The effect of antioxidant supplementation on dysmenorrhea and endometriosis-associated painful symptoms: a systematic review and meta-analysis of randomized clinical trials

Article information

Obstet Gynecol Sci. 2024;67(2):186-198
Publication date (electronic) : 2024 January 15
doi : https://doi.org/10.5468/ogs.23210
1Department of Obstetrics and Gynecology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
2Department of Obstetrics and Gynecology, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
3Department of Obstetrics and Gynecology, Al Salama Hospital, Jeddah, Saudi Arabia
4Department of Obstetrics and Gynecology, Faculty of Medicine at Rabigh, King Abdulaziz University, Rabigh, Saudi Arabia
5Department of Obstetrics and Gynecology, Faculty of Medicine, Najran University, Najran, Saudi Arabia
6Department of Obstetrics and Gynecology, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
7Department of Obstetrics and Gynecology, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
8Department of Obstetrics and Gynecology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
Corresponding author: Ahmed Abu-Zaid, MD, PhD, Department of Obstetrics and Gynecology, College of Medicine, Alfaisal University, Al Zahrawi Street, Riyadh 11533, Saudi Arabia E-mail: aabuzaid@live.com
Received 2023 August 25; Revised 2023 October 11; Accepted 2023 November 9.

Abstract

This study aimed to review randomized controlled trials (RCTs) investigating the effects of dietary antioxidant supplements on the severity of endometriosis-related pain symptoms. The PubMed/Medline, Scopus, and Web of Science databases were searched until April 2022. Additionally, we manually searched the reference lists. Endpoints were summarized as standardized mean difference (SMD) with 95% confidence intervals (CIs) in a random-effects model. The I2 statistic was used to assess heterogeneity. Ten RCTs were included in this meta-analysis. Overall, 10 studies were related to dysmenorrhea, four to dyspareunia, and four to pelvic pain. Antioxidants significantly reduced dysmenorrhea (SMD, −0.48; 95% CI, −0.82 to −0.13; I2=75.14%). In a subgroup analysis, a significant reduction of dysmenorrhea was observed only in a subset of trials that administered vitamin D (SMD, −0.59; 95% CI, −1.13 to −0.06; I2=69.59%) and melatonin (SMD, −1.40; 95% CI, −2.47 to −0.32; I2=79.15%). Meta-analysis results also suggested that antioxidant supplementation significantly improved pelvic pain (SMD, −1.51; 95% CI, −2.74 to −0.29; I2=93.96%), although they seem not to have a significant beneficial impact on the severity of dyspareunia. Dietary antioxidant supplementation seems to beneficially impact the severity of endometriosis-related dysmenorrhea (with an emphasis on vitamin D and melatonin) and pelvic pain. However, due to the relatively small sample size and high heterogeneity, the findings should be interpreted cautiously, and the importance of further well-designed clinical studies cannot be overstated.

Introduction

Endometriosis is a chronic inflammatory gynecological condition characterized by the presence and growth of tissues similar to the endometrium outside the uterine cavity. It affects at least 10% of women of reproductive age [1]. Furthermore, it frequently causes pelvic pain, dysmenorrhea, dyspareunia, and infertility, impairing patients’ quality of life [2]. Endometriosis is a multifaceted, symptomatic, pathobiological, multisystem, and heterogeneous disease. Based on the phenotype, it can be categorized into superficial peritoneal lesions, ovarian endometriomas, and deep-infiltrating endometriosis [3,4]. When addressing general endometriosis management, three therapeutic modalities are commonly available: I) medicinal treatment (e.g., painkillers, nonsteroidal anti-inflammatory drugs, combined oral contraceptives, progestins, and gonadotropin-releasing hormone analogs); II) surgery (conservative or definitive); and III) assisted reproductive technologies (such as in vitro fertilization and intra-cytoplasmic sperm injection). It is crucial to emphasize the timely administration of pain management, as therapeutic inertia portends the development of central sensitization (autonomous) [57]. Interestingly, reduction in oxidative stress is also a crucial alternative for endometriosis management. Inflammation leads to an increased production of reactive oxygen species (ROS), which play a fundamental role in the proliferation of endometriotic cells as well as in the development, persistence, and progression of the disease [8]. Dietary micro- and macro-nutrients and dietary factors are pivotal in controlling chronic diseases [9,10]. Dietary antioxidants exert beneficial neutralizing effects against free radicals and ROS produced by endometriotic cells. They demonstrate anti-inflammatory properties [11,12] and pro-apoptotic and anti-angiogenic actions and generally have a favorable safety profile. Therefore, one could speculate that they may effectively reduce pain-inducing factors and improve endometriosis-associated clinical symptoms [1315].

A growing body of literature has explored the impact of different dietary antioxidants on endometriosis-related pain [15]. Antioxidant vitamins successfully reduce the intensity of dysmenorrhea, ameliorate dyspareunia and pelvic pain, and improve the quality of life in patients with endometriosis. Consequently, therapy involving antioxidant vitamins may be considered an alternative treatment approach, independently or in conjunction with other methods, to alleviate endometriosis-related pain [16]. Zheng et al. [16] showed that supplementation with vitamin E improved endometriosis-related pelvic pain, whereas supplementation with vitamin D did not. Some systematic reviews also reported no significant effects of vitamin D on dysmenorrhea or non-cyclic pelvic pain [17]. Nevertheless, no systematic reviews or meta-analyses have comprehensively summarized the effects of dietary antioxidants on endometriosis and dysmenorrhea. To bridge this gap, we conducted a comprehensive systematic review and meta-analysis of randomized controlled trials (RCTs). We aimed to investigate the duration response and the impact of administering various dietary antioxidant supplements, including vitamins D, C, E, and A, melatonin, curcumin, omega-3 fatty acids, resveratrol, zinc, copper, chromium, and selenium, separately or in different combinations, on the severity of endometriosis-associated painful symptoms. The symptoms evaluated include dysmenorrhea, dyspareunia, and chronic pelvic pain in women of reproductive age.

Methods

This systematic review and meta-analysis of RCTs was performed according to the guidelines of the preferred reporting items for systematic reviews and meta-analyses statement and the current recommendations of the Cochrane Collaboration.

1. Search strategy and study selection

We searched electronic research databases, including Scopus, Web of Science (Science and Social Science Citation Index), and PubMed/Medline, from their inception until April 2022. In addition, we manually searched the reference lists and citations of the eight identified articles using Google Scholar. We also contacted authors who had published in this area to ensure we did not miss any relevant publications. Search terms were set by the authors and adapted for use in other databases. There were no restrictions on the language or publication dates. Document with incomplete data or the author could not be reached was discarded. The search strategy is described in detail in Supplementary Table 1. Two reviewers (S.B. and A.G.) independently screened all identified records for potentially eligible studies after reading all titles and abstracts. The final inclusion criteria were determined after reading the full texts strictly. Disagreements were resolved through discussion with a third reviewer (A.A.).

2. Eligibility criteria

The eligibility criteria of the studies were formulated according to the participants, interventions, comparisons, outcomes, and study design criteria. I) Participants: women with clinically and/or histologically confirmed endometriosis; II) intervention: supplementation with antioxidants (vitamins D, C, E, and A, melatonin, curcumin, omega-3 fatty acids, resveratrol, zinc, copper, chromium, and selenium, separately or in different combinations); III) comparators: antioxidant versus no treatment, antioxidant versus placebo; IV) outcomes: severity of dysmenorrhea, dyspareunia and/or chronic pelvic pain assessed by any pain assessment scale/tool; and V) study design: a clinical randomized controlled study. Studies meeting any of the following criteria were excluded: I) pain caused by reasons other than endometriosis; II) non-randomized clinical trials (e.g., conference abstracts, repeated publications, animal experiments, case reports, and reviews); and III) studies without available data for analysis.

3. Data extraction and assessment of the quality of included studies

Two reviewers (H.S. and M.S.A.) independently extracted the following data from the included trials: participant characteristics (e.g., age, diagnostic method, body mass index, and parity), study characteristics (e.g., first author name, publication year, region, study design, sample size, intervention type, and intervention characteristics), and study outcomes. The corresponding authors were contacted for additional information. Two reviewers (H.S. and M.S.A.) assessed the methodological quality of the included trials according to the Cochrane Handbook for Systematic Reviews of Interventions version 5.1.0 (Cochrane, London, England). Disagreements were resolved through discussion with a third reviewer (K.K.). Each trial was evaluated for seven items: random sequence generation, concealed allocation, blinding of participants and personnel (performance bias), blinding of outcome assessment (detection bias), incomplete outcome data (attrition bias), selective reporting (reporting bias), and other biases; each item was rated as “high risk”, “low risk”, or “unclear”.

4. Statistical analysis

Stata version 16 (Stata Corporation, College Station, TX, USA) was used for the statistical analyses. All continuous variables were pooled by standard mean differences (SMDs) with 95% confidence intervals (CIs). Heterogeneity was evaluated using the Higgins’ I2 statistic. I2 statistics of 0–25%, 25–50%, 50–75%, and >75% were suggestive of very low, low, moderate, and high heterogeneity, respectively. We used a random-effects model to calculate individual study SMD and corresponding 95% CIs. In addition, a chi-square test for heterogeneity was performed, and the P-values were presented. Exploration of the causes of heterogeneity was planned using variations in the antioxidant type and duration of the intervention. We performed sensitivity analyses to evaluate the robustness of pooled estimations after exclusion of every single trial through the “Leave-one-out method”. Additionally, we assessed the risk of publication bias across studies using counter-funnel plots of the outcomes. We used the GRADEpro GDT software (Evidence Prime, Hamilton, ON, USA) to formally assess the quality of evidence for selected outcomes. We evaluated the outcomes considering the following five criteria: risk of bias, inconsistency, indirectness, imprecision, and publication bias, and the level of evidence was graded as very low, low, moderate, or high. The certainty of the evidence evaluation is presented in Supplementary Table 2.

Results

1. Study selection

Initially, 134 records were identified through a database search, and eight additional records were identified through other sources. All studies were then imported into EndNote X9 software (Thomson Reuters, Philadelphia, PA, USA), 25 duplicates were removed, and 91 studies were removed after the title and abstract screening process; thus, 18 studies were screened for eligibility in more detail. Finally, after detailed title, abstract, and full-text evaluations, 10 [13,1826] RCTs were included in this systematic review and meta-analysis. A flow diagram of the complete literature search and selection of studies is shown in Fig. 1.

Fig. 1

The flow diagram of literature search and selection of studies. RCT, randomized controlled trials.

2. Study characteristics

The ethnicity of the study population varied among the studies: Iran (n=4), Brazil (n=2), Italy (n=2), Britain (n=1), and the United States (n=1). All the included trials, except one, were published in English between 2007 and 2021. A total of 541 women aged 20–40 years were examined, with individual study sample sizes ranging from 30 to 145 participants. Endometriosis was confirmed by histopathology in all trials. Seven trials reported the endometriosis stage in the patients. The characteristics of the included trials are summarized in Table 1. A summary of the risk of bias demonstrated that the methodological quality of these trials was relatively desirable (Fig. 2). All participants in each trial were randomly allocated to groups using an adequate allocation procedure. The randomization was unclear in both trials. Six trials stated that the allocation was concealed, one trial stated that it was unclear, and three trials were at high risk. Half of the trials used patient masking, and the blinding of participants and personnel was unclear in four trials. Thus, only one trial had a high risk of performance bias. Four trials reported appropriate blinding of the outcome assessment, whereas blinding was unclear in five trials. Only one trial had a high risk of detection bias. Half the trials provided an intention-to-treat analysis. Three trials were conducted for each protocol. Two trials had a high risk of attrition bias. All the trials were judged to have a low risk of reporting bias. Finally, three trials were judged as unclear for other biases. All trials recorded sufficient data on dysmenorrhea; however, five of these trials provided insufficient data on pelvic pain, and four studies did not provide sufficient information on dyspareunia.

The main characteristics of the included studies

Fig. 2

The summary of risk of bias assessments.

3. The effect of antioxidants on dysmenorrhea

Pooling results from 10 trials that compared the effect of antioxidants versus placebo in terms of reducing dysmenorrhea suggested that antioxidants significantly reduced dysmenorrhea (SMD, −0.48; 95% CI, −0.82 to −0.13; P<0.001) (Fig. 3). High heterogeneity was detected between trials (P<0.001; I2=75.14%); however, there was no indication of publication bias affecting these findings (Egger’s regression intercept, −4.77; 95% CI, −12.17 to 2.62; P=0.178). In a subgroup analysis, a significant difference was observed in the subset of trials that compared the effect of various antioxidants on dysmenorrhea: vitamin D, omega-3 fatty acids, resveratrol, melatonin, and a combination of antioxidants (Chi2, 11.52; degree of differentiation, 4; P=0.02). There was a significant reduction in dysmenorrhea in the subset of trials that administered vitamin D (SMD, −0.59; 95% CI, −1.13 to −0.06; P=0.02; I2=69.59%), and those that administered melatonin (SMD, −1.40; 95% CI, −2.47 to −0.32; P=0.03; I2=79.15%; Fig. 3). No significant reduction in the severity of dysmenorrhea was observed in the subset of trials that administered omega-3 fatty acids (SMD, 0.30; 95% CI, −0.16 to 0.76; P=0.96; I2=0%), resveratrol (SMD, −0.35; 95% CI, −0.94 to 0.23), or combination of antioxidants (SMD, −0.21; 95% CI, −0.51 to 0.09; P=0.69; I2=0%; Fig. 3). A significant reduction in dysmenorrhea was observed in the subset of trials that administered antioxidant supplementation for ≤8 weeks (SMD, −0.71; 95% CI, −1.22 to −0.21; P<0.001; I2=77.16%). Conversely, no significant reduction was observed in the subset of trials with antioxidant supplementation longer than 8 weeks (SMD, −0.13; 95% CI, −0.45 to 0.20; P=0.17; I2= 40.31%; Fig. 4). The sensitivity analysis indicated that the exclusion of any individual trial did not significantly alter the overall results of the meta-analysis, revealing the high stability of the results (Supplementary Fig. 1). A funnel plot depicts a triangular region centered on the pooled SMD, in which 95% CI of study findings should fall if there is no publication bias and no heterogeneity in the underlying true effects (Supplementary Fig. 2).

Fig. 3

Forest plot of the effect of antioxidants on dysmenorrhea pain. N, number; SD, standard deviation; CI, confidence interval; FA, fatty acid.

Fig. 4

Subgroup forest plot of the effect of antioxidants on dysmenorrhea pain. N, number; SD, standard deviation; CI, confidence interval.

4. The effect of antioxidants on dyspareunia

Total effectiveness was reported in four trials. Meta-analysis results suggested that there was no significant difference in terms of dyspareunia levels when comparing antioxidant supplementation and placebo (SMD, −0.47; 95% CI, −1.40 to 0.45; P=0.32; Fig. 5). High heterogeneity was detected between trials (I2= 91.61%; P<0.001). The sensitivity analysis indicated that excluding any individual trial did not significantly alter the overall results of the meta-analysis, revealing the high stability of the results (Supplementary Fig. 3).

Fig. 5

Forest plot of the effect of antioxidants on dyspareunia pain. N, number; SD, standard deviation; CI, confidence interval.

5. The effect of antioxidants on pelvic pain

Pelvic pain was recorded in four trials. Meta-analysis results suggested that antioxidant supplementation significantly improved endometriosis-associated pelvic pain (SMD, −1.51; 95% CI, −2.74 to −0.29; P=0.01; Fig. 6). High heterogeneity was detected between trials (I2=93.96%; P<0.001). Sensitivity analysis showed that the pooled SMD varied considerably with the omission of three trials; in particular, the exclusion of the study by Sesti et al. [26], Amini et al. [13], and Hoseinalizadeh and Cahichian. [20] which accounted for approximately 25.93%, 24.71%, and 24.30% of all weights in the meta-analysis, resulted in a pooled SMD (95% CI) of −1.45 (−3.20 to 0.29), −1.15 (−2.56 to 0.26), and −1.34 (−3.00 to 0.32), respectively (Supplementary Fig. 4).

Fig. 6

Forest plot of the effect of antioxidants on pelvic pain. N, number; SD, standard deviation; CI, confidence interval; REML, restricted maximum likelihood.

6. Quality of evidence

We were moderately confident in the outcomes of dysmenorrhea because of the uncertainty regarding the risk of bias. We had very little confidence in the outcomes of dyspareunia and pelvic pain because of some uncertainty regarding the risk of bias, inconsistency (high heterogeneity), and imprecision (non-significant results) (Supplementary Table 2).

Conclusion

Numerous studies have assessed the effectiveness of dietary and supplemental antioxidants for managing different types of pain in women with endometriosis. Previous systematic reviews and meta-analyses have reported inconsistent results. In line with our findings, systematic reviews support the positive effects of antioxidants in improving endometriosis-associated pain [16]. However, systematic reviews and meta-analyses have indicated that certain antioxidant vitamins, such as vitamin D, may not efficiently alleviate pain in patients [17]. Dysmenorrhea, dyspareunia, and chronic pelvic pain stand out as the most prevalent issues in women of reproductive age [27,28]. Our findings suggest that dietary and supplemental antioxidant intake is associated with a notable reduction in dysmenorrhea and chronic pelvic pain. However, our meta-analysis did not reveal a significant effect of antioxidant intake on dyspareunia in women with endometriosis.

Dietary antioxidants such as vitamin D, omega-3 fatty acids, and melatonin can have an interactive impact on decreasing cellular damage induced by oxidative stress and ROS [2931]. Dietary antioxidants can neutralize the ROS and oxidative damage associated with pain in endometriosis. The involvement of oxidative stress and related markers in the initiation and progression of endometriotic complications has been suggested in experimental studies [32]. Cultivated endometrial stromal cells were treated with antioxidants and oxidative stress factors. The application of antioxidant agents resulted in a dose-dependent suppression of cell growth. In contrast, control cells treated with oxidative stress factors exhibited increased endometrial stromal growth [33]. The most accepted mechanism for this effect is an antioxidant system that removes free radicals and functions through superoxide dismutases that remove the superoxide anion and glutathione peroxidase, which then removes hydrogen peroxide. In women with endometriosis, there is a diminished function of the antioxidant system activity [34].

It has been shown that the levels of most oxidative stress parameters are markedly enhanced in women with endometriosis compared with controls, and this is one of the main factors contributing to pain in these patients [35,36]. Several recent studies have shown that oxidative stress is positively associated with the migration and proliferation of endometrial cells in the peritoneal cavity, thereby increasing the probability of endometriosis and infertility [32,37]. The correlation between ROS generation and endometriosis is verified and widely investigated [38]. Moreover, it has been demonstrated that ROS and their reactive oxygen precursors are essential in the progression of pain in several etiologies [39]. Therefore, control of ROS leads to pain relief in patients with endometriosis, and it has been widely reported that dietary antioxidant supplements can effectively decrease ROS levels [40,41]. It was also observed that the increased ROS and enhanced proliferative potential in endometriotic cells were related to the full stimulation and elevated levels of phosphorylated endoplasmic reticulum kinase (ERK), as previously detected in tumor cells [42]. In addition, enhanced ERK phosphorylation has recently been reported in stromal cells of women with endometriosis [43]. Furthermore, there are links between ROS generation, ERK activation, and endometriotic cell proliferation [44]. It was also demonstrated that ERK activation is in response to different pro-inflammatory factors such as tumor necrosis factor-α and interleukin-1β [45,46]. These two pro-inflammatory markers are enhanced in the endometrium of women with endometriosis [47,48]. Pro-inflammatory markers have a pivotal role in the progression and development of pain in patients with endometriosis [49,50]. In addition, dietary antioxidant supplements have been shown to regulate pro-inflammatory cytokines efficiently [51].

Recent studies have also shown increased concentrations of other oxidative stress parameters in women with primary dysmenorrhea. Malondialdehyde (MDA) is an index of lipid peroxides [52]. MDA concentrations were higher in women with endometriosis than in healthy controls [53]. Lipid peroxide levels were higher in peritoneal fluid samples from women with endometriosis than in healthy controls [34]. Therefore, a decrease in MDA levels may help control cell proliferation in endometriosis, and it has been demonstrated that dietary antioxidants such as vitamin D and omega-3 fatty acids can decrease MDA levels [29,54].

Recent investigations have reported the elevated level of iron in different parts of the peritoneal cavity of women with endometriosis, including endometriotic lesions, macrophages, and peritoneal fluid, which basically proposes dysfunction of iron homeostasis in the peritoneal environment among this group of patients [55,56]. In women with endometriosis, elevated iron levels may result from the lysis of pelvic erythrocytes [57]. Retrograde menstruation leads to severe hemolysis of erythrocytes accompanied by a disruptive or overwhelmed peritoneal elimination system, which enhances iron levels in the peritoneal environment, leading to the progression and growth of endometrial cells [58,59]. Iron overload might have several cytotoxic effects because it reduces the equilibrium between free radical generation and the antioxidant system, which can cause oxidative stress [60]. Our subgroup analysis indicated that vitamin D and melatonin were more effective in decreasing dysmenorrhea in patients with endometriosis. It has been demonstrated that vitamin D [61] and melatonin can efficiently decrease iron toxicity complications [62,63].

The effectiveness of vitamin D in reducing dysmenorrhea in patients with endometriosis is thought to be mechanistically linked to its anti-inflammatory and immunomodulatory properties [64]. Endometriosis is characterized by endometrial-like tissue outside the uterus, causing inflammation and pain during menstruation [65,66]. Vitamin D receptors exist in various cells, including those involved in immune responses and inflammation [67]. By binding to these receptors, vitamin D may regulate the production of inflammatory mediators such as cytokines and modulate immune system activity [68]. This regulatory effect is believed to mitigate the heightened inflammatory response associated with endometriosis and subsequently reduce the severity of dysmenorrhea [69]. Although the precise mechanisms are still under investigation, emerging research suggests that maintaining optimal vitamin D levels may play a role in managing the pain and inflammation associated with endometriosis-related dysmenorrhea. On the other hand, melatonin, primarily recognized as a regulator of the sleep-wake cycle, also exhibits potent antioxidant properties [70]. Melatonin supplementation may be beneficial in the context of endometriosis-related dysmenorrhea, where oxidative stress is pivotal to exacerbating pain and inflammation [25]. Endometriosis is associated with increased ROS production, contributing to tissue damage and increased pain sensitivity [8,71]. Melatonin functions as a free radical scavenger and mitigates oxidative stress by neutralizing harmful molecules [72]. Therefore, the antioxidant effect of melatonin holds promise in reducing the severity of dysmenorrhea in patients with endometriosis [73]. By alleviating the oxidative burden, melatonin may contribute to the modulation of inflammatory responses and a subsequent decrease in pain perception, offering a supplementary avenue for managing the discomfort associated with endometriosis-related menstrual pain [74].

Additionally, in the same context, vitamins C and E, similar to vitamin D and melatonin, are recognized for their role in regulating iron levels, preventing excess, or addressing deficiency. This regulation aids in reducing oxidative stress because these vitamins function as antioxidants. However, perhaps due to the limited number of included studies that assessed the effectiveness of vitamins C and E, coupled with heterogeneous findings, we could not identify significant results for these vitamins. Therefore, further large-scale studies are required.

1. Strengths and limitations of the current review

This systematic review and meta-analysis has several limitations. The small number of included trials makes it difficult to draw a definitive conclusion regarding the effect of antioxidant supplements on dyspareunia in patients with endometriosis; therefore, these results should be interpreted cautiously. In addition, there are not enough trials in the literature to cover all dietary antioxidant supplements that may effectively reduce endometriosis-associated pain symptoms. Another limiting factor in the findings of this meta-analysis was the heterogeneity of the study populations and the types of antioxidants included in the trials. Some researchers failed to provide sufficient information on the selection criteria and data regarding where the trials were performed.

Dietary antioxidant supplementation seemed to have a beneficial effect on the severity of endometriosis-related dysmenorrhea (with an emphasis on vitamin D and melatonin) and pelvic pain. In contrast, no significant reduction in dyspareunia was observed. However, the obtained findings should be interpreted cautiously owing to the relatively small sample size and high heterogeneity between studies, and the importance of further well-designed clinical studies cannot be overstated.

Notes

Conflict of interest

All authors have no conflict of interest to declare.

Ethical approval

Not applicable.

Patient consent

Not applicable.

Funding information

None.

References

1. Gordts S, Koninckx P, Brosens I. Pathogenesis of deep endometriosis. Fertil Steril 2017;108:872–85e1.
2. Giudice LC. Clinical practice. Endometriosis. N Engl J Med 2010;362:2389–98.
3. Johnson NP, Hummelshoj L, Adamson GD, Keckstein J, Taylor HS, Abrao MS, et al. World Endometriosis Society consensus on the classification of endometriosis. Hum Reprod 2017;32:315–24.
4. Song H, Lee S, Kim MJ, Shin JE, Lee DW, Lee HN. Abdominal wall mass suspected of endometriosis: clinical and pathologic features. Obstet Gynecol Sci 2020;63:357–62.
5. Chapron C, Marcellin L, Borghese B, Santulli P. Rethinking mechanisms, diagnosis and management of endometriosis. Nat Rev Endocrinol 2019;15:666–82.
6. Taylor HS, Kotlyar AM, Flores VA. Endometriosis is a chronic systemic disease: clinical challenges and novel innovations. Lancet 2021;397:839–52.
7. Cho YJ, Yun JH, Kim SJ, Kwon HY. Nonpersistent endocrine disrupting chemicals and reproductive health of women. Obstet Gynecol Sci 2020;63:1–12.
8. Scutiero G, Iannone P, Bernardi G, Bonaccorsi G, Spadaro S, Volta CA, et al. Oxidative stress and endometriosis: a systematic review of the literature. Oxid Med Cell Longev 2017;2017:7265238.
9. Vafa M, Heshmati J, Sadeghi H, Shidfar F, Namazi N, Baradaran H, et al. Is exclusive breastfeeding and its duration related to cardio respiratory fitness in childhood? J Matern Fetal Neonatal Med 2016;29:461–5.
10. Lee HJ, Noh HK, Kim SC, Joo JK, Suh DS, Kim KH. Dietary pattern and risk of endometrioma in Korean women: a case-control study. Obstet Gynecol Sci 2021;64:99–106.
11. Morvaridzadeh M, Fazelian S, Agah S, Khazdouz M, Rahimlou M, Agh F, et al. Effect of ginger (zingiber officinale) on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Cytokine 2020;135:155224.
12. Namazi N, Heshmati J, Tarighat-Esfanjani A. Supplementation with Riboflavin (vitamin B2) for migraine prophylaxis in adults and children: a review. Int J Vitam Nutr Res 2015;85:79–87.
13. Amini L, Chekini R, Nateghi MR, Haghani H, Jamialahmadi T, Sathyapalan T, et al. The effect of combined vitamin C and vitamin E supplementation on oxidative stress markers in women with endometriosis: a randomized, triple-blind placebo-controlled clinical trial. Pain Res Manag 2021;2021:5529741.
14. Lete I, Mendoza N, de la Viuda E, Carmona F. Effectiveness of an antioxidant preparation with N-acetyl cysteine, alpha lipoic acid and bromelain in the treatment of endometriosis-associated pelvic pain: LEAP study. Eur J Obstet Gynecol Reprod Biol 2018;228:221–4.
15. Santanam N, Kavtaradze N, Murphy A, Dominguez C, Parthasarathy S. Antioxidant supplementation reduces endometriosis-related pelvic pain in humans. Transl Res 2013;161:189–95.
16. Zheng SH, Chen XX, Chen Y, Wu ZC, Chen XQ, Li XL. Antioxidant vitamins supplementation reduce endometriosis related pelvic pain in humans: a systematic review and meta-analysis. Reprod Biol Endocrinol 2023;21:79.
17. Kalaitzopoulos DR, Samartzis N, Daniilidis A, Leeners B, Makieva S, Nirgianakis K, et al. Effects of vitamin D supplementation in endometriosis: a systematic review. Reprod Biol Endocrinol 2022;20:176.
18. Abokhrais IM, Denison FC, Whitaker LHR, Saunders PTK, Doust A, Williams LJ, et al. A two-arm parallel double-blind randomised controlled pilot trial of the efficacy of Omega-3 polyunsaturated fatty acids for the treatment of women with endometriosis-associated pain (Pur-FECT1). PLoS One 2020;15:e0227695.
19. Almassinokiani F, Khodaverdi S, Solaymani-Dodaran M, Akbari P, Pazouki A. Effects of vitamin D on endometriosis-related pain: a double-blind clinical trial. Med Sci Monit 2016;22:4960–6.
20. Hoseinalizadeh H, Cahichian S. Evaluation of melatonin effect on pelvic pain in women with endometriosis referred to affiliated hospitals to Tehran Medical Sciences of Islamic Azad University. Med Sci J Islam Azad Univ 2018;28:277–82.
21. Lasco A, Catalano A, Benvenga S. Improvement of primary dysmenorrhea caused by a single oral dose of vitamin D: results of a randomized, double-blind, placebocontrolled study. Arch Intern Med 2012;172:366–7.
22. Mehdizadehkashi A, Rokhgireh S, Tahermanesh K, Eslahi N, Minaeian S, Samimi M. The effect of vitamin D supplementation on clinical symptoms and metabolic profiles in patients with endometriosis. Gynecol Endocrinol 2021;37:640–5.
23. Mendes da Silva D, Gross LA, Neto EPG, Lessey BA, Savaris RF. The use of resveratrol as an adjuvant treatment of pain in endometriosis: a randomized clinical trial. J Endocr Soc 2017;1:359–69.
24. Nodler JL, DiVasta AD, Vitonis AF, Karevicius S, Malsch M, Sarda V, et al. Supplementation with vitamin D or ω-3 fatty acids in adolescent girls and young women with endometriosis (SAGE): a double-blind, randomized, placebo-controlled trial. Am J Clin Nutr 2020;112:229–36.
25. Schwertner A, Conceição Dos Santos CC, Costa GD, Deitos A, de Souza A, de Souza IC, et al. Efficacy of melatonin in the treatment of endometriosis: a phase II, randomized, double-blind, placebo-controlled trial. Pain 2013;154:874–81.
26. Sesti F, Pietropolli A, Capozzolo T, Broccoli P, Pierangeli S, Bollea MR, et al. Hormonal suppression treatment or dietary therapy versus placebo in the control of painful symptoms after conservative surgery for endometriosis stage III–IV. A randomized comparative trial. Fertil Steril 2007;88:1541–7.
27. Kor E, Mostafavi SRS, Mazhin ZA, Dadkhah A, Kor A, Arvanagh SH, et al. Relationship between the severity of endometriosis symptoms (dyspareunia, dysmenorrhea and chronic pelvic pain) and the spread of the disease on ultrasound. BMC Res Notes 2020;13:546.
28. Sewell M, Churilov L, Mooney S, Ma T, Maher P, Grover SR. Chronic pelvic pain - pain catastrophizing, pelvic pain and quality of life. Scand J Pain 2018;18:441–8.
29. Sepidarkish M, Farsi F, Akbari-Fakhrabadi M, Namazi N, Almasi-Hashiani A, Maleki Hagiagha A, et al. The effect of vitamin D supplementation on oxidative stress parameters: a systematic review and meta-analysis of clinical trials. Pharmacol Res 2019;139:141–52.
30. Sepidarkish M, Akbari-Fakhrabadi M, Daneshzad E, Yavari M, Rezaeinejad M, Morvaridzadeh M, et al. Effect of omega-3 fatty acid plus vitamin E Co-supplementation on oxidative stress parameters: a systematic review and meta-analysis. Clin Nutr 2020;39:1019–25.
31. Morvaridzadeh M, Sadeghi E, Agah S, Nachvak SM, Fazelian S, Moradi F, et al. Effect of melatonin supplementation on oxidative stress parameters: a systematic review and meta-analysis. Pharmacol Res 2020;161:105210.
32. Van Langendonckt A, Casanas-Roux F, Donnez J. Oxidative stress and peritoneal endometriosis. Fertil Steril 2002;77:861–70.
33. Foyouzi N, Berkkanoglu M, Arici A, Kwintkiewicz J, Izquierdo D, Duleba AJ. Effects of oxidants and antioxidants on proliferation of endometrial stromal cells. Fertil Steril 2004;82:1019–22.
34. Szczepańska M, Koźlik J, Skrzypczak J, Mikołajczyk M. Oxidative stress may be a piece in the endometriosis puzzle. Fertil Steril 2003;79:1288–93.
35. Szmidt MK, Granda D, Sicinska E, Kaluza J. Primary dysmenorrhea in relation to oxidative stress and antioxidant status: a systematic review of case-control studies. Antioxidants (Basel) 2020;9:994.
36. Amreen S, Kumar P, Gupta P, Rao P. Evaluation of oxidative stress and severity of endometriosis. J Hum Reprod Sci 2019;12:40–6.
37. Ito F, Yamada Y, Shigemitsu A, Akinishi M, Kaniwa H, Miyake R, et al. Role of oxidative stress in epigenetic modification in endometriosis. Reprod Sci 2017;24:1493–502.
38. Carvalho LF, Samadder AN, Agarwal A, Fernandes LF, Abrão MS. Oxidative stress biomarkers in patients with endometriosis: systematic review. Arch Gynecol Obstet 2012;286:1033–40.
39. Hackel D, Pflücke D, Neumann A, Viebahn J, Mousa S, Wischmeyer E, et al. The connection of monocytes and reactive oxygen species in pain. PLoS One 2013;8:e63564.
40. Morvaridzadeh M, Sepidarkish M, Daneshzad E, Akbari A, Mobini GR, Heshmati J. The effect of pomegranate on oxidative stress parameters: a systematic review and meta-analysis. Complement Ther Med 2020;48:102252.
41. Fazelian S, Hoseini M, Namazi N, Heshmati J, Sepidar Kish M, Mirfatahi M, et al. Effects of L-arginine supplementation on antioxidant status and body composition in obese patients with pre-diabetes: a randomized controlled clinical trial. Adv Pharm Bull 2014;4:449–54.
42. Laurent A, Nicco C, Chéreau C, Goulvestre C, Alexandre J, Alves A, et al. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Cancer Res 2005;65:948–56.
43. Murk W, Atabekoglu CS, Cakmak H, Heper A, Ensari A, Kayisli UA, et al. Extracellularly signal-regulated kinase activity in the human endometrium: possible roles in the pathogenesis of endometriosis. J Clin Endocrinol Metab 2008;93:3532–40.
44. Ngô C, Chéreau C, Nicco C, Weill B, Chapron C, Batteux F. Reactive oxygen species controls endometriosis progression. Am J Pathol 2009;175:225–34.
45. Conde de la Rosa L, Schoemaker MH, Vrenken TE, Buist-Homan M, Havinga R, Jansen PL, et al. Superoxide anions and hydrogen peroxide induce hepatocyte death by different mechanisms: involvement of JNK and ERK MAP kinases. J Hepatol 2006;44:918–29.
46. Raymond L, Eck S, Mollmark J, Hays E, Tomek I, Kantor S, et al. Interleukin-1 beta induction of matrix metalloproteinase-1 transcription in chondrocytes requires ERK-dependent activation of CCAAT enhancer-binding protein-beta. J Cell Physiol 2006;207:683–8.
47. Lee SR, Kim SH, Lee HW, Kim YH, Chae HD, Kim CH, et al. Increased expression of glutathione by estradiol, tumor necrosis factor-alpha, and interleukin 1-beta in endometrial stromal cells. Am J Reprod Immunol 2009;62:352–6.
48. Richter ON, Dorn C, Rösing B, Flaskamp C, Ulrich U. Tumor necrosis factor alpha secretion by peritoneal macrophages in patients with endometriosis. Arch Gynecol Obstet 2005;271:143–7.
49. Kobayashi H, Yamada Y, Morioka S, Niiro E, Shigemitsu A, Ito F. Mechanism of pain generation for endometriosis-associated pelvic pain. Arch Gynecol Obstet 2014;289:13–21.
50. Malutan AM, Drugan T, Costin N, Ciortea R, Bucuri C, Rada MP, et al. Pro-inflammatory cytokines for evaluation of inflammatory status in endometriosis. Cent Eur J Immunol 2015;40:96–102.
51. Farsi F, Heshmati J, Keshtkar A, Irandoost P, Alamdari NM, Akbari A, et al. Can coenzyme Q10 supplementation effectively reduce human tumor necrosis factor-α and interleukin-6 levels in chronic inflammatory diseases? A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2019;148:104290.
52. Tsikas D. Assessment of lipid peroxidation by measuring malondialdehyde (MDA) and relatives in biological samples: analytical and biological challenges. Anal Biochem 2017;524:13–30.
53. Nasiri N, Moini A, Eftekhari-Yazdi P, Karimian L, Salman-Yazdi R, Arabipoor A. Oxidative stress statues in serum and follicular fluid of women with endometriosis. Cell J 2017;18:582–7.
54. Heshmati J, Morvaridzadeh M, Maroufizadeh S, Akbari A, Yavari M, Amirinejad A, et al. Omega-3 fatty acids supplementation and oxidative stress parameters: a systematic review and meta-analysis of clinical trials. Pharmacol Res 2019;149:104462.
55. Defrère S, Lousse JC, González-Ramos R, Colette S, Donnez J, Van Langendonckt A. Potential involvement of iron in the pathogenesis of peritoneal endometriosis. Mol Hum Reprod 2008;14:377–85.
56. Augoulea A, Alexandrou A, Creatsa M, Vrachnis N, Lambrinoudaki I. Pathogenesis of endometriosis: the role of genetics, inflammation and oxidative stress. Arch Gynecol Obstet 2012;286:99–103.
57. Van Langendonckt A, Casanas-Roux F, Donnez J. Iron overload in the peritoneal cavity of women with pelvic endometriosis. Fertil Steril 2002;78:712–8.
58. Lousse JC, Defrère S, Van Langendonckt A, Gras J, González-Ramos R, Colette S, et al. Iron storage is significantly increased in peritoneal macrophages of endometriosis patients and correlates with iron overload in peritoneal fluid. Fertil Steril 2009;91:1668–75.
59. Pirdel L, Pirdel M. Role of iron overload-induced macrophage apoptosis in the pathogenesis of peritoneal endometriosis. Reproduction 2014;147:R199–207.
60. Gazvani R, Templeton A. Peritoneal environment, cytokines and angiogenesis in the pathophysiology of endometriosis. Reproduction 2002;123:217–26.
61. Moon J. The role of vitamin D in toxic metal absorption: a review. J Am Coll Nutr 1994;13:559–64.
62. Ahammed GJ, Wu M, Wang Y, Yan Y, Mao Q, Ren J, et al. Melatonin alleviates iron stress by improving iron homeostasis, antioxidant defense and secondary metabolism in cucumber. Sci Hortic 2020;265:109205.
63. Othman AI, El-Missiry MA, Amer MA, Arafa M. Melatonin controls oxidative stress and modulates iron, ferritin, and transferrin levels in adriamycin treated rats. Life Sci 2008;83:563–8.
64. Sayegh L, Fuleihan Gel-H, Nassar AH. Vitamin D in endometriosis: a causative or confounding factor? Metabolism 2014;63:32–41.
65. Giampaolino P, Della Corte L, Foreste V, Bifulco G. Is there a relationship between vitamin D and endometriosis? An overview of the literature. Curr Pharm Des 2019;25:2421–7.
66. Yadav G, Rao M, Gothwal M, Singh P, Kathuria P, Sharma PP. Detection of nerve fibers in the eutopic endometrium of women with endometriosis, uterine fibroids and adenomyosis. Obstet Gynecol Sci 2021;64:454–61.
67. Colotta F, Jansson B, Bonelli F. Modulation of inflammatory and immune responses by vitamin D. J Autoimmun 2017;85:78–97.
68. Guillot X, Semerano L, Saidenberg-Kermanac’h N, Falgarone G, Boissier MC. Vitamin D and inflammation. Joint Bone Spine 2010;77:552–7.
69. Anastasi E, Fuggetta E, De Vito C, Migliara G, Viggiani V, Manganaro L, et al. Low levels of 25-OH vitamin D in women with endometriosis and associated pelvic pain. Clin Chem Lab Med 2017;55:e282–4.
70. Reiter RJ, Tan DX, Cabrera J, D'Arpa D, Sainz RM, Mayo JC, et al. The oxidant/antioxidant network: role of melatonin. Biol Signals Recept 1999;8:56–63.
71. Clower L, Fleshman T, Geldenhuys WJ, Santanam N. Targeting oxidative stress involved in endometriosis and its pain. Biomolecules 2022;12:1055.
72. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. J Pineal Res 2013;54:245–57.
73. Söderman L, Edlund M, Böttiger Y, Marions L. Adjuvant use of melatonin for pain management in dysmenorrhea - a randomized double-blinded, placebo-controlled trial. Eur J Clin Pharmacol 2022;78:191–96.
74. Sharifi M, Rajabpoor Nikoo N, Badehnoosh B, Shafabakhsh R, Asemi R, Reiter RJ, et al. Therapeutic effects of melatonin on endometriosis, targeting molecular pathways: current knowledge and future perspective. Pathol Res Pract 2023;243:154368.

Article information Continued

Fig. 1

The flow diagram of literature search and selection of studies. RCT, randomized controlled trials.

Fig. 2

The summary of risk of bias assessments.

Fig. 3

Forest plot of the effect of antioxidants on dysmenorrhea pain. N, number; SD, standard deviation; CI, confidence interval; FA, fatty acid.

Fig. 4

Subgroup forest plot of the effect of antioxidants on dysmenorrhea pain. N, number; SD, standard deviation; CI, confidence interval.

Fig. 5

Forest plot of the effect of antioxidants on dyspareunia pain. N, number; SD, standard deviation; CI, confidence interval.

Fig. 6

Forest plot of the effect of antioxidants on pelvic pain. N, number; SD, standard deviation; CI, confidence interval; REML, restricted maximum likelihood.

Table 1

The main characteristics of the included studies

Study Country Sample size (final analysis) Antioxidant type Dosage Duration (weeks) Age Endometriosis stage Pain type Pain assessment method

Intervention Placebo


Mean SD Mean SD
Sesti et al. (2007) [26] Italy 145 Mix - 24 29.1 3.9 31.1 4.1 III and I ↔ Dysmenorrhea
↔ Dyspareunia
Pelvic pain
VAS score

Lasco et al. (2012) [21] Italy 40 Vitamin D 300,000 IU 8 NM NM NM NM NM Dysmenorrhea VAS score

Schwertner et al. (2013) [25] Brazil 40 Melatonin 10 mg 8 36.76 6.4 37.63 5.5 All stages Dysmenorrhea VAS score

Mendes da Silva et al. (2017) [23] Brazil 44 Resveratrol 40 mg 6 35.4 7.1 32.4 7 NM ↔ Dysmenorrhea VAS score

Almassinokiani et al. (2016) [19] Iran 40 Vitamin D 50,000 IU/weekly 12 30.84 5.79 28.95 4.71 All stages ↔ Dysmenorrhea
↔ Pelvic pain
VAS score

Hoseinalizadeh and Cahichian (2018) [20] Iran 40 Melatonin 5 mg 8 NM NM NM NM III and IV Dysmenorrhea
Pelvic pain
VAS score

Abokhrais et al. (2020) [18] UK 30 Omega-3 1,000 mg/twice a day 8 35.43 8.57 36.08 9.59 All stages ↔ Dysmenorrhea
↔ Dyspareunia
-Pain
-Catastrophizing
-Questionnaire

Nodler et al. (2020) [24] USA 42 Omega-3 2,000 IU/day 24 20.01 2.7 20.1 3.5 All stages except stage III ↔ Dysmenorrhea VAS score

Amini et al. (2021) [13] Iran 60 Mix (C and E) - C, 1,000 mg/day
- E, 800 IU/day
8 35.7 5.71 38.03 6.47 I to III ↔ Dysmenorrhea
Dyspareunia
Pelvic pain
VAS score

Mehdizadehkashi et al. (2021) [22] Iran 60 Vitamin D 50,000 IU/each 2 weeks 12 34.8 7.1 35.6 7 NM Dysmenorrhoea
↔ Dyspareunia
VAS score

↓, this symbol is a sign of decreasing variables in the intervention group; ↔, this sign indicates no difference between the two groups.

SD, standard deviation; VAS, visual analog scale; IU, international unit; NM, not mentioned