You are on page 1of 16

Received: 22 July 2022 | Revised: 5 July 2023 | Accepted: 15 August 2023

DOI: 10.1111/prd.12526

REVIEW ARTICLE

Do autologous platelet concentrates (APCs) have a role in intra-­


oral bone regeneration? A critical review of clinical guidelines
on decision-­making process

Marc Quirynen | Sam Siawasch | Andy Temmerman | Simone Cortellini |


Rutger Dhondt | Wim Teughels | Anna B. Castro
Department of Oral Health Sciences, Periodontology, KU Leuven & Dentistry, University Hospitals Leuven, Leuven, Belgium

Correspondence
M. Quirynen, Department of Oral Health Sciences, Periodontology, KU Leuven, Kapucijnenvoer 7, blook a - bus 7001, 3000 Leuven, Belgium.
Email: marc.quirynen@uzleuven.be

1 | I NTRO D U C TI O N • Leukocyte-­ and PRF (L-­PRF) with leukocytes (the amount de-
pends on the protocol) and a high-­density fibrin network, or an in-
1.1 | What are autologous platelet concentrates jectable subcategory containing platelets, leucocytes, and fibrin.
(APCs)?

Autologous platelet concentrates (APCs) are biological products 1.2 | Differences between different APCs
(bioactive additives) derived from the patient's whole blood via
chair-­side centrifugation and consist of supraphysiologic concentra- 1.2.1 | 1st generation APCs
tions of platelets and growth factors (GFs). They are intended to en-
hance, accelerate, and promote tissue healing and regeneration (soft Platelet-­rich plasma (PRP)
and hard tissues) due to their potential to allow the gathering and PRP is a gel with a high concentration of autologous platelets sus-
concentration of platelets and other therapeutic blood constituents pended in a small amount of plasma after blood centrifugation.
­(fibrinogen/fibrin, growth factors, leukocytes, and circulating cells) The patient's anticoagulated blood is subjected to a first “soft”
in situ.1 spin to separate the plasma from the red blood cells (RBC; Figure 1).
Four variants of APCs have been introduced during the past The plasma fraction (top fraction) is further subjected to a second
three decennia: two platelet-­rich plasma (PRP) types representing “hard” spin to separate the platelets (PRP fraction = platelet-­rich
liquid platelet suspensions that can be transformed into a fibrin gel plasma) from the platelet-­poor plasma (PPP). The platelet pellet with
after activation, and two platelet-­rich fibrin (PRF) types that repre- some leukocytes is suspended in a lower volume of PPP and activated
sent solid platelet and fibrin clots (due to a strongly polymerized 3-­D by thrombin, calcium chloride (CaCl2), or type I collagen (e.g., from the
fibrin network). Based on the leukocyte content and fibrin structure, soft tissue in the surgical site). Through this dual centrifugation pro-
APCs have been classified into four main categories2–­5: cess, platelets are significantly enriched compared to whole blood.6
Over the past 25 years, numerous attempts have been made to
• Pure PRP (P-­PRP) without leukocytes and with a low-­density fi- standardize/improve the PRP preparation protocol. Unfortunately,
brin network, there is still a wide variation among studies with respect to the ro-
• Leukocyte-­ and PRP (L-­PRP), with leukocytes (the amount de- tational velocity, centrifugation time, blood volume, anticoagulants,
pends on the protocol) but with a low-­density fibrin network, and coagulation activators. As such, it is difficult to directly compare
• Pure PRF (P-­PRF) without leukocytes and with a high-­density fi- the reported outcomes. Whitman et al. and Marx et al. were the first
brin network, to promote the use of PRP in oral and maxillofacial surgery.7,8

M. Quirynen and S. A. M. Siawasch contributed equally to this article.

[Correction added on December 26, 2023 after first online publication: Given names for all authors have been included.]

© 2023 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.

254 | 
wileyonlinelibrary.com/journal/prd Periodontology 2000. 2023;93:254–269.
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 255

F I G U R E 1 Differences in preparation
of APCs, with 1st (PRP, PRGF) and 2nd
(L-­PRF) generation APC. L-­PRF, leukocyte-­
and platelet-­rich fibrin; P-­PGF, plasma
poor in growth factors; PPP, platelet poor
plasma; P-­RGF, plasma rich in growth
factors; PRP, platelet rich plasma; WBC,
white blood cells.

Plasma rich in growth factor (PRGF) now with human blood (14 volunteers). They reported, compared to
PRGF, a subcategory of PRP, is a plasma in a gelatinous form, en- whole blood, an 8.8 versus 2.8-­fold higher concentration of platelets
riched with platelets and growth factors. in PRP versus PRGF, and a 5.5 fold versus nearly no increase in WBC,
PRGF was introduced by Anitua et al.9,10 Patient's blood is com- respectively. PRP and PRGF need the addition of a coagulation acti-
bined with an anticoagulant (1 mL of 3.8% sodium citrate for 10 mL vator to be transformed into a gel. Their fibrin network is looser than
blood) and centrifuged at 460 g for 8 min. After centrifugation in the second-­generation APCs, which may influence their capacity
(Figure 1) three fractions are created: plasma at the top contain- to work as a scaffold.
ing mostly platelets (with two subfractions, F1 (the upper 20% in
a tube) and F2 (the next 20%)), the “buffy coat” (white blood cell
layer) in the middle (very small layer), and at the bottom the RBC 1.2.2 | 2nd generation APCs
(lower 60%). The plasma poor in growth factors fraction (PPGF, the
top part, the first fraction, F1), and the plasma rich in growth fac- Leukocyte-­and platelet-­rich fibrin (L-­PRF)
tors fraction (PRGF just above the “buffy coat”, F2) are transferred Platelet-­r ich fibrin (PRF), considered a “second generation plate-
separately into “individual” fractionation tubes. CaCl2 is added as an let concentrate”, is achieved with a simplified preparation, and
activator to both tubes to achieve platelet degranulation for the re- without biochemical manipulation of the patient's blood (no anti-
lease of growth factors. The F1 fraction (PPGF) can be transferred in coagulants, no coagulation activator). The pure platelet-­r ich fibrin
a glass receptacle and placed on top of a heating block for ±30 min (P-­PRF) subcategory only exists in a strongly activated gel form
to produce autologous fibrin (a fragile fibrin membrane) that can be and cannot be injected or used as traditional fibrin glue. There
used, for example, to seal the extraction site. The F2 fraction (PRGF) is only one commercial product in this family (Vivostat®), and its
will form in ±10 min a gelatinous substance that can be placed in the use remains limited. The leukocyte-­and platelet-­r ich fibrin (L-­PRF)
surgical site. The F2 fraction sometimes is further divided into the subcategory, however, offers many more advantages and is exten-
upper and lower half, with the lower half representing a qualitative sively used.
more interesting PRGF. For the preparation of L-­PRF, a solid form of APCs, one does
In summary: Giusto et al.11 examined, from pig blood, the concen- not require anticoagulants or thrombin (or any other gelling agent).
tration of platelets in both PRP variants. Compared to whole blood a This feature makes this product very easy to use, with a low risk
3.4-­fold higher (range 3.2–­4.2) concentration was reached with PRP, of mistakes during the preparation stage.13 The patient's blood is
and a 2.8-­fold higher (range 2.9–­4.9) concentration for PRGF. Nishi- collected in either special glass tubes or plastic tubes with a silica
yama et al.12 conducted a similar trial (with higher g-­forces for PRP), coating on the inside (to start up the coagulation cascade), in other
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
256 QUIRYNEN et al.

words, the choice of the right blood tubes is crucial. The patient's The 1st generation APCs are available in liquid solutions or an ac-
blood is immediately (within 1 min after collection) centrifuged at a tivated gel, whereas L-­PRF allows the formation of strong matrixes
g-­force of 408 (in the region where the L-­PRF clot is formed). During (membranes), allowing their use as a true barrier membrane (with a
centrifugation (≥12 min) the fibrinogen in the blood is converted into degradation of ±4 weeks). During the preparation of PRP and PRGF
a strong 3-­D fibrin matrix by the patient's thrombin. Blood platelets anticoagulants (at the start) and platelet activators (at the end) are
and leukocytes become trapped in this 3-­D fibrin net. After centrifu- required. Some of these factors can potentially have an adverse
gation, blood is separated into three fractions (Figure 1): ­effect on the coagulation process and/or lead to an immune re-
sponse.5 Moreover, anticoagulants have more recently been shown
• A clear solution of blood plasma in the upper phase, to interfere with the angiogenic and regenerative response mediated
• The buffy coat, a strong, fibrin clot (with enmeshed leukocytes by platelets. 29 For L-­PRF, however, the end product remains 100%
and platelets) in the middle, and autologous; the fibrin for example is formed via the physiologically
• An RBC fraction at the bottom of the tube (RBCs have the highest available thrombin.
density). Moreover, the preparation of PRP and to a lesser extent PRGF is
complex, but simple for L-­PRF. The number of platelets and leuko-
The L-­PRF clot can be collected from the blood tube with tweezers. cytes is clearly higher in L-­PRF membranes compared to PRGF and
After gentle removal of the remaining RBCs on the clot, it can be gently PRP gels. Also, the time of growth factor release differs significantly.
compressed into membranes (±5 min). L-­PRF is thus a 100% autologous PRP releases most growth factors within 60 min, whereas L-­PRF
biomaterial rich in fibrin, platelets, white blood cells, growth factors, shows a more gradual release (up to 2 weeks) as reported by Ko-
cytokines, and other components conducive to tissue repair.14–­17 L-­ bayashi et al.32 A PRGF scaffold shows quite similar release kinetics
PRF membranes are stable, resilient, strong, adhesive, and malleable. as L-­PRF.33
Within the L-­PRF membrane, the platelets are tightly merged, and One of the main differences between L-­PRF and PRP and espe-
18
the enmeshed leukocytes remain alive and functional. They possess cially PRGF is the presence of leukocytes in the L-­PRF. This is a key
attractive biochemical properties including hemostatic, angiogenic, factor for confronting infectious pathogens, and as such can reduce
osteogenic, anti-­inflammatory, anti-­microbial, pain-­inhibitory, and the probability of infection. They also play a key role in regulating the
wound-­healing characteristics rendering L-­PRF, at least desirable and immune response and in integrating host tissue and biomaterial.34,35
revolutionary.19–­22 The original protocol of leukocyte-­ and platelet-­
rich fibrin (L-­PRF), was introduced by Choukroun et al.23–­27 When the
original composition of the patient's whole blood is compared to the 1.3 | Differences between the original L-­PRF and
composition of an L-­PRF membrane (taking into consideration that an recent modifications
L-­PRF membrane weighs only 3% of the drawn blood), platelets and
WBC are increased 20–­25-­fold.28 Over the past decade small modi- 1.3.1 | The original L-­PRF protocol
fications to this original protocol have been introduced (CGF, A-­PRF,
A-­PRF+, T-­PRF, H-­PRF), with modification in centrifugation time and/ The original L-­PRF protocol foresees a relative centrifugal force
or speed, tubes, angle of tubes within centrifuge), but in reality, they (RCF) of 408 g (in the area where the clot is formed), and a centrifu-
all remain subtypes of L-­PRF, and as such the acronym L-­PRF applies gation time of 12 min (Table 2). 23–­27
to all of them.
In summary: Because it is a solid material, the L-­PRF membrane is
considerably easier (user-­friendly) to handle and position in a defect in 1.3.2 | CGF
comparison to a PRP gel. When the recommended procedure for the
preparation of L-­PRF is carefully followed, the content and architec- Sacco introduced in 2006 the “Concentrated Growth Factors”
ture of the resulting membrane are very consistent. L-­PRF can be used (CGF) protocol, with a centrifugation time of 13 min, changing the
as an autogenous grafting material/substitute because of its ability to RPM overtime (Medifuge centrifuge, Silfradent), in an attempt to
accelerate physiologic wound healing and new bone formation. The isolate a much larger, denser, and richer in growth factors fibrin
only drawback of L-­PRF is related to the timing during preparation; the matrix. 36,37
time between blood collection and centrifugation should be no more
than 1 min, which requires a stepwise centrifuge loading.17
1.3.3 | A-­PRF and A-­PRF+

1.2.3 | Major differences between 1st and 2nd The advanced PRF (A-­PRF) and the advanced plus PRF (A-­PRF+) pro-
generation APCs tocols were introduced to further increase the cellular content in the
L-­PRF clot and as such the release of growth factors.38,39 For A-­PRF
Table 1 summarizes the most significant differences between PRP a relative centrifugal force of 194 g, and for A-­PRF+ of 145 g, with a
and PRGF (as 1st generation) and L-­PRF (as 2nd generation) APCs. centrifugation time of 14 and 8 min, respectively, are used (Table 2).
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 257

TA B L E 1 Major differences between


Platelet concentrates PRP (1998) PRGF (2001) L-­PRF (2004)
the 1st generation (PRP, PRGF) and 2nd
generation (L-­PRF) APCs.13,18,30,31 Protocol Very complex Complex Easy
Speed rate Slow Very slow Fast
Reproducibility Possible bias Possible bias No bias
Use of anticoagulants Yes Yes No
100% autologous No No Yes
Amount obtainable Enough Poor Good
Fibrin morphology Tetramolecular Tetramolecular Trimolecular
Leukocytes amount 0–­50% 0% ≥65%
Immunomodulatory properties Poor No Yes
Neo-­angiogenic potential + ++ +++++
Osteoconductive potential Poor Poor High
(scaffolding)
Mechanical properties Enough Poor Good
(sol–­gel-­membrane)
Presence of MSCs Yes Yes Yes
Release in
TGF-­β1 ++ + +++
PDGF-­BB ++ + +++
VEGF ++ + +++
Duration of growth factor 70% in first 10 min, Primarily 1st week Up to
release ±all within 1 h 2 weeks
Costs of the protocol High Relatively high Low

TA B L E 2 Centrifuge settings for


IntraSpin® Duo Quattro® Medifuge®
the preparation of different L-­PRF
modifications for IntraSpin® (radius at L-­PRF variant RCF a
Time RPM b
RPM b
RPMb
area where clot is formed = 50 mm), Duo
Quatro® (77 mm), or Medifuge centrifuge Solid forms: clots/membranes
and of liquid forms, respectively. L-­PRF 408 g 12 min 2.700 2.180
A-­PRF 194 g 14 min 1.865 1.500
A-­PRF+ 145 g 8 min 1.610 1.300
CGF 453 g 2 min 2.700
358 g +4 min 2.400
453 g +4 min 2.700
560 g +3 min 3.000
Liquid forms of platelet concentratesc
Liquid fibrinogen 408 g 3 min 2.700 2.180
i-­PRF 60 g 3 min 820 700
C-­PRF 408 g 12 min 2.700 2.180

Note: Indicated are: the required RCF (=g-­force), the RPM (rotations/revolutions per minute), and
the time of centrifugation (in min).
a
Measured at the area where the L-­PRF clot will form.
b
IntraSpin, DuoQuattro, and Medifuge centriguge have a fixed angle rotor, the radius of centrifuge
in area where clot is formed is 50, 77, and 55.5 mm, respectively.
c
These liquid forms are prepared in inert blood tubes to delay the coagulation process.

Theoretically, less centrifugation time and/or lower centrifugation 1.3.4 | T-­PRF


speed would reduce pull-­down forces during centrifugation, which
would increase the total number of cells trapped within the clot/ Tunalı et al.39 introduced the T-­PRF (titanium-­prepared platelet rich-­
membrane. fibrin). The centrifugation protocol is identical to the original L-­PRF
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
258 QUIRYNEN et al.

protocol, but with the only difference that titanium tubes are used with an inert inner surface. The upper yellow fraction represents the
to centrifuge the blood, with the hypothesis that titanium would be liquid fibrinogen. It can be aspirated from the blood tube and has to be
more effective in activating platelets. This would result in a stronger used within 30 min, after which it will slowly start to coagulate sponta-
fibrin network. neously. This liquid is rich in fibrinogen, platelets, and leukocytes and
will form a 3-­D fibrin network.30 The latter is useful to trap a biomate-
rial to form a stronger block (sticky bone or L-­PRF bone block). The
1.3.5 | H-­PRF “clinical” benefit of this approach has recently been demonstrated.43

Miron et al.40 introduced the horizontal centrifugation protocol,


stating that, when compared to the normally used fixed-­angle cen- 1.4.3 | C-­PRF
trifuge, more platelets and leukocytes could be trapped within the
membrane with a better distribution over the entire membrane. Miron et al.44 introduced a novel harvesting technique called C-­PRF
(concentrated-­PRF) in an attempt to isolate the liquid buffy coat layer
(0.3 to 0.5 mL directly above the RBC layer) immediately after blood
1.3.6 | Overall impact centrifugation. This part indeed contains high concentrations of leuko-
cytes and platelets (±20 times higher than in the blood). This flowable
So far, contradictory laboratory data have been reported on the ben- platelet-­rich fibrin can be used alone or in combination with bioma-
eficial effect of the above-­mentioned centrifugation modifications terials. The “clinical” relevance of C-­PRF still needs to be confirmed.
in terms of cellular content and/or release in growth factors.17,38,40,41
Moreover, also the “clinical” benefits of these so-­called “improved”
clots/membranes still need to be confirmed. RCTs comparing these 1.5 | How to reach the desired g-­force in a
modifications directly to each other are indeed desirable. centrifuge?

The separation (layer formation) in the blood tube during centrifugation


1.4 | Differences between i-­PRF, liquid fibrinogen, (which is based on blood cell's density) depends on several factors in-
and C-­PRF? cluding: the time of spinning, the speed (rotations/revolutions per min-
ute, RPM), and the g-­force (also called the relative centrifugal force, RCF).
The flowable 2nd generation APCs (Table 2), the fibrin glues, also The RCF can be calculated with the following formula:
known as fibrin sealants, have been introduced to be used in com- RCF = 11.18 × radius × (RPM/1000)2. For this equation, the radius is
bination with a bone substitute or to have an injectable alternative the distance in centimeters from the center of the rotor to the sam-
(e.g., for joints (including the TMJ), chronic wounds). ple, and RPM is the rotor speed expressed as revolutions per minute.
It is especially important to realize the significance of these pa-
rameters. It clarifies, for instance, why not each centrifuge (each of
1.4.1 | i-­PRF them with its own radius) will lead to the same separation, and in the
case of L-­PRF to the same clot or membrane.45,46 Moreover, the RCF
i-­PRF (injectable PRF) is a liquid formulation, also prepared without differs slightly within the tube. Table 2 summarizes the g-­force cal-
additives (without anti-­coagulants), but using plastic hydrophobic culated for the region in which the L-­PRF clot is formed for tree fre-
inert tubes to slow down spontaneous clotting. Blood is centrifuged quently studied centrifuges (IntraSpin®, Duo Quattro®, Medifuge®).
at 60 g, for 3 min only. The upper liquid fraction represents the i-­
PRF. This flowable platelet-­rich fibrin can be either utilized alone
or combined with various biomaterials. Compared to the patient's 1.6 | Is there an impact of age or gender on the
whole blood, i-­PRF contains slightly more platelets and leukocytes outcome of APCs and how much blood can we donate
(±1.3 × higher).42 The “clinical” benefits of i-­PRF in oral surgery still without complications?
need to be confirmed.
Our blood is a mixture of about 55% plasma and 45% blood cells.
It accounts for about 7% to 8% of the total body weight and rep-
1.4.2 | Liquid fibrinogen resents approximately 5 L. A healthy adult can lose almost 20%
of blood volume (1 L) before the first symptoms appear (restless-
Liquid fibrinogen is also flowable.43 It has the significant advantage that ness), and approximately 40% of volume (2 L) before a hypov-
it can be prepared with the same setting as for L-­PRF (thus within the olemic shock sets in.
same centrifugation procedure, no need for a 2nd centrifugation/blood The RBC accounts for about 40% to 45% of the blood volume,
collection). For liquid fibrinogen, the spinning time is only 3 min at 2.700 the WBC as well as the platelets for less than 1%. Eventhough RBCs
RPM with a relative centrifugal force of 408 g, again using plastic tubes are smaller than WBCs, they are pushed to the bottom of blood
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 259

tubes during centrifugation, because they are more dense (higher transforming growth factor-­β (TGF-­β), fibroblast growth factor
specific mass/weight). (FGF), insulin-­like growth factor (IGF-­I and IGF-­II), vascular endo-
The relative proportion of RBC, measured via a hematocrit test, thelial growth factor (VEGF). The dense bodies contain adenosine
differs between men and women. It is normally 40.7% to 50.3% for diphosphate (ADP), adenosine triphosphate (ATP), ionized calcium,
men (4.7–­6.1 million cells), and 36.1% to 44.3% for women (4.2–­ histamine, serotonin, and adrenaline. These bioactive molecules
5.4 million cells per μL). On an individual level, the hematocrit value play important roles in different areas of regenerative medicine, in-
decreases in men in their sixth decade and in women in their seventh cluding bone remodeling, wound healing, hair regrowth, nerve re-
decade, and the change becomes more prominent with advancing generation, aging facial skin, acne scarring, androgenic alopecia, and
age, especially in men.47,48 The lower hematocrit in women and el- diabetic wounds.31
derly people explain the increased size of their L-­PRF membranes.49 Signaling molecules of importance during bone healing (Table 3)
Yajamanya et al.50 observed that the fibrin network of L-­PRF mem- can be categorized into three groups: (i) the pro-­inflammatory cy-
branes was less dense as patient age increased. Whether these ob- tokines [interleukin-­1, interleukin-­6 , and tumor necrosis factor-­α
servations have a clinical impact is still unclear. (TNF-­α)], (ii) the TGF-­β superfamily (including bone morphoge-
netic proteins) and other growth factors (PDGF, FGF, and IGF),
and (iii) the angiogenic factors [VEGF, angiopoietins 1 and 2, and
1.7 | How do APCs work? matrix metalloproteinases, that degrade bone and cartilage and
enable vessel invasion].
1.7.1 | Platelets

During wound healing, platelets arrive among the first cells at a wound 1.7.2 | Leukocytes
site. Besides their procoagulant effects, platelets form a rich source
of important growth factors. They contain more than 300 biologically At first sight, the presence of leukocytes may be controversial, due
active molecules that are released upon activation from platelet alpha to a potential risk of stimulating an inflammatory process.10 On the
granules and dense bodies. Alpha granules and dense bodies (also contrary, several research groups insisted on the need for some leu-
known as dense granules or delta granules) are specialized secretory kocyte population, even in the injectable PRP, to increase the pro-
organelles. Activated platelet-­derived factors serve as messengers and duction of growth factors, the release of anti-­pain mediators, and
regulators that influence a variety of cell–­cell and cell-­extracellular ma- a natural anti-­infectious activity.51–­53 Leukocytes can also regulate
trix (ECM) interactions and modify the pericellular microenvironment. cell proliferation and cell differentiation. In addition, they are the
The alpha granules contain fibrinogen, fibronectin, factor V and basic cells responsible for wound healing and the first cells to start
von Willebrand's factor, platelet-­derived growth factor (PDGF), neo-­angiogenesis.54–­59

TA B L E 3 Production and action of signaling molecules (adapted from Upadhayaya et al.).31

Factors Produced by Action

Cytokines Macrophages & mesenchymal Exert chemotactic activity on inflammatory cells, enhance cellular matrix
cells synthesis and stimulate angiogenesis
Matrix metalloproteinases Macrophages, smooth muscle Degrade cartilage and bone; this allows angiogenic factors to regulate vessel
cells & osteoblasts ingrowth by either the vascular endothelial growth factor-­dependent
pathway or the angiopoietin-­dependent pathway
Transforming growth Osteoblasts Stimulates the expression of bone matrix proteins and suppresses the
factor-­β (TGF-­β) degrading activity of matrix metalloproteinases and other enzymes;
also induces the differentiation or proliferation of osteoblastic cells
while inhibiting the formation of osteoclast precursors and, in greater
concentrations, may exert an inhibitory effect on mature osteoclasts
Platelet-­derived growth Platelets, monocytes, Potent mitogen for mesenchymal cells, powerful chemotactic agent for
factor (PGDF) macrophages, endothelial inflammatory cells and a stimulus for osteoblasts and macrophages
cells & osteoblasts
Fibroblast growth factor Monocytes, macrophages, Chondrogenesis and bone resorption
(FGF) mesenchymal cells,
chondrocytes & osteoblast
Insulin-­like growth factor Bone matrix, endothelial cells, Bone matrix formation and in the later stages of endochondral bone
(IGF) osteoblasts & chondrocytes formation
Vascular endothelial growth Macrophages, smooth muscle Induces the migration and proliferation of endothelial cells using
factor (VEGF) cells & osteoblasts transmembrane adhesion proteins (intergrins); also induces relaxation in
the cell-­to-­cell contact of endothelial cells, resulting in hyperpermeability
of blood vessels
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
260 QUIRYNEN et al.

Neutrophils are recruited to the site of injury within minutes pathological pain. During inflammation, these cytokines counteract
following trauma and are the hallmark of acute inflammation. Their the effects of the pro-­inflammatory mediators naturally generated
primary role is to phagocytize debris, microbes, and necrotic tissue, in the early stages of inflammation. This might explain the signifi-
in order to clean the wound and prevent infection. They also pro- cant reduction in complications after 3rd molar extraction. Recent
duce inflammatory cytokines and growth factors.9 After completing data indicate that L-­PRF has anti-­inflammatory activity and shifts the
these functions during the early inflammatory phase of healing, the macrophage polarization from an M1 toward an M2 phenotype.63
neutrophils die by apoptosis.
Monocytes are the second type of leukocytes that are recruited
to the wound site and mature to become macrophages. When ap- 1.7.3 | L-­PRF Fibrin matrix
plying L-­PRF to a wound, before circulating monocytes can extrava-
sate the blood vessels, L-­PRF could already supply monocytes. Since Although leukocytes and platelets play a vital role in the L-­PRF healing
monocytes must attach to the extracellular matrix for differentiation, capacity, it has also been suggested that it is the fibrin matrix that sup-
entrapment of the monocytes in the L-­PRF fibrin clot would already ports the healing (including the gradual release of growth factors) and
60
induce their maturation to macrophages. Thereby, perhaps the early is also responsible for the therapeutic potential. L-­PRF might serve as
and supplemented macrophage levels from L-­PRF could shorten the a resorbable membrane for guided bone regeneration (GBR), prevent-
inflammatory phase and wound healing. Macrophages phagocytose ing the migration of non-­desirable cells into the bone defect, provid-
the dead neutrophils and further debride the wound, which is of ing a space that allows the immigration of osteogenic and angiogenic
course critical for normal wound healing. Together with the neutro- cells, and permitting the underlying blood clot to mineralize.34,64
phils and platelets, macrophages also prevent infections by eliminating
microbes. They also play an essential role in bone repair. Macrophages
apparently direct osteogenic cell signals and promote bone mineral- 2 | D O A P C S PL AY A “ S I G N I FI C A NT ”
ization during in vitro studies. Inducing angiogenesis is a key role of RO LE I N B O N E R EG E N E R ATI O N?
macrophages. New blood vessels are of course essential for delivering
nutrients, inflammatory cells, and oxygen. Moreover, macrophages To evaluate the “adjunctive” benefit of APCs in bone regeneration
also participate in the formation of granulation tissue and the removal within the oral cavity, a narrative review of the literature has been
of necrotic tissue. Macrophages secrete collagenase, which promotes conducted on their impact during alveolar ridge preservation (APR),
the cleaning of the wound. Other critical roles of macrophages include sinus floor augmentation, and guided tissue regeneration (GTR).
the release of growth factors and cytokines, recruitment of other cell Only papers, published before June 2022, where APCs were used
types to the wound, and stimulation of angiogenesis. as “sole” substitutes (thus not in combination with bone substitutes,
Like platelets and macrophages, other leukocytes also secrete statins, or enamel matrix derivatives) and in humans were consid-
many growth factors important in healing, including: TGF-­β1, PDGF, ered. However, because of the low number of papers on PRP and
VEGF, IGF, epidermal growth factor (EGF), TGF-­α , and basic fibro- PRGF as the “sole” substitute in GTR, for this indication, a combina-
blast growth factor (bFGF).55,61 tion of APCs with a bovine bone substitute was also considered.
In addition to growth factors, leukocytes express many pro- For alveolar ridge preservation and guided tissue regeneration,
teinases, such as serine and matrix metalloproteinases, which only randomized controlled trials (RCTs) or controlled clinical trials
also have critical roles in wound healing. 56 Proteinases have an- (CCTs) were considered. For sinus floor, augmentation case series
timicrobial activities and contribute to the migration of polymor- were also included.
phonuclear leukocytes and modulate inflammatory responses. There is considerable confusion in the literature when the term
Proteinases control the inflammatory response by deactivated PRF is employed, as it is often not specified whether the authors
inflammatory cells to limit injury to surrounding tissues during refer to P-­PRF or L-­PRF. However, all the studies identified in this
wound healing. Another crucial role of proteinases is to control review mentioned a protocol where leukocytes were present in the
the activity of growth factors, such as TGF-­β, PDGF, and bFGF. platelet concentrates, thus representing L-­PRF or one of its modifi-
Matrix metalloproteinases also have important roles in cartilage cations (CGF, A-­PRF, A-­PRF+, T-­PRF, H-­PRF).
and bone remodeling and angiogenesis.
L-­PRF membranes also have an antibacterial capacity, for exam-
ple against methicillin-­resistant Staphylococcus aureus and Esche- 2.1 | What is the impact of APCs as “sole” bone
richia coli, and against several periodontopathogens. 20,62 Leukocytes substitutes during “alveolar ridge preservation (ARP)”?
increase the antibacterial impact and might reduce the probability of
infection up to 10 times. After tooth extraction, the alveolar ridge undergoes a remodeling
Leukocytes also present anti-­nociceptive effects through dif- in both horizontal and vertical directions. After 6 months, according
ferent chemokines, anti-­inflammatory cytokines (IL-­4, IL-­10, and to Tan et al.,65 the vertical resorption ranges from 11% to 22% and
IL-­13) and opioid peptides (b-­endorphin, metenkephalin, and dynor- the horizontal resorption from 29% to 63%. However, the exten-
phin-­A), and can therefore promote a clinically relevant inhibition of sion of alveolar bone resorption may vary depending on tooth type
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 261

as observed by Couso-­Queiruga et al.66 with more horizontal bone • Significantly less alveolar ridge resorption,73,81–­84
resorption in molar versus non-­molar sites (3.6 vs. 2.5 mm). An ad- • Histologically improved bone quality and higher radiographically
ditional bone loss of 2.4 mm in width and 1.1 mm in height has been bone density,81–­83,85
67
reported in sites with a compromised buccal bone wall. This often • Faster soft tissue healing,84,86–­88
jeopardizes the placement of implants in an optimal position. • Less post-­extraction pain and swelling,86,87
Many surgical techniques have been introduced to minimize this • Lower incidence of alveolitis.86,87
68–­70
alveolar bone resorption. Different bone grafts/bone substi-
tutes have been used to fill extraction sockets, with or without the Another important advantage is that primary wound closure is not
addition of a soft tissue graft and/or a soft tissue substitute to seal required, and even not advised since the release of a mucoperiosteal
the alveolus. Biological additives, for instance, APCs, have also been flap and the use of releasing incisions will reduce the local blood supply
proposed as substitutes for bone regeneration. The absence of bone and as such jeopardize the regeneration process.85 This offers the ad-
substitute remnants after healing when only APCs are used has been ditional advantage of not losing gingival width and vestibulum depth.
seen as an important advantage.71–­73 However, the results are often less favorable when the following mod-
ifications are made to the above-­mentioned protocol: incomplete socket
fill with, for example, only one L-­PRF clot/membrane/plug, no sealing of
2.1.1 | PRP extraction socket entrance with an L-­PRF membrane, use of clots instead
of membranes/plugs, improper condensation of L-­PRF in the extraction
74,75
Two RCTs evaluated the use of PRP for ridge preservation. Alissa socket, removal of the face part of the clot, multiple extractions next to
et al.74 reported less pain, better soft tissue healing, and a denser trabec- each other, extraction site covered by a removable denture.89–­92
ular bone pattern when PRP was compared to unassisted healing. How-
ever, there were insufficient data to support the use of PRP to prevent
bone resorption. Nisar et al.,75 on the contrary, observed favorable out- 2.1.4 | Explanation for the beneficial effects when
comes in terms of preservation of the alveolar bone height but failed to applying L-­PRF during alveolar ridge preservation
show any significant results in terms of alveolar bone width resorption.
The included articles proposed the following potential explanations
for beneficial effects obtained with L-­PRF:
2.1.2 | PRGF
• The formation of a natural fibrin matrix that supports clot forma-
Several RCTs have analyzed the effect of PRGF on hard and soft tis- tion and covers the clot to prevent mechanical dislodgement, 23–­25
sues after tooth extraction. Despite some controversy data,76 PRGF • The increased potential for bone regeneration by the released
has been reported to provide the following potential advantages: growth factors, including a reduced healing time,30
• The enhancement of the neo-­angiogenesis and vasculogenesis,19
9,77,78
• Significantly faster socket closure/healing • The immune regulation node with inflammation retro-­control
79
• Less post-­extraction pain abilities, 26
79
• Significantly improved soft tissue healing • The sealing effect of the L-­PRF (in order to delay/prevent the in-
• Histologically greater percentage of newly formed bone77,80 growth of epithelium),
• The antibacterial activity of L-­PRF constructs, 20
The use of PRGF may be recommended when early healing is • The hemostatic effect.
needed, for instance, in hospitalized patients.

2.2 | What is the impact of APCs as “sole” bone


2.1.3 | L-­PRF substitutes during sinus floor elevation?

A large number of RCTs and CCTs have examined the potential It still remains an open question whether a bone substitute is really
benefits of applying L-­PRF in the socket immediately after tooth necessary for a sinus floor elevation via a transcrestal or a 1-­stage
extraction. Several protocols have been introduced using either the lateral window approach, applying the “tent pole technique” with the
non-­condensed L-­PRF clots or the condensed membranes/plugs. implant(s) keeping up the elevated Schneiderian membrane. Several
Moreover, the number of L-­PRF clots/plugs/membranes per socket clinical trials indeed reported a favorable outcome for a sinus aug-
varied significantly between studies. mentation without grafting (blood only) including solicitude regarding
When using the “correct” protocol (including the use of “con- remaining graft particles, reduced rates of complications, and lower
densed” membranes/plugs, the application of sufficient numbers of costs.93–­100 However, such a graft-­free procedure still remains contro-
them, and sealing of the alveolus with L-­PRF membranes) highly sig- versial. A crucial aspect for such a procedure is the absence of a per-
nificant improvements have been reported including: foration of the sinus membrane, which unfortunately occurs relatively
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
262 QUIRYNEN et al.

frequently. This is of course not a concern in the case of the use of for the window) with the use of a bone substitute (DBBM) and a col-
example an APC membrane. lagen membrane over the window, and reported similar outcomes.

2.2.1 | Transcrestal approach, APCs only 2.3 | What is the impact of APCs as “sole” bone
substitute for bone regeneration in intra-­bony defects
PRP during open flap debridement (OFD)?
For PRP no studies could be identified.
Deep crater-­shaped defects because of severe periodontitis are
PRGF often treated via non-­surgical periodontal therapy followed by peri-
PRGF has been explored in one clinical trial, reporting a vertical odontal regeneration. Ideally, new bone, cementum, and periodontal
bone gain of 3.7 mm after 1 year, and of 4.2 mm after 3 years.101 ligament will be formed. Wound stability (for stable blood clot for-
mation), and space provision (for new tissue formation within the
L-­PRF defect) are mandatory to obtain periodontal regeneration.
Five prospective case series reported on transcrestal sinus floor el- The regenerative surgery can be accompanied by the use of dif-
evation with L-­PRF as the sole substitute.64,102–­105 In general, a bone ferent biomaterials, including APCs.114–­118 The rational for applying
gain of 3.2–­4.4 mm could be obtained. In three out of the five stud- APCs is twofold. Firstly, APCs can serve as a stable scaffold ensuring
ies, the implant survival rate after 1 year of loading was >97%. In the space provision/wound stability, and at the same time be a source of
study of Molemans et al., with data from post-­graduate students, the crucial cells for wound cleaning and healing/repair. APCs for exam-
implant survival rate was 91%.104 Testori et al.105 reported a 5-­year ple promote the neo-­angiogenesis, cell migration, and bone forma-
cumulative survival rate of 93.3%. Similar observations were made in tion. Secondly, APCs can be used as a barrier membrane to cover the
two retrospective studies applying CGF as the only substitute.106,107 intra-­bony defects. An APC barrier will prevent the apical migration
Cho et al.108 compared the use of 5 mL of saline with 5 mL of of gingival epithelial and connective tissue cells. This will provide the
L-­PRF as filling material after hydraulic transcrestal sinus lifting and regenerative cells more time to restore the periodontal tissues.119,120
observed significantly more bone gain in the L-­PRF group. In total, 22 RCTs reported on the adjunctive benefits of using
It was suggested that you would need 1 L-­PRF membrane per APCs as a sole biomaterial in the treatment of intra-­bony defects
mm sinus augmentation. during OFD. It is, however, important to realize that different strat-
egies have been applied (Figure 2). In the included studies, PRP and
PRGF were only used as a scaffold (Figure 2C), whereas L-­PRF has
2.2.2 | 1-­stage Window approach, APCs only been tested for all three strategies (Figure 2B–­D).

PRP/PRGF
We were not able to find a paper on the use of PRP or PRGF as a 2.3.1 | PRP
single substitute during a 1-­stage sinus floor elevation.
Only one RCT evaluated the adjunctive benefit of applying PRP in
L-­PRF 3-­wall intra-­bony defects. In comparison to OFD alone, the addition
However, six clinical studies explored the outcome of L-­PRF for of PRP resulted in significantly more: probing pocket depth reduc-
104,109–­113
this indication. L-­PRF clots or membranes have been tion (0.8 mm), intra-­bony defect reduction (2.6 mm), and intra-­bony
used in these studies. The overall mean bone augmentation defect fill (55%).121
ranged from 5.3 to 10.4 mm and the implant survival rate was al-
ways 100%, even up to 6 years follow-­u p. Several authors sug-
gested using the bony window (folded inside) on top of the apices 2.3.2 | PRGF
of the implants as a new floor for the sinus. The simultaneously
inserted implants of course functioned as tent poles to keep the The beneficial effect of PRGF was explored in one RCT in which only
sinus membrane up. It was again recommended to use 1 L-­PRF 2-­wall defects were treated. This study could not reveal any statistically
membrane per mm sinus augmentation. significant difference, but it is unclear if smokers were excluded or not.122
112
Dominiak et al. compared in an RCT the amount of bone gain
between either a xenograft alone (Cerabone) or L-­PRF alone for a 1-­
stage lateral window approach. The survival rate of implants in both 2.3.3 | L-­PRF
groups was 100%, with no implant mobility or pain. At the 36-­month
follow-­up visit, the xenograft group showed significantly more bone As barrier membrane
height gain (4.5 mm vs. 3.4 mm for the A-­PRF group). Merli et al.113 The benefit of covering the intra-­bony defect with L-­PRF as a bar-
compared in another RCT the use of CGF (to fill the sinus and to seal rier membrane during OFD was explored in three RCTs.123–­125 In all
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 263

studies a statistically significant benefit with L-­PRF was found for (0.8–­1.4 mm), gain in attachment level (1.0–­1.6 mm), and bone defect
probing pocket depth reduction (±1.1 mm), two studies reported sig- fill (several parameters).
nificantly more gain in attachment level (±1.0 mm), and one study
observed significantly more reduction of the intra-­bony defect
depth (±2.0 mm). It remains unclear if there is more benefit when 2.3.4 | PRP versus L-­PRF
using multiple membrane layers versus a single layer.
Two studies compared the benefits of L-­PRF with those of PRP,
As defect filler as filler of intra-­bony defects, either covered or not with a barrier
Seven RCTs evaluated the added value of applying L-­PRF during the membrane during OFD, and reported slightly better outcomes in
126–­132
OFD procedure to fill the intra-­bony defects. L-­PRF was ap- favor of the L-­PRF, especially when the intra-­bony defects were also
plied in a clot (four studies) or membrane (three studies) form. As far covered.121,142
as mentioned all studies included 2 or 3 wall defects. The follow-­up
period was 9–­12 months, and smokers were excluded. Statistically
significant adjunctive benefits were found in favor of L-­PRF in terms 2.4 | What is the impact of APCs + “xenograft” for
of probing pocket depth reduction (0.9–­2.4 mm), gain in attachment bone regeneration in intra-­bony defects during open
level (0.8–­3.3 mm), and bone fill (several parameters). A clear differ- flap debridement?
ence between membrane and clot could not be made.
To prevent collapse and preserve the space for regeneration, a com-
As bony defect filler + barrier membrane bination of APCs and another (more stable) grafting material could
In 10 RCTs L-­PRF was used both as filler and as barrier membrane be another treatment option for intra-­bony defects around teeth.
during OFD of 2 or 3 wall defects.121,133–­141 Again, L-­PRF as ad- In total seven RCTs explored the beneficial effects of combin-
junct resulted in statistically more probing pocket depth reduction ing APCs with a xenograft for treatment of intra-­bony defects in

F I G U R E 2 Different protocols for the using APCs only in intra-­bony defects during OFD: (A) open flap debridement as control versus:
(B) the coverage of the defect only with a APC membrane as barrier (yellow orange layer in drawing), (C) the filling of the bony crater only
(yellow orange in drawing), or (D) the application of an APC to both fill and cover the defect.

F I G U R E 3 Different protocols for


using a xenograft in combination with
APCs in intra-­bony defects during OFD:
(A) OFD + xenograft alone as defect filler
(blue stars) serving as control treatment,
versus: (B) OFD + xenograft mixed
with an APC to fill the defect, and (C)
OFD + xenograft mixed with an APC to
fill the defect plus an APC membrane to
cover the defect.
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
264 QUIRYNEN et al.

comparison to a xenograft alone. Again different strategies have 3.2 | Sinus floor elevation
been applied (Figure 3).
Several case series indicated that L-­PRF can be successfully used
as a single substitute for a transcrestal approach, with high-­implant
2.4.1 | PRP survival rates and acceptable vertical bone gain (3–­4 mm). It also
has the benefit of reducing post-­op pain, and the long-­term vertical
Three RCTs made the comparison between xenograft mixed bone resorption is limited.106,107,152 As such it can be recommended
with PRP versus xenograft alone for the treatment of intra-­bony in clinical practice. For PRP and PRGF, there is nearly no data for its
defects.143–­145 Two studies found statistically significant adjunctive application, probably because of their weak physical characteristics.
benefits when adding PRP concerning probing pocket depth reduc- L-­PRF is also the only APC used as a single substitute during
tion (≥1.0 mm) and gain in attachment level (≥0.9 mm). One of those a 1-­stage lateral window approach. Several case series reported
studies reported additionally significant bone fill with PRP. The third high-­implant survival rates and good vertical bone gain with this ap-
study however could not find any statistical differences.145 proach. The use of L-­PRF, of course, reduces the costs of the treat-
ment, has the advantage of being 100% autogenous, and results in
a well-­vascularized bone without remnants of bone substitutes. Un-
2.4.2 | PRGF fortunately, so far only two RCTs compared the use of L-­PRF (as a
single substitute) with the application of standard bone substitutes.
For PRGF no studies could be identified. However, both studies indicated similar implant survival rates, but a
slightly lower vertical bone gain when using L-­PRF only.112,113

2.4.3 | L-­PRF
3.3 | Bone regeneration in intra-­bony defects
Four RCTs evaluated the benefit of combining a xenograft with L-­
PRF.146–­149 They all reported significantly more gain in the attach- A large number of papers evaluated the benefits of using APCs dur-
ment (0.9–­1.2 mm) and intra-­bony defect depth reduction (11%) ing open flap debridement. The large variety of protocols (APC as
when L-­PRF was mixed with the xenograft. filler material/as barrier membrane/or a combi of both, with/without
additional bone substitute) makes it difficult to give clear recommen-
dations, on which strategy is most beneficial. However, one can con-
3 | DISCUSSION clude that most applications resulted in significant improvements
in probing pocket depth reduction, clinical attachment gain, and
APCs have a number of benefits including: the release of growth intra-­bony defect fill when compared with OFD alone. The latter is
factor, a fibrous structure suitable for cell-­attachment, an improved confirmed by several systematic reviews including meta-­analyses.120
angiogenesis, an antibacterial capacity, and a differentiation of stem Unfortunately, there are no RCTs comparing the regenerative capac-
cells toward the osteoblast. These characteristics should facilitate ity of APCs to more standard biomaterials like enamel matrix deri-
wound healing/repair, and support soft-­ and hard-­tissue regenera- vates, hyaluronic acid, or bone substitutes.
tion. In the past decade, a large number of papers have investigated
the adjunctive impact of APCs in periodontal surgery.
4 | CO N C LU S I O N

3.1 | Alveolar ridge preservation L-­PRF is easy to prepare, simple to manage, not expensive, and
100% autogenous. As indicated in this review it offers a number
The benefits of PRPs on alveolar ridge preservation are controver- of benefits in ARP, sinus floor elevation, and the regeneration of
sial, probably because of different preparation protocols. Therefore, infra-­b ony craters around teeth. RCTs comparing their impact with
its application has become very limited.74,75,150 PRGF has shown to the “standard” periodontal treatments are scares or lacking, but
be effective,77,80 although some studies could not find any differ- the data presented in this paper are promising and justifies their
76
ence with unassisted healing. clinical use. An important question is whether their application
A series of studies supported the use of L-­PRF for ARP, not only offers additional benefits in more demanding conditions (elderly
accelerating soft tissue healing,84,86–­88 but also limiting alveolar patients, smokers, and patients with compromised wound heal-
ridge resorption.73,81,83,151 One should however realize that an in- ing). The clinical reports on the benefits of APCs in the healing
adequate use of L-­PRF (insufficient membranes apply, instability of of chronic non-­responding wounds (e.g., diabetic foot, venous leg
the wound, etc.) might jeopardize a promising bone regeneration. L-­ ulcer, pressure ulcers) seem to indicate that one can expect a bene-
PRF also significantly improved the PROMs including pain, swelling fit in these unfavorable conditions. The data on the 1st generation
as well as post-­op bleeding. APCs for the above-­m entioned indications are non-­conclusive.
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 265

C O N FL I C T O F I N T E R E S T S TAT E M E N T Choukroun's platelet-­rich fibrin clot and membrane. J Periodontol.


All (co)-­authors declare that they have no conflict of interest. The 2010;81(4):546-­555.
15. Anitua E, Alkhraisat MH, Orive G. Perspectives and challenges
department of periodontology owns research chairs from sev-
in regenerative medicine using plasma rich in growth factors. J
eral implant companies: Dentsply Sirona, Straumann, and Henry Control Release. 2012;157(1):29-­38.
Schein. 16. Li Q, Pan S, Dangaria SJ, et al. Platelet-­rich fibrin promotes peri-
odontal regeneration and enhances alveolar bone augmentation.
Biomed Res Int. 2013;2013:638043.
DATA AVA I L A B I L I T Y S TAT E M E N T
17. Castro AB, Andrade C, Li X, Pinto N, Teughels W, Quirynen M.
Data sharing is not applicable to this article as no new data were cre- Impact of g force and timing on the characteristics of platelet-­rich
ated or analyzed in this study. fibrin matrices. Sci Rep. 2021;11(1):6038.
18. Yu S, Wang Y, Miron RJ, Zhang Y. Structure, barrier function, and
bioactivity of platelet-­rich fibrin following thermal processing.
ORCID
Tissue Eng Part C Methods. 2021;27(11):605-­615.
S. A. M. Siawasch https://orcid.org/0000-0002-5860-761X 19. Ratajczak J, Vangansewinkel T, Gervois P, et al. Angiogenic
properties of ‘Leukocyte-­ and platelet-­rich Fibrin’. Sci Rep.
REFERENCES 2018;8(1):14632.
20. Castro AB, Herrero ER, Slomka V, Pinto N, Teughels W, Quirynen
1. Yung YL, Fu SC, Cheuk YC, et al. Optimisation of platelet concen-
M. Antimicrobial capacity of leucocyte-­and platelet rich fibrin
trates therapy: composition, localisation, and duration of action.
against periodontal pathogens. Sci Rep. 2019;9(1):8188.
Asia Pac J Sports Med Arthrosc Rehabil Technol. 2017;7:27-­36.
21. Kargarpour Z, Nasirzade J, Strauss FJ, et al. Platelet-­rich fi-
2. Dohan Ehrenfest DM, Rasmusson L, Albrektsson T. Classification
brin suppresses in vitro osteoclastogenesis. J Periodontol.
of platelet concentrates: from pure platelet-­rich plasma (P-­PRP)
2020;91(3):413-­421.
to leucocyte-­ and platelet-­ rich fibrin (L-­PRF). Trends Biotechnol.
22. Pitzurra L, Jansen IDC, de Vries TJ, Hoogenkamp MA, Loos
2009;27(3):158-­167.
BG. Effects of L-­PRF and A-­PRF+ on periodontal fibroblasts
3. Dohan Ehrenfest DM, Andia I, Zumstein MA, Zhang CQ, Pinto
in in vitro wound healing experiments. J Periodontal Res.
NR, Bielecki T. Classification of platelet concentrates (platelet-­rich
2020;55(2):287-­295.
plasma-­PRP, platelet-­rich fibrin-­PRF) for topical and infiltrative
23. Choukroun J, Diss A, Simonpieri A, et al. Platelet-­rich fibrin (PRF):
use in orthopedic and sports medicine: current consensus, clin-
a second-­generation platelet concentrate. Part IV: clinical effects
ical implications and perspectives. Muscles Ligaments Tendons J.
on tissue healing. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
2014;4(1):3-­9.
2006a;101(3):e56-­e60.
4. Varshney S, Dwivedi A, Pandey V. Antimicrobial effects of various
24. Choukroun J, Diss A, Simonpieri A, et al. Platelet-­rich fibrin (PRF):
platelet rich concentrates-­vibes from in-­vitro studies-­a systematic
a second-­generation platelet concentrate. Part V: histologic eval-
review. J Oral Biol Craniofac Res. 2019;9(4):299-­3 05.
uations of PRF effects on bone allograft maturation in sinus lift.
5. Pietruszka P, Chruścicka I, Duś-­Ilnicka I, Paradowska-­Stolarz A.
Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2006b;101(3):29
PRP and PRF-­subgroups and divisions when used in dentistry. J
9-­3 03.
Pers Med. 2021;11(10):944.
25. Dohan DM, Choukroun J, Diss A, et al. Platelet-­rich fibrin (PRF):
6. Oudelaar BW, Peerbooms JC, Huis In't Veld R, Vochteloo AJH.
a second-­generation platelet concentrate. Part I: technological
Concentrations of blood components in commercial platelet-­rich
concepts and evolution. Oral Surg Oral Med Oral Pathol Oral Radiol
plasma separation systems: a review of the literature. Am J Sports
Endod. 2006a;101(3):e37-­e 44.
Med. 2019;47(2):479-­487.
26. Dohan DM, Choukroun J, Diss A, et al. Platelet-­rich fibrin (PRF):
7. Whitman DH, Berry RL, Green DM. Platelet gel: an autologous al-
a second-­generation platelet concentrate. Part II: platelet-­related
ternative to fibrin glue with applications in oral and maxillofacial
biologic features. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
surgery. J Oral Maxillofac Surg. 1997;55(11):1294-­1299.
2006b;101(3):e45-­e50.
8. Marx RE, Carlson ER, Eichstaedt RM, Schimmele SR, Strauss JE,
27. Dohan DM, Choukroun J, Diss A, et al. Platelet-­rich fibrin (PRF): a
Georgeff KR. Platelet-­rich plasma: growth factor enhancement
second-­generation platelet concentrate. Part III: leucocyte activa-
for bone grafts. Oral Surg Oral Med Oral Pathol Oral Radiol Endod.
tion: a new feature for platelet concentrates? Oral Surg Oral Med
1998;85(6):638-­6 46.
Oral Pathol Oral Radiol Endod. 2006c;101(3):e51-­e55.
9. Anitua E. Plasma rich in growth factors: preliminary results of use
28. Andrade C, Camino J, Nally M, Quirynen M, Martínez B, Pinto N.
in the preparation of future sites for implants. Int J Oral Maxillofac
Combining autologous particulate dentin, L-­PRF, and fibrinogen to
Implants. 1999;14(4):529-­535.
create a matrix for predictable ridge preservation: a pilot clinical
10. Anitua E, Sánchez M, Orive G, Andía I. The potential impact of
study. Clin Oral Investig. 2020;24(3):1151-­1160.
the preparation rich in growth factors (PRGF) in different medical
29. Oneto P, Zubiry PR, Schattner M, Etulain J. Anticoagulants inter-
fields. Biomaterials. 2007;28(31):4551-­4560.
fere with the Angiogenic and regenerative responses mediated by
11. Giusto G, Vercelli C, Iussich S, Tursi M, Perona G, Gandini M.
platelets. Front Bioeng Biotechnol. 2020;8:223.
Comparison of the effects of platelet-­rich or growth factor-­rich
30. Castro AB, Cortellini S, Temmerman A, et al. Characterization of
plasma on intestinal anastomosis healing in pigs. BMC Vet Res.
the leukocyte-­and platelet-­rich fibrin block: release of growth fac-
2017;13(1):188.
tors, cellular content, and structure. Int J Oral Maxillofac Implants.
12. Nishiyama K, Okudera T, Watanabe T, et al. Basic characteristics of
2019;34(4):855-­864.
plasma rich in growth factors (PRGF): blood cell components and
31. Upadhayayaa V, Arorab A, Goyal A. Bioactive platelet aggre-
biological effects. Clin Exp Dent Res. 2016;2(2):96-­103.
gates: Prp, Prgf, Prf, Cgf and sticky bone. IOSR J Dent Med Sci.
13. Giannini S, Cielo A, Bonanome L, et al. Comparison between PRP,
2017;16(5):5-­11.
PRGF and PRF: lights and shadows in three similar but different
32. Kobayashi E, Flückiger L, Fujioka-­Kobayashi M, et al. Comparative
protocols. Eur Rev Med Pharmacol Sci. 2015;19(6):927-­930.
release of growth factors from PRP, PRF, and advanced-­PRF. Clin
14. Dohan Ehrenfest DM, Del Corso M, Diss A, Mouhyi J, Charrier
Oral Investig. 2016;20(9):2353-­2360.
JB. Three-­ dimensional architecture and cell composition of a
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
266 QUIRYNEN et al.

33. Anitua E, Zalduendo MM, Alkhraisat MH, Orive G. Release 52. Bielecki T, Dohan Ehrenfest DM, Everts PA, Wiczkowski A.
kinetics of platelet-­derived and plasma-­derived growth fac- The role of leukocytes from L-­PRP/L-­PRF in wound healing and
tors from autologous plasma rich in growth factors. Ann Anat. immune defense: new perspectives. Curr Pharm Biotechnol.
2013;195(5):461-­466. 2012;13(7):1153-­1162.
34. Miron RJ, Choukroun J. Platelet Rich Fibrin in Regenerative Dentistry: 53. Moojen DJ, Everts PA, Schure RM, et al. Antimicrobial activity of
Biological Background and Clinical Indications. John Wiley & Sons; platelet-­leukocyte gel against Staphylococcus aureus. J Orthop Res.
2017. 2008;26(3):404-­410.
35. Bai MY, Wang CW, Wang JY, Lin MF, Chan WP. Three-­dimensional 54. Martin P, Leibovich SJ. Inflammatory cells during wound repair: the
structure and cytokine distribution of platelet-­rich fibrin. Clinics good, the bad and the ugly. Trends Cell Biol. 2005;15(11):599-­607.
(Sao Paulo). 2017;72(2):116-­124. 55. Tsirogianni AK, Moutsopoulos NM, Moutsopoulos HM. Wound
36. Rodella LF, Favero G, Boninsegna R, et al. Growth factors, CD34 healing: immunological aspects. Injury. 2006;37(Suppl 1):S5-­S12.
positive cells, and fibrin network analysis in concentrated growth 56. Barrick B, Campbell EJ, Owen CA. Leukocyte proteinases in
factors fraction. Microsc Res Tech. 2011;74(8):772-­777. wound healing: roles in physiologic and pathologic processes.
37. Lee HM, Shen EC, Shen JT, Fu E, Chiu HC, Hsia YJ. Tensile strength, Wound Repair Regen. 1999;7(6):410-­422.
growth factor content and proliferation activities for two platelet 57. Lingen MW. Role of leukocytes and endothelial cells in the devel-
concentrates of platelet-­rich fibrin and concentrated growth fac- opment of angiogenesis in inflammation and wound healing. Arch
tor. J Dent Sci. 2020;15(2):141-­146. Pathol Lab Med. 2001;125(1):67-­71.
38. Fujioka-­Kobayashi M, Miron RJ, Hernandez M, Kandalam U, Zhang 58. Adamson R. Role of macrophages in normal wound healing: an
Y, Choukroun J. Optimized platelet-­rich fibrin with the low-­speed overview. J Wound Care. 2009;18(8):349-­351.
concept: growth factor release, biocompatibility, and cellular re- 59. Eming SA, Hammerschmidt M, Krieg T, Roers A. Interrelation of
sponse. J Periodontol. 2017;88(1):112-­121. immunity and tissue repair or regeneration. Semin Cell Dev Biol.
39. Tunalı M, Özdemir H, Küçükodacı Z, et al. A novel platelet con- 2009;20(5):517-­527.
centrate: titanium-­prepared platelet-­rich fibrin. Biomed Res Int. 60. Schultz GS, Wysocki A. Interactions between extracellular ma-
2014;2014:209548. trix and growth factors in wound healing. Wound Repair Regen.
40. Miron RJ, Chai J, Zheng S, Feng M, Sculean A, Zhang Y. A novel 2009;17(2):153-­162.
method for evaluating and quantifying cell types in platelet rich 61. Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-­C anic
fibrin and an introduction to horizontal centrifugation. J Biomed M. Growth factors and cytokines in wound healing. Wound Repair
Mater Res A. 2019;107(10):2257-­2271. Regen. 2008;16(5):585-­601.
41. Ghanaati S, Booms P, Orlowska A, et al. Advanced platelet-­rich fi- 62. Cieslik-­Bielecka A, Dohan Ehrenfest DM, Lubkowska A, Bielecki T.
brin: a new concept for cell-­based tissue engineering by means of Microbicidal properties of leukocyte-­ and platelet-­rich plasma/fi-
inflammatory cells. J Oral Implantol. 2014;40(6):679-­689. brin (L-­PRP/L-­PRF): new perspectives. J Biol Regul Homeost Agents.
42. Varela HA, Souza JCM, Nascimento RM, et al. Injectable platelet 2012;26(2 Suppl 1):43S-­52S.
rich fibrin: cell content, morphological, and protein characteriza- 63. Nasirzade J, Kargarpour Z, Hasannia S, Strauss FJ, Gruber R.
tion. Clin Oral Investig. 2019;23(3):1309-­1318. Platelet-­rich fibrin elicits an anti-­inflammatory response in macro-
43. Cortellini S, Castro AB, Temmerman A, et al. Leucocyte-­ and phages in vitro. J Periodontol. 2020;91(2):244-­252.
platelet-­rich fibrin block for bone augmentation procedure: a 64. Toffler M, Toscano N, Holtzclaw D. Osteotome-­mediated sinus
proof-­of-­concept study. J Clin Periodontol. 2018;45(5):624-­634. floor elevation using only platelet-­rich fibrin: an early report on
44. Miron RJ, Chai J, Zhang P, et al. A novel method for harvest- 110 patients. Implant Dent. 2010;19(5):447-­456.
ing concentrated platelet-­rich fibrin (C-­PRF) with a 10-­fold 65. Tan WL, Wong TL, Wong MC, Lang NP. A systematic review
increase in platelet and leukocyte yields. Clin Oral Investig. of post-­extractional alveolar hard and soft tissue dimensional
2020;24(8):2819-­2828. changes in humans. Clin Oral Implants Res. 2012;23(Suppl 5):1-­21.
45. Pinto N, Quirynen M. Letter to the editor: RE: optimized 66. Couso-­Q ueiruga E, Stuhr S, Tattan M, Chambrone L, Avila-­Ortiz
platelet-­rich fibrin with the low-­speed concept: growth factor G. Post-­extraction dimensional changes: a systematic review and
release, biocompatibility, and cellular response. J Periodontol. meta-­analysis. J Clin Periodontol. 2021;48(1):126-­144.
2019;90(2):119-­121. 67. García-­González S, Galve-­Huertas A, Aboul-­Hosn Centenero
46. Miron RJ, Pinto NR, Quirynen M, Ghanaati S. Standardization of S, Mareque-­Bueno S, Satorres-­Nieto M, Hernández-­Alfaro F.
relative centrifugal forces in studies related to platelet-­rich fibrin. Volumetric changes in alveolar ridge preservation with a compro-
J Periodontol. 2019b;90(8):817-­820. mised buccal wall: a systematic review and meta-­analysis. Med
47. Woodman R, Ferrucci L, Guralnik J. Anemia in older adults. Curr Oral Patol Oral Cir Bucal. 2020;25(5):e565-­e575.
Opin Hematol. 2005;12(2):123-­128. 68. Barootchi S, Wang HL, Ravida A, et al. Ridge preservation tech-
48. Zeng SM, Yankowitz J, Widness JA, Strauss RG. Etiology of differ- niques to avoid invasive bone reconstruction: a systematic review
ences in hematocrit between males and females: sequence-­based and meta-­analysis: Naples consensus report working group C. Int J
polymorphisms in erythropoietin and its receptor. J Gend Specif Oral Implantol (Berl). 2019;12(4):399-­416.
Med. 2001;4(1):35-­4 0. 69. Troiano G, Zhurakivska K, Lo Muzio L, Laino L, Cicciù M, Lo Russo
49. Miron RJ, Dham A, Dham U, Zhang Y, Pikos MA, Sculean A. The L. Combination of bone graft and resorbable membrane for alveo-
effect of age, gender, and time between blood draw and start of lar ridge preservation: a systematic review, meta-­analysis, and trial
centrifugation on the size outcomes of platelet-­rich fibrin (PRF) sequential analysis. J Periodontol. 2018;89(1):46-­57.
membranes. Clin Oral Investig. 2019;23(5):2179-­2185. 70. Vignoletti F, Matesanz P, Rodrigo D, Figuero E, Martin C, Sanz M.
50. Yajamanya SR, Chatterjee A, Babu CN, Karunanithi D. Fibrin net- Surgical protocols for ridge preservation after tooth extraction.
work pattern changes of platelet-­rich fibrin in young versus old A systematic review. Clin Oral Implants Res. 2012;23(Suppl
age group of individuals: a cell block cytology study. J Indian Soc 5):22-­3 8.
Periodontol. 2016;20(2):151-­156. 71. Dragonas P, Katsaros T, Avila-­Ortiz G, Chambrone L, Schiavo JH,
51. Mishra A, Randelli P, Barr C, Talamonti T, Ragone V, Cabitza Palaiologou A. Effects of leukocyte-­platelet-­rich fibrin (L-­PRF) in
P. Platelet-­rich plasma and the upper extremity. Hand Clin. different intraoral bone grafting procedures: a systematic review.
2012;28(4):481-­491. Int J Oral Maxillofac Surg. 2019;48(2):250-­262.
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 267

72. Dragonas P, Schiavo JH, Avila-­Ortiz G, Palaiologou A, Katsaros 89. Suttapreyasri S, Leepong N. Influence of platelet-­rich fibrin on al-
T. Plasma rich in growth factors (PRGF) in intraoral bone graft- veolar ridge preservation. J Craniofac Surg. 2013;24(4):1088-­1094.
ing procedures: a systematic review. J Craniomaxillofac Surg. 90. Girish Kumar N, Chaudhary R, Kumar I, Arora SS, Kumar N, Singh
2019;47(3):443-­453. H. To assess the efficacy of socket plug technique using platelet
73. Temmerman A, Vandessel J, Castro A, et al. The use of leucocyte rich fibrin with or without the use of bone substitute in alveolar
and platelet-­rich fibrin in socket management and ridge preser- ridge preservation: a prospective randomised controlled study.
vation: a split-­mouth, randomized, controlled clinical trial. J Clin Oral Maxillofac Surg. 2018;22(2):135-­142.
Periodontol. 2016;43(11):990-­999. 91. Srinivas B, Das P, Rana MM, Qureshi AQ, Vaidya KC, Ahmed
74. Alissa R, Esposito M, Horner K, Oliver R. The influence of platelet-­ Raziuddin SJ. Wound healing and bone regeneration in
rich plasma on the healing of extraction sockets: an explorative Postextraction sockets with and without platelet-­rich fibrin. Ann
randomised clinical trial. Eur J Oral Implantol. 2010;3(2):121-­134. Maxillofac Surg. 2018;8(1):28-­3 4.
75. Nisar N, Nilesh K, Parkar MI, Punde P. Extraction socket preser- 92. Areewong K, Chantaramungkorn M, Khongkhunthian P. Platelet-­
vation using a collagen plug combined with platelet-­rich plasma rich fibrin to preserve alveolar bone sockets following tooth ex-
(PRP): a comparative clinico-­radiographic study. J Dent Res Dent traction: a randomized controlled trial. Clin Implant Dent Relat Res.
Clin Dent Prospects. 2020;14(2):139-­145. 2019;21(6):1156-­1163.
76. Farina R, Bressan E, Taut A, Cucchi A, Trombelli L. Plasma rich in 93. Lundgren S, Andersson S, Sennerby L. Spontaneous bone forma-
growth factors in human extraction sockets: a radiographic and tion in the maxillary sinus after removal of a cyst: coincidence or
histomorphometric study on early bone deposition. Clin Oral consequence? Clin Implant Dent Relat Res. 2003;5(2):78-­81.
Implants Res. 2013;24(12):1360-­1368. 94. Thor A, Sennerby L, Hirsch JM, Rasmusson L. Bone formation at
77. Anitua E, Murias-­Freijo A, Alkhraisat MH, Orive G. Clinical, radio- the maxillary sinus floor following simultaneous elevation of the
graphical, and histological outcomes of plasma rich in growth fac- mucosal lining and implant installation without graft material: an
tors in extraction socket: a randomized controlled clinical trial. Clin evaluation of 20 patients treated with 44 Astra tech implants. J
Oral Investig. 2015;19(3):589-­600. Oral Maxillofac Surg. 2007;65(7 Suppl 1):64-­72.
78. Cocero N, Caratori E, Martini S, Carossa S. Can plasma rich in 95. Si MS, Shou YW, Shi YT, Yang GL, Wang HM, He FM. Long-­term
growth factors expedite healing of Postextraction tooth sockets in outcomes of osteotome sinus floor elevation without bone grafts:
patients undergoing urgent liver transplantation? J Oral Maxillofac a clinical retrospective study of 4–­9 years. Clin Oral Implants Res.
Surg. 2021;79(2):305-­312. 2016;27(11):1392-­1400.
79. Mozzati M, Gallesio G, di Romana S, Bergamasco L, Pol R. Efficacy 96. Borges FL, Dias RO, Piattelli A, et al. Simultaneous sinus mem-
of plasma-­rich growth factor in the healing of postextraction sock- brane elevation and dental implant placement without bone graft:
ets in patients affected by insulin-­dependent diabetes mellitus. J a 6-­month follow-­up study. J Periodontol. 2011;82(3):403-­412.
Oral Maxillofac Surg. 2014;72(3):456-­462. 97. Nedir R, Nurdin N, Khoury P, et al. Osteotome sinus floor elevation
80. Stumbras A, Januzis G, Gervickas A, Kubilius R, Juodzbalys G. with and without grafting material in the severely atrophic max-
Randomized and controlled clinical trial of bone healing after al- illa. A 1-­year prospective randomized controlled study. Clin Oral
veolar ridge preservation using xenografts and allografts versus Implants Res. 2013;24(11):1257-­1264.
plasma rich in growth factors. J Oral Implantol. 2020;46(5):515-­525. 98. Nedir R, Nurdin N, Abi Najm S, El Hage M, Bischof M. Short im-
81. Alzahrani AA, Murriky A, Shafik S. Influence of platelet rich fibrin plants placed with or without grafting into atrophic sinuses: the
on post-­ extraction socket healing: a clinical and radiographic 5-­year results of a prospective randomized controlled study. Clin
study. Saudi Dent J. 2017;29(4):149-­155. Oral Implants Res. 2017;28(7):877-­886.
82. Zhang Y, Ruan Z, Shen M, et al. Clinical effect of platelet-­rich fi- 99. Nedir R, Nurdin N, Khoury P, Bischof M. Short implants placed
brin on the preservation of the alveolar ridge following tooth ex- with or without grafting in atrophic sinuses: the 3-­year results of
traction. Exp Ther Med. 2018;15(3):2277-­2286. a prospective randomized controlled study. Clin Implant Dent Relat
83. Canellas JVDS, da Costa RC, Breves RC, et al. Tomographic and Res. 2016;18(1):10-­18.
histomorphometric evaluation of socket healing after tooth 100. Duan DH, Fu JH, Qi W, Du Y, Pan J, Wang HL. Graft-­free maxil-
extraction using leukocyte-­ and platelet-­rich fibrin: a random- lary sinus floor elevation: a systematic review and meta-­analysis. J
ized, single-­blind, controlled clinical trial. J Craniomaxillofac Surg. Periodontol. 2017;88(6):550-­564.
2020;48(1):24-­32. 101. Anitua E, Flores J, Alkhraisat MH. Transcrestal sinus lift using
84. Ma F, Lin Y, Sun F, Jiang X, Wei T. The impact of autologous con- platelet concentrates in association to short implant placement:
centrated growth factors on the alveolar ridge preservation after a retrospective study of augmented bone height remodeling. Clin
posterior tooth extraction: a prospective, randomized controlled Implant Dent Relat Res. 2016;18(5):993-­1002.
clinical trial. Clin Implant Dent Relat Res. 2021;23(4):579-­592. 102. Diss A, Dohan DM, Mouhyi J, Mahler P. Osteotome sinus floor el-
85. Hauser F, Gaydarov N, Badoud I, Vazquez L, Bernard JP, Ammann evation using Choukroun's platelet-­rich fibrin as grafting material:
P. Clinical and histological evaluation of postextraction platelet-­ a 1-­year prospective pilot study with microthreaded implants. Oral
rich fibrin socket filling: a prospective randomized controlled Surg Oral Med Oral Pathol Oral Radiol Endod. 2008;105(5):572-­579.
study. Implant Dent. 2013;22(3):295-­3 03. 103. Kanayama T, Horii K, Senga Y, Shibuya Y. Crestal approach to sinus
86. Marenzi G, Riccitiello F, Tia M, di Lauro A, Sammartino G. Influence floor elevation for atrophic maxilla using platelet-­rich fibrin as the
of leukocyte-­and platelet-­rich fibrin (L-­PRF) in the healing of sim- only grafting material: a 1-­year prospective study. Implant Dent.
ple Postextraction sockets: a Split-­mouth study. Biomed Res Int. 2016;25(1):32-­38.
2015;2015:369273. 104. Molemans B, Cortellini S, Jacobs R, Pinto N, Teughels W, Quirynen
87. de Almeida Barros Mourão CF, Calasans-­Maia MD, de Mello M. Simultaneous sinus floor elevation and implant placement
Machado RC, de Brito Resende RF, Alves GG. The use of platelet-­ using leukocyte-­and platelet-­rich fibrin as a sole graft material. Int
rich fibrin as a hemostatic material in oral soft tissues. Oral J Oral Maxillofac Implants. 2019;34(5):1195-­1201.
Maxillofac Surg. 2018;22(3):329-­333. 105. Testori T, Panda S, Clauser T, et al. Short implants and platelet-­rich
88. Liu YH, To M, Okudera T, et al. Advanced platelet-­rich fibrin (A-­ fibrin for transcrestal sinus floor elevation: a prospective multi-
PRF) has an impact on the initial healing of gingival regeneration center clinical study. J Biol Regul Homeost Agents. 2019;33(6 Suppl.
after tooth extraction. J Oral Biosci. 2022;64(1):141-­147. 2):121-­135.
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
268 QUIRYNEN et al.

106. Kim JM, Sohn DS, Bae MS, Moon JW, Lee JH, Park IS. Flapless two-­wall intrabony periodontal defects: a randomized clinical trial.
transcrestal sinus augmentation using hydrodynamic piezoelectric J Dent Res Dent Clin Dent Prospects. 2019;13(1):51-­56.
internal sinus elevation with autologous concentrated growth fac- 123. Arabaci T, Albayrak M. Titanium-­prepared platelet-­rich fibrin
tors alone. Implant Dent. 2014;23(2):168-­174. provides advantages on periodontal healing: a randomized split-­
107. Chen Y, Cai Z, Zheng D, et al. Inlay osteotome sinus floor eleva- mouth clinical study. J Periodontol. 2018;89(3):255-­264.
tion with concentrated growth factor application and simultane- 124. Ustaoğlu G, Uğur Aydin Z, Özelçi F. Comparison of GTR, T-­PRF and
ous short implant placement in severely atrophic maxilla. Sci Rep. open-­flap debridement in the treatment of intrabony defects with
2016;6:27348. endo-­perio lesions: a randomized controlled trial. Med Oral Patol
108. Cho YS, Hwang KG, Jun SH, Tallarico M, Kwon AM, Park CJ. Oral Cir Bucal. 2020;25(1):e117-­e123.
Radiologic comparative analysis between saline and platelet-­ 125. Kizildağ A, Taşdemir U, Arabaci T, Özmen Ö, Kizildağ CA, Iyilikci
rich fibrin filling after hydraulic transcrestal sinus lifting without B. Evaluation of new bone formation using autogenous tooth
adjunctive bone graft: a randomized controlled trial. Clin Oral bone graft combined with platelet-­rich fibrin in Calvarial defects. J
Implants Res. 2020;31(11):1087-­1093. Craniofac Surg. 2019;30(6):1662-­1666.
109. Mazor Z, Horowitz RA, Del Corso M, Prasad HS, Rohrer MD, 126. Rosamma Joseph V, Raghunath A, Sharma N. Clinical effectiveness
Dohan Ehrenfest DM. Sinus floor augmentation with simultane- of autologous platelet rich fibrin in the management of infrabony
ous implant placement using Choukroun's platelet-­rich fibrin as periodontal defects. Singapore Dent J. 2012;33(1):5-­12.
the sole grafting material: a radiologic and histologic study at 6 127. Pradeep AR, Garg V, Kanoriya D, Singhal S. Platelet-­rich fibrin with
months. J Periodontol. 2009;80(12):2056-­2064. 1.2% Rosuvastatin for treatment of Intrabony defects in chronic
110. Simonpieri A, Choukroun J, Del Corso M, Sammartino G, Dohan periodontitis: a randomized controlled clinical trial. J Periodontol.
Ehrenfest DM. Simultaneous sinus-­lift and implantation using 2016;87(12):1468-­1473.
microthreaded implants and leukocyte-­ and platelet-­rich fibrin 128. Martande SS, Kumari M, Pradeep AR, Singh SP, Suke DK,
as sole grafting material: a six-­year experience. Implant Dent. Guruprasad CN. Platelet-­rich fibrin combined with 1.2% ator-
2011;20(1):2-­12. vastatin for treatment of Intrabony defects in chronic peri-
111. Tajima N, Ohba S, Sawase T, Asahina I. Evaluation of sinus floor odontitis: a randomized controlled clinical trial. J Periodontol.
augmentation with simultaneous implant placement using platelet-­ 2016;87(9):1039-­1046.
rich fibrin as sole grafting material. Int J Oral Maxillofac Implants. 129. Chandradas ND, Ravindra S, Rangaraju VM, Jain S, Dasappa S.
2013;28(1):77-­83. Efficacy of platelet rich fibrin in the treatment of human intrabony
112. Dominiak S, Karuga-­Kuźniewska E, Popecki P, Kubasiewicz-­Ross P. defects with or without bone graft: a randomized controlled trial.
PRF versus xenograft in sinus augmentation in case of HA-­coating J Int Soc Prev Community Dent. 2016;6(Suppl 2):S153-­S169.
implant placement: a 36-­month retrospective study. Adv Clin Exp 130. Chatterjee A, Pradeep AR, Garg V, Yajamanya S, Ali MM, Priya
Med. 2021;30(6):633-­6 40. VS. Treatment of periodontal intrabony defects using autologous
113. Merli M, Moscatelli M, Merli M, Mariotti G, Pagliaro U, Nieri M. platelet-­rich fibrin and titanium platelet-­rich fibrin: a randomized,
Lateral sinus floor elevation in the severely atrophied maxilla: con- clinical, comparative study. J Investig Clin Dent. 2017;8(3):e12231.
centrated growth factors versus bone substitutes. A controlled 131. Patel GK, Gaekwad SS, Gujjari SK, Veerendra Kumar SC. Platelet-­
clinical trial. Int J Periodontics Restorative Dent. 2022;42(1):65-­72. rich fibrin in regeneration of Intrabony defects: a randomized con-
114. Cortellini P, Bowers GM. Periodontal regeneration of intrabony trolled trial. J Periodontol. 2017;88(11):1192-­1199.
defects: an evidence-­based treatment approach. Int J Periodontics 132. Yajamanya SR, Chatterjee A, Hussain A, Coutinho A, Das S,
Restorative Dent. 1995;15(2):128-­145. Subbaiah S. Bioactive glass versus autologous platelet-­rich fibrin
115. Palmer RM, Cortellini P, Group B of European Workshop on for treating periodontal intrabony defects: a comparative clinical
Periodontology. Periodontal tissue engineering and regeneration: study. J Indian Soc Periodontol. 2017;21(1):32-­36.
consensus report of the sixth European workshop on periodontol- 133. Thorat M, Pradeep AR, Pallavi B. Clinical effect of autologous
ogy. J Clin Periodontol. 2008;35(8 Suppl):83-­86. platelet-­rich fibrin in the treatment of intra-­bony defects: a con-
116. Cortellini P, Tonetti MS. Clinical concepts for regenerative therapy trolled clinical trial. J Clin Periodontol. 2011;38(10):925-­932.
in intrabony defects. Periodontol 2000. 2015;68(1):282-­3 07. 134. Sharma A, Pradeep AR. Treatment of 3-­wall intrabony defects in
117. Reynolds MA, Kao RT, Camargo PM, et al. Periodontal regenera- patients with chronic periodontitis with autologous platelet-­rich
tion –­intrabony defects: a consensus report from the AAP regen- fibrin: a randomized controlled clinical trial. J Periodontol. 2011a;8
eration workshop. J Periodontol. 2015;86(2 Suppl):S105-­S107. 2(12):1705-­1712.
118. Kao RT, Nares S, Reynolds MA. Periodontal regeneration –­ in- 135. Ajwani H, Shetty S, Gopalakrishnan D, et al. Comparative evalua-
trabony defects: a systematic review from the AAP regeneration tion of platelet-­rich fibrin biomaterial and open flap debridement
workshop. J Periodontol. 2015;86(2 Suppl):S77-­S104. in the treatment of two and three wall intrabony defects. J Int Oral
119. Zhou S, Sun C, Huang S, et al. Efficacy of adjunctive bioactive ma- Health. 2015;7(4):32-­37.
terials in the treatment of periodontal intrabony defects: a system- 136. Pradeep AR, Nagpal K, Karvekar S, Patnaik K, Naik SB, Guruprasad
atic review and meta-­analysis. Biomed Res Int. 2018;2018:8670832. CN. Platelet-­rich fibrin with 1% metformin for the treatment of in-
120. Miron RJ, Moraschini V, Fujioka-­Kobayashi M, et al. Use of trabony defects in chronic periodontitis: a randomized controlled
platelet-­rich fibrin for the treatment of periodontal intrabony de- clinical trial. J Periodontol. 2015;86(6):729-­737.
fects: a systematic review and meta-­analysis. Clin Oral Investig. 137. Bajaj P, Pradeep AR, Agarwal E, et al. Comparative evaluation of
2021;25(5):2461-­2478. autologous platelet-­rich fibrin and platelet-­rich plasma in the treat-
121. Pradeep AR, Rao NS, Agarwal E, Bajaj P, Kumari M, Naik SB. ment of mandibular degree II furcation defects: a randomized con-
Comparative evaluation of autologous platelet-­rich fibrin and trolled clinical trial. J Periodontal Res. 2013;48(5):573-­581.
platelet-­rich plasma in the treatment of 3-­wall intrabony defects 138. Kanoriya D, Pradeep AR, Singhal S, Garg V, Guruprasad CN.
in chronic periodontitis: a randomized controlled clinical trial. J Synergistic approach using platelet-­rich fibrin and 1% alendronate
Periodontol. 2012;83(12):1499-­1507. for Intrabony defect treatment in chronic periodontitis: a random-
122. Khalifehzadeh S, Haghanifar S, Jenabian N, Kazemi S, Hajiahmadi ized clinical trial. J Periodontol. 2016;87(12):1427-­1435.
M. Clinical and radiographic evaluation of applying 1% metformin 139. Pradeep AR, Bajaj P, Rao NS, Agarwal E, Naik SB. Platelet-­rich fibrin
biofilm with plasma rich in growth factor (PRGF) for treatment of combined with a porous hydroxyapatite graft for the treatment of
|

16000757, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/prd.12526 by National Institutes Of Health, Wiley Online Library on [31/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUIRYNEN et al. 269

3-­wall Intrabony defects in chronic periodontitis: a randomized tomography scan-­assisted volumetric analysis. J Indian Soc
controlled clinical trial. J Periodontol. 2017;88(12):1288-­1296. Periodontol. 2020;24(2):156-­162.
140. Thorat M, Baghele ON, Rakhewar PS. Adjunctive effect of autol- 148. Qiao J, Duan J, Zhang Y, Chu Y, Sun C. The effect of concentrated
ogous platelet-­rich fibrin in the treatment of intrabony defects growth factors in the treatment of periodontal intrabony defects.
in localized aggressive periodontitis patients: a randomized con- Future Sci OA. 2016;2(4):FS136.
trolled Split-­mouth clinical trial. Int J Periodontics Restorative Dent. 149. Xu Y, Qiu J, Sun Q, et al. One-­year results evaluating the effects of
2017;37(6):e302-­e309. concentrated growth factors on the healing of intrabony defects
141. Pham TAV. Intrabony defect treatment with platelet-­r ich treated with or without bone substitute in chronic periodontitis.
fibrin, guided tissue regeneration and open-­f lap debride- Med Sci Monit. 2019;25:4384-­4389.
ment: a randomized controlled trial. J Evid Based Dent Pract. 150. Del Fabbro M, Bortolin M, Taschieri S. Is autologous platelet
2021;21(3):101545. concentrate beneficial for post-­extraction socket healing? A sys-
142. Suchetha A, Lakshmi P, Bhat D, Mundinamane DB, Soorya KV, tematic review. Int J Oral Maxillofac Surg. 2011;40(9):891-­900.
Bharwani GA. Platelet concentration in platelet concentrates and doi:10.1016/j.ijom.2011.04.009
periodontal regeneration-­unscrambling the ambiguity. Contemp 151. Anwandter A, Bohmann S, Nally M, Castro AB, Quirynen M, Pinto
Clin Dent. 2015;6(4):510-­516. N. Dimensional changes of the post extraction alveolar ridge,
143. Hanna R, Trejo PM, Weltman RL. Treatment of intrabony de- preserved with leukocyte-­ and platelet rich fibrin: a clinical pilot
fects with bovine-­derived xenograft alone and in combination study. J Dent. 2016;52:23-­29.
with platelet-­rich plasma: a randomized clinical trial. J Periodontol. 152. Chen H, Zhou L, Wu D, Zhang J, Zheng Y, Chen Y. Osteotome sinus
2004;75(12):1668-­1677. floor elevation with concentrated growth factor and simultaneous
144. Ouyang XY, Qiao J. Effect of platelet-­rich plasma in the treatment implant placement with or without bone grafting: a retrospective
of periodontal intrabony defects in humans. Chin Med J (Engl). study. Int J Oral Maxillofac Surg. 2022;51(8):1078-­1084.
2006;119(18):1511-­1521.
145. Döri F, Kovács V, Arweiler NB, et al. Effect of platelet-­rich plasma on
the healing of intrabony defects treated with an anorganic bovine
bone mineral: a pilot study. J Periodontol. 2009;80(10):1599-­1605.
How to cite this article: Quirynen M., Siawasch SAM,
146. Sezgin Y, Uraz A, Taner IL, Çulhaoğlu R. Effects of platelet-­rich
fibrin on healing of intra-­bony defects treated with anorganic bo- Temmerman A., et al. Do autologous platelet concentrates
vine bone mineral. Braz Oral Res. 2017;31:e15. (APCs) have a role in intra-­oral bone regeneration? A critical
147. Goyal J, Sachdeva S, Salaria SK, Vakil N, Mittal A. Comparative review of clinical guidelines on decision-­making process.
assessment of periodontal regeneration in periodontal intraos-
Periodontol 2000. 2023;93:254-269. doi:10.1111/prd.12526
seous defects treated with PepGen P-­15 unaided or in blend
with platelet-­rich fibrin: a clinical and high-­ resolution computed

You might also like