Next Article in Journal
Comparative Study on the Mechanism of Macrophage Activation Induced by Polysaccharides from Fresh and Dried Longan
Previous Article in Journal
Potential Neuroprotective Effect of Melatonin in the Hippocampus of Male BTBR Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Extended Treatment with Micron-Size Oral Palmitoylethanolamide (PEA) in Chronic Pain: A Systematic Review and Meta-Analysis

1
Department of Anesthesia, Intensive Care and Pain Therapy, Verona University Hospital, 37134 Verona, Italy
2
Innovative Statistical Research, 35100 Padua, Italy
3
Department of Medicine and Public Health, Section of Forensic Medicine, University of Verona, 37134 Verona, Italy
4
Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy
5
Paolo Procacci Foundation, 00193 Rome, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(11), 1653; https://doi.org/10.3390/nu16111653
Submission received: 23 April 2024 / Revised: 23 May 2024 / Accepted: 25 May 2024 / Published: 28 May 2024
(This article belongs to the Section Clinical Nutrition)

Abstract

:
Palmitoylethanolamide (PEA) emerged over the years as a promising approach in the management of chronic pain. Despite the fact that the efficacy of micron-size PEA formulations appears to be time-dependent, the optimal timing has not yet been elucidated. This systematic review and meta-analysis aim to estimate the possible advantage of an extended treatment in the relief of chronic pain. The literature search was conducted consulting scientific databases, to identify clinical trials in which micron-size PEA was administered for at least 60 days, and pain assessed by the Visual Analogue Scale (VAS) or Numeric Rating Scale (NRS). Nine studies matched the required criteria, for a total of 742 patients involved. The meta-analysis showed a statistically and clinically significant pain intensity reduction after 60 days of micron-size PEA supplementation, compared to 30 days (1.36 points, p < 0.01). The secondary analysis revealed a weighted NRS/VAS score decrease of 2.08 points within the first month of treatment. These two obtained scores corresponded to a 35.1% pain intensity reduction within the first month, followed by a further 35.4% during the second month. Overall, these results confirm the clinically relevant and time-depended pain-relieving effect of micron-size PEA and therefore the advantage of an extended treatment, especially in patient with incomplete pain management.

1. Introduction

According to the International Association for the Study of Pain (IASP), chronic pain is defined as “pain that persists or recurs for longer than three months” [1]. It affects between a third and half of the population worldwide [2]. Complete relief from chronic pain is often unrealistic and integrated pain management using multimodal therapies is currently the advocated strategy [3]. These therapies may include exercise, physical therapy, massage, cognitive and behavioral therapy, mind–body practices, neuroablative or neuromodulative treatments and drugs [4]. To date, the pharmacological options for analgesia are limited to opioids, nonsteroidal anti-inflammatory drugs (NSAIDs), acetaminophen, antidepressants and anticonvulsants, which provide variable efficacy and common adverse events (AEs) [5]. A growing body of evidence suggests that neuroinflammation plays an important role in the induction and maintenance of chronic pain [6]. In response to peripheral and central injury, glial cells, in particular microglia, show an increased activity in several pain pathways. After trauma or nerve injury, microglia increase the expression of markers and receptors, and release inflammatory mediators that contribute to neuronal excitability and pain generation [7]. Microglia also respond to inflammatory signals coming from mast cells, whose activation can modify the sensory transmission via a wide spectrum of pain mediators which can directly interact with sensory nerve terminals [7,8,9]. Sensory neurons, by releasing neuropeptides, may in turn increase mast cells activation/degranulation, which results in nociceptor sensitization, reduced pain threshold at the site of inflammation, dysfunctional pain signaling and hyperalgesia [10]. The involvement of mast cells and microglia in the development of hyperalgesia has been already demonstrated in different conditions such as chronic low back pain, visceral or pelvic pain, migraine, trigemino-cervical pain, lumbosacral pain and neurological disorders like Parkinson’s disease, multiple sclerosis and stroke [11,12,13].
While the excess of peripheral nociceptive stimulation leads to peripheral sensitization, which is essential for the development of pain, chronic pain is the result of neuronal plasticity and the so called “central sensitization”. This phenomenon derives from multiple and complex interactions between the nervous system, the immune system cells and other non-neuronal and non-immune cells, which play a fundamental role in both the amplification and chronicization of pain, also potentially influencing its different emotional and cognitive components [14]. Once central sensitization occurs, treatments for the associated syndromes require a multimodal approach that includes physical therapy, behavioral pain psychology and pharmacological agents specifically targeting neuroinflammation, pain modulation and pain pathways amplification [15]. Based on this evidence, the modulation of neuroinflammation emerged as an efficacious therapeutic option for pain control [16]. Among neuromodulators, the endogenous lipid mediator palmitoylethanolamide (PEA), a member of the N-acylethanolamine family, naturally present in several food sources, has been reported to exert anti-allodynic and anti-hyperalgesic effects by down-modulating mast cell activation and controlling glial cell behaviors [9,10]. These immuno-regulatory and analgesic properties of PEA are mediated through direct and indirect biological pathways, involving cannabinoid receptors type 1 (CB1) and type 2 (CB2), cannabinoid-related G protein-coupled receptors 55 (GPR55) and 19 (GPR119), transient receptor potential vanilloid type 1 (TRPV1) channels, and nuclear peroxisome proliferator-activated receptor-alpha (PPAR-a) [17,18]. PEA is endogenously produced in all tissues of the human body in response to stressful conditions, inflammatory stimuli, injury or pain [19]. When these conditions are protracted, PEA may be depleted and exogenous PEA administration may become important to restore its protective, anti-inflammatory and analgesic effects [20]. Although PEA is present in several dietary sources, its levels in foods are too low to represent an adequate intake in pathological conditions and a further exogenous supplementation must be considered [21,22,23]. However, exogenous PEA administration may significantly counteract neuroinflammation at the cellular level only in micronized (mPEA, 2–10 μm range) or ultra-micronized (umPEA, 0.8–6 μm range) forms, while in the native state (naïve PEA), due to the large particle size (from 100 up to 2000 μm range), PEA showed a poor absorption that significantly reduced its distribution and bioavailability, providing a low biological effect [23,24,25,26]. Also, it was demonstrated that mPEA and umPEA, in combination with natural compounds (co-micronized or co-ultra-micronized forms, respectively), such as the antioxidant polydatin (e.g., mPEAPol), demonstrated a synergistic effects and stronger biological activity [21]. From a clinical perspective, many publications reported the efficacy of all these micron-size PEA formulations (micronized, ultra-micronized, co-micronized and co-ultra-micronized) in chronic pain conditions of different etiologies [16,27,28,29,30,31,32,33,34,35]. Furthermore, several clinical trials have highlighted the time-dependent efficacy of these preparations in the management of chronic pain [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55], but none of them have quantified the advantage of extending the supplementation beyond the first month of treatment. The aim of this systematic review and meta-analysis was therefore to estimate this improvement in different chronic pain conditions.

2. Materials and Methods

2.1. Search Strategy

This review adhered to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) guidelines [56]. The search technique was performed according to the PICO (Population/Participants, Interventions, Comparisons, Outcome) approach. We searched in the PubMed and Scopus databases until September 2023 using the following keywords: “micronized Palmitoylethanolamide” or “ultra-micronized Palmitoylethanolamide” AND “chronic pain” or “chronic pelvic pain” or “neuropathic pain” or “neuropathy” or “neuralgia”. A supplementary search examined the references of the papers selected after the initial search. No restrictions regarding the publication date or language were imposed.

2.2. Inclusion and Exclusion Criteria

We searched studies including patients of both genders with different ages and chronic pain of various etiologies, submitted to different preparations of micron-size PEA alone or as an add-on to treatment with other pain medications. The eligible studies had to include: (i) pain intensity measurement using either the Visual Analogue Scale (VAS, 0–100 mm) or the Numeric Rating Scale (NRS, 0–10 points) and (ii) the assessment of pain at baseline, 30 days and 60 days after the start of micron-size PEA’s administration.
Narrative or systematic reviews, meta-analyses, book chapters, abstracts, congress communications and case reports were excluded from the analysis.
The initial search results underwent a thorough deduplication process, wherein two authors separately examined the titles and abstracts to identify and exclude papers that were not relevant. Afterward, the entire text was scrutinized to determine whether items met the qualifying criteria. The authors of three publications were contacted and requested to deliver additional data relevant for the analysis and not reported in the published papers [53,54,55].

2.3. Endpoints

The primary endpoint was the assessment of the further change in pain intensity, using VAS or NRS scores, after two months of micron-size PEA administration, compared to a 30-day treatment period (T30–T60 interval). The secondary endpoints were the assessment of the weighted change in pain scores from baseline to the first month (T0–T30), and the percentage changes in NRS/VAS scores during the two different intervals T0–T30 and T30–T60.

2.4. Risk of Bias

The evaluation of the potential for bias in each study included in the meta-analysis was performed autonomously by two authors. The Cochrane risk of bias (RoB) instrument, RoB2, was used to assess the quality of randomized clinical trials [57]. The Risk of Bias in non-randomized studies of Interventions (ROBINS-I) tool was adopted for non-randomized studies [58]. The evaluation covered different areas (5 areas for randomized trials and 7 for non-randomized studies) that addressed potential biases related to the randomization process (RoB D1), confounding (ROBINS-I D1), participant selection (ROBINS-I D2), intervention classification (ROBINS-I D3), deviation from intended interventions (RoB D2 and ROBINS-I D4), missing data (RoB D3 and ROBINS-I D5), outcome measurement (RoB D4 and ROBINS-I D6) and selection of reported results (RoB D5 and ROBINS-I D7). These evaluations classify each study’s risk of bias as low, moderate, serious, critical or with some concerns. The RoB assessment visualization was created using the Cochrane Risk-of-bias visualization (Robvis) program [59].

2.5. Statistical Analysis

A random effects model, which handles the non-homogeneity among the selected studies, was employed to estimate the primary efficacy outcome [60]. In instances wherein studies reported multiple interventional groups, the outcomes of each group were treated as independent entities. In such cases where the standard error of the mean (SEM) was presented, the standard deviation (SD) was computed using the relevant mathematical formula. When the SD of the NRS/VAS score change from the 30-day endpoint to the T60 final follow-up was not provided in the original articles, a conservative repeated measures correlation of 0.4 was employed for calculation [61], with data from the study by Schweiger et al. [49] utilized to validate this choice, resulting in a repeated measures correlation of 0.43. The average pain intensity change (estimated effect) and its corresponding 95% confidence interval (CI) were computed for each study. Heterogeneity among studies was assessed using I2 and τ2 statistics. The meta-analysis was executed using R 4.3.1 software, with statistical significance set at p < 0.05. The weighted average pain intensity score at each evaluation time point was calculated for secondary outcomes, considering the weight assigned to each study resulting from the meta-analysis. The corresponding SEM was determined from the SD by the software. The treatment effect was expressed as the change in the weighted average NRS/VAS score between baseline, T30 and T60 endpoints (T30 vs. T0, T60 vs. T0, T60 vs. T30). The change in pain intensity between T0–T30 and T30–T60, computed as a percentage, served to estimate the effect at each follow-up relative to the preceding one (T30 vs. T0, T60 vs. T30) and, in comparison to the overall impact (T60–T0) achieved during the course of the studies.

3. Results

3.1. Search Results

A total of 160 studies were identified from the initial search. Among these, 116 studies were excluded after screening and removing duplications. Then, 44 studies were assessed for eligibility. After abstract screening, eight studies were excluded, and 36 studies were analyzed to evaluate the required criteria. Of these, nine studies were deemed eligible and included in the systematic review [40,42,49,50,51,52,53,54,55]. The flowchart of the study selection process is reported in Figure 1.

3.2. Description of the Included Studies

Out of the nine selected studies, one is a randomized clinical trial [53], while eight are uncontrolled studies with various designs (observational prospective or retrospective, pilot studies or case series) [40,42,49,50,51,52,54,55]. Five studies focused on chronic pelvic pain [40,52,53,54,55], three on chronic low back pain of different origins [i.e., nonsurgical lumbar radiculopathies [50], FBSS (failed back surgery syndrome, to date modified as persistent spinal pain syndrome, PSPS) [51], lumbosciatica [42] and one study focused on chronic pain related to fibromyalgia syndrome (FMS) [49]. The selected studies involved a total of 742 patients. Among these, 614 patients were treated with umPEA (NORMAST®600), 81 with mPEAPol (PELVILEN®), 30 with mPEAPol + umPEA (PELVILEN® DUAL ACT) and 17 with a combined treatment started with umPEA (NORMAST®600) and followed by mPEAPol (PELVILEN®). All these preparations are released on the market by a single manufacturing company (Epitech Group SpA, Saccolongo, Padova, Italy). The dosages of these preparations, generally used as an add-on supplement to pain medications, ranged from 600 mg to 1800 mg per day for umPEA and from 800 mg + 80 mg to 1200 mg + 120 mg per day for mPEAPol. UmPEA was generally prescribed twice daily during the first period of treatment, followed by once a day until the final follow-up [42,49,50,51,55], whereas mPEAPol and the mPEAPol + umPEA combination were administered twice daily for the entire study duration [40,52,53,54,55]. Pain intensity was assessed using the VAS score in six studies (66.7%) [40,49,50,51,52,55] and the NRS score in three studies (33.3%) [42,53,54]. Mild AEs, mainly of gastrointestinal type, were reported in only three studies and generally judged by the authors as unrelated to the treatment [40,49,54]. However, in all the included studies, these micron-size formulations have been considered safe and with a significant tolerability profile, and no pharmacological interaction with the concomitant analgesic and/or anti-inflammatory therapies were reported. The main characteristics of the included studies are presented in Table 1.

3.3. Risk of Bias

All studies were identified as having a moderate risk of bias, since they all showed moderate bias in the outcome measurement domain. Six trials exhibited bias attributable to confounding factors [40,42,49,50,51,52], one in participant selection [51] and two in the classification of intervention [49,50]. In all studies, the bias related to deviations from the intended intervention as well as to missing data was deemed low. Concerns about the selection of reported results were rated at a moderate risk for most of the studies. No sensitivity analysis was performed since none of the studies had a high risk of bias. The ROB summary is presented in Figure 2 and Figure 3.

3.4. Primary Endpoint: T60 vs. T30 Pain Intensity Change

Regarding the primary endpoint, the meta-analysis showed that the extension of micron-size PEA supplementation to 60 days led to a further pain reduction effect compared to the first 30 days, therefore favoring the 60-day intervention. The estimate average pain intensity reduction was of 1.36 points (95% CI: 0.98; 1.74; p < 0.01) on the NRS/VAS scale between T30 and T60. This overall effect was statistically significant, although the heterogeneity among the included studies was high (I2 = 95%; τ2 = 0.34; p < 0.01). The forest plot of the meta-analysis is presented in Figure 4.

3.5. Secondary Endpoints: T30 vs. T0 Pain Intensity Change, and Percentage Variation over Time

Table 2 shows the number of patients and the average pain intensity scores at T0, T30 and T60 for each study considered in the meta-analysis.
Regarding the secondary endpoints, the data analysis showed a weighted reduction of 2.08 points from baseline to the first month (T30). This reduction was equivalent to a decrease of 35.1% in the NRS/VAS score during the first 30 days of treatment (T0–T30 interval). The decrease of 1.36 points, highlighted during the second month (T30–T60 period) by the meta-analysis, corresponded to a further reduction of 35.4% in pain intensity (Table 3 and Figure 5). Comparing the reduction in pain score obtained during the T0–T30 and T30–T60 intervals with the overall improvement observed during the 2-month period (T0–T60, 3.44 points, 100%), the NRS/VAS score reduction was of 60.4% after the first month and of 39.6% after 60 days of micron-size PEA supplementation (Table 3 and Figure 5).

4. Discussion

To date, several clinical studies have demonstrated the role of oral PEA in its micron-size formulations in the management of various painful conditions, with an efficacy that appears to be time-dependent [36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55]. Also, this time-dependency effect was highlighted in a recent meta-analysis, suggesting a treatment of at least four weeks with PEA [63]. Despite this evidence, the optimal timing and treatment duration have not been clearly elucidated or explored in detail until now. Our systematic review and meta-analysis, focused primarily on whether an extended supplementation with micron-size oral PEA could lead to additional pain relief, showed a statistically significant efficacy of this supplementation after 60 days compared to the 30-day treatment. The decrease in pain intensity was also accompanied by an improvement in QoL, without relevant AEs or pharmacological interaction with the concomitant analgesic therapies during the entire considered period. Furthermore, in two out the nine considered studies, the improvement in pain intensity was accompanied by a reduced need of medications, suggesting that the supplementation with oral micron-size PEA may have an analgesics’ sparing effect, reducing also the incidence of drugs-related AEs [40,55]. From a pathophysiological point of view, the benefits of an extended treatment with oral micron-size PEA in chronic pain may be explained by considering the specific role of the neuroinflammation in promoting and maintaining pain in different conditions. It is well known that neuroinflammation plays a key role in the onset and evolution of chronic pain and probably also in the transition from acute to chronic pain [64]. In fact, the so-called “non-neuronal cells” (microglia and mast cells) are considered the main responsible actors of this process [10]. Their prolonged activation leads to the uncontrolled release of pro-inflammatory mediators, resulting in changes in pain signaling pathways and chronic pain development [65,66]. The antinociceptive properties of oral PEA mainly depend on the down-regulation of these non-neuronal cells, through direct and indirect mechanisms [23]. It is also established that the timing of intervention is crucial in such situations and that early treatment is therefore essential [67,68].
The secondary analysis conducted in this work made it possible to estimate a weighted decrease of 2.08 points in the NRS/VAS score between the initial measurement and the 30-day follow-up, followed by a further decrease of 1.36 points in pain scores during the second month (T30–T60 interval). These results are consistent with the literature observations of the time-dependent effectiveness of micron-size PEA, as previously reported. However, one may argue that the reduction in small points in pain scores, while statistically significant, may not be clinically relevant for the patient. Although the clinically significant change in pain intensity measured by the scales or the minimum percentage of pain reduction from baseline was not clearly defined in the literature, some data are available in this context. Farrar et al., evaluating data from 2724 patients suffering from chronic pain of different etiologies and enrolled in 10 clinical trials, estimated that a reduction of approximately 2 points or of approximately 30% in the NRS score represent a clinically relevant difference in patients’ QoL [69]. The authors also demonstrated that, in studies with greater variability in baseline pain, such as those with no minimum baseline pain requirements, clinical relevance should be defined in terms of percent change, as the relationship between the percentage change and the patient’s impression of the change should be more coherent [69]. Furthermore, in some diseases like fibromyalgia syndrome, a limited improvement in perceived pain or QoL was also deemed by the authors to be clinically relevant, particularly in a patients’ population that has a very poor response to available conventional treatments [49,70]. Based on these findings, since in only four of the nine included studies the requirement for patient eligibility was an NRS/VAS score ≥ 5, to define whether the change in pain intensity was clinically relevant, the percentage reduction in the NRS/VAS score was also considered in our analysis. A pain intensity improvement of 35.1% was obtained during the first 30 days of micron-size PEA treatment, and a further 35.4% improvement was achieved during the second month, compared to the previous one, demonstrating that the supplementation with oral micron-size PEA allowed for a clinically significant and continued pain reduction over time.
Our analysis has some limitations, the main one of which is the uncontrolled nature of the included studies. Unfortunately, only a relatively small number of publications met the strict inclusion criteria, and all were inherently biased. Several studies considered just a few patients, who in general also underwent different concomitant therapies. In addition, the meta-analysis revealed a high heterogeneity of the selected studies, probably due to the different pathological conditions and/or to the difference in the oral micron-size PEA daily dosage. Taken together, all these factors may limit the statistical power of the final result.

5. Conclusions

This meta-analysis supports the advantage of extending the supplementation with oral micron-size PEA beyond the first month of treatment and the overall beneficial effect of this supplementation in the management of chronic pain. Micron-size PEA could therefore represent a possible adjunctive approach, with a noticeable tolerability profile, for patients experiencing pain associated with chronic diseases and already undergoing debilitating pharmacological therapies. However, due to the limitations of the analysis, more in-depth methodological studies are needed to corroborate these observations.

Author Contributions

Conceptualization, V.S. and E.P.; methodology, C.S.; software, C.S.; validation, G.V., E.P. and G.F.; formal analysis, A.M.; investigation, L.P.; resources, S.S.; data curation, C.S.; writing—original draft preparation, V.S.; writing—review and editing, V.S.; visualization, G.D.B.; supervision, G.V. and B.M.; project administration, E.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kang, Y.; Trewern, L.; Jackman, J.; McCartney, D.; Soni, A. Chronic Pain: Definitions and Diagnosis. BMJ 2023, 381, e076036. [Google Scholar] [CrossRef]
  2. Latina, R.; De Marinis, M.G.; Giordano, F.; Osborn, J.F.; Giannarelli, D.; Di Biagio, E.; Varrassi, G.; Sansoni, J.; Bertini, L.; Baglio, G.; et al. Epidemiology of Chronic Pain in the Latium Region, Italy: A Cross-Sectional Study on the Clinical Characteristics of Patients Attending Pain Clinics. Pain. Manag. Nurs. 2019, 20, 373–381. [Google Scholar] [CrossRef]
  3. Hanna, M.; Perrot, S.; Varrassi, G. Critical Appraisal of Current Acute LBP Management and the Role of a Multimodal Analgesia: A Narrative Review. Pain. Ther. 2023, 12, 377–398. [Google Scholar] [CrossRef]
  4. Collins, A. Pain Management in Primary Care: A Review of the Updated CDC Guideline. Nurse Pract. 2024, 49, 13–19. [Google Scholar] [CrossRef]
  5. Artukoglu, B.B.; Beyer, C.; Zuloff-Shani, A.; Brener, E.; Howard Bloch, M. Efficacy of Palmitoylethanolamide for Pain: A Meta-Analysis. Pain. Physician 2017, 20, 353–362. [Google Scholar]
  6. Paladini, A.; Fusco, M.; Cenacchi, T.; Schievano, C.; Piroli, A.; Varrassi, G. Palmitoylethanolamide, a Special Food for Medical Purposes, in the Treatment of Chronic Pain: A Pooled Data Meta-Analysis. Pain. Physician 2016, 19, 11–24. [Google Scholar] [PubMed]
  7. Bettoni, I.; Comelli, F.; Colombo, A.; Bonfanti, P.; Costa, B. Non-Neuronal Cell Modulation Relieves Neuropathic Pain: Efficacy of the Endogenous Lipid Palmitoylethanolamide. CNS Neurol. Disord. Drug Targets 2013, 12, 34–44. [Google Scholar] [CrossRef]
  8. Skaper, S.D.; Facci, L.; Giusti, P. Glia and Mast Cells as Targets for Palmitoylethanolamide, an Anti-Inflammatory and Neuroprotective Lipid Mediator. Mol. Neurobiol. 2013, 48, 340–352. [Google Scholar] [CrossRef]
  9. Skaper, S.D.; Giusti, P.; Facci, L. Microglia and Mast Cells: Two Tracks on the Road to Neuroinflammation. FASEB J. 2012, 26, 3103–3117. [Google Scholar] [CrossRef] [PubMed]
  10. Skaper, S.D.; Facci, L.; Fusco, M.; Della Valle, M.F.; Zusso, M.; Costa, B.; Giusti, P. Palmitoylethanolamide, a naturally occurring disease-modifying agent in neuropathic pain. Inflammopharmacology 2014, 22, 79–94. [Google Scholar] [CrossRef] [PubMed]
  11. Keppel Hesselink, J.M. New Targets in Pain, Non-Neuronal Cells, and the Role of Palmitoylethanolamide. Open Pain. J. 2012, 5, 12–23. [Google Scholar] [CrossRef]
  12. Chen, G.; Zhang, Y.Q.; Qadri, Y.J.; Serhan, C.N.; Ji, R.R. Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 2018, 100, 1292–1311. [Google Scholar] [CrossRef]
  13. Wasner, G. Central Pain Syndromes. Curr. Pain. Headache Rep. 2010, 14, 489–496. [Google Scholar] [CrossRef]
  14. Vergne-Salle, P.; Bertin, P. Chronic pain and neuroinflammation. Jt. Bone Spine 2021, 88, 105222. [Google Scholar] [CrossRef]
  15. Bettini, L.; Moore, K. Central Sensitization in Functional Chronic Pain Syndromes: Overview and Clinical Application. Pain. Manag. Nurs. 2016, 17, 333–338. [Google Scholar] [CrossRef]
  16. Gatti, A.; Lazzari, M.; Gianfelice, V.; Di Paolo, A.; Sabato, E.; Sabato, A.F. Palmitoylethanolamide in the Treatment of Chronic Pain Caused by Different Etiopathogenesis. Pain. Med. 2012, 13, 1121–1130. [Google Scholar] [CrossRef]
  17. Domínguez, C.M.; Martín, A.D.; Ferrer, F.G.; Puertas, M.I.; Muro, A.L.; González, J.M.; Prieto, J.P.; Taberna, I.R. N-Palmitoylethanolamide in the Treatment of Neuropathic Pain Associated with Lumbosciatica. Pain. Manag. 2012, 2, 119–124. [Google Scholar] [CrossRef]
  18. Cruccu, G.; Di Stefano, G.; Marchettini, P.; Truini, A. Micronized Palmitoylethanolamide: A Post Hoc Analysis of a Controlled Study in Patients with Low Back Pain—Sciatica. CNS Neurol. Disord. Drug Targets 2019, 18, 491–495. [Google Scholar] [CrossRef]
  19. Capra, A.P.; Ardizzone, A.; Crupi, L.; Calapai, F.; Campolo, M.; Cuzzocrea, S.; Esposito, E. Efficacy of Palmitoylethanolamide and Luteolin Association on Post-Covid Olfactory Dysfunction: A Systematic Review and Meta-Analysis of Clinical Studies. Biomedicines 2023, 11, 2189. [Google Scholar] [CrossRef]
  20. Clayton, P.; Hill, M.; Bogoda, N.; Subah, S.; Venkatesh, R. Palmitoylethanolamide: A Natural Compound for Health Management. Int. J. Mol. Sci. 2021, 22, 5305. [Google Scholar] [CrossRef]
  21. Peritore, A.F.; Siracusa, R.; Crupi, R.; Cuzzocrea, S. Therapeutic Efficacy of Palmitoylethanolamide and Its New Formulations in Synergy with Different Antioxidant Molecules Present in Diets. Nutrients 2019, 11, 2175. [Google Scholar] [CrossRef] [PubMed]
  22. Casale, R.; Symeonidou, Z.; Ferfeli, S.; Micheli, F.; Scarsella, P.; Paladini, A. Food for Special Medical Purposes and Nutraceuticals for Pain: A Narrative Review. Pain. Ther. 2021, 10, 225–242. [Google Scholar] [CrossRef] [PubMed]
  23. Petrosino, S.; Moriello, A.S. Palmitoylethanolamide: A Nutritional Approach to Keep Neuroinflammation within Physiological Boundaries. A Systematic Review. Int. J. Mol. Sci. 2020, 21, 9526. [Google Scholar] [CrossRef] [PubMed]
  24. Impellizzeri, D.; Bruschetta, G.; Cordaro, M.; Crupi, R.; Siracusa, R.; Esposito, E.; Cuzzocrea, S. Micronized/Ultramicronized Palmitoylethanolamide Displays Superior Oral Efficacy Compared to Non micronized Palmitoylethanolamide in a Rat Model of Inflammatory Pain. J. Neuroinflammation 2014, 11, 136. [Google Scholar] [CrossRef] [PubMed]
  25. Petrosino, S.; Cordaro, M.; Verde, R.; Schiano Moriello, A.; Marcolongo, G.; Schievano, C.; Siracusa, R.; Piscitelli, F.; Peritore, A.F.; Crupi, R.; et al. Oral Ultramicronized Palmitoylethanolamide: Plasma and Tissue Levels and Spinal Anti-Hyperalgesic Effect. Front. Pharmacol. 2018, 9, 249. [Google Scholar] [CrossRef] [PubMed]
  26. Indraccolo, U.; Favilli, A.; Dell’anna, A.; Di Francesco, A.; Dionisi, B.; Giugliano, E.; Murina, F.; Stocco, E. Looking for Responders among Women with Chronic Pelvic Pain Treated with a Comicronized Formulation of Micronized Palmitoylethanolamide and Polydatin. Biomed. Res. Int. 2022, 2022, 8620077. [Google Scholar] [CrossRef] [PubMed]
  27. Cremon, C.; Stanghellini, V.; Barbaro, M.R.; Cogliandro, R.F.; Bellacosa, L.; Santos, J.; Vicario, M.; Pigrau, M.; Alonso Cotoner, C.; Lobo, B.; et al. Randomized Clinical Trial: The Analgesic Properties of Dietary Supplementation with Palmitoylethanolamide and Polydatin in Irritable Bowel Syndrome. Aliment. Pharmacol. Ther. 2017, 45, 909–922. [Google Scholar] [CrossRef] [PubMed]
  28. Tartaglia, E.; Armentano, M.; Giugliano, B.; Sena, T.; Giuliano, P.; Loffredo, C.; Mastrantonio, P. Effectiveness of the Association N-Palmitoylethanolamine and Transpolydatin in the Treatment of Primary Dysmenorrhea. J. Pediatr. Adolesc. Gynecol. 2015, 28, 447–450. [Google Scholar] [CrossRef] [PubMed]
  29. Truini, A.; Biasiotta, A.; Di Stefano, G.; La Cesa, S.; Leone, C.; Cartoni, C.; Federico, V.; Petrucci, M.; Cruccu, G. Palmitoylethanolamide Restores Myelinated-Fibre Function in Patients with Chemotherapy-Induced Painful Neuropathy. CNS Neurol. Disord. Drug Targets 2011, 10, 916–920. [Google Scholar] [CrossRef]
  30. Canteri, L.; Petrosino, S.; Guida, G. Reducción Del Consumo de Antiinflamatorios y Analgésicos En El Tratamiento Del Dolor Neuropático Crónico En Pacientes Afectados Por Lumbociatalgia de Tipo Compresivo y En Tratamiento Con Normast® 300 Mg. Dolor 2010, 25, 227–234. [Google Scholar]
  31. Guida, G.; De Martino, M.; De Fabiani, A.; Cantieri, L.; Alexandre, A.; Vassallo, G.M.; Rogai, M.; Lanaia, F.; Petrosino, S. La Palmitoiletanolamida (Normast ®) En El Dolor Neuropático Crónico Por Lumbociatalgia de Tipo Compresivo: Estudio Clínico Multicéntrico. Dolor 2010, 25, 35–42. [Google Scholar]
  32. Murina, F.; Graziottin, A.; Felice, R.; Radici, G.; Tognocchi, C. Vestibulodynia: Synergy Between Palmitoylethanolamide + Transpolydatin and Transcutaneous Electrical Nerve Stimulation. J. Low. Genit. Tract. Dis. 2013, 17, 111–116. [Google Scholar] [CrossRef] [PubMed]
  33. Dalla Volta, G.; Zavarize, P.; Ngonga, G.; Carli, D. Ultramicronized Palmitoylethanolamide Reduces Frequency and Pain Intensity in Migraine. A Pilot Study. Int. J. Neurol. Brain Disord. 2016, 3, 1–5. [Google Scholar] [CrossRef]
  34. Conigliaro, R.; Drago, V.; Foster, P.; Schievano, C.; Di Marzo, V. Use of Palmitoylethanolamide in the Entrapment Neuropathy of the Median in the Wrist. Minerva Med. 2011, 102, 141–147. [Google Scholar] [PubMed]
  35. Del Giorno, R.; Skaper, S.; Paladini, A.; Varrassi, G.; Coaccioli, S. Palmitoylethanolamide in Fibromyalgia: Results from Prospective and Retrospective Observational Studies. Pain. Ther. 2015, 4, 169–178. [Google Scholar] [CrossRef] [PubMed]
  36. Cocito, D.; Peci, E.; Ciaramitaro, P.; Merola, A.; Lopiano, L. Short-Term Efficacy of Ultramicronized Palmitoylethanolamide in Peripheral Neuropathic Pain. Pain. Res. Treat. 2014, 2014, 854560. [Google Scholar] [CrossRef] [PubMed]
  37. Putzu, G.A. Efficacy of Ultramicronized Palmitoylethanolamide on the Clinical Symptoms of Charcot-Marie-Tooth Neuropathy. Archiv Neurol. Neurosurgery 2016, 1, 12–14. [Google Scholar]
  38. Marini, I.; Cavallaro, M.; Bartolucci, M.; Alessandri-Bonetti, A.; Gatto, M.; Cordaro, M.; Checchi, L. Can Celecoxib Enhance Palmitoylethanolamide’s Effect in the Treatment of Temporo-Mandibular Arthralgia in Osteoarthritis Patients? J. Transl. Sci. 2018, 5, 1–4. [Google Scholar]
  39. Chirchiglia, D.; Cione, E.; Caroleo, M.C.; Wang, M.; Di Mizio, G.; Faedda, N.; Giacolini, T.; Siviglia, S.; Guidetti, V.; Gallelli, L. Effects of Add-on Ultramicronized N-Palmitol Ethanol Amide in Patients Suffering of Migraine with Aura: A Pilot Study. Front. Neurol. 2018, 9, 674. [Google Scholar] [CrossRef] [PubMed]
  40. Lo Monte, G.; Soave, I.; Marci, R. Utilizzo Della Palmitoiletanolamide Micronizzata (PEA)-Transpolidatina Nel Trattamento Del Dolore Perlvico Cronico in Donne Affette Da Endometriosi. Risultati Preliminari. Minerva Ginecol. 2013, 65, 453–463. [Google Scholar]
  41. Marini, I.; Bartolucci, M.L.; Bortolotti, F.; Gatto, M.R.; Bonetti, G.A. Palmitoylethanolamide Versus a Nonsteroidal Anti-Inflammatory Drug in the Treatment of Temporomandibular Joint Inflammatory Pain. J. Orofac. Pain. 2012, 26, 99–104. [Google Scholar] [PubMed]
  42. Scaturro, D.; Asaro, C.; Lauricella, L.; Tomasello, S.; Varrassi, G.; Mauro, G.L. Combination of Rehabilitative Therapy with Ultramicronized Palmitoylethanolamide for Chronic Low Back Pain: An Observational Study. Pain. Ther. 2020, 9, 319–326. [Google Scholar] [CrossRef] [PubMed]
  43. Chirchiglia, D.; Paventi, S.; Seminara, P.; Cione, E.; Gallelli, L. N-Palmitoyl Ethanol Amide Pharmacological Treatment in Patients With Nonsurgical Lumbar Radiculopathy. J. Clin. Pharmacol. 2018, 58, 733–739. [Google Scholar] [CrossRef] [PubMed]
  44. Passavanti, M.B.; Fiore, M.; Sansone, P.; Aurilio, C.; Pota, V.; Barbarisi, M.; Fierro, D.; Pace, M.C. The Beneficial Use of Ultramicronized Palmitoylethanolamide as Add-on Therapy to Tapentadol in the Treatment of Low Back Pain: A Pilot Study Comparing Prospective and Retrospective Observational Arms. BMC Anesthesiol. 2017, 17, 171. [Google Scholar] [CrossRef] [PubMed]
  45. Sommariva, M.; Schievano, C.; Saleh, O. Micronized Palmitoylethanolamide Reduces Bladder Chronic Pelvic Pain Due to Different Etiologies and Improves Bladder Functions. Pelviperineology 2017, 36, 92–96. [Google Scholar]
  46. Cervigni, M.; Nasta, L.; Schievano, C.; Lampropoulou, N.; Ostardo, E. Micronized Palmitoylethanolamide-Polydatin Reduces the Painful Symptomatology in Patients with Interstitial Cystitis/Bladder Pain Syndrome. Biomed. Res. Int. 2019, 2019, 9828397. [Google Scholar] [CrossRef] [PubMed]
  47. Orefice, N.S.; Alhouayek, M.; Carotenuto, A.; Montella, S.; Barbato, F.; Comelli, A.; Calignano, A.; Muccioli, G.G.; Orefice, G. Oral Palmitoylethanolamide Treatment Is Associated with Reduced Cutaneous Adverse Effects of Interferon-Β1a and Circulating Proinflammatory Cytokines in Relapsing–Remitting Multiple Sclerosis. Neurotherapeutics 2016, 13, 428–438. [Google Scholar] [CrossRef] [PubMed]
  48. Evangelista, M.; Cilli, V.; De Vitis, R.; Militerno, A.; Fanfani, F. Ultra-Micronized Palmitoylethanolamide Effects on Sleep-Wake Rhythm and Neuropathic Pain Phenotypes in Patients with Carpal Tunnel Syndrome: An Open-Label, Randomized Controlled Study. CNS Neurol. Disord. Drug Targets 2018, 17, 291–298. [Google Scholar] [CrossRef]
  49. Schweiger, V.; Martini, A.; Bellamoli, P.; Donadello, K.; Schievano, C.; Balzo, G.D.; Sarzi-Puttini, P.; Parolini, M.; Polati, E. Ultramicronized Palmitoylethanolamide (Um-PEA) as Add-on Treatment in Fibromyalgia Syndrome (FMS): Retrospective Observational Study on 407 Patients. CNS Neurol. Disord. Drug Targets 2019, 18, 326–333. [Google Scholar] [CrossRef]
  50. Chirchiglia, D.; Chirchiglia, P.; Signorelli, F. Nonsurgical Lumbar Radiculopathies Treated with Ultramicronized Palmitoylethanolamide (UmPEA): A Series of 100 Cases. Neurol. Neurochir. Pol. 2018, 52, 44–47. [Google Scholar] [CrossRef]
  51. Paladini, A.; Varrassi, G.; Bentivegna, G.; Carletti, S.; Piroli, A.; Coaccioli, S. Palmitoylethanolamide in the Treatment of Failed Back Surgery Syndrome. Pain. Res. Treat. 2017, 2017, 1486010. [Google Scholar] [CrossRef] [PubMed]
  52. Giugliano, E.; Cagnazzo, E.; Soave, I.; Lo Monte, G.; Wenger, J.M.; Marci, R. The Adjuvant Use of N-Palmitoylethanolamine and Transpolydatin in the Treatment of Endometriotic Pain. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 168, 209–213. [Google Scholar] [CrossRef] [PubMed]
  53. Di Francesco, A.; Pizzigallo, D. Use of Micronized Palmitoylethanolamide and Trans-Polydatin in Chronic Pelvic Pain Associated with Endometriosis. An Open-Label Study. Giorn. It Ost Gin 2014, 36, 353–357. [Google Scholar] [CrossRef]
  54. Dell’Anna, A.; De Marzi, C.A. Mast Cells and Microglia: New Therapeutic Targets for the Treatment of Endometriosis-Associated Symptomatology. Giorn. It Ost Gin 2017, 39, 193–201. [Google Scholar]
  55. Stochino Loi, E.; Pontis, A.; Cofelice, V.; Pirarba, S.; Fais, M.F.; Daniilidis, A.; Melis, I.; Paoletti, A.M.; Angioni, S. Effect of Ultramicronized-Palmitoylethanolamide and Co-Micronizedpalmitoylethanolamide/Polydatin on Chronic Pelvic Pain and Quality of Life in Endometriosis Patients: An Open-Label Pilot Study. Int. J. Womens Health 2019, 11, 443–449. [Google Scholar] [CrossRef] [PubMed]
  56. Page, M.J.; E McKenzie, J.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; A Akl, E.; E Brennan, S.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  57. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A Revised Tool for Assessing Risk of Bias in Randomised Trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [PubMed]
  58. Sterne, J.A.C.; Hernán, M.A.; Reeves, B.C.; Savović, J.; Berkman, N.D.; Viswanathan, M.; Henry, D.; Altman, D.G.; Ansari, M.T.; Boutron, I.; et al. ROBINS-I: A Tool for Assessing Risk of Bias in Non-Randomised Studies of Interventions. BMJ 2016, 355, i4919. [Google Scholar] [CrossRef] [PubMed]
  59. McGuinness, L.A.; Higgins, J.P.T. Risk-of-Bias VISualization (Robvis): An R Package and Shiny Web App for Visualizing Risk-of-Bias Assessments. Res. Synth. Methods 2021, 12, 55–61. [Google Scholar] [CrossRef]
  60. Borenstein, M.; Hedges, L.V.; Higgins, J.P.T.; Rothstein, H.R. A Basic Introduction to Fixed-Effect and Random-Effects Models for Meta-Analysis. Res. Synth. Methods 2010, 1, 97–111. [Google Scholar] [CrossRef]
  61. Gu, S.; Shi, J.; Tang, Z.; Sawhney, M.; Hu, H.; Shi, L.; Fonseca, V.; Dong, H. Comparison of Glucose Lowering Effect of Metformin and Acarbose in Type 2 Diabetes Mellitus: A Meta-Analysis. PLoS ONE 2015, 10, e0126704. [Google Scholar]
  62. PRISMA—Tansparent Reporting of Systematic Reviews and Meta-Analyses. Available online: http://prisma-statement.org (accessed on 29 September 2023).
  63. Lang-Illievich, K.; Klivinyi, C.; Lasser, C.; Brenna, C.T.A.; Szilagyi, I.S.; Bornemann-Cimenti, H. Palmitoylethanolamide in the Treatment of Chronic Pain: A Systematic Review and Meta-Analysis of Double-Blind Randomized Controlled Trials. Nutrients 2023, 15, 1350. [Google Scholar] [CrossRef] [PubMed]
  64. Echeverria-Villalobos, M.; Tortorici, V.; Brito, B.E.; Ryskamp, D.; Uribe, A.; Weaver, T. The role of neuroinflammation in the transition of acute to chronic pain and the opioid-induced hyperalgesia and tolerance. Front. Pharmacol. 2023, 14, 1297931. [Google Scholar] [CrossRef] [PubMed]
  65. Tsuda, M. Microglia in the CNS and Neuropathic Pain. Adv. Exp. Med. Biol. 2018, 1099, 77–91. [Google Scholar] [PubMed]
  66. Mai, L.; Liu, Q.; Huang, F.; He, H.; Fan, W. Involvement of Mast Cells in the Pathophysiology of Pain. Front. Cell Neurosci. 2021, 15, 665066. [Google Scholar] [PubMed]
  67. Polati, E.; Martini, A.; Schweiger, V. Ultramicronized palmitoylethanolamide treatment in central neuropathic pain following longstanding spinal cord injury: Try to extinguish the fire after everything was burned. Pain 2017, 158, 763–764. [Google Scholar] [CrossRef] [PubMed]
  68. Della Rocca, G.; Gamba, D. Chronic Pain in Dogs and Cats: Is There Place for Dietary Intervention with Micro-Palmitoylethanolamide? Animals 2021, 11, 952. [Google Scholar] [CrossRef] [PubMed]
  69. Farrar, J.T.; Young, J.P.B.; Lamoreaux, L.; Werth, J.L.; Poole, R.M. Clinical Importance of Changes in Chronic Pain Intensity Measured on an 11-Point Numerical Pain Rating Scale. Pain 2001, 94, 149–158. [Google Scholar] [CrossRef]
  70. Bennett, R.M.; Bushmakin, A.G.; Cappelleri, J.C.; Zlateva, G.; Sadosky, A.B. Minimal clinically important difference in the fibromyalgia impact questionnaire. J. Rheumatol. 2009, 36, 1304–1311. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the search strategy and studies selection according to PRISMA [62].
Figure 1. Flowchart of the search strategy and studies selection according to PRISMA [62].
Nutrients 16 01653 g001
Figure 2. Robvis traffic light plot for the risk of bias of the studies included in the meta-analysis [40,42,49,50,51,52,53,54,55].
Figure 2. Robvis traffic light plot for the risk of bias of the studies included in the meta-analysis [40,42,49,50,51,52,53,54,55].
Nutrients 16 01653 g002
Figure 3. Robvis summary plot for the risk of bias of the studies included in the meta-analysis.
Figure 3. Robvis summary plot for the risk of bias of the studies included in the meta-analysis.
Nutrients 16 01653 g003
Figure 4. Forest plot of the effect of a 60-day supplementation with micron-size PEA on chronic pain intensity reduction. Positive mean values (>0) represent the additional effect after the first 30 days of supplementation. Squares display the estimate impact, the size of each square reflects the weight assigned to each study. Horizontal lines represent the 95% CI for each estimate effect. The diamond width, which indicates the total 95% CI, represents the overall effect of intervention estimate using a random statistical model. I2 and τ2 statistics measure the heterogeneity [40,42,49,50,51,52,53,54,55].
Figure 4. Forest plot of the effect of a 60-day supplementation with micron-size PEA on chronic pain intensity reduction. Positive mean values (>0) represent the additional effect after the first 30 days of supplementation. Squares display the estimate impact, the size of each square reflects the weight assigned to each study. Horizontal lines represent the 95% CI for each estimate effect. The diamond width, which indicates the total 95% CI, represents the overall effect of intervention estimate using a random statistical model. I2 and τ2 statistics measure the heterogeneity [40,42,49,50,51,52,53,54,55].
Nutrients 16 01653 g004
Figure 5. NRS/VAS score improvement during the first and the second month of micron-size PEA treatment and the corresponding percentage of pain intensity reduction after each time point compared to the previous one.
Figure 5. NRS/VAS score improvement during the first and the second month of micron-size PEA treatment and the corresponding percentage of pain intensity reduction after each time point compared to the previous one.
Nutrients 16 01653 g005
Table 1. Study characteristics.
Table 1. Study characteristics.
ReferenceStudy DesignNumber of PatientsChronic Pain ConditionPain ToolTreatmentConcomitant TherapiesMain Results
Chirchiglia
et al., 2018
[50]
Retrospective, case-series100
(73 males, 27 females)
Low back pain
(nonsurgical lumbar radiculopathies with or without sciatica)
VAS1st cycle: 1200 mg/day sublingual umPEA for 10 days; 1200 mg/day umPEA tablets for 20 days
2nd cycle: 600 mg/day umPEA tablets for 1 month
1st cycle: Paracetamol 500 mg + codeine 30 mg/day for 4 days, then for 1 month as needed
2nd cycle: Paracetamol 500 mg + codeine 30 mg/day as needed
Significant decrease in pain intensity after one month of treatment with a further improvement after the second cycle. Greater effect on patients with mild and moderate pain
Paladini
et al., 2017
[51]
Observational, open-label35
(15 males, 20 females)
Low back pain
(failed back surgery syndrome)
VAS1200 mg/day umPEA for 1 month; subsequently 600 mg/day for the second monthTapentadol 150 mg/day
Pregabalin 300 mg/day
Further and significant decrease in pain intensity after umPEA supplementation
Scaturro
et al., 2020
[42]
Prospective, observational120
(37 males, 83 females)
Low back pain
(lumbosciatica/
lumbocruralgia)
NRS1200 mg/day umPEA for 20 days; subsequently 600 mg/day for 40 daysPregabalin 150 mg/day or oxycodone 10 mg/day,
daily functional rehabilitation, relaxation massage
Significant reduction in chronic pain to a not clinically relevant intensity; improvement in QoL; decrease in pain-dependent disability
Lo Monte
et al., 2013
[40]
Pilot,
open-label
24 femalesChronic pelvic pain
(endometriosis)
VAS800 + 80 mg/day mPEAPol for 90 daysAnalgesics and hormonal therapiesSignificant reduction in pelvic pain, dysmenorrhea and dyspareunia; improvement in QoL; decrease NSAIDs consumption
Giugliano
et al., 2013
[52]
Prospective,
open-label
47 females
Group A: 19 recto-vaginal endometriosis;
Group B: 28 ovarian endometriosis
Chronic pelvic pain
(endometriosis)
VAS800 + 80 mg/day mPEAPol for 90 daysEstrogen–progestin pill (13 patients group A, 18 patients
group B), anti-inflammatory drugs (6 patients group A, 10 patients group B)
Significant reduction in pelvic pain, dysmenorrhea, dyspareunia regardless of lesion site already after 30 days, reaching the maximum relief after 60 days
Di Francesco
and Pizzigallo, 2014
[53]
Three-arm, randomized,
open-label
30 females
Group A: 10 mPEAPol;
Group B: 10 leuprorelin acetate;
Group C: 10 ethinylestradiol + drospirenone
Chronic pelvic pain
(endometriosis)
NRS800 + 80 mg/day mPEAPol for 6 monthsNASignificant decrease in painful symptoms in all groups regardless of the treatment; improvement in QoL; not interference with woman fertility
Dell’Anna
and De Marzi, 2017
[54]
Observational, open-label17 femalesChronic pelvic pain
(endometriosis)
NRS600 mg/day umPEA +
1200 + 120 mg/day mPEAPol for 4 months
Analgesics and
anti-inflammatory drugs
as needed
Significant reduction in pelvic pain, dysmenorrhea, dyschezia, dyspareunia and dysuria; improvement in QoL; not interference with fertility
Stochino Loi
et al., 2019
[55]
Single arm,
non-randomized,
open-label
30 femalesChronic pelvic pain
(endometriosis)
VAS1200 mg/day umPEA for 10 days; subsequently 800 + 80 mg/day mPEAPol for 80 daysKetoprofen 80 mg, maximum twice dailySignificant reduction in pelvic pain, dyspareunia, dysmenorrhea, dyschezia; improvement in QoL and psychological well-being; significant reduction in the ketoprofen consumption
Schweiger
et al., 2019
[49]
Retrospective,
observational
359
(23 males, 336 females)
Fibromyalgia syndrome (FMS)VAS1800 mg/day umPEA for 10 days; subsequently 1200 mg/day for 20 days; followed by 600 mg/day until the 15th monthsVarious FMS drug treatments
(SSRI, SNRI, GBPs, TCA, BZD, OPI, NSAIDs, MR, ACT)
Significant improvement in pain intensity and QoL
SSRIs: Serotonin selective reuptake inhibitors; SNRIs: Serotonin noradrenaline selective inhibitors, GBPs: Gabapentinoids, TCAs: Tricyclic antidepressants, BZDs: Benzodiazepines, OPIs: Opiates, NSAIDs: Non-steroidal anti-inflammatory drugs; MRs: Muscle relaxants, ACT: acetaminophen; NA: not available, QoL: quality of life.
Table 2. Number of patients and outcomes of each study included in the meta-analysis.
Table 2. Number of patients and outcomes of each study included in the meta-analysis.
ReferenceT0
n
T0
NRS/VAS
Mean ± SD
T30
n
T30
NRS/VAS
Mean ± SD
T60
n
T60
NRS/VAS Mean ± SD
Giugliano et al., 2013 [52]
Group A
195.8 ± 2.8193.8 ± 2.4192.9 ± 1.7
Giugliano et al., 2013 [52]
Group B
284.6 ± 2.4282.7 ± 1.7281.6 ± 1.6
Chirchiglia et al., 2018 [50]
Group Mild
143.5 ± 0.75141.7 ± 0.37140 ± 0
Chirchiglia et al., 2018 [50]
Group Moderate
145.3 ± 0.37142.6 ± 0.37140 ± 0
Chirchiglia et al., 2018 [50]
Group Severe
728.7 ± 0.85726.4 ± 1.27724.6 ± 2.46
Paladini et al., 2017 [51]354.3 ± 0.65352.7 ± 0.53351.7 ± 0.65
Scaturro et al., 2020 [42]1206.3 ± 1.101203.7 ± 0.991202.0 ± 0.99
Lo Monte et al., 2013 [40]245.1 ± 2.65233.0 ± 2.09231.9 ± 1.65
Di Francesco
and Pizzigallo, 2014 [53]
105.3 ± 3.6394.8 ± 2.5894.0 ± 2.40
Dell’Anna and De Marzi, 2017 [54]177.8 ± 1.53166.1 ± 2.04154.9 ± 2.56
Stochino Loi et al., 2019 [55]307.2 ± 1.2304.1 ± 0.3282.9 ± 0.2
Schweiger et al., 2019 [49]3597.58 ± 1.523036.3 ± 1.982485.9 ± 2.09
n: number of patients; SD: standard deviation.
Table 3. Summary of the effects measured.
Table 3. Summary of the effects measured.
T0T30T60
NRS/VAS score (Weighted mean ± SEM)5.93 ± 0.473.85 ± 0.472.49 ± 0.45
Effect vs. T0 (score) 2.083.44
Effect vs. T30 (score) 1.36
Effect vs. previous follow-up (%) 35.135.4
SEM: standard error of the mean.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Schweiger, V.; Schievano, C.; Martini, A.; Polati, L.; Del Balzo, G.; Simari, S.; Milan, B.; Finco, G.; Varrassi, G.; Polati, E. Extended Treatment with Micron-Size Oral Palmitoylethanolamide (PEA) in Chronic Pain: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 1653. https://doi.org/10.3390/nu16111653

AMA Style

Schweiger V, Schievano C, Martini A, Polati L, Del Balzo G, Simari S, Milan B, Finco G, Varrassi G, Polati E. Extended Treatment with Micron-Size Oral Palmitoylethanolamide (PEA) in Chronic Pain: A Systematic Review and Meta-Analysis. Nutrients. 2024; 16(11):1653. https://doi.org/10.3390/nu16111653

Chicago/Turabian Style

Schweiger, Vittorio, Carlo Schievano, Alvise Martini, Luca Polati, Giovanna Del Balzo, Salvatore Simari, Beatrice Milan, Gabriele Finco, Giustino Varrassi, and Enrico Polati. 2024. "Extended Treatment with Micron-Size Oral Palmitoylethanolamide (PEA) in Chronic Pain: A Systematic Review and Meta-Analysis" Nutrients 16, no. 11: 1653. https://doi.org/10.3390/nu16111653

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