorter progression-free survival (PFS) (13.10 months vs. 1.63 months, P < 0.001) and overall survival (OS) (14.83 months vs. 25.43 months, P < 0.001). HCIPS has also been found to be an independent prognostic factor in this study. In addition, the stratified analysis found a significant correlation between low HCIPS and shorter OS in patients with tumor size ≥ 5 cm ( P of interaction = 0.032). The C-index and 95% CI of the nomograms for PFS and OS were 0.730 (0.680–0.779) and 0.758 (0.711–0.804), respectively. Conclusions As a new score established based on HCC patients receiving ICIs, HCIPS was significantly correlated with clinical outcomes in patients with ICIs and might serve as a new biomarker to predict HCC patients who cloud benefit from ICIs.
... Regarding COVID -19 (Figure 3), leukocyte counts are generally decreased early in COVID-19 [8], suggesting that activated platelets and injured vascular endothelial cells may play an important role in the onset of thrombosis through CD-147 [53]. However, CWA-APTT and CWA-sTF/FIXa showed hypercoagulability in patients with COVID-19 [41] suggesting that thrombin burst (Figure 2) [83] which is enhanced by activated platelets, causes hypercoagulability in this state. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) has spread, with thrombotic complications being increasingly frequently reported. Although thrombosis is frequently complicated in septic patients, there are some differences in the thrombosis noted with COVID-19 and that noted with bacterial infections. The incidence (6–26%) of thrombosis varied among reports in patients with COVID-19; the incidences of venous thromboembolism and acute arterial thrombosis were 4.8–21.0% and 0.7–3.7%, respectively. Although disseminated intravascular coagulation (DIC) is frequently associated with bacterial infections, a few cases of DIC have been reported in association with COVID-19. Fibrin-related markers, such as D-dimer levels, are extremely high in bacterial infections, whereas soluble C-type lectin-like receptor 2 (sCLEC-2) levels are high in COVID-19, suggesting that hypercoagulable and hyperfibrinolytic states are predominant in bacterial infections, whereas hypercoagulable and hypofibrinolytic states with platelet activation are predominant in COVID-19. Marked platelet activation, hypercoagulability and hypofibrinolytic states may cause thrombosis in patients with COVID-19.
Article
Full-text available
We analyzed the utility for a clot waveform analysis (CWA) of small tissue factor induced FIX activation (sTF/FIXa) assay in patients with major orthopedic surgery (including total hip arthroplasty [THA] and total knee arthroplasty [TKA]) receiving edoxaban for the prevention of venous thromboembolism (VTE). The sTF/FIXa assay using recombinant human TF in platelet-rich plasma (PRP) and platelet-poor plasma (PPP) was performed using a CWA in the above patients to monitor the efficacy of edoxaban administration. Of 147 patients (109 THA and 38 TKA), 21 exhibited deep vein thrombosis (DVT), and 15 had massive bleeding. Increased peak heights of the CWA-sTF/FIX were observed in almost patients after surgery and prolonged peak heights of the CWA-sTF/FIX were observed in almost patients treated with edoxaban. The peak heights and times of the CWA-sTF/FIX were significantly higher and shorter, respectively, in PRP than in PPP. There were no significant differences in parameters of the CWA-sTF/FIXa between the patients with and without DVT or between those with and without massive bleeding. The peak time of CWA-sTF/FIXa were significantly longer in TKA patients than in THA patients on day 1 after surgery. The second derivative peak height of the CWA-sTF/FIXa was significantly lower in TKA patients than in THA patients on day 4. The CWA-sTF/FIX reflected hemostatic abnormalities after surgery and the administration of edoxaban, and the results were better in PRP than PPP. Further studies separately analyzing the THA and TKA subgroups should be conducted.
Article
Full-text available
Turbidity analysis is widely used as a quantitative technique in hereditary dysfibrinogenemia. We aimed to compare several coagulation triggers in hereditary dysfibrinogenemia and control plasmas. We included 20 patients with hereditary dysfibrinogenemia, 19 with hotspot mutations Aα Arg35His (n = 9), Aα Arg35Cys (n = 2), γ Arg301His (n = 6), γ Arg301Cys (n = 2), and one with Aα Phe27Tyr, and a commercial pooled normal plasma. Fibrin polymerization was activated by bovine or human thrombin or tissue factor (TF), in the presence or absence of tissue type plasminogen activator. The lag time (min), slope (mOD/s), maximum absorbance (MaxAbs, mOD), and area under the curve (AUCp, OD s) were calculated from the fibrin polymerization curves and the time for 50% clot degradation (T50, min), AUCf (OD s) and the overall fibrinolytic potential from fibrinolysis curves. The lag time was significantly shorter and AUC increased in Aα Arg35His patients with bovine thrombin as compared with human thrombin. The MaxAbs and AUCp were significantly higher in γArg301His patients with bovine thrombin compared with human thrombin. Fibrin polymerization parameters of patients' samples were closer to those of control when assessed with TF compared with both human and bovine thrombin. T50 and overall fibrinolytic potential were similar in all samples regardless of the coagulation trigger used, however, with TF the AUCf of Aα Arg35His and γ Arg301His groups were significantly decreased compared with control. Bovine and human thrombin cannot be used equally for studying fibrin polymerization in hotspot hereditary dysfibrinogenemia or control plasmas.
Article
Full-text available
The activated partial thromboplastin time (APTT)–clot waveform analysis (CWA) was previously reported to be associated with the early detection of disseminated intravascular coagulation and was also reported to be able to measure very low levels of coagulation factor VIII activity. The software program for the analysis for the APTT-CWA allows the associated first and second derivative curves (first and second DCs) to be displayed. The first and second DC reflect the velocity and acceleration, respectively. The height of the first DC reflects the “thrombin burst” and bleeding risk, while that of the second DC is useful for detecting any coagulation factor deficiency and abnormal enhancement of coagulation by phospholipids. Activated partial thromboplastin time-CWA aids in making a differential diagnosis which is difficult to do using only the routine APTT. The CWA is currently used for many applications in the clinical setting, including the monitoring of hemophilia patients and patients receiving anticoagulant therapy and the differential diagnosis of diseases.
Article
Introduction Bleeding after major hepatobiliary pancreatic (HBP) surgery may be serious. Although postoperative abnormality of the hemostatic system are important elements that affect bleeding, routine activated partial thromboplastin time (APTT) assessment is considered inadequate to predict massive bleeding (MB). Recently, APTT-clot waveform analysis (CWA) was reported to be useful for detecting coagulation disorders. Methods APTT-CWA was performed using the ACL-TOP analyzer in 188 patients who underwent four major HBP surgeries (distal pancreatectomy, hepatectomy, subtotal stomach-preserving pancreatoduodenectomy (SSPPD), and SSPPD with combined resection and reconstruction of the portal vein) to analyze its usefulness in predicting the risk of bleeding. Results Seventy (37.2%) patients developed MB and the incidence of MB was highest among patients who underwent hepatectomy. There were no significant differences in routine APTT, the first derivative peak (DP) time and 1/2 fibrin formation peak time between patients with MB and those without MB, throughout the postoperative course. On the other hand, the first and second DP height were significantly lower in patients with MB than in those without MB and lowest in patients who underwent hepatectomy. Conclusion APTT-CWA was able to detect the detailed changes in the hemostatic system after major HBP surgery. The patterns of APTT-CWA after major HBP surgery differed among various surgical procedures according to invasiveness. The lower first and the second DP height, which were frequently observed in hepatectomy patients, may be useful for predicting the risk of MB.
Article
Introduction Disseminated intravascular coagulation (DIC) is a feared complication of various systemic illnesses. We aimed to evaluate the laboratory requesting practices of clinicians, especially concerning the laboratory parameters, included in the International Society of Thrombosis and Haemostasis (ISTH) DIC score. Methods A retrospective descriptive study was performed and included data from DIC screen requests analysed at Universitas National Health Laboratory Service (NHLS) laboratory, Bloemfontein, South Africa, for one calendar year. Laboratory request forms were analysed, recording the pretest diagnosis and whether the diagnosis was associated with DIC. Parameters of the DIC screen, prothrombin time, activated partial thromboplastin time, thrombin time, d‐dimer and fibrinogen were used to calculate the ITSH DIC score and diagnose heparin contamination. The platelet count, currently not part of the DIC screen test set, was also recorded. Results A total of 778 DIC screen requests were processed. One hundred and eighty‐three requests were excluded due to laboratory‐defined rejection criteria, heparin contamination or for lacking an ISTH score parameter. Of the remaining 595 complete requests, 283 (47.7%) were laboratory‐defined overt DIC. The pretest diagnosis was not predictive of either a positive or negative finding of overt DIC. The contribution of fibrinogen to assigning overt DIC was questionable. Conclusion The number of DIC screen requests received highlights the need for laboratory evidence of DIC. To improve laboratory DIC testing, the authors suggest critical evaluation of the contribution of the pretest diagnosis and fibrinogen in a prospective study and adding the platelet count in our local DIC test set.
Article
Objective This article aims to analyze the phenotype and genotype of an inherited dysfibrinogenemia pedigree associated with a heterozygous mutation in the FGA gene, and to investigate the pathogenesis of this disease. Clinical Presentation The proband of interest is a 29-year-old woman. She was in her 37 weeks of gestation. Routine coagulation tests showed low fibrinogen activity (0.91 g/L; normal range: 2.0–4.0 g/L) and normal fibrinogen antigen (FIB:Ag) level (2.09 g/L; normal range: 2.0–4.0 g/L). Techniques The prothrombin time, activated partial thromboplastin time, thrombin time, and activity of plasma fibrinogen (FIB:C) were detected by the one-stage clotting method. The FIB:Ag, D-dimer, and fibrinogen degradation products were tested by the immunoturbidimetry method. To identify the novel missense mutation, fibrinogen gene sequencing and molecular modeling were performed. We used ClustalX-2.1-win and online bioinformatic software to analyze the conservation and possible effect of the amino acid substitution on fibrinogen. Results Phenotypic analysis revealed that the FIB:C of the proband was significantly reduced while the FIB:Ag was normal. Sequencing analysis detected a heterozygous C.2185G > A point mutation in the FGA gene (AαGlu710Lys). Bioinformatic and modeling analyses indicated that the mutation probably caused harmful effects on fibrinogen. Conclusion The heterozygous mutation of Glu710Lys in the FGA gene was identified that could cause the reduction of the FIB structure stability and result in the dysfibrinogenemia.
Article
Background Factor VIII (FVIII) is activated by thrombin‐catalyzed cleavage at three sites. Previous reports indicated that the A2 domain contained thrombin‐interactive sites responsible for cleavage at Arg³⁷². We have also found that the A1 domain of FVIII bound to the anion‐binding exosite I of thrombin. The present study focused, therefore, on thrombin interaction with A1 residues 337‐372 containing clustered acidic and hirugen‐like sequences. Aim To identify specific thrombin‐interactive site(s) within the A1 acidic region of FVIII. Methods and Results The synthetic peptide of residues 337‐353 with sulfated Tyr³⁴⁶ (337‐353S) significantly blocked thrombin‐catalyzed FVIII activation and cleavage at Arg³⁷², while a corresponding peptide of residues 354‐372 had no significant effect. Treatment with 1‐ethyl‐3‐(3‐dimethylaminopropyl)‐carbodiimide to cross‐link thrombin and 340‐350S suggested that the 344‐349 clustered acidic region was involved in thrombin interaction. Alanine‐substituted FVIII mutants, Y346A and D347A/D348A/D349A, depressed thrombin‐catalyzed activation and cleavage at Arg³⁷², with peak activation at ~ 50% and cleavage rates of ~ 10% to 20% compared to wild type (WT). The peak level of thrombin‐catalyzed activation and the cleavage rate at Arg³⁷² using FVIII mutants with 337‐346 residues substituted with hirugen‐sequences (MKNNEEAEDY337‐346GDFEEIPEEY) were ~ 1.5‐ and ~ 2.5‐fold of WT, respectively. Surface plasmon resonance‐based analysis demonstrated that the Kd for active‐site modified thrombin interactions using Y346A and D347A/D348A/D349A mutants was ~ 3‐ to 6‐fold higher than that of WT, and that the hirugen‐hybrid mutant facilitated association kinetics ~ 1.8‐fold of WT. Conclusion Residues 346‐349 with sulfated Tyr provided a thrombin‐interactive site responsible for activation and cleavage at Arg³⁷². A hirugen‐hybrid A1 mutant showed more efficient thrombin‐catalyzed cleavage at Arg³⁷².
Article
Introduction Physiological coagulation is considered to activate coagulation factor IX (FIX) by a small amount of tissue factor (TF) and activated coagulation factor VII (FVIIa) with the presence of platelets. A Clot waveform analysis (CWA) may be useful for evaluating physiological coagulation. Material and methods A CWA using a small amount of TF (CWA/sTF) was performed in platelet-rich plasma (PRP), platelet-poor plasma (PPP), several phospholipids (PLs) and patients with lupus anticoagulant (LA), idiopathic thrombocytopenic purpura (ITP) or inhibitor for FVIII. Results The CWA/sTF without PLs showed a shorter peak time and higher peak height in PRP than in PPP. The effect of PRP on the CWA/sTF depended on the platelet count, and PLs showed a similar effect on the CWA/sTF results in PPP. The peak time of the CWA/sTF in PRP was prolonged in patient with ITP. The CWA/sTF in PRP showed a prolonged peak time and decreased peak height of the second derivative in patient with LA. Both a shortened peak time and elevated peak height were observed in the CWA/sTF of patient with inhibitor after treatment with activated recombinant human FVII. Conclusion A CWA can be conducted using a small amount of TF and platelets or PL without contact activation and may be able to detect not only hemostatic abnormalities but also changes in platelet counts.
Article
Background In hemophilia bypass therapy, a platelet‐dependent mechanism is believed to be primarily responsible for recombinant factor VIIa (rFVIIa)’s hemostatic effect. rFVIIa may also possibly interact with other cells through its binding to endothelial cell protein C receptor (EPCR) or cell surface phospholipids. Objectives We aim to investigate the relative contribution of platelet‐dependent and platelet‐independent mechanisms in rFVIIa‐mediated thrombin generation in hemophilic conditions at the injury site. Methods Platelets were depleted in acquired and genetic hemophilia mice using anti‐platelet antibodies. The mice were subjected to the saphenous vein injury, and the hemostatic effect of pharmacological concentrations of rFVIIa was evaluated by measuring thrombin generation at the injury site. Results Administration of anti‐mouse CD42 antibodies to mice depleted platelets by more than 95%. As expected, hemophilia mice, compared to wild‐type mice, generated only a small fraction of thrombin at the injury site. The depletion of platelets in hemophilia mice further reduced thrombin generation. However, when pharmacological doses of rFVIIa were administered to hemophilia mice, substantial amounts of thrombin were generated even in the platelet‐depleted hemophilia mice. No differences in thrombin generation were detected among FVIII‐/‐, EPCR‐deficient FVIII‐/‐, and EPCR‐overexpressing FVIII‐/‐ mice depleted of platelets or not. Evaluation of platelets by flow cytometry as well as immunoblot analysis showed no detectable expression of EPCR. Conclusions Our data suggest that pharmacological concentrations of rFVIIa generate thrombin in hemophilia in both platelet‐dependent and platelet‐independent mechanisms.
Chapter
The purpose of this review is to describe structure and function of the multiple proteins of the coagulation system and their subcomponent domains. Coagulation is the process by which flowing liquid blood plasma is converted to a soft, viscous gel entrapping the cellular components of blood including red cells and platelets and thereby preventing extravasation of blood. This process is triggered by the minimal proteolysis of plasma fibrinogen. This transforms the latter to sticky fibrin monomers which polymerize into a network. The proteolysis of fibrinogen is a function of the trypsin-like enzyme termed thrombin. Thrombin in turn is activated by a cascade of trypsin-like enzymes that we term coagulation factors. In this review we examine the mechanics of the coagulation cascade with a view to the structure-function relationships of the proteins. We also note that two of the factors have no trypsin like protease domain but are essential cofactors or catalysts for the proteases. This review does not discuss the major role of platelets except to highlight their membrane function with respect to the factors. Coagulation testing is a major part of routine diagnostic clinical pathology. Testing is performed on specimens from individuals either with bleeding or with thrombotic disorders and those on anticoagulant medications. We examine the basic in-vitro laboratory coagulation tests and review the literature comparing the in vitro and in vivo processes. In vitro clinical testing typically utilizes plasma specimens and non-physiological or supraphysiological activators. Because the review focuses on coagulation factor structure, a brief overview of the evolutionary origins of the coagulation system is included.