Next Article in Journal
Sex-Specific Differences in Pre-Stroke Characteristics Reveal Vulnerability of Elderly Women
Next Article in Special Issue
Personalized Medicine in the Field of Inflammatory Skin Disorders
Previous Article in Journal
Dynamics of Soluble Factors and Double-Negative T Cells Associated with Response to Renal Denervation in Resistant Hypertension Patients
Previous Article in Special Issue
Dealing with Corticosteroid and High-Dose Cyclosporine Therapy in a Pyoderma Gangrenosum Patient Contracting a COVID-19 Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effectiveness of Platelet-Rich Plasma Therapy in Androgenic Alopecia—A Meta-Analysis

by
Simona Roxana Georgescu
1,2,
Andreea Amuzescu
2,*,
Cristina Iulia Mitran
3,
Madalina Irina Mitran
3,
Clara Matei
1,
Carolina Constantin
4,5,
Mircea Tampa
1,* and
Monica Neagu
4,5,6
1
Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
2
Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
3
Department of Microbiology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
4
Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
5
Colentina Clinical Hospital, 020125 Bucharest, Romania
6
Faculty of Biology, University of Bucharest, 76201 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(3), 342; https://doi.org/10.3390/jpm12030342
Submission received: 2 February 2022 / Revised: 22 February 2022 / Accepted: 22 February 2022 / Published: 24 February 2022
(This article belongs to the Special Issue Personalized Medicine in the Field of Inflammatory Skin Disorders)

Abstract

:
Platelet-rich plasma (PRP) represents a novel therapy tested and is used more and more frequently in dermatology and cosmetic surgery for a variety of conditions, including androgenic alopecia (AGA), a common condition with a complex pathogenesis involving genetic factors, hormonal status and inflammation. We performed an extensive literature search which retrieved 15 clinical trials concerning the use in AGA of PRP therapy, alone or in combination, in male, female or mixed patient groups. A quantitative statistical meta-analysis of n = 17 trial groups proved significant increases in hair density from 141.9 ± 108.2 to 177.5 ± 129.7 hairs/cm2 (mean ± SD) following PRP (p = 0.0004). To the best of our knowledge, this is the first meta-analysis that proved a statistically significant correlation between the number of PRP treatments per month and the percentage change in hair density (r = 0.5, p = 0.03), as well as a negative correlation between the mean age of treatment group and the percentage change in hair density (r = −0.56, p = 0.016). Other factors considered for analysis were the PRP preparation method, amount used per treatment, hair diameter, terminal hairs and pull test. We conclude that PRP represents a valuable and effective therapy for AGA in both males and females if patients are rigorously selected.

1. Introduction

Alopecia is a common condition that affects a large part of the population, particularly Caucasian males, the most common type being androgenic alopecia (AGA), a progressive disorder with significant psychosocial effects that can lead to depression. There are several theories trying to explain the complex multifactorial pathogeny of this condition. One of them postulates that chronic perifollicular microinflammation amplifies the expression of pro-inflammatory cytokines, leading to oxidative stress. The oxidative profile acts in combination with high levels of androgens, genetic predisposition and environmental factors such as stress, affecting the corticotropin-releasing hormone pathway and cortisol levels, hence generating the condition [1]. Gene variants that lead to increased activity of 5-α-reductase or increased sensitivity of androgen receptors are deemed to play an important role [2]. There is no single key mechanism involved in the disease, but a plethora of interconnected mechanisms that need to be treated simultaneously for a successful result. The reestablishment of healthy hair cell follicles can be influenced by certain growth factors that can stimulate the anagen phase of hair cycle, favoring cell proliferation, improvement of extracellular matrix and neoangiogenesis [3,4]. Recent studies have evidenced inflammatory infiltrates located in perifollicular/perivascular areas with histiocytes and lymphocytes. These findings, together with the implication of certain inflammatory genes (e.g., CASP7, TNF), strongly suggest a relationship between AGA and inflammatory pathways [1].
The close relationship between chronic inflammation and oxidative stress is well known [5,6,7,8]. There are many studies that show the involvement of oxidative stress in alopecia areata [9,10,11,12]. Recent research indicates an imbalance between oxidants and antioxidants in AGA [13,14]. The skin appendages, especially the hair follicle, can be exposed to elevated levels of oxidative stress. Reactive oxygen species (ROS) accumulate in the hair follicle and the antioxidant defense capacity is exceeded, resulting in premature aging of dermal papilla cells and eventually to the loss of their function [13,15]. Furthermore, it has been hypothesized that ROS may mediate the response of cells to growth factors [13]. In the light of oxidative stress involvement in AGA, Kaya Erdogan et al. consider that antioxidant therapies may be useful in the treatment of these patients [14].
Beyond classical AGA therapies such as minoxidil or finasteride, several new strategies have been proposed in recent years, each of them claiming long-term beneficial effects on hair follicle homeostasis [1,16], and offering multiple choices for personalized treatments. Among these new therapies, platelet-rich plasma (PRP) has been tested in several randomized case-control clinical trials [17,18]. The procedure involves collecting autologous blood from the patient under sterile conditions, on anticoagulant, and separating a platelet-rich plasma fraction by centrifugation. After a first spin, the second intermediate layer of plasma can be collected with a sterile syringe, dropping out the first layer of PPP (Platelet-Poor Plasma or buffy coat) and the red blood cells at the bottom of the tube. Some practitioners use a second spin to concentrate the PRP, as well as an activator of platelet α-granules release such as calcium or thrombin before injection into the scalp. There is a wide variation of protocols for preparation of PRP [3].
PRP therapy is already extensively used in dermatology and plastic surgery [19], being successfully applied for facial rejuvenation, treatment of wrinkles, scars, striae distensae, atrophic acne, vitiligo, facelift surgery, periorbital rejuvenation, dermal augmentation, as well as in other medical specialties such as orthopedic and trauma surgery, ocular surgery, stomatology, wound healing, urology, etc. [20,21]. Due to the important effects of growth factors present in PRP on skin rejuvenation, fibroblast and macrophage chemotaxis, fibroblast proliferation and extracellular matrix synthesis [22], this treatment is expected to exert beneficial effects on hair follicle degeneration, particularly in AGA. Autologous platelet rich fibrin (PRF), another related therapy, has been successfully used in both solid and liquid form administered intralesionally to accelerate healing in a case of facial pyoderma gangrenosum, due to its capacity to promote the prolonged release of multiple growth factors including TGF, EGF, FGF, KGF, CTGF, TNF-α [23], as well as in oral and maxillofacial surgery to cure medication-related osteonecrosis [24,25].
Until now, the only two effective treatments of AGA approved by the FDA are minoxidil for both men and women and finasteride for men [26,27,28]. Other treatment options are low-level laser therapy or hair transplant surgery, both with limited effectiveness, as well as synthetic PRP analogues such as growth factors biomimetic cocktails [16] or hair follicle dermal papilla stem cell cloning [29]. Considering their potential adverse reactions and other limiting factors, published results of clinical trials have to be carefully assessed via meta-analyses like the present one, which can be expanded for finding new possible therapy targets.

2. Materials and Methods

2.1. Literature Search and Selection of Studies

A search of the literature was performed by interrogating the PubMed database (on 27 July 2021) with the keyword combination “PRP AND alopecia AND clinical trial”, a sufficiently specific search string that avoids sampling biases. We selected for meta-analysis the clinical trials that included treatment groups, using as therapy PRP alone, combined with or compared to other procedures. The exclusion criteria comprised clinical trials including patients with other types of alopecia (e.g., alopecia areata, seborrheic dermatitis, iatrogenic alopecia, a.s.o.). The stepwise procedure for selecting clinical trials included in the meta-analysis observed the methods, stages, as well as selected checklist items of the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines and its subsequent updates [30,31].

2.2. Data Analysis

A number of qualitative or quantitative assessment variables (e.g., hair density, diameter, results of pull test, total number and frequency of PRP treatments, total amount of PRP per treatment, mean age of each group) were retrieved from each published clinical trial included in this meta-analysis. Data are expressed as means ± SD. The normality of the data samples was verified with the Kolmogorov-Smirnov, D’Agostino-Person and Shapiro-Wilk tests; normally distributed data were compared with Student’s t test, and data sets that did not pass normality tests with its non-parametric versions: Mann-Whitney for independent data sets and Wilcoxon signed rank test for paired data. Most variables included in analysis were normally distributed, therefore we used directly Student’s t test for independent samples (two-tailed), except for the absolute change in hair density, where we used its non-parametric variant, the non-directional Mann-Whitney test. We also performed linear correlation and regression analysis, indicating the values of Pearson’s product-momentum correlation coefficient r, its statistical significance p, and the linear regression function. Statistical significance was assessed using a critical level p = 0.05. Forrest plots were generated with the clinical trials meta-analysis software Review Manager (RevMan) Version 5.4.1 The Cochrane Collaboration, 2020 [32].

3. Results

The literature search performed according to the criteria described within the Methods section resulted in the identification of 15 clinical trials, including studies on the effects of PRP treatment of AGA suitable for inclusion in our meta-analysis. The PRISMA flowchart describing the successive steps of the selection procedure is shown in Figure 1, while Table 1 presents the main features of the selected clinical trials. Most of them (86.7%) were randomized, 46.7% were double-blind, and 26.7% were single-blind. We found marked differences regarding the study design: 2 studies applied PRP/placebo on half-scalp and 3 studies on individual areas of the scalp in same patients, resulting in better case-control matching. Two studies included separate PRP and negative control (placebo) groups, while two other studies comprised two groups with different PRP administration protocols, differing in timing or preparation quality; three studies compared effects of PRP alone with those of PRP combined with minoxidil, or different PRP/minoxidil combinations, or other combinations of synthetic growth factors similar to those retrieved in PRP [16].
Table 1. Features of the clinical trials selected and included in the meta-analysis (in alphabetical order).
Table 1. Features of the clinical trials selected and included in the meta-analysis (in alphabetical order).
StudyRando-
Mized
Double-
Blind
GroupsMean Age &/Age
Interval
SexmL PRP/
Treatment
Change in Hair Density (Hairs/cm2)
Initial–Final
Alves & Grimalt 2016 [26]YYn = 22, PRP/placebo on half-scalp
3 PRP treatments (1/month)
39 (21–62)M/F3165.7–179.9
Bayat et al. 2019 [33]NNn = 19, 3 PRP treatments
(0, 4, 8 weeks)
36.26 (28–40)M530.11–38.58
Bruce et al. 2020 [34]YNarm A–PRP then minoxidil (n = 19)
3 PRP treatments over 4 weeks
8 weeks wash-out then minoxidil 12 weeks
56 (20–79)F5134–145
arm B–minoxidil then PRP (n = 18)
minoxidil 12 weeks, 8 weeks wash-out then 3 PRP treatments over 4 weeks
56 (20–79)F5139–153
Butt et al. 2018 [35]NNn = 30, 2 PRP treatments (1/month)28.7 (19–47)M/F434.18–50.2
Butt et al. 2019 [36]YNPRP group (n = 11)
2 PRP treatments at 4 weeks
26.45M/F552.64–63.72
stromal vascular fraction-PRP group (n = 11), same treatment timing33.27M/F537.66–57.11
Gentile et al. 2015 [37]YYn = 23, PRP/placebo on half-scalp
3 PRP treatments (1/month)
34.74 (19–63)M9161.2–207.1
Hausauer & Jones 2018 [38]Ysingle-blindPRP group 1 (n = 20)
3 PRP treatments (1/month) + 1 booster treatment after 3 months
40.1 (18–60)M/F5160.4–207.1
PRP group 2 (n = 19)
2 PRP treatments at 3 months
46.85 (18–60)M/F5177.6–190.6
Kapoor et al. 2020 [16]Ysingle-blindgroup B (PRP) (n = 25)
8 PRP treatments at 3 weeks
25–50M1.5167.2–176.1
Mapar et al. 2016 [39]Ysingle-blindn = 17, PRP/placebo on squares
2 PRP treatments (1/month)
37.2 (25–45)M1.587.29–85.06
Pakhomova & Smirnova 2020 [40]Ysingle-blindgroup I (PRP) (n = 25)
4 PRP treatments (1/month)
29.7 (18–53)M4381.5–426.1
group II (PRP + minoxidil) (n = 22)
4 PRP treatments (1/month)
M4408.4–539.6
Puig et al. 2016 [41]YYPRP group (n = 15) 1 treatment>18F10no signifcant differences
placebo group (n = 11) 1 treatment
Rodrigues et al. 2020 [42]YYPRP group (n = 15) 4 treatments
at 15 days intervals
32 (18–50)M2140–180
placebo group (n = 11) 4 treatments
at 15 days intervals
Shapiro et al. 2020 [43]YYn = 35, PRP/placebo on squares
3 PRP treatments (1/month)
36.5 (18–58)M/F5151–170.96
Singh et al. 2020 [44]YYPRP group (n = 20)
3 treatments (1/month)
26.5M3–3.593.73–143.2
PRP+minoxidil group (n = 20)
3 treatments (1/month)
25.6M3–3.590.05–150.45
Tawfick & Osman 2017 [45]YYn = 30, PRP/placebo on areas
4 PRP treatments (1/week)
29.3 (20–45)F0.973.66–150.94
The inclusion and exclusion criteria used within the clinical trials selected for analysis are listed in Table S1. Most studies used the Norwood-Hamilton scale for assessment of male patients with AGA and the Ludwig scale for female patients. Exclusion criteria were also largely similar among studies, including the use of other topical or systemic hair growth medications, other causes of alopecia, other general diseases (endocrine, inflammatory, autoimmune, neoplasia, platelet and other bleeding disorders), and risk factors such as alcohol use or smoking.
There were also important differences in the PRP preparation methods, as shown in Table S2. The amount of peripheral blood used for one PRP treatment ranged between 9 and 60 mL, centrifugation was done as a single-step or two-steps, and calcium chloride was the most frequently used agent for α-granule release by platelet fractions. The most widely used monitoring methods were digital photography and phototrichogram analysis using the standard TrichoScan method.
Figure 2 presents the absolute and relative changes in average hair density for n = 17 study groups included in this meta-analysis. The mean hair density within these groups varied from an initial value of 141.9 ± 108.2 hairs/cm2 (mean ± SD) to 177.5 ± 129.7 hairs/cm2 at the last evaluation, a statistically significant increase as proved by a two-tailed Wilcoxon signed rank test (p = 0.0004). The same study groups were analyzed for effects on hair density using a Forrest plot shown in Figure 3; the main difference between baseline and post-therapy levels was 36.84 hairs/cm2 (95% confidence interval 22.63–51.06).
We have also assessed the correlations between the total number of PRP treatments and the absolute or relative change in hair density: both were statistically non-significant (r = 0.176, p = 0.47 for absolute and r = −0.115, p = 0.64 for relative changes, Figure 4). However, the percentage change in hair density was significantly correlated with the frequency of PRP treatments (r = 0.50, p = 0.03), but the absolute change in hair density was not (r = 0.135, p = 0.58) (Figure 5). We also found no significant correlation between the amount of PRP administered per treatment and the absolute (r = −0.069, p = 0.78) or relative change in hair density (r = −0.215, p = 0.38), while the mean ages of the treatment groups were significantly correlated with the absolute (r = −0.44, p = 0.07) and relative (r = −0.56, p = 0.016) change in hair density (Figure 6).
We verified if the sex composition of study groups exerted an effect on initial values, absolute and relative changes in hair density, using data exposed in Supplementary Table S3. Thus, the initial hair density, the absolute and the relative (%) change in hair density did not show significant differences between study groups formed exclusively from males vs. groups of females or with mixed gender composition.
Although relatively few studies of those included in the meta-analysis provided relevant data, we assessed the effects of PRP therapy on the mean hair diameter and the results of the pull test. Therefore, for n = 8 study groups the mean hair diameter increased by 15.67 µm after therapy (95% confidence interval 9.77–21.57) (Figure 7), while the results of the pull test decreased on average by 5.32 (95% confidence interval 2.84–7.80) for n = 5 study groups (Figure 8).
8 of the 15 studies included in the analysis (53.3%) reported use of patient self-assessment questionnaires, and 5 of them (33.3%) used physician assessments. Patient self-assessment comprised most often the degree of satisfaction following PRP therapy, usually at several time points, and divided on four levels (e.g., highly satisfied, satisfied, dissatisfied, highly dissatisfied). One study reported the percentage improvement after therapy on a 5-level scale [44], and another one on a 1-10 scale [45]. One self-assessment questionnaire included supplementary items such as evaluation of results (Yes, No, Unsure/maybe), recommendation to other patients (Yes, No, Maybe), and motivation to continue (Yes, No, Maybe) [38]. Physician assessments were based on analysis of global photographs of the scalp or four-level satisfaction questionnaires.

4. Discussion

AGA is a challenging disorder for both healthcare providers and patients. Current therapeutic options for AGA may generate biochemical abnormalities and clinical adverse effects. Therefore, new personalized therapies are necessary according to the characteristics of the patients, with fewer side effects and satisfactory results [46,47,48,49]. An increasing body of evidence emphasizes an important dermal and follicular response to certain growth factors such as platelet-derived growth factor, transforming growth factor beta, etc. [17,50,51]. In line with this, recent research has focused on evaluating the effectiveness of PRP in AGA.
The present meta-analysis succeeded in retrieving a sufficiently large number of clinical trials including PRP treatments for AGA in male, female, or mixed gender groups to allow pertinent quantitative analysis of results. By comparison, a relatively recent similar meta-analysis, Giordano, Romeo and Lankinen (2017) [3], found only 6 out of 16 clinical trials of PRP in AGA accurately reporting changes in hair density following treatment. A similar attempt of meta-analysis, Kramer & Keaney (2018) [52], focusing on technical details of PRP preparation, concluded that only 21% of the 19 studies selected for analysis provided complete quantitative assessment of initial and final cellular composition of PRP preparations. Data reporting seems to have improved in recent clinical trials [17,53], so we were able to retrieve accurate quantitative estimates of changes in hair density obtained with reliable standardized methods in 14 out of 15 studies (Table 1), as well as sufficient details concerning PRP preparation or delivery protocols.
The amount of PRP administered per treatment varied between 0.9 and 10 mL (Table 1 and Table S2), and it was distributed at multiple subcutaneous injection points, each receiving 100–150 µL. Interestingly, some studies reported significant increases in hair density even for the placebo treatment (e.g., from 151.04 ± 41.99 to 166.72 ± 37.13 hairs/cm2, mean ± SD [43]). Although the authors explained such effects as resulting from a possible diffusion of the growth factors from the PRP-injected to the placebo-injected area, it is also plausible that microstimulation by needle puncture itself exerts beneficial effects on hair growth; this principle is currently applied in microneedling, another novel therapy used for AGA [54,55].
Among the 15 clinical trials selected for this meta-analysis, only two included histopathological and/or immunohistochemical assessment of PRP treatment effects. Gentile et al. (2015) showed significant increases in epidermal thickness and number of hair follicles after PRP injection on hematoxylin-eosin-stained scalp biopsies; they also assessed Ki67 proliferation index of hair follicle bulge cells and basal cells of epidermis [37]. Pakhomova and Smirnova (2020) proved by immunohistochemistry post-PRP treatment increases in areas of CD34 and β-catenin reactivity, as well as almost doubling of Ki67 proliferative index [40]. Rodrigues et al. (2018) used the multiplex method (Luminex®) to assess VEGF, PDGF and EGF concentrations in PRP preparations and correlate with their platelet counts, but also showed a lack of correlation with clinical effects on hair density [42].
Although there is no consensus concerning the molecular mechanisms and signaling pathways involved in PRP therapy, histological examination of biopsy samples from treated vs. control areas and immunohistochemical markers suggest multiple changes such as increased epidermal cell proliferation, particularly at hair follicles dermal papillae, augmentation of blood capillary network, and strengthening of the basal dermal extracellular matrix [43]. Platelet-secreted factors present in PRP preparations seem to enhance hair growth by stimulating multiple signaling pathways involved in the hair follicle cell cycle, such as the protein kinase B (Akt) pathway [18]. Upon Akt phosphorylation, subsequent Wnt/β-catenin transcription upregulation induces stem cell differentiation into hair follicle cells [56]. In vitro studies also showed that PRP causes papillary cells of the skin to grow by activating the extracellular signal-regulated kinases (ERK) [49]. Another important effect of PRP is reduction of hair follicle microinflammation associated with hair loss during AGA [1,57]. PRP growth and transcription factors signal the follicle to enter the anagen versus catagen phase, playing a role in regeneration and renewal; these effects partly overlap those of other effective AGA therapies such as minoxidil [58] or finasteride [59], justifying combined therapy approaches [44].
Due to these remarkable properties and activation of multiple signaling pathways by platelet-secreted factors, PRP raised a lot of interest as a potentially beneficial therapy in various medical fields, such as dermatology and cosmetology, ophthalmology, neurology, sports medicine, stomatology, gynaecology and reproductive medicine. A similar remedy is PRF, which is even easier to prepare in either leukocyte-rich or leukocyte-poor varieties, and includes cytokines and growth factors that are released for up to several weeks, resulting in enhanced wound healing [60,61].
Our study presents a number of limitations, such as those related to the extent of literature search, as well as the lack of accurate reporting of quantitative assessment variables and details concerning parameters of study groups, particularly in earlier published clinical trials. There is also a high degree of variability concerning PRP preparation and administration protocols, selection of experimental groups and general design of the trials, objective assessment methods. Possibly standardization requirements and larger patient samples will facilitate comparisons between objective outcomes of future studies.

5. Conclusions

PRP represents a relatively new therapeutic approach widely used in different areas of dermatology and plastic surgery [19,21,22], although its underlying mechanisms of action are not completely understood. The present meta-analysis proves the valuable role of PRP therapy in AGA for patients of both sexes, but also points to the need for personalized therapeutic indications and approaches. Although the total number of PRP treatments and the amount of PRP injected per treatment do not appear to influence the outcome, an increased frequency of application (number of treatments per month) results in larger increases in hair density. Other important factors are the age of patients and implicitly duration since alopecia onset. For best results it is advisable to apply a complex combined therapy protocol as early as possible.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/jpm12030342/s1, Table S1: Inclusion and exclusion criteria for the clinical trials included in the meta-analysis, Table S2: Methods of PRP preparation, injection and of patient monitoring for the clinical trials included in the meta-analysis, Table S3: Influence of sex composition of study groups on hair density (initial and after PRP treatment).

Author Contributions

All authors have equally contributed to this paper. Conceptualization, S.R.G., A.A. and M.T.; methodology, S.R.G., A.A. and C.M.; validation, C.I.M., M.I.M., C.C. and M.N.; formal analysis, A.A., M.T. and C.M.; investigation, A.A., C.I.M., and M.I.M.; resources, C.M.; data curation, C.C. and C.M.; writing—original draft preparation, A.A. and M.T.; writing—review and editing, M.T., C.I.M., M.I.M., C.C. and M.N.; visualization, S.R.G., C.M. and C.C.; supervision, S.R.G., M.N. and M.T.; project administration, S.R.G. and M.N. All authors have read and agreed to the published version of the manuscript.

Funding

The article processing charges are funded by Carol Davila University of Medicine and Pharmacy within the project Publish not perish.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AGAandrogenic alopecia
CASP7caspase 7
CDcluster of differentiation
CTGFconnective tissue growth factor
EGFepidermal growth factor
ERKextracellular signal-regulated kinases
FDAUnited States Food and Drug Administration
FGFfibroblast growth factor
KGFkeratinocyte growth factor
PDGFplatelet-derived growth factor
PPPplatelet-poor plasma
PRFplatelet-rich fibrin
PRISMAPreferred Reporting Items for Systematic Reviews and Meta-Analyses
PRPplatelet-rich plasma
ROSreactive oxygen species
TGFtransforming growth factor
TNF-αtumor necrosis factor α
VEGFvascular endothelial growth factor

References

  1. Sadick, N.S.; Callender, V.D.; Kircik, L.H.; Kogan, S. New Insight into the Pathophysiology of Hair Loss Trigger a Paradigm Shift in the Treatment Approach. J. Drugs Dermatol. 2017, 16, s135–s140. [Google Scholar]
  2. Gupta, A.K.; Mays, R.R.; Dotzert, M.S.; Versteeg, S.G.; Shear, N.H.; Piguet, V. Efficacy of non-surgical treatments for androgenetic alopecia: A systematic review and network meta-analysis. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 2112–2125. [Google Scholar] [CrossRef] [PubMed]
  3. Giordano, S.; Romeo, M.; Lankinen, P. Platelet-rich plasma for androgenetic alopecia: Does it work? Evidence from meta analysis. J. Cosmet. Dermatol. 2017, 16, 374–381. [Google Scholar] [CrossRef]
  4. Gupta, A.K.; Carviel, J.L. Meta-analysis of efficacy of platelet-rich plasma therapy for androgenetic alopecia. J. Dermatolog. Treat. 2017, 28, 55–58. [Google Scholar] [CrossRef] [PubMed]
  5. Dandekar, A.; Mendez, R.; Zhang, K. Cross talk between ER stress, oxidative stress, and inflammation in health and disease. Methods Mol. Biol. 2015, 1292, 205–214. [Google Scholar]
  6. Georgescu, S.R.; Tampa, M.; Mitran, C.I.; Mitran, M.I.; Caruntu, C.; Caruntu, A.; Lupu, M.; Matei, C.; Constantin, C.; Neagu, M. Tumour Microenvironment in Skin Carcinogenesis. Adv. Exp. Med. Biol. 2020, 1226, 123–142. [Google Scholar] [PubMed]
  7. Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid. Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef] [Green Version]
  8. Spychalowicz, A.; Wilk, G.; Śliwa, T.; Ludew, D.; Guzik, T.J. Novel therapeutic approaches in limiting oxidative stress and inflammation. Curr. Pharm. Biotechnol. 2012, 13, 2456–2466. [Google Scholar] [CrossRef]
  9. Acharya, P.; Mathur, M.C. Oxidative stress in alopecia areata: A systematic review and meta-analysis. Int. J. Dermatol. 2020, 59, 434–440. [Google Scholar] [CrossRef]
  10. Dizen-Namdar, N.; Emel Kocak, F.; Kidir, M.; Sarici, G.; Tak, H.; Altuntas, I. Evaluation of Serum Paraoxonase, Arylesterase, Prolidase Activities and Oxidative Stress in Patients with Alopecia Areata. Skin Pharmacol. Physiol. 2019, 32, 59–64. [Google Scholar] [CrossRef]
  11. Georgescu, S.R.; Ene, C.D.; Tampa, M.; Matei, C.; Benea, V.; Nicolae, I. Oxidative stress-related markers and alopecia areata through latex turbidimetric immunoassay method. Mater. Plast. 2016, 53, 522–526. [Google Scholar]
  12. Mustafa, A.I.; Khashaba, R.A.; Fawzy, E.; Baghdady, S.M.A.; Rezk, S.M. Cross talk between oxidative stress and inflammation in alopecia areata. J. Cosmet. Dermatol. 2021, 20, 2305–2310. [Google Scholar] [CrossRef] [PubMed]
  13. Cwynar, A.; Olszewska-Słonina, D.M.; Czajkowski, R. The impact of oxidative stress in androgenic alopecia in women. Postepy Dermatol. Alergol. 2020, 37, 119–120. [Google Scholar] [CrossRef] [PubMed]
  14. Kaya Erdogan, H.; Bulur, I.; Kocaturk, E.; Yildiz, B.; Saracoglu, Z.N.; Alatas, O. The role of oxidative stress in early-onset androgenetic alopecia. J. Cosmet. Dermatol. 2017, 16, 527–530. [Google Scholar] [CrossRef] [PubMed]
  15. Jadkauskaite, L.; Coulombe, P.A.; Schäfer, M.; Dinkova-Kostova, A.T.; Paus, R.; Haslam, I.S. Oxidative stress management in the hair follicle: Could targeting NRF2 counter age-related hair disorders and beyond? Bioessays 2017, 39, 1700029. [Google Scholar] [CrossRef]
  16. Kapoor, R.; Shome, D.; Vadera, S.; Ram, M.S. QR 678 & QR678 Neo Vs PRP-A randomised, comparative, prospective study. J. Cosmet. Dermatol. 2020, 19, 2877–2885. [Google Scholar] [PubMed]
  17. Gentile, P.; Garcovich, S. Systematic Review of Platelet-Rich Plasma Use in Androgenetic Alopecia Compared with Minoxidil®, Finasteride®, and Adult Stem Cell-Based Therapy. Int. J. Mol. Sci. 2020, 21, 2702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Gupta, A.K.; Versteeg, S.G.; Rapaport, J.; Hausauer, A.K.; Shear, N.H.; Piguet, V. The Efficacy of Platelet-Rich Plasma in the Field of Hair Restoration and Facial Aesthetics-A Systematic Review and Meta-analysis. J. Cutan. Med. Surg. 2019, 23, 185–203. [Google Scholar] [CrossRef]
  19. Leo, M.S.; Kumar, A.S.; Kirit, R.; Konathan, R.; Sivamani, R.K. Systematic review of the use of platelet-rich plasma in aesthetic dermatology. J. Cosmet. Dermatol. 2015, 14, 315–323. [Google Scholar] [CrossRef]
  20. Bucur, M.; Constantin, C.; Neagu, M.; Zurac, S.; Dinca, O.; Vladan, C.; Cioplea, M.; Popp, C.; Nichita, L.; Ionescu, E. Alveolar blood clots and platelet-rich fibrin induce in vitro fibroblast proliferation and migration. Exp. Ther. Med. 2019, 17, 982–989. [Google Scholar] [CrossRef]
  21. Elghblawi, E. Platelet-rich plasma, the ultimate secret for youthful skin elixir and hair growth triggering. J. Cosmet. Dermatol. 2017, 17, 423–430. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, M.; Park, G.; Zhou, B.; Luo, D. Applications and efficacy of platelet-rich plasma in dermatology: A clinical review. J. Cosmet. Dermatol. 2018, 17, 660–665. [Google Scholar] [CrossRef] [PubMed]
  23. Fortunato, L.; Barone, S.; Bennardo, F.; Giudice, A. Management of Facial Pyoderma Gangrenosum Using Platelet-Rich Fibrin: A Technical Report. J. Oral Maxillofac. Surg. 2018, 76, 1460–1463. [Google Scholar] [CrossRef] [PubMed]
  24. Maluf, G.; Caldas, R.J.; Silva Santos, P.S. Use of Leukocyte- and Platelet-Rich Fibrin in the Treatment of Medication-Related Osteonecrosis of the Jaws. J. Oral Maxillofac. Surg. 2018, 76, 88–96. [Google Scholar] [CrossRef]
  25. Soydan, S.S.; Uckan, S. Management of bisphosphonate-related osteonecrosis of the jaw with a platelet-rich fibrin membrane: Technical report. J. Oral Maxillofac. Surg. 2014, 72, 322–326. [Google Scholar] [CrossRef]
  26. Alves, R.; Grimalt, R. Randomized Placebo-Controlled, Double-Blind, Half-Head Study to Assess the Efficacy of Platelet-Rich Plasma on the Treatment of Androgenetic Alopecia. Dermatol. Surg. 2016, 42, 491–497. [Google Scholar] [CrossRef]
  27. Varothai, S.; Bergfeld, W.F. Androgenetic alopecia: An evidence-based treatment update. Am. J. Clin. Dermatol. 2014, 15, 217–230. [Google Scholar] [CrossRef]
  28. Motofei, I.G.; Rowland, D.L.; Baconi, D.L.; Tampa, M.; Sârbu, M.I.; Păunică, S.; Constantin, V.D.; Bălălău, C.; Păunică, I.; Georgescu, S.R. Androgenetic alopecia; drug safety and therapeutic strategies. Expert Opin. Drug Saf. 2018, 17, 407–412. [Google Scholar] [CrossRef]
  29. Talavera-Adame, D.; Newman, D.; Newman, N. Conventional and novel stem cell based therapies for androgenic alopecia. Stem Cells Cloning 2017, 10, 11–19. [Google Scholar] [CrossRef] [Green Version]
  30. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. J. Clin. Epidemiol. 2009, 62, 1006–1012. [Google Scholar] [CrossRef]
  31. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. J. Clin. Epidemiol. 2021, 134, 178–189. [Google Scholar] [CrossRef] [PubMed]
  32. The Cochrane Collaboration. Review Manager (RevMan); Vesion 5.3; The Nordic Cochrane Centre: Copenhagen, Denmark, 2014. [Google Scholar]
  33. Bayat, M.; Yazdanpanah, M.J.; Hamidi Alamdari, D.; Banihashemi, M.; Salehi, M. The effect of platelet-rich plasma injection in the treatment of androgenetic alopecia. J. Cosmet. Dermatol. 2019, 18, 1624–1628. [Google Scholar] [CrossRef] [PubMed]
  34. Bruce, A.J.; Pincelli, T.P.; Heckman, M.G.; Desmond, C.M.; Arthurs, J.R.; Diehl, N.N.; Douglass, E.J.; Bruce, C.J.; Shapiro, S.A. A Randomized, Controlled Pilot Trial Comparing Platelet-Rich Plasma to Topical Minoxidil Foam for Treatment of Androgenic Alopecia in Women. Dermatol. Surg. 2020, 46, 826–832. [Google Scholar] [CrossRef] [PubMed]
  35. Butt, G.; Hussain, I.; Ahmed, F.J.; Choudhery, M.S. Efficacy of platelet-rich plasma in androgenetic alopecia patients. J. Cosmet. Dermatol. 2018, 18, 996–1001. [Google Scholar] [CrossRef] [PubMed]
  36. Butt, G.; Hussain, I.; Ahmad, F.J.; Choudhery, M.S. Stromal vascular fraction-enriched platelet-rich plasma therapy reverses the effects of androgenetic alopecia. J. Cosmet. Dermatol. 2019, 19, 1078–1085. [Google Scholar] [CrossRef]
  37. Gentile, P.; Garcovich, S.; Bielli, A.; Scioli, M.G.; Orlandi, A.; Cervelli, V. The Effect of Platelet-Rich Plasma in Hair Regrowth: A Randomized Placebo-Controlled Trial. Stem Cells Transl. Med. 2015, 4, 1317–1323. [Google Scholar] [CrossRef]
  38. Hausauer, A.K.; Jones, D.H. Evaluating the Efficacy of Different Platelet-Rich Plasma Regimens for Management of Androgenetic Alopecia: A Single-Center, Blinded, Randomized Clinical Trial. Dermatol. Surg. 2018, 44, 1191–1200. [Google Scholar] [CrossRef]
  39. Mapar, M.A.; Shahriari, S.; Haghighizadeh, M.H. Efficacy of platelet-rich plasma in the treatment of androgenetic (male-patterned) alopecia: A pilot randomized controlled trial. J. Cosmet. Laser Ther. 2016, 18, 452–455. [Google Scholar] [CrossRef]
  40. Pakhomova, E.E.; Smirnova, I.O. Comparative Evaluation of the Clinical Efficacy of PRP-Therapy, Minoxidil, and Their Combination with Immunohistochemical Study of the Dynamics of Cell Proliferation in the Treatment of Men with Androgenetic Alopecia. Int. J. Mol. Sci. 2020, 21, 6516. [Google Scholar] [CrossRef]
  41. Puig, C.J.; Reese, R.; Peters, M. Double-Blind, Placebo-Controlled Pilot Study on the Use of Platelet-Rich Plasma in Women With Female Androgenetic Alopecia. Dermatol. Surg. 2016, 42, 1243–1247. [Google Scholar] [CrossRef]
  42. Rodrigues, B.L.; Montalvão, S.A.L.; Cancela, R.B.B.; Silva, F.A.R.; Urban, A.; Huber, S.C.; Júnior, J.; Lana, J.; Annichinno-Bizzacchi, J.M. Treatment of male pattern alopecia with platelet-rich plasma: A double-blind controlled study with analysis of platelet number and growth factor levels. J. Am. Acad. Dermatol. 2018, 80, 694–700. [Google Scholar] [CrossRef]
  43. Shapiro, J.; Ho, A.; Sukhdeo, K.; Yin, L.; Lo Sicco, K. Evaluation of platelet-rich plasma as a treatment for androgenetic alopecia: A randomized controlled trial. J. Am. Acad. Dermatol. 2020, 83, 1298–1303. [Google Scholar] [CrossRef]
  44. Singh, S.K.; Kumar, V.; Rai, T. Comparison of efficacy of platelet-rich plasma therapy with or without topical 5% minoxidil in male-type baldness: A randomized, double-blind placebo control trial. Indian J. Dermatol. Venereol. Leprol. 2020, 86, 150–157. [Google Scholar] [CrossRef]
  45. Tawfik, A.A.; Osman, M.A.R. The effect of autologous activated platelet-rich plasma injection on female pattern hair loss: A randomized placebo-controlled study. J. Cosmet. Dermatol. 2017, 17, 47–53. [Google Scholar] [CrossRef]
  46. Motofei, I.G.; Rowland, D.L.; Georgescu, S.R.; Tampa, M.; Baconi, D.; Stefanescu, E.; Baleanu, B.C.; Balalau, C.; Constantin, V.; Paunica, S. Finasteride adverse effects in subjects with androgenic alopecia: A possible therapeutic approach according to the lateralization process of the brain. J. Dermatolog. Treat. 2016, 27, 495–497. [Google Scholar] [CrossRef]
  47. Motofei, I.G.; Rowland, D.L.; Georgescu, S.R.; Tampa, M.; Paunica, S.; Constantin, V.D.; Balalau, C.; Manea, M.; Baleanu, B.C.; Sinescu, I. Post-Finasteride Adverse Effects in Male Androgenic Alopecia: A Case Report of Vitiligo. Skin Pharmacol. Physiol. 2017, 30, 42–45. [Google Scholar] [CrossRef]
  48. Pereira, A.F.J.R.; Coelho, T.O.A. Post-finasteride syndrome. An. Bras. Dermatol. 2020, 95, 271–277. [Google Scholar] [CrossRef]
  49. Vasserot, A.P.; Geyfman, M.; Poloso, N.J. Androgenetic alopecia: Combing the hair follicle signaling pathways for new therapeutic targets and more effective treatment options. Expert Opin. Ther. Targets 2019, 23, 755–771. [Google Scholar] [CrossRef]
  50. Emer, J. Platelet-Rich Plasma (PRP): Current Applications in Dermatology. Skin Ther. Lett. 2019, 24, 1–6. [Google Scholar]
  51. Gupta, A.K.; Cole, J.; Deutsch, D.P.; Everts, P.A.; Niedbalski, R.P.; Panchaprateep, R.; Rinaldi, F.; Rose, P.T.; Sinclair, R.; Vogel, J.E.; et al. Platelet-Rich Plasma as a Treatment for Androgenetic Alopecia. Dermatol. Surg. 2019, 45, 1262–1273. [Google Scholar] [CrossRef] [Green Version]
  52. Kramer, M.E.; Keaney, T.C. Systematic review of platelet-rich plasma (PRP) preparation and composition for the treatment of androgenetic alopecia. J. Cosmet. Dermatol. 2018, 17, 666–671. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, S.; Qi, F.; Gong, Y.; Zhang, C.; Zhao, S.; Yang, X.; He, Y. Platelet-Rich Plasma in Female Androgenic Alopecia: A Comprehensive Systematic Review and Meta-Analysis. Front. Pharmacol. 2021, 12, 642980. [Google Scholar] [CrossRef]
  54. Litchman, G.; Nair, P.A.; Badri, T.; Kelly, S.E. Microneedling; StatPearls Publishing LLC: Treasure Island, FL, USA, 2020. [Google Scholar]
  55. Ocampo-Garza, S.S.; Fabbrocini, G.; Ocampo-Candiani, J.; Cinelli, E.; Villani, A. Micro needling: A novel therapeutic approach for androgenetic alopecia, A Review of Literature. Dermatol. Ther. 2020, 33, e14267. [Google Scholar] [CrossRef] [PubMed]
  56. Gupta, A.K.; Carviel, J. A Mechanistic Model of Platelet-Rich Plasma Treatment for Androgenetic Alopecia. Dermatol. Surg. 2016, 42, 1335–1339. [Google Scholar] [CrossRef] [PubMed]
  57. Mahé, Y.F.; Michelet, J.F.; Billoni, N.; Jarrousse, F.; Buan, B.; Commo, S.; Saint-Léger, D.; Bernard, B.A. Androgenetic alopecia and microinflammation. Int. J. Dermatol. 2000, 39, 576–584. [Google Scholar] [CrossRef]
  58. Han, J.H.; Kwon, O.S.; Chung, J.H.; Cho, K.H.; Eun, H.C.; Kim, K.H. Effect of minoxidil on proliferation and apoptosis in dermal papilla cells of human hair follicle. J. Dermatol. Sci. 2004, 34, 91–98. [Google Scholar] [CrossRef]
  59. Tosti, A.; Piraccini, B.M. Finasteride and the hair cycle. J. Am. Acad. Dermatol. 2000, 42, 848–849. [Google Scholar] [CrossRef]
  60. Cecerska-Heryć, E.; Goszka, M.; Serwin, N.; Roszak, M.; Grygorcewicz, B.; Heryć, R.; Dołęgowska, B. Applications of the regenerative capacity of platelets in modern medicine. Cytokine Growth Factor Rev. 2021, 6101, 00090–00093. [Google Scholar] [CrossRef]
  61. Inbarajan, A.; Veeravalli, P.T.; Seenivasan, M.K.; Natarajan, S.; Sathiamurthy, A.; Ahmed, S.R.; Vaidyanathan, A.K. Platelet-Rich Plasma and Platelet-Rich Fibrin as a Regenerative Tool. J. Pharm. Bioallied Sci. 2021, 13, S1266–S1267. [Google Scholar]
Figure 1. PRISMA flowchart showing the algorithm used for selection of the studies included in the current meta-analysis.
Figure 1. PRISMA flowchart showing the algorithm used for selection of the studies included in the current meta-analysis.
Jpm 12 00342 g001
Figure 2. Effects of PRP treatments on absolute and percentage change in hair density within the clinical trials selected for meta-analysis. (a) absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) percentage changes in hair density relative to initial values. The legend is the same for both graphs. Data for experimental groups where only PRP treatment was applied are marked with squares, those where minoxidil treatment was added are marked with hollow star symbols.
Figure 2. Effects of PRP treatments on absolute and percentage change in hair density within the clinical trials selected for meta-analysis. (a) absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) percentage changes in hair density relative to initial values. The legend is the same for both graphs. Data for experimental groups where only PRP treatment was applied are marked with squares, those where minoxidil treatment was added are marked with hollow star symbols.
Jpm 12 00342 g002
Figure 3. Forrest plot showing the effects of PRP treatment on hair density in selected study groups.
Figure 3. Forrest plot showing the effects of PRP treatment on hair density in selected study groups.
Jpm 12 00342 g003
Figure 4. Absence of significant correlations between the number of PRP treatments and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, values of correlation coefficients r and their probabilities p are marked on each graph.
Figure 4. Absence of significant correlations between the number of PRP treatments and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, values of correlation coefficients r and their probabilities p are marked on each graph.
Jpm 12 00342 g004
Figure 5. Correlations between the frequency of PRP treatments and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) lack of significant correlation of absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) significant correlation with percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, values of correlation coefficients r and their probabilities p are marked on each graph.
Figure 5. Correlations between the frequency of PRP treatments and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) lack of significant correlation of absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) significant correlation with percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, values of correlation coefficients r and their probabilities p are marked on each graph.
Jpm 12 00342 g005
Figure 6. Correlations between the mean age of experimental groups and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) almost significant correlation with the absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) significant correlation with percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, the values of the correlation coefficients r, and their probabilities p are marked on each graph.
Figure 6. Correlations between the mean age of experimental groups and absolute or percentage change in hair density within the clinical trials selected for meta-analysis. (a) almost significant correlation with the absolute changes in hair density (hairs/cm2) at the end of evaluation period. (b) significant correlation with percentage changes in hair density relative to initial values (same legend for both graphs). The regression lines are plotted in red, and the corresponding equations, the values of the correlation coefficients r, and their probabilities p are marked on each graph.
Jpm 12 00342 g006
Figure 7. Forrest plot showing the effects of PRP treatment on mean hair diameter in selected study groups.
Figure 7. Forrest plot showing the effects of PRP treatment on mean hair diameter in selected study groups.
Jpm 12 00342 g007
Figure 8. Forrest plot showing effects of PRP treatment on results of pull test in selected study groups.
Figure 8. Forrest plot showing effects of PRP treatment on results of pull test in selected study groups.
Jpm 12 00342 g008
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Georgescu, S.R.; Amuzescu, A.; Mitran, C.I.; Mitran, M.I.; Matei, C.; Constantin, C.; Tampa, M.; Neagu, M. Effectiveness of Platelet-Rich Plasma Therapy in Androgenic Alopecia—A Meta-Analysis. J. Pers. Med. 2022, 12, 342. https://doi.org/10.3390/jpm12030342

AMA Style

Georgescu SR, Amuzescu A, Mitran CI, Mitran MI, Matei C, Constantin C, Tampa M, Neagu M. Effectiveness of Platelet-Rich Plasma Therapy in Androgenic Alopecia—A Meta-Analysis. Journal of Personalized Medicine. 2022; 12(3):342. https://doi.org/10.3390/jpm12030342

Chicago/Turabian Style

Georgescu, Simona Roxana, Andreea Amuzescu, Cristina Iulia Mitran, Madalina Irina Mitran, Clara Matei, Carolina Constantin, Mircea Tampa, and Monica Neagu. 2022. "Effectiveness of Platelet-Rich Plasma Therapy in Androgenic Alopecia—A Meta-Analysis" Journal of Personalized Medicine 12, no. 3: 342. https://doi.org/10.3390/jpm12030342

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop