ArticlePDF Available

Comparison between manufacturing sites shows differential adhesion, activation, and GPIbα expression of cryopreserved platelets: CRYOPRESERVED PLATELETS IN SHEAR FLOW

Authors:
  • Australian Red Cross Blood Service, Sydney, Australia

Abstract and Figures

BACKGROUND Transfusion of cryopreserved platelets (cryoplatelets) is not common but may replace standard liquid‐preserved platelets (PLTs) in specific circumstances. To better understand cryoplatelet function, frozen concentrates from different manufacturing sites were compared. STUDY DESIGN AND METHODS Cryoplatelets from Denver, Colorado (DEN); Sydney, Australia (SYD); and Ghent, Belgium (GHE) were compared (n = 6). A paired noncryopreserved control was included in Ghent. Microfluidic‐flow chambers were used to study PLT adhesion and fibrin deposition in reconstituted blood. Receptor expression was measured by flow cytometry. Coagulation in static conditions was evaluated by rotational thromboelastometry (ROTEM). RESULTS Regardless of the manufacturing site, adhesion of cryoplatelets under shear flow (1000/sec) was significantly (p < 0.05) reduced compared to control. Expression of GPIbα was decreased in a subpopulation of cryoplatelets comprising 45% ± 11% (DEN), 63% ± 9% (GHE), and 94% ± 6% (SYD). That subpopulation displayed increased annexin V binding and decreased integrin activation. PLT adhesion, agglutination, and aggregation were moreover decreased in proportion to that subpopulation. Fibrin deposition under shear flow was normal but initiated faster (546 ± 163 sec GHE) than control PLTs (631 ± 120 sec, p < 0.01), only in the absence of tissue factor. In static conditions, clotting time was faster, but clot firmness decreased compared to control. Coagulation was not different between manufacturing sites. CONCLUSION Cryopreservation results in a subset of PLTs with enhanced GPIbα shedding, increased phosphatidylserine expression, reduced integrin response, and reduced adhesion to collagen in microfluidic models of hemostasis. The proportion of this phenotype is different between manufacturing sites. The clinical effects, if any, will need to be verified.
Content may be subject to copyright.
ORIGINAL RESEARCH
Comparison between manufacturing sites shows differential
adhesion, activation, and GPIbαexpression of cryopreserved
platelets
Katrijn R. Six,
1,2
Willem Delabie,
1
Katrien M.J. Devreese,
2,3
Lacey Johnson,
4
Denese C. Marks,
4,5
Larry J. Dumont,
6,7
Veerle Compernolle,
1,2,8
and Hendrik B. Feys
1,2
BACKGROUND: Transfusion of cryopreserved
platelets (cryoplatelets) is not common but may replace
standard liquid-preserved platelets (PLTs) in specic
circumstances. To better understand cryoplatelet
function, frozen concentrates from different
manufacturing sites were compared.
STUDY DESIGN AND METHODS: Cryoplatelets from
Denver, Colorado (DEN); Sydney, Australia (SYD); and
Ghent, Belgium (GHE) were compared (n = 6). A paired
noncryopreserved control was included in Ghent.
Microuidic-ow chambers were used to study PLT
adhesion and brin deposition in reconstituted blood.
Receptor expression was measured by ow cytometry.
Coagulation in static conditions was evaluated by
rotational thromboelastometry (ROTEM).
RESULTS: Regardless of the manufacturing site, adhesion
of cryoplatelets under shear ow (1000/sec) was
signicantly (p < 0.05) reduced compared to control.
Expression of GPIbαwas decreased in a subpopulation of
cryoplatelets comprising 45% 11% (DEN), 63% 9%
(GHE), and 94% 6% (SYD). That subpopulation
displayed increased annexin V binding and decreased
integrin activation. PLT adhesion, agglutination, and
aggregation were moreover decreased in proportion to that
subpopulation. Fibrin deposition under shear ow was
normal but initiated faster (546 163 sec GHE) than control
PLTs (631 120 sec, p < 0.01), only in the absence of
tissue factor. In static conditions, clotting time was faster, but
clot rmness decreased compared to control. Coagulation
was not different between manufacturing sites.
CONCLUSION: Cryopreservation results in a subset of
PLTs with enhanced GPIbαshedding, increased
phosphatidylserine expression, reduced integrin
response, and reduced adhesion to collagen in
microuidic models of hemostasis. The proportion of this
phenotype is different between manufacturing sites. The
clinical effects, if any, will need to be veried.
ABBREVIATIONS: CRP-XL = cross-linked collagen-related
peptides; cryoplatelet(s) = cryopreserved platelet(s); DEN =
cryoplatelets from Denver, Colorado; GHE = cryoplatelets from
Ghent, Belgium; PC(s) = platelet concentrate(s); ROTEM =
rotational thromboelastometry; RT = room temperature; TF = tissue
factor; SYD = cryoplatelets from Sydney, Australia.
From the
1
Transfusion Research Center, Belgian Red Cross-
Flanders, Ghent, Belgium; the
2
Faculty of Medicine and Health
Sciences, Department of Diagnostic Sciences, Ghent University,
Ghent, Belgium; and the
3
Coagulation Laboratory, Department of
Laboratory Medicine, Ghent University Hospital, Ghent, Belgium;
4
Research & Development, Australian Red Cross Blood Service,
Sydney, Australia; and the
5
Sydney Medical School, University of
Sydney, Sydney, Australia;
6
Blood Systems Research Institute,
Denver, Colorado; the
7
Geisel School of Medicine at Dartmouth,
Lebanon, New Hampshire, USA; and the
8
Blood Service of the
Belgian Red Cross-Flanders, Mechelen, Belgium.
Address reprint requests to: Hendrik B. Feys, Ottergemsesteen-
weg 413, Ghent 9000, Belgium; e-mail: hendrik.feys@rodekruis.be.
This is an open access article under the terms of the Creative
Commons Attribution-NonCommercial License, which permits use,
distribution and reproduction in any medium, provided the original
work is properly cited and is not used for commercial purposes.
This research was supported by the Foundation for Scientic
Research of the Belgian Red Cross Flanders. KS is a fellow of the
Special Research Fund of Ghent University. The Australian govern-
ment funds the Australian Red Cross Blood Service to provide blood
and blood products to the Australian community.
The views, opinions, and/or ndings contained in this report
are those of the author (s) and should not be construed as an of-
cial Department of Defense position, policy, or decision unless so
designated by other documentation.
Received for publication December 22, 2017; revision received
March 22, 2018; and accepted March 22, 2018.
doi:10.1111/trf.14828
© 2018 The Authors. Transfusion published by Wiley
Periodicals, Inc. on behalf of AABB.
TRANSFUSION 2018;9999;112
TRANSFUSION 1
The demand for platelet concentrates (PCs) has
increased progressively over the years
1
due to an
aging population and an increased number of
patients suffering from chronic thrombocytope-
nia. Platelets (PLTs) are transfused for prophylaxis in
patients with hematologic malignancies or to treat active
bleeding.
2
Room temperature (RT) storage of PCs results in
better transfusion yields than cold-stored PLTs.
3
However, it
also limits the shelf life to between 4 and 7 days, resulting
in a logistic challenge that all blood banks continuously
face.
4
In addition, these conditions give rise to a substantial
risk of bacterial transmission. Consequently, cold storage
and cryopreservation have remained of interest as an alter-
native to standard PC banking
57
for nonprophylactic use,
particularly for remote areas with low-quality transportation
infrastructure or distant rural communities or during mili-
tary operations when transport of liquid-stored PCs is ham-
pered.
8
In selected cases, cryopreserved platelets
(cryoplatelets) could serve as backup product in emergen-
cies for an acutely bleeding patient when supply of regular
liquid stored PC is exhausted.
Several cryopreservation methods have been published
aiming to minimize PLT damage. Dimethyl sulfoxide
(DMSO) is the most commonly used cryoprotectant,
although several others have been investigated.
5,911
Most
protocols include hyperconcentration of PLTs before freez-
ing to reduce the amount of DMSO transfused. This
requires reconstitution after thawing, most commonly in
plasma or saline.
12
Cryoplatelets have been transfused in healthy volun-
teers, generally yielding lower count increments compared
to liquid-stored PLTs.
5,13,14
This is caused by biochemical
changes during cryopreservation that can be detected
in vitro including decreased PLT aggregation, decient sig-
nal transduction, altered morphology, differential expression
of certain membrane markers, and microparticle
release.
1418
Recent studies suggest that cryoplatelets are
procoagulant,
19,20
but these experiments were performed in
static conditions when hemostasis was not restricted by
convective hydrodynamic forces. Studies of PLT adhesive
and procoagulant function of cryoplatelets under shear ow
are lacking. In addition, comparison between cryopreserva-
tion procedures and cryopreserved products manufactured
in different facilities is not available. This study combines
both questions in a comparison of cryoplatelets from three
unrelated international blood institutions, using
microuidic-ow chamber perfusion and real-time video
microscopy.
MATERIALS AND METHODS
Study design
Platelet concentrates were prepared and cryopreserved in
three different manufacturing sites: Denver, CO, (DEN);
Sydney, Australia (SYD); and Ghent, Belgium (GHE). Pri-
mary products from DEN and SYD were from apheresis and
stored in plasma with ACD-A. Those from GHE were from
pooling of six buffy coats in CPD-plasma with 65% (vol/vol)
additive solution (SSP+, Macopharma). Cryopreservation
protocols were similar for all three manufacturing sites and
were described previously.
14,21
In brief, a 27% (vol/vol)
DMSO with 0.9% NaCl (wt/vol) in water was added to the
PC to achieve a nal concentration of 5% to 6% (vol/vol)
DMSO. These PCs were hyperconcentrated by centrifuga-
tion, resuspended in the remaining supernatant, and then
stored at 80C. Cryopreserved DEN and SYD products
were shipped (World Courier) on dry ice to GHE where
these were immediately transferred to a 80C freezer
until use.
Concentrates from DEN were thawed for 8 minutes at
37C and were then held undisturbed for 30 minutes before
resuspension in 25 mL of 0.9% (wt/vol) NaCl in water at
RT. Those from SYD and GHE were resuspended in 250 mL
of ABO/D-matched plasma (30C) immediately after thaw-
ing for 5 minutes at 37C. After resuspension, SYD and GHE
PLTs were held undisturbed for 1 hour at RT before experi-
ments. Paired liquid-preserved control PCs from DEN and
SYD could not be transported to GHE because of the logistic
challenge. Therefore, paired control PCs were only included
in Ghent (GHE CTR). These were shamtreated with all
preparation and handling methods but without freezing,
therefore controlling for confounding effects of DMSO,
hyperconcentration, resuspension, and incubation. Prelimi-
nary experiments using microuidic-ow chambers found
no difference between sham-treated and untreated control
PLTs (data not shown). GHE CTR PLTs were resuspended
in 250 mL of ABO/D plasma and incubated for 1 hour at
RT, as for the paired GHE cryoplatelets. From every
manufacturing site, six unrelated cryoplatelet units were
included (n = 6).
Blood reconstitution for microuidic-ow chambers
and thromboelastometry
Fresh blood samples were taken from healthy consenting
nonmedicated volunteers in hirudin vacutainers (REF
08128812001, Diapharma Group Inc.) or in sodium citrate
vacutainers (REF 366575, BD Diagnostics). Hirudin vacutai-
ners were used for microuidic-ow chambers studying
PLT adhesion alone, in the absence of coagulation.
22
Citrate
was used for microuidic-ow chambers with recalcication
to study PLT adhesion in combination with brin deposition
(i.e., coagulation).
23
Blood cells were separated by centrifugation (13 min at
250 ×gwithout brake) generating PLT-rich plasma and
concentrated red blood cells (RBCs). PLT-poor plasma was
obtained by centrifugation of that PLT-rich plasma for
10 minutes at 4500 ×g. Blood reconstitution was performed
by mixing the RBCs with the corresponding PLT-poor
2 TRANSFUSION
SIX ET AL
plasma and with PLTs from the various cryo- or control
groups, aiming for a mean of 40% hematocrit and 250 ×10
9
PLTs/L. Complete blood and PLT counts after reconstitution
were determined with an automated hematology analyzer
(PocH-1000i, Sysmex, Kobe, Japan). The PLT count before
cryopreservation was with the hematology analyzers from
the respective manufacturing sites: PocH-1000i, Sysmex
(GHE); XE 2100D, Sysmex (DEN); and CellDyn Ruby, Abbott
Diagnostics (SYD).
Perfusion-ow chamber experiments
Adhesion of PLTs to immobilized collagen was examined by
measuring surface coverage as a function of time during
perfusion of reconstituted, hirudin anticoagulated blood in
microuidic channels coated with collagen as described.
22
Briey, microuidic-ow chambers were coated with 50 μg/
mL of collagen (horm collagen Type III, Takeda Pharma) in
an isotonic glucose solution (pH 2.7) overnight at 4C.
Reconstituted blood was labeled for 10 minutes at 37C with
3,30-dihexyloxacarbo-cyanine iodide in a nal concentration
of 1 μmol/L (Sigma-Aldrich, St Louis, MO). Labeling ef-
ciency was tested using ow cytometry and was not differ-
ent between PCs. A microuidic pump (Mirus evo
nanopump, Cellix Ltd.) was used to generate a wall shear
stress of 50 dyne/cm
2
corresponding to a wall shear rate of
1000/sec. PLT deposition was recorded as a function of per-
fusion time using real-time video microscopy and accompa-
nying acquisition software (Carl Zeiss, Oberkochen,
Germany). PLT surface coverage (%) was retrieved by image
data analysis (Zen 2 [blue edition], Carl Zeiss). A linear
regression of PLT surface coverage with time was used to
calculate the PLT adhesion rate (Prism Version 6.07,
GraphPad Software Inc.).
Adhesion of PLTs in combination with brin deposition
was examined by measuring median uorescence increase
as a function of time during perfusion of reconstituted and
recalcied blood under perfusion ow, as described.
23
Instead of hirudin, citrate was used to anticoagulate fresh
blood for reconstitution. The reconstituted blood was mixed
during perfusion with one tenth volume of recalcication
buffer (HBS; 10 mmol/L HEPES, 155 mmol/L NaCl, pH 7.4)
containing 100 mmol/L CaCl
2
and 37.5 mmol/L MgCl
2
. Sep-
arately operated syringe pumps (Exigo syringe pump, Cellix)
were used. PLT and brin deposition under ow was exam-
ined in channels coated with only collagen for contact acti-
vation or with collagen plus puried recombinant human
lipidated tissue factor (TF, Dade Innovin) at approximately
100 pmol/L in HBS (Siemens Healthcare GmbH) for TF-
based activation. Before perfusion, the reconstituted blood
was spiked with 70 μg/mL of Alexa Fluor 405-labeled brin-
ogen (Sigma-Aldrich) in addition to 3,30-dihexyloxacarbo-
cyanine iodide to label PLTs as above. For investigation of
TF-mediated coagulation, reconstituted blood samples were
supplemented with 4 μmol/L corn trypsin inhibitor
(Enzyme Research Laboratories) to inhibit FXIIa-based con-
tact activation. Image acquisition and analysis were as
described.
23
In brief, PLT adhesion rate (/sec) was mea-
sured by linear regression of the green uorescent signal
increase as a function of perfusion time. The variables
retrieved for brin deposition included coagulation rate
(/sec), which is the linear portion of brin deposition kinet-
ics and clotting time (sec), which is the lag time indicating
the moment of coagulation onset. This analysis takes into
account thrombus growth in the z-plane. The outcome vari-
ables were extracted from the raw uorescence data using a
software plugin developed in MatLab (MathWorks). All ow
chamber studies were performed in duplicate and the mean
of two technical repeats was used.
Flow cytometry
Expression of P-selectin (phycoerythrin-anti-CD62P, Life
Technologies), phosphatidylserine (peridinin-chlorophyll-
Cy5.5 Annexin V, BD Biosciences), activated integrin α
IIb
β
3
(uorescein-labeled PAC1, BD Biosciences) and GPIbα
(uorescein-labeled anti-CD42b, Life Technologies) was
analyzed with an acoustic focusing ow cytometer (Attune,
Life Technologies). PLTs were incubated with the labeled
antibodies for 10 minutes in buffer 10 mmol/L
HBS/1 mmol/L MgSO
4
at RT. Samples were diluted 20-fold
immediately before readout. Phosphatidylserine measure-
ment was in buffer supplemented with 2 mmol/L CaCl
2
.
Integrin α
IIb
β
3
activation was measured with PAC1 for
resting PLTs or after activation using cross-linked collagen-
related peptides (CRP-XL; 0.25 μg/mL, University of Cam-
bridge). Threshold gates were set including 0.5% of 10,000
events incubated with corresponding isotype antibody con-
trols. For phosphatidylserine controls, a sample containing
labeled annexin V was prepared without CaCl
2
. The per-
centage of positive events or median uorescent intensities
were determined for 10,000 events staining positive for
CD61 (allophycocyanin-labeled anti-CD61, Life Technolo-
gies). The number of microparticles was determined using
calibration beads (Biocytex) and the accompanying gating
strategy provided by the manufacturer. Microparticles were
dened as GPIbαpositive events with sizes smaller than
0.9 μm. The result is expressed as the fraction of microparti-
cles per 10,000 all-size events positive for GPIbα.
Western blotting to determine GPIbαectodomain
shedding
Samples (1 mL) were taken from the PLT bag immediately
before freezing at 80C and immediately after thawing.
These experiments were performed on GHE PLTs only for
logistic reasons. Metalloproteinase activity was quenched
immediately with 50 mmol/L EDTA. Centrifugation was per-
formed at 4500 ×gfor 1 minute at RT. The PLT pellet was
resuspended in reducing sample buffer (60 mmol/L Tris,
10% [vol/vol] glycerol, 2% [wt/vol] SDS, 0.01% [wt/vol]
TRANSFUSION 3
CRYOPRESERVED PLATELETS IN SHEAR FLOW
bromophenol blue, and 40 mmol/L dithiothreitol). The
supernatant was 0.2 μmltered rst and then prepared for
electrophoresis by addition of reducing sample buffer. Equal
volumes of sample were loaded onto 4% to 15% polyacryl-
amide Tris-Glycine TGX gels (Bio-Rad). The GPIbαprotein
or its ectodomain were detected with the mouse monoclo-
nal anti-GPIbα(Clone 8H211) antibody (Antibodies Online)
and a secondary peroxidase conjugated polyclonal antibody
(Cell Signaling Technologies). Antibodies were prepared in
Tris-buffered saline (pH 7.4, 25 mmol/L Tris with
150 mmol/L NaCl, and 2 mmol/ KCl) with 5% (wt/vol)
skimmed milk. Membranes were developed in a imaging
system (ChemiDoc MP, Bio-Rad). Densitometry was per-
formed using (ImageLab v4.0.1, Bio-Rad).
PLT aggregation
Platelet aggregation and agglutination were examined with
light transmission at 37C (Chrono-Log, Helena Laborato-
ries). Ristocetin-induced PLT agglutination was with
1.5 mg/mL ristocetin (American Biochemical and Pharma-
ceuticals). Aggregation was with a combination of 10 μmol/L
protease activated receptor-1 activating peptide (PAR1AP,
Sigma-Aldrich), 20 μmol/L 2-(methylthio)adenosine
5-diphosphate trisodium salt hydrate (MeSADP, Santa Cruz
Biotechnology) and 5 μmol/L epinephrine (Sigma-Aldrich).
Aggregation cuvettes contained 250 ×10
9
PLTs/L in a nal
volume of 500 μL, diluted in their respective supernatant
(obtained by centrifugation at 4500 ×gfor 10 min). The sig-
nal was calibrated using the respective PLT-free superna-
tants. Maximal amplitude (%) is reported.
Thrombin generation assay
Generation of thrombin in vitro was analyzed with the
thrombin generation assay kit from Technoclone GmbH
according to the manufacturers instructions with minor
modications. Samples were prepared in 96-well micro-
plates. PCs were diluted to 10 ×10
9
,50×10
9
,or
250 ×10
9
/L in a xed 40% (vol/vol) volume of heterologous
human pooled plasma and 5% (vol/vol) of saline (0.9%
[wt/vol] NaCl in water) and supplemented with 1 pmol/L
TF, 4.0 μmol/L corn trypsin inhibitor, 0.5 μmol/L uoro-
genic substrate (Z-G-G-RAMC) and 7.5 mmol/L CaCl
2
(nal
concentrations). Samples were immediately analyzed in a
microplate reader (Innite F200PRO, Tecan Group Ltd.)
with lter settings for excitation at 360 nm and emission at
460 nm. The uorescent signal was recorded as a function
of time for a total of 120 minutes at 37C. The raw signal
was converted to thrombin concentrations based on a cali-
bration kit and a script in Excel (Microsoft) provided by the
manufacturer.
24
Rotational thromboelastometry
Rotational thromboelastometry (ROTEM) was performed on
reconstituted and citrate anticoagulated blood using a
ROTEM delta analyzer (Tem Innovations) according to the
manufacturers recommendations. ROTEM provides
dynamic information on the speed of coagulation initiation,
kinetics of clot growth, clot strength, and breakdown of the
clot. Contact activation is with ellagic acid and phospho-
lipids. Activation of the TF pathway with lipidated TF was
performed as described previously.
25
The following vari-
ables were analyzed: clotting time (sec) to examine speed of
brin formation, clot formation time (sec) to examine clot
formation kinetics, and maximum clot rmness (mm) to
examine rmness of the clot. Time between in vitro recon-
stitution of blood and analysis was 120 minutes at
maximum.
Statistical analysis
Results are reported as mean with standard deviation (SD).
Comparison between sites was by one-way analysis of vari-
ance (ANOVA) with Tukeys multiple comparisons correc-
tion. Comparison of paired GHE and GHR-CTR
concentrates was with a two-tailed paired t-test. Compari-
son of thrombin generation output data by varying PLT con-
centration and production site was with two-way ANOVA
with Tukeys multiple comparisons correction. Results from
statistical analysis are depicted on top of the panels. All sta-
tistical analyses were performed using computer software
(Prism, Version 6, GraphPad Software Inc.).
RESULTS
PLT recovery and storage time
Table 1 shows the mean PLT content of the products in the
study before and after cryopreservation. Mean recovery was
more than 90% for all manufacturing sites. It should be
noted that the PLT count after thawing was determined
using the GHE hematology analyzer while for the PLT count
before freezing cell counters from the respective
manufacturing sites were used. Storage duration was lon-
gest for SYD and shortest for GHE cryoplatelets.
PLT adhesion and brin deposition under
perfusion ow
In reconstituted blood, the adhesion rate of cryoplatelets
measured by surface coverage was decreased at least three-
fold compared to control (p < 0.05; green signal in Video S1
[available as supporting information in the online version of
this paper] and Fig. 1A). SYD PLTs were more affected
(p < 0.05) than GHE or DEN. Similar observations were
made in the presence of restored calcium levels both in
conditions with TF (green signal in Video S2 [available as
supporting information in the online version of this paper]
and Fig. 1B) and without TF (green signal in Video S3
[available as supporting information in the online version
of this paper] and Fig. 1C). Cryoplatelets also bound to
control surfaces without collagen, leading to localized
4 TRANSFUSION
SIX ET AL
brin deposition (Fig. 1D). This was not observed with
control PLTs.
The rate of brin deposition following adhesion of
cryoplatelets was variable, but not different from control
PLTs in the presence of TF (violet signal in Video S2 and
Fig. 2A). In the absence of TF, brin deposition rates were
slightly decreased for cryoplatelets (violet signal in
Video S3, Fig. 2B). The clotting time was not different
when TF was present (Fig. 2C). In the absence of TF,
however, clotting time decreased for cryoplatelets (GHE,
546 163 sec) compared to control PLTs (GHE-CTR,
631 120 seconds; Fig. 2D). This indicates that cryoplate-
lets enhance contact activation of coagulation under ow
compared to control. There were no signicant differ-
ences between SYD, DEN, and GHE PLTs for brin
deposition under hydrodynamic ow, despite the differ-
ences in PLT binding (Fig. 1).
GPIbαectodomain shedding in a subset of
cryoplatelets
Almost all events in the cryopreserved PCs were positive for
GPIbα(Fig. 3A), similar to control PLTs. Although control
PLTs had uniformly bright GPIbαexpression, the cryoplate-
lets also contained a dim GPIbαsubpopulation, comprising
45% 11% (DEN), 63% 9% (GHE), and 94% 6% (SYD)
of all GPIbαpositive events (Figs. 3B and 3C). The dim and
bright subpopulations were distinguishable but not always
entirely resolved in different cryoplatelet preparations
(Fig. S1, available as supporting information in the online
TABLE 1. PLT recovery and storage duration*
Center
PLT count (×10
9
)
Recovery (%) Storage (days)Before cryo After cryo
DEN 374.6 40.7 354.9 71.4 94.1 10.8 558 138
SYD 524.2 17.3 504.9 67.5 98.6 7.7 758 12
GHE 345.8 29.1 316.7 17.2 92.1 8.6 21 12
*Data are given as mean SD (n = 6).
Fig. 1. Platelet adhesion under shear ow. Perfusion of blood reconstituted with cryopreserved and control PLTs was at 1000/sec. (A) The PLT
surface coverage as a function of perfusion time (% SC/Time) in channels coated with only collagen (Video S1). This condition was with hirudin-
anticoagulated blood without TF. (B, C) The rate of PLT adhesion in citrated blood with restored Ca
2+
levels. These channels were coated with
(B) collagen and TF (Video S2) or with (C) collagen only (Video S3). (D) PLT (green) and brin (violet) accumulation in a part of the channel not
coated with collagen. Images were acquired at endpoint (magnication, 100×). Comparison was between DEN, SYD, and GHE (open bars). A
separate paired comparison between GHE and control noncryoplatelets was also performed (GHE CTR, closed bars). Data are shown as mean SD
(n = 6). Statistical analysis results are shown as *p < 0.05, **p < 0.01, or not signicant (ns). [Color gure can be viewed at wileyonlinelibrary.com]
TRANSFUSION 5
CRYOPRESERVED PLATELETS IN SHEAR FLOW
version of this paper). Ristocetin induced PLT agglutination
was signicantly decreased in cryoplatelets (Fig. 3D) and
SYD PLTs were more affected (P < 0.001) than GHE or DEN.
The ristocetin response was associated with the fraction of
GPIbα-bright PLTs (Fig. 3C).
Western blotting of PLT lysate and of the PLT-free
supernatant indicated signicant GPIbαectodomain shed-
ding after cryopreservation (Fig. 4). The signal of full-length
GPIbαin PLT cell lysates signicantly decreased (Fig. 4A).
The signal of GPIbαectodomain in PLT-free supernatant
correspondingly increased three- to sevenfold in cryoplate-
lets compared to before cryopreservation (Figs. 4A and 4B).
This signicant variation in the level of ectodomain shed-
ding conrms the variation of GPIbαexpression found in
ow cytometry (Fig. S1).
Platelet aggregation was signicantly reduced for cryo-
platelets (Fig. 5A) compared to control, despite a strong ago-
nist blend of PAR1AP, MeSADP, and epinephrine. Integrin
α
IIb
β
3
activation by CRP-XL was also signicantly reduced
compared to control (Fig. 5B). Again, SYD PLTs were more
affected (p < 0.01) than GHE or DEN. The aggregation and
integrin activation response furthermore followed the fraction
of GPIbα-bright PLTs (Fig. 3C). In addition, the bright GPIbα
subpopulation had normal integrin activation, while the
GPIbα-dim subpopulation did not (representative data in
Fig. 5C). Similarly, the bright subpopulation had low levels of
annexin V binding, while the dim one had high levels of
annexin V binding (representative data in Fig. 5D). This
resulted overall in higher number of annexin Vpositive
events (Fig. S2A). In line with this, the percentage of micro-
particles was increased after cryopreservation (Fig. S2B). The
highest number was found in SYD cryoplatelet preparations.
The refractive properties of the GPIbα-dim subpopulation
were altered. In forward mode, less light (median intensity,
1.9 ×10
5
0.2 ×10
5
) was scattered compared to the GPIbα-
bright subpopulation either in GHE cryoplatelets (median
intensity, 3.3 ×10
5
0.5 ×10
5
)orGHE-CTRcontrolPLTs
(median intensity, 3.3 ×10
5
0.4 ×10
5
;Fig.S3).Collectively,
these data suggest that the GPIbα-dim subpopulation is indic-
ative of the partial damage caused by cryopreservation.
Function of cryoplatelets in reconstituted whole
blood under static conditions
Coagulation under static conditions in vitro was examined with
thromboelastometry. Reconstituted whole blood containing
Fig. 2. Coagulation under shear ow. Perfusion of blood reconstituted with cryopreserved and control PLTs was at 1000/sec with real-time
restoration of Ca
2+
levels. Fibrin accumulation was followed over time. Accumulation rate (/sec) of brin in channels coated with (A) collagen
and TF or with (B) only collagen. The clotting time (sec) indicates the moment of coagulation onset in channels coated with (C) collagen and
TF or with (D) only collagen. Comparison was between DEN, SYD, and GHE (all open symbols). A separate paired comparison between GHE
and control noncryoplatelets was also performed (GHE CTR, closed symbols). Data are shown as mean SD (n = 6). Statistical analysis results
are shown as *p < 0.05, **p < 0.01, or not signicant (ns).
6 TRANSFUSION
SIX ET AL
GHE cryoplatelets showed decreased clotting times com-
pared to GHE-CTR indicating that initiation of coagulation
was faster irrespective of TF (47 6 sec vs. 58 5 sec;
Fig. 6A) or contact activation (158 30 sec vs. 203 11 sec;
Fig. 6B). In line with this, the lag time between initiating
thrombin generation and initial thrombin formation was
short for all cryoplatelet preparations (Fig. 6C). Peak throm-
bin concentrations were signicantly higher (Fig. 6D). Both
lag time and peak thrombin were dependent on the dose of
PLTs as well, irrespective of cryopreservation. Data between
SYD, DEN, and GHE were not different for these parame-
ters, in line with the coagulation experiments under shear
ow. Despite shortening lag times with cryoplatelets, how-
ever, the subsequent clot formation rate and clot rmness
were reduced compared to control (Fig. 7). This was inde-
pendent of TF.
DISCUSSION
Freezing cells damages their integrity, often irreversibly
decreasing the cells ability to function optimally.
26
Excessive release of microparticles and decreased responses
to agonists in vitro are known indicators of PLT
cryopreservation-induced damage.
4,18
Despite these
changes, no serious adverse events were documented in a
recent Phase I dose-escalation trial in hematology-oncology
patients.
27
Older studies even suggested clinical efcacy of
cryoplatelets, although count increments were consistently
lower in comparison to liquid-preserved PLTs.
28,29
Cryopla-
telets are promising as they can be shipped over great dis-
tances, but this will require signicant standardization of
the production process. In addition, a deep understanding
of cryoplatelet biochemical quality and its markers is
required. This study has examined cryoplatelets produced
in different countries and shipped to one investigation site,
with a view to understanding cryoplatelet variability and
function under conditions of shear ow.
A key nding in this study was the presentation of a
GPIbα-dim subpopulation representing 45% to 94% in cryo-
platelets, but less than 5% in controls. This observation was
reported rst by Barnard et al.
17
in a seminal paper on
receptor density of cryoplatelets. Cryoplatelet preparations
with an abundant GPIbα-dim subpopulation displayed
Fig. 3. GPIbαexpression and PLT activation. (A) The percentage of GPIbα-positive events determined in ow cytometry. (B) Representative
histogram of GPIbαsignals in ow cytometry in paired control GHE CTR (red) and cryopreserved GHE (black) PCs denoting dim and bright
GPIbαsubpopulations. (C) Within the GPIbα-positive population, events were gated for dim or bright GPIbαexpression based on the
respective histograms as per B. The population of dim GPIbα(hatched bars) is overlaid with the population of bright GPIbα(open bars)
expressing events. (D) Ristocetin-induced PLT agglutination was measured and data are shown as maximal amplitude (%). Comparison was
between DEN, SYD, and GHE (all open symbols). A separate paired comparison between GHE and control noncryoplatelets was also
performed (GHE CTR, closed symbols). Data are shown as mean SD (n = 6). Statistical analysis results are shown as **p < 0.01,
***p < 0.001, ****p < 0.0001, or not signicant (ns). [Color gure can be viewed at wileyonlinelibrary.com]
TRANSFUSION 7
CRYOPRESERVED PLATELETS IN SHEAR FLOW
reduced or absent aggregation, poor integrin activation, and
reduced PLT adhesion under shear. In addition, the GPIbα-
dim subpopulation itself was unresponsive to agonists and
had increased annexin V binding and altered refractive
properties.
Platelets can actively decrease the GPIbαreceptor
number on the plasma membrane by internalization and by
ectodomain shedding. For instance, PLT activation by
PAR1AP decreases GPIbαlevels to below 60% of resting
control PLTs. This does not affect their subsequent adhesion
to collagen in ow chambers because internalized GPIbα
receptors are quickly restored upon contact with the immo-
bilized collagen surface.
30
These observations imply that
GPIbαinternalization is not sufcient to sustainably
decrease PLT adhesion to collagen. The GPIbαdecrease
during cryopreservation therefore is not governed by recep-
tor internalization but by ectodomain shedding,
15
in a sub-
set of PLTs. Contrary to GPIbαinternalization by PLT
activation, ectodomain shedding is irreversible. Moreover,
the GPIbα-dim PLTs have GPIbαlevels well below 50% of
normal, which is a critical value for accomplishing adhesion
Fig. 4. Ectodomain shedding of GPIbα. (A) A representative
Western blot of the GPIbαreceptor in the PLT cell lysate (full
length) or the PLT supernatant (ectodomain) is shown. All samples
were loaded on the same gel, but not adjacent to each other as
indicated by the panel borders. Molecular weight markers are in
kilodalton (kDa). Paired samples before and after cryopreservation
are shown. (B) Densitometry of the GPIbαectodomain signal was
performed for samples before and after cryopreservation. The signal
after cryopreservation is expressed relative to that before. Data from
individual biologic repeats (n = 9) are shown. Mean is shown as a
horizontal line and error bars represent the SD.
Fig. 5. Platelet activation and GPIbαexpression levels. (A) PLT aggregation in response to a mix of PAR1AP, MeSADP, and epinephrine.
Maximal amplitude (%) is shown. (B) The percentage of PLTs expressing activated integrin α
IIb
β
3
in response to stimulation with CRP-XL
measured by binding of PAC1. (C) A representative dot plot of PAC1 binding versus GPIbαexpression. The plot is divided by a quadrant
indicating subpopulations expressing high or low signals of PAC1, GPIbα, or both. (D) A representative dot plot of annexin V binding versus
GPIbαexpression. The plot is divided as in C. Comparison was between DEN, SYD, and GHE (all open symbols). A separate paired
comparison between GHE and control noncryoplatelets was also performed (GHE CTR, closed symbols). Data are shown as mean SD
(n = 6). Statistical analysis results are shown as *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, or not signicant (ns).
8 TRANSFUSION
SIX ET AL
to collagen under ow.
3032
Consequently, these PLTs con-
tribute most if not all to the decrease in adhesion and by
extrapolation to the decrease in overall cryoplatelet function.
Furthermore, that fraction was remarkably variable
between production sites as well as between PLT prepara-
tions within a given site (Fig. S1). This suggests that cryopla-
telet quality may be improved if the number of
dysfunctional PLTs can be consistently controlled at low
levels. It is currently not clear, however, what makes certain
PLTs more susceptible to cryopreservation-induced dam-
age. Maybe differences in primary PLT concentration and
age, cryoprocess steps, (preliminary) exposure to tempera-
ture cycles
33
, and/or (bio)chemicals contribute. Whether
shedding inhibitors could prevent cryopreservation-induced
damage depends on what comes rst. If GPIbαshedding is
secondary to a damaging event, then shedding inhibitors
will not help but in the case that shedding sparks down-
stream lesions, inhibitors may be able to mitigate damage.
For age-related storage lesion of liquid PLTs, specic inhibi-
tion of GPIbαshedding rescued the rapid clearance of aged
PLTs to some extent.
34
Future research can always exploit
the GPIbα-dim subpopulation as a biomarker for the extent
of damage. The impact of directed modications in the
cryoprocess can be easily monitored using this particular
marker in ow cytometry.
Cryopreservation-induced damage was particularly
obvious in assays that specically study PLTs, like integrin
activation, microparticle release, and aggregation. This is in
line with previous publications
1418
, which were mostly
under static conditions (or extremely low shear) that do not
always mimic the biophysical environment of blood vessel
injury. Our data now conrm that in shear ow, cryoplatelet
adhesion rates to immobilized collagen were also signi-
cantly decreased compared to control. This was both with
and without anticoagulation, indicating that cryoplatelets
are less adhesive irrespective of simultaneous thrombin and
brin formation. Unlike controls, cryoplatelets also bound
to uncoated parts of the channel during perfusion. This sug-
gests that cryoplatelets are less selective than control PLTs
for binding to nonbiologic substrates. This requires further
investigation because cryoplatelets may be lost by nonspeci-
c adhesion to transfusion bags, lines, or needles.
Fig. 6. Coagulation initiation in static conditions. Blood was reconstituted with cryopreserved (open symbols) or control PLTs (closed symbols)
and studied in the presence of Ca
2+
by ROTEM using (A) TF or (B) ellagic acid to initiate coagulation. The clotting time (sec) is the lag time
from adding the activating agent to the blood until the elastogram tracing reached 2 mm. (C, D) Thrombin generation was measured in the
presence of TF and increasing PLT concentrations of 10 ×10
9
/L (lled bars), 50 ×10
9
/L (hatched bars), and 250 ×10
9
/L (open bars). (C) The
lag time (min) measures the time until threshold thrombin concentrations are reached. Statistics are indicated for the conditions with
250 ×10
9
PLTs/L. (D) Peak thrombin (nmol/L) measures the peak amount of thrombin formed. Statistical signicance is indicated for the
conditions with 250 ×10
9
PLTs/L. Cryoplatelets from DEN, SYD, and GHE were used and the latter was paired to control noncryoplatelets
GHE CTR (dashed line). Data are shown as mean SD (n = 6). Statistical analysis results are shown as *p < 0.05, ***p < 0.001,
****p < 0.0001, or not signicant (ns).
TRANSFUSION 9
CRYOPRESERVED PLATELETS IN SHEAR FLOW
Cryopreservation-induced damage was less prominent
in assays that focus on coagulation. In ROTEM, cryoplatelets
had more rapid clotting times compared to controls even
though clot formation times and maximum clot rmness
were decreased. Further, in terms of thrombin generation,
cryoplatelets demonstrated faster clotting times than con-
trols and higher peak thrombin, all in a dose-dependent
manner. Johnson and colleagues
19,35
had previously shown
that the presence of cryoplatelet supernatant shortens time
to coagulation presumably by the activity of microparticles,
but these experiments were conducted in static conditions,
allowing coagulation factors to diffuse freely in bulk. In this
case, the cascade can quickly assemble tenase and pro-
thrombinase complexes on the abundant negatively charged
phospholipid surface provided by damaged cryoplatelets
and microparticles. Under shear ow, factor availability
depends on convective hydrodynamic forces rather than dif-
fusion.
36
Our data conrm, however, that also under shear
ow, coagulation is not affected by cryopreservation of
PLTs. In contrast, contact-induced coagulation initiation in
the absence of TF was even shorter than control. This is
remarkable because with or without TF, signicantly fewer
PLTs were adhering to the surface during perfusion. Clinical
trials are required to investigate whether the procoagulant
role of cryoplatelets is sufcient to stop bleeding in patients.
The effect of cryopreservation for SYD cryoplatelets was
greater than that for the other manufacturing sites particu-
larly in PLT assays. The GPIbα-dim subpopulation was
more abundant than in the other sites, the aggregation
response was lower, integrin activation was less, and PLT
adhesion under shear was the lowest with SYD cryoplate-
lets. Despite this, the differences between production sites
were less obvious in PLT-based coagulation assays. It is not
clear why SYD PLTs were particularly more damaged than
those from the other sites. SYD and GHE PLTs were resus-
pended the same way with similar plasma so differences in
reconstitution procedure are not likely. Irregularities in
deep-freeze conditions could be one of the reasons, but dif-
ferences in primary product preparation are also possible.
The SYD PLTs were collected by apheresis in plasma, while
GHE PLTs were prepared by pooling of buffy coats in plasma
with SSP+. The SYD PLTs were frozen for 2 years, which is
the maximum reported shelf-life.
37
Although preliminary data
suggest that cryoplatelets stored for 4 years are equivalent to
those stored for 2 years,
38
the effect of extended storage on
the GPIbαsubpopulation and PLT function have not been
reported. The SYD PLTs also had the highest initial PLT con-
tent in comparison to the other sites, which effectively
reduces the amount of DMSO per PLT unit. These variables
might have contributed to the observations.
Fig. 7. Coagulation propagation in static conditions. Blood was reconstituted with cryopreserved (open symbols) or control PLTs (closed
symbols) and studied in the presence of Ca
2+
by ROTEM using (A, C) TF or (B, D) ellagic acid to initiate coagulation. (A, B) The clot formation
time (sec) was measured between the clotting time (Figs. 5A and 5B) and the moment a clot rmness of 20 mm has been reached. (C, D)
Maximum clot rmness (MCF) reects the absolute strength of the clot. Comparison was between DEN, SYD, and GHE (all open symbols). A
separate paired comparison between GHE and control noncryoplatelets was also performed (GHE CTR, closed symbols). Data are shown as
mean SD (n = 6). Statistical analysis results are shown as *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, or not signicant (ns).
10 TRANSFUSION
SIX ET AL
We conclude that cryoplatelets are less adhesive in
shear ow conditions, in proportion to a subset of damaged
GPIbα-dim PLTs, which decrease the effective PLT dose.
The size of the affected subpopulation may be indicative of
the extent of damage. Different production sites may have
different cryoplatelet qualities especially in PLT function
assays. Finally, even though fewer cryoplatelets bind colla-
gen under ow, these nonetheless enhance contact activa-
tion, provide sufcient support for TF activation and normal
coagulation under shear ow.
ACKNOWLEDGMENTS
The authors thank Michael Luypaert and Esha Wauters for their
technical support in performing ROTEM analyses.
AUTHOR CONTRIBUTIONS
KS, VC, LJ, LJD, DCM, and HBF designed research, provided
cryopreserved platelet products, and performed quality con-
trol experiments; KS and WD performed experiments; KD
was responsible for the thromboelastometry assays; HBF
and KS wrote the manuscript; VC, LJ, LJD, DCM, and HBF
supervised the study; and all authors critically reviewed and
amended the manuscript.
CONFLICT OF INTEREST
The authors have disclosed no conicts of interest.
REFERENCES
1. Desborough MJ, Smethurst PA, Estcourt LJ, et al. Alternatives
to allogeneic platelet transfusion. Br J Haematol 2016;175:
381-92.
2. Liumbruno G, Bennardello F, Lattanzio A, et al. Recommenda-
tions for the transfusion of plasma and platelets. Blood Trans-
fus 2009;7:132-50.
3. Murphy S, Gardner FH. Effect of storage temperature on main-
tenance of platelet viabilitydeleterious effect of refrigerated
storage. N Engl J Med 1969;280:1094-8.
4. Johnson L, Tan S, Wood B, et al. Refrigeration and cryopreser-
vation of platelets differentially affect platelet metabolism and
function: a comparison with conventional platelet storage con-
ditions. Transfusion 2016;56:1807-18.
5. Valeri CR, Feingold H, Marchionni LD. A simple method for
freezing human platelets using 6% dimethylsulfoxide and stor-
age at 80C. Blood 1974;43:131-6.
6. Cap AP. Platelet storage: a license to chill! Transfusion 2016;56:
13-6.
7. Reddoch KM, Pidcoke HF, Montgomery RK, et al. Hemostatic
function of apheresis platelets stored at 4C and 22C. Shock
2014;41(Suppl 1):54-61.
8. Hess JR, Lelkens CC, Holcomb JB, et al. Advances in military,
eld, and austere transfusion medicine in the last decade.
Transfus Apher Sci 2013;49:380-6.
9. Balduini CL, Mazzucco M, Sinigaglia F, et al. Cryopreservation
of human platelets using dimethyl sulfoxide and glycerol-glu-
cose: effects on "in vitro" platelet function. Haematologica
1993;78:101-4.
10. Nie Y, de Pablo JJ, Palecek SP. Platelet cryopreservation using a
trehalose and phosphate formulation. Biotechnol Bioeng 2005;
92:79-90.
11. Taylor MA. Cryopreservation of platelets: an in-vitro compari-
son of four methods. J Clin Pathol 1981;34:71-5.
12. Valeri CR, Ragno G, Khuri S. Freezing human platelets with
6 percent dimethyl sulfoxide with removal of the supernatant
solution before freezing and storage at 80 degrees C without
postthaw processing. Transfusion 2005;45:1890-8.
13. Spector JI, Yarmala JA, Marchionni LD, et al. Viability and
function of platelets frozen at 2 to 3 C per minute with 4 or
5 per cent DMSO and stored at 80 C for 8 months. Transfu-
sion 1977;17:8-15.
14. Dumont LJ, Cancelas JA, Dumont DF, et al. A randomized con-
trolled trial evaluating recovery and survival of 6% dimethyl
sulfoxide-frozen autologous platelets in healthy volunteers.
Transfusion 2013;53:128-37.
15. Johnson LN, Winter KM, Reid S, et al. Cryopreservation of
buffy-coat-derived platelet concentrates in dimethyl sulfoxide
and platelet additive solution. Cryobiology 2011;62:100-6.
16. Valeri CR, Macgregor H, Ragno G. Correlation between in vitro
aggregation and thromboxane A2 production in fresh, liquid-
preserved, and cryopreserved human platelets: effect of ago-
nists, pH, and plasma and saline resuspension. Transfusion
2005;45:596-603.
17. Barnard MR, MacGregor H, Ragno G, et al. Fresh, liquid-pre-
served, and cryopreserved platelets: adhesive surface receptors
and membrane procoagulant activity. Transfusion 1999;39:880-8.
18. Waters L, Padula MP, Marks DC, et al. Cryopreserved platelets
demonstrate reduced activation responses and impaired sig-
naling after agonist stimulation. Transfusion 2017;57:2845-57.
19. Johnson L, Coorey CP, Marks DC. The hemostatic activity of
cryopreserved platelets is mediated by phosphatidylserine-
expressing platelets and platelet microparticles. Transfusion
2014;54:1917-26.
20. Cid J, Escolar G, Galan A, et al. In vitro evaluation of the hemo-
static effectiveness of cryopreserved platelets. Transfusion
2016;56:580-6.
21. Johnson L, Reade MC, Hyland RA, et al. In vitro comparison of
cryopreserved and liquid platelets: potential clinical implica-
tions. Transfusion 2015;55:838-47.
22. Van Aelst B, Feys HB, Devloo R, et al. Microuidic ow cham-
bers using reconstituted blood to model hemostasis and plate-
let transfusion in vitro. J Vis Exp 2016;109:e53823.
23. Six KR, Devloo R, Van Aelst B, et al. A microuidic ow cham-
ber model for platelet transfusion and hemostasis measures
platelet deposition and brin formation in real-time. J Vis Exp
2017;120:e55351.
TRANSFUSION 11
CRYOPRESERVED PLATELETS IN SHEAR FLOW
24. Hemker HC, Kremers R. Data management in thrombin gener-
ation. Thromb Res 2013;131:3-11.
25. Chitlur M, Sorensen B, Rivard GE, et al. Standardization of
thromboelastography: a report from the TEG-ROTEM working
group. Haemophilia 2011;17:532-7.
26. Iwatani M, Ikegami K, Kremenska Y, et al. Dimethyl sulfoxide
has an impact on epigenetic prole in mouse embryoid body.
Stem Cells 2006;24:2549-56.
27. Slichter SJ, Dumont LJ, Cancelas JA, et al. Treatment of bleed-
ing in severely thrombocytopenic patients with transfusion of
dimethyl sulfoxide (DMSO) cryopreserved platelets (CPP) is
safe - report of a phase 1 dose escalation safety trial. Blood
2016;128:1030.
28. Daly PA, Schiffer CA, Aisner J, et al. Successful transfusion of
platelets cryopreserved for more than 3 years. Blood 1979;54:
1023-7.
29. Towell BL, Levine SP, Knight WA 3rd, et al. A comparison of
frozen and fresh platelet concentrates in the support of throm-
bocytopenic patients. Transfusion 1986;26:525-30.
30. van Zanten GH, Heijnen HF, Wu Y, et al. A fty percent reduc-
tion of platelet surface glycoprotein Ib does not affect platelet
adhesion under ow conditions. Blood 1998;91:2353-9.
31. Cauwenberghs N, Ajzenberg N, Vauterin S, et al. Characteriza-
tion of murine anti-glycoprotein Ib monoclonal antibodies that
differentiate between shear-induced and ristocetin/botrocetin-
induced glycoprotein Ib-von Willebrand factor interaction.
Haemostasis 2000;30:139-48.
32. Kageyama S, Matsushita J, Yamamoto H. Effect of a humanized
monoclonal antibody to von Willebrand factor in a canine
model of coronary arterial thrombosis. Eur J Pharmacol 2002;
443:143-9.
33. Hoffmeister KM, Felbinger TW, Falet H, et al. The clearance
mechanism of chilled blood platelets. Cell 2003;112:87-97.
34. Chen W, Liang X, Syed AK, et al. Inhibiting GPIbalpha shed-
ding preserves post-transfusion recovery and hemostatic func-
tion of platelets after prolonged storage. Arterioscler Thromb
Vasc Biol 2016;36:1821-8.
35. Raynel S, Padula MP, Marks DC, et al. Cryopreservation alters
the membrane and cytoskeletal protein prole of platelet
microparticles. Transfusion 2015;55:2422-32.
36. Zhu S, Lu Y, Sinno T, et al. Dynamics of thrombin generation
and ux from clots during whole human blood ow over col-
lagen/tissue factor surfaces. J Biol Chem 2016;291:23027-35.
37. Valeri CR, Srey R, Lane JP, et al. Effect of WBC reduction and
storage temperature on PLTs frozen with 6 percent DMSO for
as long as 3 years. Transfusion 2003;43:1162-7.
38. Noorman F, Strelitski R, Badloe J. Frozen platelets can be
stored for 4 years at 80C without affecting in vitro recovery,
morphology, receptor expression, or coagulation prole. Trans-
fusion 2014;54:74A.
SUPPORTING INFORMATION
Additional Supporting Information may be found in the
online version of this article.
Fig. S1 Platelet subpopulations by GPIbαexpression.
GPIbαexpression was determined by ow cytometry on
10,000 platelets. Isotype negative control (blue) was com-
pared to (A) non cryopreserved control platelets of GHE-
CTR (red) and to all GHE cryopreserved platelets (black).
Three different GHE products are shown in panels B to
D. The dotted line represents the arbitrary gate determined
for dim (left) and bright (right) GPIbαsubpopulations. The
histograms were normalized to display relative count.
Fig. S2 Annexin V binding and microparticles. (A) The
percentage of GPIbαpositive events binding annexin V.
(B) Microparticles were dened as events smaller than
0.9 μm. The relative number of microparticles to the total
number of GPIbαpositive events is depicted. Comparison
was between cryopreserved platelets from Denver, CO
(DEN), Sydney (Australia, SYD) and Ghent (Belgium, GHE)
(open bars). A separate paired comparison was between
cryopreserved platelets (GHE) and control non-
cryopreserved platelets (GHE CTR, lled bars). Data are
shown as mean SD (n = 6). Statistical analysis results are
shown as ** P < 0.01, *** P < 0.001, **** P < 0.0001 or not
signicant (ns).
Fig. S3 Scatter properties of cryopreserved platelets. The
median intensity (MI) of the forward scattered light (FSC) in
ow cytometry for subpopulations of cryoplatelets (open
bars) or control platelets (closed bars) based on the GPIbα
signal as shown in Fig. S1. All samples were from the Bel-
gian (GHE) site. Data are shown as mean SD (n = 6). Sta-
tistical analysis results are shown as ** P < 0.01 or not
signicant (ns).
12 TRANSFUSION
SIX ET AL
... Some groups have found a decrease in this parameter after cryopreservation, while others have found no difference among different platelet frozen methods. [55][56][57] Such discrepancies could be explained, among others, by the agonist used to perform hemostatic assay, e.g. kaolin or tissue factor, the differences in the methods of preparation, platelet concentration 58,59 and resuspension media. ...
... 64 It has been reported that platelet morphology is significantly affected after cryopreservation. 56,57,64,65 However, most of the evidences regard structural changes and do not investigate how cryopreservation quantitatively alter the biological components of PLT. 64,66,67 Here, we obtained important clues on the macromolecular changes induced by cryopreservation on membrane lipids, protein content and increasing of peroxidation processes. ...
... According to previous studies, 27,57,69 we found that cryopreservation produced an increase secretion of platelet derived EVs accompanied by an increase of PS externalization compared to fresh components. Both increases declined over time after thawing ( Figure 3, 3 h and 6 h). ...
Article
Full-text available
Plain Language Summary What is the context? Transfusion of Fresh platelets (Fresh-PLT) with prophylaxis purposes is common in onco-hematological patients. Cryopreservation is an alternative storage method that allows to extend platelet component shelf life and build supplies usable in case of emergency. It is well established that cryopreservation affects platelet function questioning their use in onco-hematological patients. It is still unknown how platelet impairment, induced by cryopreservation, occurs over time after thawing, nor how the by-products of PLT deterioration may impact on cancer cell behavior. What is new? In this study, we deeply characterized the functional and morphological changes induced by cryopreservation on platelets by comparing Fresh-PLT with Cryo-PLT at 1 h, 3 h and 6 h after thawing. Afterwards, we evaluated the effect of PLT supernatants on cancer cell behavior in vitro. The data presented show that within 3 hours after thawing Cryo-PLT undergo to irreversible macromolecular changes accompanied by increase of peroxidation processes and protein misfolding. After thawing the clot formation is reduced but still supported at all-time points measured, combined with unchanged phosphatidylserine expression and extracellular vesicles release over time. Cryo-PLT supernatants do not sustain proliferation and migration of cancer cells. WHAT is the impact? Cryo-PLT may be considered a precious back-up product to be used during periods of Fresh-PLT shortage to prevent bleeding in non-hemorrhagic patients. It is desirable to make it logistically feasible to transfuse cryopreserved platelets within 1 hour of thawing to maintain the platelets in their best performing condition.
... 6 In vitro studies show impaired aggregation and lower clot strength in thromboelastography. [7][8][9][10][11][12][13][14][15][16][17] Despite less adhesion, DTC show the same fibrin formation in flow models under shear stress. 18,19 Despite a lower in vivo recovery [20][21][22] DTC have been shown to be safe and effective. [22][23][24] DTC and RSP have similar clinical effects in thrombocytopenic patients 23,24 trauma 25 and cardiac surgery. ...
... Most published in vitro studies on DTC report freezethaw recoveries between 75% and 90% 1,7,8,12,14,17,21 and a few studies report freeze thaw recoveries below 70% 11,15 or above 90%. 19,37,41 Surprisingly, products stored <0.5 years at T A B L E 3 KX/XS DMSO-1 and XS DMSO-2 frozen DTC, comparison fresh versus cold-stored thawed plasma, analyzer freeze = analyzer thaw a ), ≥0. platelets was associated with a higher TEG-MA, a higher proportion of small activated platelets was associated with a shorter R time. ...
... The only in vitro measurements that have shown equivalence of DTC and RSP are (micro)fluidic models that can measure fibrin deposition under shear flow. 18,19 Although in vitro recovery may reflect product loss due to freezethaw damage and therefore is a good parameter to assess the effect of storage duration, it is not a predictor of in vivo efficacy as it is unknown whether high or low recovery would affect the hemostatic properties of DTC in vivo, especially when it has been shown that there was no evidence of a dose response based on bleeding reduction and the number of DTC units transfused. 24 Therefore, the lower in vitro recovery of very short-stored DTC does not disqualify these products for clinical use. ...
Article
Full-text available
Background: The Netherlands Armed Forces (NLAF) are using -80°C deep-frozen thrombocyte concentrate (DTC) since 2001. The aim of this study is to investigate the effect of storage duration and alterations in production/measurement techniques on DTC quality. It is expected that DTC quality is unaffected by storage duration and in compliance with the European guidelines for fresh and cryopreserved platelets. Study design and methods: Pre-freeze and post-thaw product platelet content and recovery were collected to analyze the effects of dimethyl sulfoxide (DMSO) type, duration of frozen storage (DMSO-1 max 12 years and DMSO-2 frozen DTC max 4 years at -80°C) and type of plasma used to suspend DTC. Coagulation characteristics of thawed DTC, plasma and supernatant of DTC (2× 2500 G) were measured with Kaolin thromboelastography (TEG) and phospholipid (PPL) activity assay. Results: Platelet content and recovery of DTC is ±10%-15% lower in short-stored products and remained stable when stored beyond 0.5 years. Thawed DTC (n = 1724) were compliant to the European guidelines (98.1% post-thaw product recovery ≥50% from original product, 98.3% ≥200 × 109 platelets/unit). Compared to DMSO-1, products frozen with DMSO-2 showed ±8% reduced thaw-freeze recovery, a higher TEG clot strength (MA 58 [6] vs. 64 [8] mm) and same ±11 s PPL clotting time. The use of cold-stored thawed plasma instead of fresh thawed plasma did not influence product recovery or TEG-MA. Discussion: Regardless of alterations, product quality was in compliance with European guidelines and unaffected by storage duration up to 12 years of -80°C frozen storage.
... Functionally, cryopreserved platelets display enhanced thrombin generation potential with elevated peak thrombin and faster clotting times compared to RT-stored platelets 11-13 . In contrast, they have a reduced capacity to aggregate in response to agonists and adhere to collagen 7,11,14 . This divergence in function may be related to the development of platelet subpopulations within the component. ...
... Furthermore, cryopreserved components have been shown to be clinically safe and potentially have enhanced haemostatic function in actively bleeding patients [43][44][45][46][47][48] . Although the exact mechanism mediating the therapeutic benefit is incompletely understood, the changes induced by cryopreservation seem to enhance their procoagulant potential [10][11][12]14 . ...
Article
Full-text available
Cryopreservation of platelets, at − 80 °C with 5–6% DMSO, results in externalisation of phosphatidylserine and the formation of extracellular vesicles (EVs), which may mediate their procoagulant function. The phenotypic features of procoagulant platelets overlap with other platelet subpopulations. The aim of this study was to define the phenotype of in vitro generated platelet subpopulations, and subsequently identify the subpopulations present in cryopreserved components. Fresh platelet components (n = 6 in each group) were either unstimulated as a source of resting platelets; or stimulated with thrombin and collagen to generate a mixture of aggregatory and procoagulant platelets; calcium ionophore (A23187) to generate procoagulant platelets; or ABT-737 to generate apoptotic platelets. Platelet components (n = 6) were cryopreserved with DMSO, thawed and resuspended in a unit of thawed plasma. Multi-colour panels of fluorescent antibodies and dyes were used to identify the features of subpopulations by imaging flow cytometry. A combination of annexin-V (AnnV), CD42b, and either PAC1 or CD62P was able to distinguish the four subpopulations. Cryopreserved platelets contained procoagulant platelets (AnnV+/PAC1−/CD42b+/CD62P+) and a novel population (AnnV+/PAC1−/CD42b+/CD62P−) that did not align with the phenotype of aggregatory (AnnV−/PAC1+/CD42b+/CD62P+) or apoptotic (AnnV+/PAC1−/CD42b−/CD62P−) subpopulations. These data suggests that the enhanced haemostatic potential of cryopreserved platelets may be due to the cryo-induced development of procoagulant platelets, and that additional subpopulations may exist.
... The rate of GPIba proteolysis can be significantly increased in situations with physical stress like cold temperature [36,37] or pathological hydrodynamic shear forces [38,39]. This shedding has been linked to poor posttransfusion recovery [39,40]. ...
Article
Purpose of review Ectodomain shedding has been investigated since the late 1980s. The abundant and platelet specific GPIbα receptor is cleaved by ADAM17 resulting in the release of its ectodomain called glycocalicin. This review will address the role of glycocalicin as an end-stage marker of platelet turnover and storage lesion and will consider a potential function as effector in processes beyond hemostasis. Recent findings Glycocalicin has been described as a marker for platelet senescence, turnover and storage lesion but is not routinely used in a clinical setting because its diagnostic value is nondiscriminatory. Inhibition of glycocalicin shedding improves posttransfusion recovery but little is known (yet) about potential hemostatic improvements. In physiological settings, GPIbα shedding is restricted to the intracellular GPIbα receptor subpopulation suggesting a role for shedding or glycocalicin beyond hemostasis. Summary So far, all evidence represents glycocalicin as an end-stage biomarker of platelet senescence and a potential trigger for platelet clearance. The extensive list of interaction partners of GPIbα in fields beyond hemostasis opens new possibilities to investigate specific effector functions of glycocalicin.
... Moreover, CSP with PAS-C can ensure platelet function and metabolism, reduce platelet activation and improve platelet survival.100 In CP with DMSO, GPIbα shedding and enhanced PS exposure may be associated with decreased adhesion and increased coagulation function respectively.101 PMP levels in CP are 10-15 folds higher than those at room temperature, which potentially promotes their coagulation.102 ...
Article
Full-text available
Background Platelet concentrate (PC) transfusions are crucial in prevention and treatment of bleeding in infection, surgery, leukemia, and thrombocytopenia patients. Although the technology for platelet preparation and storage has evolved over the decades, there are still challenges in the demand for platelets in blood banks because the platelet shelf life is limited to 5 days due to bacterial contamination and platelet storage lesions (PSLs) at 20–24°C under constant horizontal agitation. In addition, the relations between some adverse effects of platelet transfusions and PSLs have also been considered. Therefore, understanding the mechanisms of PSLs is conducive to obtaining high quality platelets and facilitating safe and effective platelet transfusions. Objective This review summarizes developments in mechanistic research of PSLs and their relationship with clinical practice, providing insights for future research. Methods Authors conducted a search on PubMed and Web of Science using the professional terms “PSL” and “platelet transfusion.” The obtained literature was then roughly categorized based on their research content. Similar studies were grouped into the same sections, and further searches were conducted based on the keywords of each section. Results Different studies have explored PSLs from various perspectives, including changes in platelet morphology, surface molecules, biological response modifiers (BMRs), metabolism, and proteins and RNA, in an attempt to monitor PSLs and identify intervention targets that could alleviate PSLs. Moreover, novel platelet storage conditions, including platelet additive solutions (PAS) and reconsidered cold storage methods, are explored. There are two approaches to obtaining high‐quality platelets. One approach simulates the in vivo environment to maintain platelet activity, while the other keeps platelets at a low activity level in vitro under low temperatures. Conclusion Understanding PSLs helps us identify good intervention targets and assess the therapeutic effects of different PSLs stages for different patients.
... Studies on platelet concentrates demonstrate a constitutive but slow rate of GPIbα receptor shedding as a function of storage time [11]. The rate of proteolysis can be significantly increased during strong and sustained platelet activation [12] or in situations of physical stress like temperature shock [13,14]. GPIbα shedding is primarily catalyzed by a disintegrin and metalloproteinase (ADAM) 17 [15][16][17], also known as tumor necrosis factor α (TNF-α) -converting enzyme (TACE). ...
Article
Full-text available
Background: ADAM17 catalyzes platelet glycoprotein (GP) Ibα ectodomain shedding, thereby releasing glycocalicin in plasma. The spatiotemporal control over the enzyme-substrate interaction and the biological consequences of GPIbα shedding are poorly understood. Ojbectives: To determine the spatiotemporal control over GPIbα shedding by ADAM17. Methods: Transmission electron microscopy with immunogold staining, immunoprecipitation and quantitative western blotting were used. Results: Immunogold staining showed that all ADAM17 antigen is expressed intracellularly, irrespective of platelet activation. ADAM17 clustered in patches on a tortuous membrane system different from α- and dense granules. Mild activation by platelet adhesion to immobilized fibrinogen did not cause GPIbα shedding, while strong and sustained stimulation using thrombin and collagen (analogues) did. Glycocalicin release kinetics was considerably slower than typical hemostasis, starting at 20 minutes and reaching plateau after 3 hours of strong stimulation. Inhibition of the ADAM17 scissile bond specifically in GPIbα receptors that reside on the platelet's extracellular surface, did not prevent shedding which is in line with the strict intracellular location of ADAM17. Instead, shedding was restricted to a large GPIbα subpopulation that is inaccessible on resting platelets but becomes partially accessible following platelet stimulation. The data furthermore show that proteinaceous, water-soluble ADAM17 inhibitors cannot inhibit GPIbα shedding, while membrane permeable small molecule ADAM inhibitors can. Conclusions: The data show that platelets harbor two distinct GPIbα subpopulations, one that presents at the platelet's surface known for its role in primary hemostasis and one that provides substrate for proteolysis by ADAM17 with kinetics that suggest a role beyond hemostasis.
... CPPs from different manufacturing sites and continents differ in specific in vitro characteristics. 120 Two randomized controlled pilot trials from Australia and New Zealand were recently reported 110,121 ; as pilot trials, their primary end points were protocol feasibility and safety. The authors reported no significant difference in bleeding but significantly more fresh-frozen plasma and platelet transfusion in the cryopreserved group in one of the trials. ...
Article
Over 100 years ago, Duke transfused whole blood to a thrombocytopenic patient to raise the platelet count and prevent bleeding. Since then, platelet transfusions have undergone numerous modifications from whole blood-derived platelet-rich plasma to apheresis-derived platelet concentrates. Similarly, the storage time and temperature have changed. The mandate to store platelets for a maximum of 5-7 days at room temperature has been challenged by recent clinical trial data, ongoing difficulties with transfusion-transmitted infections, and recurring periods of shortages, further exacerbated by the COVID-19 pandemic. Alternative platelet storage approaches are as old as the first platelet transfusions. Cold-stored platelets may offer increased storage times (days) and improved hemostatic potential at the expense of reduced circulation time. Frozen (cryopreserved) platelets extend the storage time to years but require storage at -80 °C and thawing before transfusion. Lyophilized platelets can be powder-stored for years at room temperature and reconstituted within minutes in sterile water but are probably the least explored alternative platelet product to date. Finally, whole blood offers the hemostatic spectrum of all blood components but has challenges, such as ABO incompatibility. While we know more than ever before about the in vitro properties of these products, clinical trial data on these products are accumulating. The purpose of this review is to summarize the findings of recent preclinical and clinical studies on alternative, donor-derived platelet products.
Article
Background Current procedures for thawing and issuing of cryopreserved platelets (CPPs) are laborious and have remained challenging in emergency settings such as blood banks and military operations. In this prospective study, a novel processing method designed to facilitate the rapid issuance of CPPs with no postthaw handling required was developed and functionally characterized in parallel with standard CPPs manufactured. Study Design and Methods Double‐dose plateletpheresis units ( n = 42) were cryopreserved at −80°C in 5%–6% dimethyl sulfoxide to produce matched pairs thawed successively over a 27‐month period for comparison between two processing arms. In contrast to the standard CPPs manufactured as standalone units, platelets were frozen in tandem with resuspending plasma in a distinct partition as a single unit in the novel method, herein referred to as tandem CPPs. Postthaw (PT) CPPs from both arms were assessed at PT0‐, 12‐, and 24‐h to measure platelet recovery, R‐time (time to clot initiation; min), and maximum amplitude (MA; clot strength; mm) using thromboelastography. Results In the overall dataset, mean platelet recovery was higher ( p < .0005) for tandem CPPs (83.9%) compared with standard CPPs (73.3%) at PT0; mean R‐times were faster ( p < .0005) for tandem CPPs (2.5–3.6 min) compared with standard CPPs (3.0–3.8 min); mean MA was higher for tandem CPPs (57.8–59.5 mm) compared with standard CPPs (52.1–55.8 mm) at each postthaw time point ( p < .05). Conclusion Robust temporal dynamics of superior hemostatic functionality were established for tandem CPPs over extended cryopreservation up to 27 months and 24 h of postthaw storage.
Article
Background Platelet cryopreservation extends the shelf‐life to at least 2 years. However, platelets are altered during the freeze/thaw process. Downscaling platelet cryopreservation by freezing in tubes would enable rapid screening of novel strategies to improve the quality of cryopreserved platelets (CPPs). The aim of this study was to characterize the effect of freezing conditions on the in vitro phenotype and function of platelets frozen in a low volume compared to standard CPPs. Methods Platelets were prepared for cryopreservation using 5%–6% DMSO and processed using standard protocols or aliquoted into 2 mL tubes. Platelets were hyperconcentrated to 25 mL (standard CPPs) or 200 μL (tubes) before freezing at −80°C ( n = 8). Six insulators/controlled rate freezing containers were used to vary the freezing rate of platelets in tubes. Platelets were thawed, resuspended in plasma, and then assessed by flow cytometry and thromboelastography. Results The use of different insulators for tubes changed the freezing rate of platelets compared to platelets frozen using the standard protocol ( p < .001). However, this had no impact on the recovery of the platelets ( p = .87) or the proportion of platelets expressing GPIbα ( p = .46) or GPVI ( p = .07), which remained similar between groups. A lower proportion of platelets frozen in tubes externalized phosphatidylserine compared to standard CPPs ( p < .001). The clot‐forming ability (thromboelastography) of platelets was similar between groups ( p > .05). Conclusion Freezing platelets in tubes modified the freezing rate and altered some platelet characteristics. However, the functional characteristics remained comparable, demonstrating the feasibility of downscaling platelet cryopreservation for high‐throughput exploratory investigations.
Article
Background: Manufacturing methods for dimethyl sulfoxide (DMSO)-cryopreserved platelets (CPPs) are manual and labor intensive. Thawing and prepare-for-transfusion steps are in an open system that requires transfusion within 4 h. A fill-and-finish system (CUE) can automate the manufacturing process. A newly configured bag system allows freezing, thawing, and use of resuspension solutions while maintaining the functionally closed system, and extending the post-thaw shelf life beyond 4 h. Our objective is to evaluate the feasibility of the CUE system and the functionally closed bag system. Study design and methods: DMSO was volumetrically added to double-dose apheresis platelets, concentrated, and delivered to a 50- or 500-mL ethylene-vinyl acetate (EVA) bag by the CUE (n = 12). The functionally closed bag system contained 25 mL platelet additive solution 3 (PAS-3) in a 50-mL EVA bag. Control CPP (n = 2) were manually prepared. PAS-3 and CPP were thawed together. CPP were stored up to 98 h (20-24°C) and tested using a standard assay panel. Results: CUE prepared CPP met the design targets: volume, platelet content, and DMSO concentration. CUE CPP P-selectin was high. CD42b, phosphatidylserine (PS) expression, and live cell percentage were favorable compared to controls and favorably maintained over storage. The thrombin generation potency was slightly reduced compared to controls. The 50 mL EVA bag maintained pH for up to 30 h, and the 500 mL EVA bag beyond 76 h. Discussion: The CUE system presents a technically feasible method to prepare CPP. A functionally closed bag system with resuspension solution was successful and can extend the post-thaw storage time of CPP.
Article
Full-text available
Glycoprotein (GP) Ib is an adhesion receptor on the platelet surface that binds to von Willebrand Factor (vWF). vWF becomes attached to collagens and other adhesive proteins that become exposed when the vessel wall is damaged. Several investigators have shown that during cardiopulmonary bypass (CPB) surgery and also during platelet activation in vitro by thrombin or thrombin receptor activating peptide (TRAP) GPIb disappears from the platelet surface. Such a disappearance is presumed to lead to a decreased adhesive capacity. In the present study, we show that a 65% decrease in platelet surface expression of GPIb, due to stimulation of platelets in Orgaran anticoagulated whole blood with 15 μmol/L TRAP, had no effect on platelet adhesion to both collagen type III and the extracellular matrix (ECM) of human umbilical vein endothelial cells under flow conditions in a single-pass perfusion system. In contrast to adhesion, ristocetin-induced platelet agglutination was highly dependent on the presence of GPIb. Immunoelectron microscopic studies showed that GPIb almost immediately returned to the platelet surface once platelets had attached to collagen. In a subsequent series of experiments, we showed that when less than 50% of GPIb was blocked by an inhibitory monoclonal antibody against GPIb (6D1), platelet adhesion under flow conditions remained unaffected.
Article
Full-text available
To determine the duration of storage for cryopreserved platelets, 14 transfusions of random-donor, pooled platelets, stored in the vapor phase of liquid nitrogen for a mean period of 1157 days (range 1060- 1240), were analyzed. Twelve of these transfusions were compared in a paired fashion with fresh, random-donor, pooled platelets given within a few days to the same thrombocytopenic recipients. Platelets had been frozen using 5% dimethylsulfoxide as a cryoprotective agent either at a controlled rate of -1 degrees C/min to -80 degrees C or by simply placing them in the vapor phase (-120 degrees C) of a liquid nitrogen freezer. The mean freeze-thaw loss for the 14 transfusions was 22%, and the mean corrected 1-hr increment in platelet count was 12,600/microliter. In the 12 paired observations, the mean corrected 1- hr increment for frozen platelets was 11,800/microliter and 25,900 for fresh platelets, giving a frozen/fresh recovery of 46%. Random donor platelets can be cryopreserved by these methods for greater than 3 yr with satisfactory post-transfusion increments. This suggests that a reservoir of frozen platelets, either random-donor for emergency transfusion or of known HLA-type for transfusion to alloimmunized patients, can be established and stored for at least 3 yr.
Article
Full-text available
Microfluidic models of hemostasis assess platelet function under conditions of hydrodynamic shear, but in the presence of anticoagulants, this analysis is restricted to platelet deposition only. The intricate relationship between Ca²⁺-dependent coagulation and platelet function requires careful and controlled recalcification of blood prior to analysis. Our setup uses a Y-shaped mixing channel, which supplies concentrated Ca²⁺/Mg²⁺ buffer to flowing blood just prior to perfusion, enabling rapid recalcification without sample stasis. A ten-fold difference in flow velocity between both reservoirs minimizes dilution. The recalcified blood is then perfused in a collagen-coated analysis chamber, and differential labeling permits real-time imaging of both platelet and fibrin deposition using fluorescence video microscopy. The system uses only commercially available tools, increasing the chances of standardization. Reconstitution of thrombocytopenic blood with platelets from banked concentrates furthermore models platelet transfusion, proving its use in this research domain. Exemplary data demonstrated that coagulation onset and fibrin deposition were linearly dependent on the platelet concentration, confirming the relationship between primary and secondary hemostasis in our model. In a timeframe of 16 perfusion min, contact activation did not take place, despite recalcification to normal Ca²⁺ and Mg²⁺ levels. When coagulation factor XIIa was inhibited by corn trypsin inhibitor, this time frame was even longer, indicating a considerable dynamic range in which the changes in the procoagulant nature of the platelets can be assessed. Co-immobilization of tissue factor with collagen significantly reduced the time to onset of coagulation, but not its rate. The option to study the tissue factor and/or the contact pathway increases the versatility and utility of the assay.
Article
Full-text available
Coagulation kinetics are well established for purified blood proteases or human plasma clotting isotropically. However, less is known about thrombin generation kinetics and transport within blood clots formed under hemodynamic flow. Using microfluidic perfusion (wall shear rate, 200 (s-1)) of corn trypsin inhibitor-treated whole blood over a 250-micron long patch of type I fibrillar collagen/lipidated TF (~ 1 TF molecule/μm(2)), we measured thrombin released from clots using thrombin-antithrombin (TAT) immunoassay. The majority (>85 %) of generated thrombin was captured by intrathrombus fibrin since TAT was largely undetectable in the effluent, unless Gly-Pro-Arg-Pro (GPRP) was added to block fibrin polymerization. With GPRP present, the flux of thrombin increased to ~0.5 x 10(-12) nmole/μm(2)-sec over the first 500 sec of perfusion and then further increased by ~2 to 3-fold over the next 300 sec. The increased thrombin flux after 500 sec was blocked by anti-FXIa antibody (O1A6), consistent with thrombin-feedback activation of FXI. Over the first 300 sec, about 18000 molecules of thrombin were generated per surface TF molecule for the 250-micron long coating. A single layer of platelets (obtained with αIIbβ3 antagonism preventing continued platelet deposition) was largely sufficient for thrombin production. Also, the overall thrombin generating potential of a 1000-micron long coating became less efficient on a per μm(2) basis, likely due to distal boundary layer depletion of platelets. Overall, thrombin is robustly generated within clots by the extrinsic pathway, followed by late-stage FXIa contributions, with fibrin localizing thrombin via its antithrombin-I activity as a potentially self-limiting hemostatic mechanism.
Article
Full-text available
Objective: The platelet storage lesion accelerates platelet clearance after transfusion, but the underlying molecular mechanism remains elusive. Although inhibiting sheddase activity hampers clearance of platelets with storage lesion, the target platelet protein responsible for ectodomain shedding-induced clearance is not definitively identified. Monoclonal antibody 5G6 was developed recently to bind specifically human platelet receptor glycoprotein (GP)Ibα and inhibit its shedding but not shedding of other receptors. Here, the role of GPIbα shedding in platelet clearance after transfusion was addressed. Approach and results: Both human leukoreduced apheresis-derived platelets and transgenic mouse platelets expressing human GPIbα were stored at room temperature in the presence and absence of 5G6 Fab fragment. At various time points, aliquots of stored platelets were analyzed and compared. 5G6 Fab inhibited GPIbα shedding in both platelets during storage and preserved higher level of GPIbα on the platelet surface. Compared with age-matched control platelets, 5G6 Fab-stored platelets exhibited similar levels of platelet activation, degranulation, and agonist-induced aggregation. 5G6 Fab-stored human GPIbα platelets exhibited significantly higher post-transfusion recovery and in vivo hemostatic function in recipient mice than control platelets. Consistently, 5G6 Fab-stored, 8-day-old human platelets produced similar improvement in post-transfusion recovery in immunodeficient mice and in ex vivo thrombus formation compared with collagen under shear flow. Conclusions: Specific inhibition of GPIbα shedding in the stored platelets improves post-transfusion platelet recovery and hemostatic function, providing clear evidence for GPIbα shedding as a cause of platelet clearance. These results suggest that specific inhibition of GPIbα shedding may be used to optimize platelet storage conditions.
Article
Availability of platelets (plts) is severely limited by shelf life in some military as well as civilian settings. Additionally, some bleeding, thrombocytopenic patients do not have a therapeutic response to a standard plt transfusion. Methods for cryopreservation of apheresis plts for up to two years in 6% DMSO at <-65°C (CPP) have been developed and evaluated by in vitro assays and by in vivo infusions in non-human primates, in a few controlled human trials, and in field military operations. However, FDA has not yet approved CPP for routine use. Autologous radiolabeled CPP in healthy volunteers (n=32) had average plt recoveries of 33 ± 10% and survivals of 7.5 ± 1.2 days, and these results were 52 ± 12% and 89 ± 15%, respectively, of the same volunteers' fresh radiolabeled plts. The in vitro phenotype of CPP showed higher granule secretion, phosphotidylserine expression, and plt microparticles and poorer responses to common plt agonists compared to standard room temperature stored plts. These data suggest that transfused CPP might lead to an accelerated and enhanced clotting process in vivo. Our objective was to evaluate the safety of transfused CPP in a Phase-1 dose escalation trial. Eligibility criteria were hospitalized thrombocytopenic hematology/oncology patients with active World Health Organization (WHO) Grade 2 or greater bleeding that was, in most patients, uncontrolled by standard plts, no history of unprovoked thrombotic events, and no indication of active DIC. Fifty-nine patients consented and 28 were transfused with ½ CPP unit (n=5), one CPP unit (n=7), two CPP units (n=6), and three CPP units (n=6). One thawed single apheresis CPP unit contained 2.5 x 1011 ± 4.2 x 1010 plts in 50 ± 4 ml. In addition, one standard apheresis plt unit was randomly given to patients enrolled in each cohort (n=4). The study conformed to the Declaration of Helsinki and was listed on clinical trials.gov (NCT02078284). Patients were monitored for six days post-transfusion for adverse events (AEs) including clinical assessments for signs or symptoms of thrombosis and specific laboratory assays: prothrombin time (PT), partial thromboplastin time (aPTT), D-dimer, fibrinogen, prothrombin fragments 1 + 2 (F1+2), antithrombin, thrombin antithrombin (TAT), thrombin generation (TGT), and thromboelastography (TEG). All safety data were reviewed by an independent data safety monitoring committee prior to escalation to the next higher dose cohort. No thrombotic events occurred after transfusion of CPP units. Five serious AEs were reported, and none were associated with the CPP transfusion but, rather, were related to worsening of the patients' underlying medical conditions. Of 38 AEs, 5 were, at least, possibly related to a CPP transfusion and included DMSO skin odor following a ½ CPP unit and three CPP units (n=2), mild fever and chills in the same patient after one CPP unit (n=2), and moderate headache the next day following transfusion of three CPP units (n=1). As expected in this clinically-ill patient population, D-dimer, fibrinogen, F1+2, aPTT, and TAT averaged higher than the upper limit of normal prior to transfusion and remained similar following transfusion. TGT and TEG were suppressed pre-transfusion and were improved towards normal levels following transfusion of either CPP or standard plts. There was no induction of a post-transfusion hypercoagulable state in any patient based on laboratory results. Modest increases in corrected plts count increments (x 103/mm3) were observed following CPP transfusion (one CPP unit gave CCI's of 2.3 ± 3.5; two CPP units 4.2 ± 2.8; and three CPP units 5.6 ± 2.3) compared with 21.1 ± 3.6 after one unit of standard apheresis plts. Notably, all patients had stabilization or improvement of their bleeding following a CPP transfusion including one patient with Grade 4 CNS bleeding who had resolution of neurologic symptoms with no further plt transfusions, and four patients (17%) had WHO bleeding downgraded. In conclusion, the infusion of up to three sequential units of CPP in patients with severe thrombocytopenia and active bleeding was safe without any evidence of thrombotic complications despite CPP having a procoagulant phenotype resulting from the cryopreservation process. CPP may be efficacious to stop bleeding in thrombocytopenic patients as suggested by stabilization or downgrading of WHO bleeding grades. Phase 2/3 efficacy clinical trials are now indicated. Disclosures Slichter: NHLBI / NIH: Research Funding; Genentech: Research Funding; Cerus Corporation: Research Funding; Terumo BCT: Research Funding; Cellphire: Research Funding; Department of Defense / US Army Medical Research and Material Command: Research Funding; Megakaryon: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Stock options. Dumont:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Cancelas:New Health Sciences, Inc.: Membership on an entity's Board of Directors or advisory committees; US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding; National Institutes of Health: Research Funding; Terumo BCT: Research Funding; Cerus Corporation: Research Funding; Haemonetics, Inc.: Research Funding; Citra Labs, Inc.: Research Funding; Cellphire, Inc.: Membership on an entity's Board of Directors or advisory committees; William & Lawrence Hughes Foundation: Research Funding; Leukemia & Lymphoma Society of North America: Research Funding. Gernsheimer:Department of Defense: Research Funding; NHLBI / NIH: Research Funding. Szczepiorkowski:Fresenius Kabi: Consultancy, Membership on an entity's Board of Directors or advisory committees; Grifols: Consultancy, Research Funding; Terumo BCT: Consultancy; Cerus Corporation: Research Funding; Erydel: Research Funding; Citra Labs: Research Funding; US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding; American Association of Blood Banks: Other: President-Elect. Dunbar:Verax Biomedical: Membership on an entity's Board of Directors or advisory committees; US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Jones:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Rugg:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Prakash:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Hmel:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding. Ransom:US Army Medical Research and Material Command (Award W81XWH-15-C-0047) / Department of Defense: Research Funding.
Article
Background: Room temperature-stored (20-24°C) platelets (PLTs) have a shelf life of 5 days, making it logistically challenging to supply remote medical centers with PLT products. Cryopreservation of PLTs in dimethyl sulfoxide (DMSO) and storage at -80°C enables an extended shelf life up to 2 years. Although cryopreserved PLTs have been widely characterized under resting conditions, their ability to undergo agonist-induced activation is yet to be fully explored. Study design and methods: Buffy coat PLTs were cryopreserved at -80°C with 5% to 6% DMSO and sampled before freezing and after thawing. PLTs were analyzed under resting conditions and after agonist stimulation with adenosine diphosphate, collagen, or thrombin receptor-activating peptide-6. The expression of activation markers, microparticle formation, and calcium mobilization were analyzed by flow cytometry. Soluble PLT proteins present in the PLT supernatant were examined by enzyme-linked immunosorbent assay. Protein phosphorylation was investigated with Western blotting. Results: After cryopreservation, PLTs displayed increased surface activation markers and higher basal calcium levels. Cryopreserved PLTs demonstrated diminished aggregation responses. Additionally, cryopreserved PLTs showed a limited ability to become activated (as measured by CD62P and phosphatidylserine exposure and cytokine release) after agonist stimulation. A reduction in the abundance and phosphorylation of key signaling proteins (Akt, Src, Lyn, ERK, and p38) was seen in cryopreserved PLTs. Conclusions: Cryopreservation of PLTs induces dramatic changes to the basal PLT phenotype and renders them largely nonresponsive to agonist stimulation, likely due to the alterations in signal transduction. Therefore, further efforts are required to understand how cryopreserved PLTs achieve their hemostatic effect once transfused.
Article
Allogeneic platelet transfusions are widely used for the prevention and treatment of bleeding in thrombocytopenia. Recent evidence suggests platelet transfusions have limited efficacy and are associated with uncertain immunomodulatory risks and concerns about viral or bacterial transmission. Alternatives to transfusion are a well-recognised tenet of Patient Blood Management, but there has been less focus on different strategies to reduce bleeding risk by comparison to platelet transfusion. Direct alternatives to platelet transfusion include agents to stimulate endogenous platelet production (thrombopoietin mimetics), optimising platelet adhesion to endothelium by treating anaemia or increasing von Willebrand factor levels (desmopressin), increasing formation of cross-linked fibrinogen (activated recombinant factor VII, fibrinogen concentrate or recombinant factor XIII), decreasing fibrinolysis (tranexamic acid or epsilon aminocaproic acid) or using artificial or modified platelets (cryopreserved platelets, lyophilised platelets, haemostatic particles, liposomes, engineered nanoparticles or infusible platelet membranes). The evidence base to support the use of these alternatives is variable, but an area of active research. Much of the current randomised controlled trial focus is on evaluation of the use of thrombopoietin mimetics and anti-fibrinolytics. It is also recognised that one alternative strategy to platelet transfusion is choosing not to transfuse at all.
Article
Background: Alternatives to room temperature storage of platelets (PLTs) may be beneficial to extend the limited shelf life and support transfusion logistics in rural and military areas. The aim of this study was to assess the morphologic, metabolic, and functional aspects of PLTs stored at room temperature or in refrigerated conditions or cryopreserved. Study design and methods: A three-arm pool-and-split study was carried out using buffy coat-derived PLTs stored in 30% plasma/70% SSP+. The three matched treatment arms were room temperature stored (20-24°C), cold-stored (2-6°C), and cryopreserved (-80°C with dimethyl sulfoxide). Liquid-stored PLTs were tested over a 21-day period, while cryopreserved PLTs were examined immediately after thawing and after 6 and 24 hours of storage at room temperature. Results: Cold-stored and cryopreserved PLTs underwent a significant shape change, although the cryopreserved PLTs appeared to recover from this during subsequent storage. Glycolytic metabolism was reduced in cold-stored PLTs, but accelerated in cryopreserved PLTs, while oxidative phosphorylation was negatively affected by both storage conditions. PLT aggregation was potentiated by cold storage and diminished by cryopreservation in comparison to room temperature-stored PLTs. Cold storage and cryopreservation resulted in faster clot formation (R-time; thromboelastography), which was associated with an increase in microparticles. Conclusion: Cold storage and cryopreservation of PLTs led to morphologic and metabolic changes. However, storage under these conditions appears to maintain or even enhance certain aspects of in vitro PLT function.