Next Article in Journal
Breast Cancer Detection via Multi-Tiered Self-Contrastive Learning in Microwave Radiometric Imaging
Previous Article in Journal
Decoding the Puzzle of Male Infertility: The Role of Infection, Inflammation, and Autoimmunity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endometriosis in Adolescence: A Narrative Review of the Psychological and Clinical Implications

1
Department of Human Neuroscience, Sapienza University of Rome, 00185 Rome, Italy
2
Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy
3
Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
4
Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Diagnostics 2025, 15(5), 548; https://doi.org/10.3390/diagnostics15050548
Submission received: 23 January 2025 / Revised: 19 February 2025 / Accepted: 21 February 2025 / Published: 24 February 2025
(This article belongs to the Section Pathology and Molecular Diagnostics)

Abstract

:
Endometriosis is a chronic, inflammatory condition where endometrial-like tissue grows outside the uterus, affecting around 10% of women of reproductive age. This condition is associated with debilitating symptoms, including dysmenorrhea, dyspareunia, chronic pelvic pain, fatigue, and infertility. Adolescents with endometriosis face unique challenges, as the disease is often misdiagnosed or undiagnosed for an average of 7–10 years due to its complex and multifactorial nature. Consequently, patients frequently suffer from worsening symptoms and significant psychological distress, including anxiety, depression, and social withdrawal. While there is no definitive cure for endometriosis, treatment approaches typically involve hormonal therapies, lifestyle adjustments (such as diet and exercise), and psychological support. Recent studies emphasize the profound impact of endometriosis on the mental health of adolescents, highlighting the need for a more holistic treatment approach that integrates both medical and psychological care. This narrative review explores the psychological and psychosocial effects of endometriosis in adolescents, examining the biological and psychological mechanisms linking the disease to mental health outcomes. It also discusses current therapeutic strategies, such as cognitive behavioral therapy, mindfulness, and peer support, and underscores the importance of early diagnosis and multidisciplinary care to mitigate both the physical and emotional burdens of the condition. This integrated approach is critical in improving the overall well-being and quality of life for adolescents living with endometriosis.

1. Introduction

Endometriosis is an estrogen-dependent chronic inflammatory condition of unclear etiology, characterized by the presence of endometrium-like epithelium and/or stroma outside the uterus [1]. Primarily affecting women of reproductive age, it can begin as early as the onset of menarche during adolescence and may persist beyond menopause [2,3]. Although its exact prevalence remains unknown, it is estimated that approximately 10% of women and girls of reproductive age are affected, representing roughly 190 million individuals worldwide [4]. Around 67% of endometriosis patients report experiencing symptoms before the age of 20 [5]. The diagnostic and therapeutic pathway for adolescent endometriosis involves several critical steps, from symptom recognition to multidisciplinary follow-up (Figure 1). The most common symptom of endometriosis during adolescence is severe menstrual pain, which interferes with daily activities, social relationships, and school attendance and is poorly responsive to non-steroidal anti-inflammatory drugs and/or oral contraceptives [6,7,8]. Other common symptoms include chronic pelvic pain (CPP), dyspareunia, dysuria, dysmenorrhea, abnormal menstruation, infertility, and gastrointestinal discomfort [9]. On average, patients spend about 10 years searching for a diagnosis.
During adolescence, the diagnosis of endometriosis is often delayed, and the risk of misdiagnosis is high due to the overlap of symptoms with normal menstruation, the varied presentation in younger patients, and the lack of awareness in the medical community [10,11,12]. The prevalence of endometriosis in adolescence varies depending on the study in the literature, the population being examined, and the diagnostic methods used. However, it remains high and represents a significant issue in this age group (Table 1). A definitive diagnosis can be made through laparoscopy; however, given the well-recognized delays in diagnosis, a clinical diagnosis focusing on history, physical examination, and imaging has proven to be a valuable adjunct to diagnosis [13,14,15]. Adenomyosis, historically considered a disease of multiparous women, is increasingly recognized in adolescents, often in conjunction with endometriosis. It is characterized by the ectopic presence of endometrial glands and stroma within the myometrium, leading to dysmenorrhea, menorrhagia, and chronic pelvic pain—symptoms overlapping with those of endometriosis [16]. Despite the limited availability of pediatric-specific data, studies suggest that adenomyosis exacerbates pain severity in adolescents diagnosed with endometriosis. The preferred diagnostic approach in young patients relies on transvaginal or transabdominal ultrasound (US) and magnetic resonance imaging (MRI). Since adenomyosis often coexists with endometriosis, therapeutic strategies largely overlap. However, data on adenomyosis in adolescents remain limited, as most studies focus on adult women [17]. Deep infiltrating endometriosis (DIE) is a severe form of endometriosis with deep infiltration below the peritoneum, commonly affecting the rectovaginal space, bladder, and uterosacral ligaments. Though more frequent in adults, early-stage DIE can occur in adolescents, making early detection crucial [18]. Symptoms include severe dysmenorrhea, bowel dysfunction, dysuria, and deep dyspareunia. MRI is the preferred imaging tool, offering better detection of deep nodules compared to ultrasound. While there is a strong overlap with endometriosis, DIE is more aggressive and often needs a multidisciplinary approach with targeted surgery in order to ensure long-term symptom control and fertility preservation in adolescents [18]. Several treatment options are available for endometriosis, including both surgical and pharmacological approaches, although a definitive cure is still lacking [19]. Treatment responses vary among individuals, with some patients experiencing persistent or recurring symptoms that are debilitating [20,21]. The recent literature suggests that endometriosis negatively impacts the mental well-being of adult patients, affecting their work environment, family relationships, social life, and self-esteem, and contributing to depression and anxiety [22,23,24,25,26,27].
Adolescence is a critical period for emotional and psychological development, and the onset of a chronic illness such as endometriosis can significantly affect adolescents’ well-being and social life. This narrative review was conducted to synthesize current knowledge on the psychological and clinical implications of endometriosis in adolescence.

2. Methods

For this narrative review, we conducted an electronic literature search using MEDLINE/PubMed, Scopus, and Web of Science databases to identify articles on endometriosis in adolescence published between January 1999 and January 2025. The following keywords and Medical Subject Headings (MeSH) terms were used in various combinations to identify relevant studies: “Endometriosis” (unique ID: D004715), “Adolescent” (unique ID: D000293), “Mental health” (unique ID: D008603), “Quality of life” (unique ID: D011788), “Diagnosis” (unique ID: D003933), “Therapy” (unique ID: D013812), and “Ultrasonography” (unique ID: D014463). Eligible studies included original research articles (randomized and non-randomized clinical trials, prospective observational studies, retrospective cohort studies, and case–control studies); review articles focusing on the psychological and clinical aspects of endometriosis in adolescence; and studies evaluating diagnostic and treatment strategies, including psychological interventions. Exclusion criteria were as follows: non-English-language manuscripts; articles lacking detailed information on diagnostic or treatment approaches relevant to endometriosis, in particular for adolescence; and conference abstracts, letters to the editor, and case reports. The literature selection was conducted independently by two authors (F.Pa. and R.P.) who screened and reviewed all the studies meeting the inclusion criteria. A total of 215 articles were included, covering pathogenetic theories, epidemiology, psychological implications, imaging-based diagnosis, and treatment options, providing a comprehensive overview of endometriosis in adolescence.

3. Integrated Insights into Endometriosis and Emotional Well-Being

3.1. Psychological Impact on Adolescence

Endometriosis symptoms, primarily chronic severe pelvic pain and dysmenorrhea, which are often severe and unpredictable, can significantly affect the daily life and basic functioning of adolescents [35,36]. Severe dysmenorrhea, in particular, has been associated with an increased risk of absenteeism and the need for pain medication [3,37,38,39,40]. Absenteeism rates are higher in adolescent patients who also report gastrointestinal symptoms, such as vomiting and diarrhea during menstruation [40]. Furthermore, dysmenorrhea has been shown to significantly decrease subjective sleep quality, sleep efficiency, and rapid eye movement due to elevated nocturnal body temperatures and higher morning estrogen concentrations [41]. Sleep disturbances further contribute to reduced quality of life in these patients, exacerbating the impact of pain on daytime functioning [42].
CPP and severe dysmenorrhea have been correlated with negative perceptions of general health and psychological distress in adolescents, who also experience high rates of somatization [3,43]. CPP is the primary driver of anxiety and depressive symptoms [44], and the presence of a mental health condition may exacerbate the perception of pain [24,45]. Adolescents with CPP are more likely to experience moderate-to-severe mood disturbances compared to their peers without chronic pain conditions [43,46].
Adolescents with endometriosis face various physical, hormonal, and emotional changes associated with growth. A diagnosis of a chronic, painful condition such as endometriosis can disrupt this critical developmental phase [47,48]. The negative impact of endometriosis on school attendance, academic performance, and social engagement can adversely affect self-esteem and self-confidence, potentially leading to emotional dysregulation and long-lasting psychological effects [2]. This association may be more pronounced in individuals reporting a higher symptom burden [46,49]. Decreased social interaction and withdrawal from activities can be attributed to factors such as pain, bleeding, fatigue, depression, feelings of isolation, body image dissatisfaction, low self-esteem, and lack of self-confidence. These factors correlate with diminished perceived social support and lower self-rated emotional well-being [49].
Increased emotional vulnerability leads to difficulties coping with the illness, as well as feelings of frustration, helplessness, anger, and depression due to adolescents’ inability to understand their condition and its progression [50,51]. Girls with endometriosis often experience intense feelings of worthlessness, guilt, and frustration due to disease-related limitations on daily activities, social functioning, independence, and interpersonal relationships [52,53,54]. Negative emotions may be exacerbated by social stigma and the belief that others, including healthcare professionals, perceive their symptoms as “all in their heads”, normalize their pain as part of being a woman, or attribute it to psychological causes [55,56,57,58,59]. The societal normalization of menstrual pain often leads adolescents to hide their symptoms, feeling misunderstood and unrecognized in their struggle [59].

3.2. Interplay of Biological Mechanisms Between Endometriosis and Mental Health

Hormonal factors, immunological dysregulation, and chronic inflammation, influenced by both hereditary and environmental factors, play a central role in the pathogenesis of endometriosis. Patients with endometriosis often exhibit elevated levels of estradiol and progesterone [60]. The increased synthesis of estrogen, combined with progesterone resistance and hyperexpression of estrogen receptors, promotes the proliferation and suppresses apoptosis of endometriotic lesions [61]. This hormonal dysregulation also affects the central nervous system (CNS), particularly influencing the synthesis of brain-derived neurotrophic factor (BDNF) [62], as well as other neurotransmitters such as serotonin, increasing the vulnerability to mood disorders in women with endometriosis.
BDNF plays a critical role in the development, survival, and plasticity of neurons in the CNS. Its concentration increases progressively throughout adolescence [63,64,65,66]. Furthermore, BDNF is involved in the regulation and transmission of nociceptive signals in chronic pain syndromes, contributing to the hyperexcitability of spinal nerves and the persistence of pain [67]. Chronic pain, as previously mentioned, is a primary contributor to the development of anxiety and depression in adolescents with endometriosis [68]. Although BDNF is primarily synthesized in the brain, elevated levels of BDNF have been detected in the plasma, peritoneal fluid, and endometrial tissue of women with endometriosis compared to healthy controls, sustained by the local interaction of inflammatory factors and estrogen [69,70,71,72].
Endometriotic lesions are surrounded by an inflammatory microenvironment, driven by cytokines and chemokines (e.g., tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β) that promote the growth, invasion, and survival of ectopic endometrial cells. This inflammatory milieu not only contributes to tissue damage but also fosters the persistence and expansion of lesions. The microenvironment, rich in immune cells, cytokines, and extracellular matrix components, supports the local immune response, which may lead to fibrosis, scarring, and the formation of adhesions. In this setting, immune cells such as T cells, B cells, and natural killer cells fail to properly recognize and eliminate ectopic endometrial cells, while macrophages and dendritic cells are overactivated, thereby promoting sustained inflammation. Elevated levels of pro-inflammatory cytokines, including ILs and TNF-α, are linked to increased pelvic pain and other symptoms of endometriosis and may also affect brain function, contributing to the development of mood disturbances [73,74].
In the early stages of the disease, endometriotic cell survival is supported by altered energy metabolism, which is characterized by the activation of aerobic glycolysis pathways [75]. Another significant factor in the pathogenesis of endometriosis is dysbiosis in the gut, vaginal, and uterine microbiomes [76,77,78,79,80,81]. These microbiomes are typically characterized by reduced microbial diversity and an overabundance of Gram-negative bacteria, including genera such as Shigella and Escherichia, as well as Prevotella species [82,83,84]. Protective microbes in the gut, such as Clostridia, Ruminococcus, and Lachnospiraceae, are diminished in women with endometriosis, leading to a reduction in short-chain fatty acids, which are important for maintaining intestinal integrity [85]. Altered epithelial permeability causes bacterial translocation, which serves as a trigger for inflammation, activating both local and systemic immune responses [86].
Pro-inflammatory cytokines such as IL-6, TNF-α, and IL-1β have been linked to depression, anxiety, and decreased quality of life [87,88,89]. These cytokines are particularly elevated in patients with gastrointestinal symptoms such as constipation, bloating, flatulence, vomiting, and nausea [82]. The gut microbiome and vaginal microbiome are interconnected, and microbial dysbiosis in one area can influence microbial balance in the other [73]. Moreover, the microbiome plays a role in modulating pain pathways through the gut–brain axis [90], influencing microglial and astrocytic activity, which leads to increased glutamate levels and decreased gamma-aminobutyric acid levels in central synaptic neurotransmission. This imbalance contributes to pain hypersensitivity and its persistence [91,92].
Finally, the role of the microbiome is bidirectional as it is influenced by mental health and pain perception while also influencing them [93]. Gut inflammation can stimulate the afferent vagal nerve, which in turn affects the hypothalamic–pituitary–adrenal axis, microglial activation, and the kynurenine pathway, inducing symptoms associated with depression such as fatigue, decreased motivation, anhedonia, cognitive impairment, and sleep disruption [94].

3.3. Non-Invasive Diagnostic Biomarkers and Digital Tools for Monitoring and Support

Advancements in non-invasive diagnostic biomarkers and digital health technologies are transforming the care of adolescents with endometriosis. The traditional reliance on invasive procedures such as laparoscopy has delayed diagnosis for many, highlighting the urgent need for alternative methods [95,96]. US is the first-line imaging modality for diagnosing adolescent endometriosis, particularly in detecting ovarian endometriomas. Transvaginal US is widely used in adults [97], but in adolescents, transabdominal or transrectal US may be preferable for virgin patients. US can detect endometriomas with high specificity but has limited sensitivity for detecting DIE [31]. MRI is a valuable adjunct in evaluating deep endometriotic lesions, particularly in cases of rectovaginal and bladder involvement. High-resolution imaging is essential for pre-surgical assessment, guiding minimally invasive approaches to avoid excessive ovarian tissue loss [98]. Thus, a multimodal imaging approach—US as a first step, followed by MRI if needed—is recommended to improve early diagnosis and treatment planning in adolescent patients. Non-invasive biomarkers are emerging as promising tools, particularly blood-based markers such as Cancer Antigen 125, which, while widely studied, have limited specificity in early or mild cases [99]. More promising are microRNAs (miRNAs), small non-coding RNA molecules that regulate gene expression and remain stable in blood samples. Certain miRNAs, such as the miR-200 family, show significant potential for early diagnosis, offering a non-invasive alternative for identifying endometriosis [100,101]. Proteomic and metabolomic analyses are further advancing this field, identifying inflammatory markers such as IL-6 and TNF-α [102] as well as altered metabolic profiles in endometriosis patients [103,104,105]. These biomarkers could lead to the development of simple blood or urine tests that detect the condition with greater accuracy. Menstrual fluid also presents an accessible, non-invasive diagnostic medium, as studies have identified unique genetic, epigenetic, and protein signatures in menstrual effluent from patients with endometriosis [106,107]. The application of next-generation sequencing technologies enables comprehensive analysis of these samples, offering an alternative to invasive tissue biopsy [108]. Additionally, exosomes and extracellular vesicles, which are released by cells into bodily fluids and carry proteins, lipids, and nucleic acids, are being studied for their diagnostic potential in endometriosis. These small vesicles contain distinct molecular signatures that could serve as reliable biomarkers for the disease [109,110]. Combining multiple biomarkers into diagnostic panels, such as integrating miRNAs, cytokines, and metabolic profiles, may improve the sensitivity and specificity of non-invasive testing, particularly for adolescents (Table 2).
Alongside biomarker development, digital tools are reshaping how endometriosis is monitored and managed, addressing both the physical and psychological challenges of the disease [139]. Mobile health applications allow adolescents to track symptoms such as pain, menstrual patterns, and emotional well-being. These apps provide real-time data to clinicians, enabling more personalized treatment adjustments, while also offering educational resources to improve disease literacy and reduce stigma [140,141]. Wearable devices such as smartwatches and fitness trackers enhance this capability by monitoring physiological parameters such as sleep quality, stress levels, and activity patterns. For instance, heart rate variability data collected by these devices can provide indirect insights into pain severity and emotional distress, supporting targeted interventions [142]. Telemedicine has emerged as a critical tool for improving access to care, particularly for adolescents who may face barriers such as geographic isolation or discomfort discussing symptoms in traditional settings. Virtual consultations enable regular follow-ups and facilitate multidisciplinary collaboration among healthcare providers [143,144]. Digital communities and online support groups further extend this support by connecting adolescents with peers who share similar experiences, reducing feelings of isolation and providing emotional validation [145,146].
Artificial intelligence (AI) is increasingly being integrated into endometriosis care, with machine learning algorithms analyzing symptom patterns, imaging data, and biomarker profiles to predict disease likelihood. AI-powered chatbots provide adolescents with real-time guidance on managing symptoms and understanding treatment options [147,148]. Virtual reality (VR) offers another innovative avenue for care, particularly in pain management. Immersive VR experiences distract patients from chronic pain, reduce stress, and promote relaxation. Preliminary studies suggest that VR interventions can improve both physical and psychological outcomes, making them a valuable complement to traditional treatments [149]. The integration of these digital tools with emerging biomarkers has the potential to transform clinical practice. Collaborative efforts between researchers, clinicians, and technology developers are essential to validate and optimize these advancements, ensuring that they are accessible to all patients. By combining these innovations with traditional medical and psychological interventions, healthcare providers can offer more personalized, effective, and holistic care, addressing the unique challenges of adolescents with endometriosis.

3.4. Psychological and Mind–Body Interventions

Medical treatments for endometriosis typically involve non-steroidal anti-inflammatory drugs for pain management and hormonal therapies aimed at reducing estrogen levels and controlling menstrual cycles. Surgery for ovarian endometriosis should be carefully considered in adolescents; conservative surgical techniques with long-term hormonal suppression provide the best symptom control while preserving fertility (Table 3).
While these treatments can alleviate physical symptoms, their impact on mental health remains less clear, especially in adolescents. Some studies suggest that hormonal therapies may help reduce depressive and anxiety symptoms by alleviating pain [45], whereas others highlight potential side effects, such as mood swings, fatigue, and decreased libido, which could negatively affect psychological well-being [179]. Furthermore, while hormonal treatments help control symptoms [179], they may not directly address the emotional and psychological challenges of living with a chronic condition such as endometriosis [45].
The timely diagnosis of endometriosis in adolescents is paramount in preventing long-term psychological and physical sequelae. Delays in diagnosis—averaging 7–10 years—are often attributed to the normalization of menstrual pain, inadequate awareness among healthcare providers, and the challenges in differentiating endometriosis from other pelvic pain conditions in younger patients [180,181]. Prolonged diagnostic timelines increase the likelihood of disease progression, worsening symptoms, and the accumulation of negative psychological effects such as chronic anxiety, social withdrawal, and depressive episodes [182,183]. Early recognition and diagnosis allow for prompt intervention, mitigating pain and preventing the cascading effects of chronic illness on adolescents’ education, social development, and emotional health [184,185].
Personalized care models, integrating both medical and psychological treatments, emerge as essential strategies for managing endometriosis in adolescence. Tailoring hormonal therapies, such as combined oral contraceptives or progestins, to the patient’s symptomatology and medical history is critical for optimizing symptom control while minimizing side effects that may exacerbate mood disturbances [152,186]. Additionally, the integration of psychological therapies, such as cognitive behavioral therapy (CBT), addresses the psychosocial burden of the disease by improving pain management, coping skills, and emotional resilience [187,188]. Adolescents diagnosed early are also better candidates for educational interventions, allowing healthcare providers to enhance their understanding of endometriosis and empower them to advocate for their care [181].
Moreover, multidisciplinary care involving pediatricians, gynecologists, psychologists, and pain specialists is increasingly recognized as a gold standard for improving outcomes. This team-based approach facilitates comprehensive management, combining physical treatments with emotional and social support to address the multifaceted nature of endometriosis [181,183].
Future research should focus on refining diagnostic algorithms and exploring innovative non-invasive diagnostic biomarkers to expedite diagnosis in younger populations [189,190]. By identifying adolescents at high risk of endometriosis early and implementing individualized, holistic treatment plans, the long-term mental and physical impact of the disease can be significantly reduced, improving quality of life and psychological well-being [184,191].
Given the strong association between endometriosis and mental health issues, early psychological and mind–body interventions have demonstrated effectiveness in reducing both pain and symptoms of anxiety and depression in adolescents [192]. Specifically, CBT is particularly effective in addressing both the psychological and physical symptoms of endometriosis, helping patients manage pain and improve emotional well-being [193,194].
CBT, which focuses on identifying and changing negative thought patterns, helps adolescents cope with the emotional distress associated with chronic pain and illness [193,194]. Mindfulness-based interventions, including relaxation techniques and mindfulness meditation, have been shown to improve emotional regulation and mental health and reduce pain perception, anxiety, and depression symptoms [195,196].
Yoga has also been highlighted as a promising intervention for adolescents with endometriosis. A study conducted by Gonçalves et al. demonstrated that yoga could improve sleep quality and reduce pain intensity, offering a non-pharmacological complement to traditional treatments [197]. A wide range of studies examined the impact of dietary interventions on managing endometriosis symptoms, consistently suggesting that dietary changes can be beneficial in alleviating pain and other related symptoms (Table 4). Interventions commonly explored include antioxidant-rich diets, vitamin D supplementation, gluten-free diets, and omega-3/6 fatty acids, as well as specific dietary approaches such as low-FODMAP and low-nickel diets [198,199].
While most studies report significant reductions in pain and symptom severity, the generalizability of these findings may be limited due to factors such as high heterogeneity, small sample sizes, and varying methodologies. Overall, the growing body of literature supports the potential of dietary modifications as a complementary strategy in managing endometriosis symptoms, although further research is needed to confirm optimal dietary patterns and interventions.
Finally, educating family members and the broader community about the psychological impact of endometriosis and promoting open communication is essential in reducing feelings of isolation and stigma. The chronic nature of the disease, combined with its often invisible symptoms, can lead to social withdrawal and a lack of understanding from family members and peers [43]. Peer support groups, whether in person or online, can offer adolescents a sense of community and validation, helping them cope with the emotional challenges associated with the disease [200].
These groups foster connections with others who understand their experiences, which is particularly beneficial for reducing the social and psychological burden of the disease. Furthermore, promoting mental health awareness within the community and among healthcare providers can ensure that adolescents receive comprehensive care that addresses both physical and emotional needs [201].
Table 4. Dietary interventions and their effects on endometriosis symptoms.
Table 4. Dietary interventions and their effects on endometriosis symptoms.
CategoryDiet/Nutritional FactorKey ComponentsReported Effects on Endometriosis
Specific dietsMediterranean diet
(Ott et al., 2012 [202])
Rich in olive oil, fish, nuts, legumes, fruits, and vegetablesLinked to lower endometriosis risk, reduced pelvic pain, and inflammation
Low-FODMAP diet
(Moore et al., 2017 [203]; van Haaps et al., 2023 [204])
Excludes fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs)Reduces gastrointestinal and abdominal symptoms common in endometriosis (especially if associated with irritable bowel syndrome)
Low-nickel diet
(Borghini et al., 2020 [205])
Excludes foods that contain a high amount of nickelMay be recommended to reduce gastrointestinal, extra-intestinal, and gynecological symptoms (especially if nickel allergic contact mucositis is present)
Gluten-free diet
(Marziali et al., 2012 [206])
Excludes any foods that contain glutenMay reduce pain in gluten-sensitive individuals with endometriosis
Specific nutrientsOmega-3 fatty acids
(Nodler et al., 2020 [207])
Found in fatty fish, flaxseed, and walnutsReduces inflammation, pain, and prostaglandin production
Antioxidants
(Mier-Cabrera et al., 2009 [208]; Santanam et al., 2013 [209])
Vitamins C and E, selenium, and zincMitigates oxidative stress, reduces pain, and enhances symptom relief
Vitamin D
(Nodler et al., 2020 [207]; Qiu et al., 2020 [210])
Found in fortified foods, sunlight exposure, and fatty fishRegulates immune function. There is a negative relationship between vitamin D levels and the severity of endometriosis
Food groupsDairy products
(Harris et al., 2013 [211]; Nodler et al., 2020 [212]; Qi et al., 2021 [213])
Milk, yogurt, and cheeseAn optimal intake of total dairy may be associated with a decreased risk of endometriosis
Red meat
(Parazzini et al., 2004 [214]; Yamamoto et al., 2018 [215])
High intake of processed and red meatsAssociated with a higher risk of developing endometriosis and worsened symptoms
Soy products
(Tsuchiya et al., 2007 [216])
Isoflavones (phytoestrogens) found in soy milk and tofu Higher urinary levels of isoflavones may be associated with a reduced risk of advanced but not minimal–mild-stage endometriosis
Dietary fibers
(Parazzini et al., 2004 [214]; Harris et al., 2018 [217])
Fruits and vegetablesReduction in the risk of endometriosis with higher consumption of (not all, some) fruits and vegetables

4. Conclusions

This narrative review provides a comprehensive and multidisciplinary perspective on adolescent endometriosis, integrating clinical, psychological, and emerging biomarker-based insights. One of its major strengths lies in the broad scope of the discussion, which encompasses not only traditional medical and surgical management but also the psychosocial impact of the disease. By addressing the interplay between chronic pain, hormonal dysregulation, and mental health, this review highlights the need for a holistic, patient-centered approach to treatment. Furthermore, the inclusion of non-invasive diagnostic strategies, digital health tools, and psychological interventions offers an innovative outlook on emerging trends in the management of adolescent endometriosis. This broad perspective is particularly valuable given the complex nature of the disease and the frequent diagnostic delays experienced by young patients.
However, as a narrative review, this study does not include a quantitative meta-analysis of treatment outcomes, which could have provided statistical comparisons of therapeutic efficacy. The heterogeneity of included studies—varying in population size, diagnostic criteria, and treatment approaches—may introduce bias and limit the generalizability of findings. Additionally, much of the available literature on endometriosis treatment is derived from adult populations, making it challenging to draw definitive conclusions about the safety and efficacy of specific interventions in adolescents. Longitudinal studies focused on adolescent-specific outcomes are needed to further clarify the long-term effects of hormonal therapies and the impact of psychological interventions on disease progression and quality of life.
Despite these limitations, this review underscores the importance of early diagnosis, personalized treatment, and a multidisciplinary care model. By synthesizing current knowledge and identifying areas for future research, it serves as a valuable resource for clinicians, researchers, and policymakers striving to improve outcomes for adolescents with endometriosis.

Author Contributions

Conceptualization, F.P. (Fabiola Panvino) and R.P.; methodology, F.P. (Fabiola Panvino) and R.P.; writing—original draft preparation, F.P. (Fabiola Panvino) and R.P.; writing—review and editing, F.P. (Francesco Pisani), F.T., G.F., M.F., I.A. and L.T.; supervision, F.P. (Francesco Pisani), F.T., G.F., M.F., I.A. and L.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank the IBBC-CNR and the Sapienza University of Rome in Rome, Italy.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tomassetti, C.; Johnson, N.P.; Petrozza, J.; Abrao, M.S.; Einarsson, J.I.; Horne, A.W.; Lee, T.T.; Missmer, S.; Vermeulen, N.; Zondervan, K.T.; et al. An International Terminology for Endometriosis, 2021. J. Minim. Invasive Gynecol. 2021, 28, 1849–1859. [Google Scholar] [CrossRef] [PubMed]
  2. Brosens, I.; Gordts, S.; Benagiano, G. Endometriosis in adolescents is a hidden, progressive and severe disease that deserves attention, not just compassion. Hum. Reprod. 2013, 28, 2026–2031. [Google Scholar] [CrossRef] [PubMed]
  3. Suvitie, P.A.; Hallamaa, M.K.; Matomäki, J.M.; MäFkinen, J.I.; Perheentupa, A.H. Prevalence of Pain Symptoms Suggestive of Endometriosis Among Finnish Adolescent Girls (TEENMAPS Study). J. Pediatr. Adolesc. Gynecol. 2016, 29, 97–103. [Google Scholar] [CrossRef]
  4. Zondervan, K.T.; Becker, C.M.; Missmer, S.A. Endometriosis. N. Engl. J. Med. 2020, 382, 1244–1256. [Google Scholar] [CrossRef]
  5. American College of Obstetricians and Gynecologists. ACOG Committee Opinion #310: Endometriosis in Adolescents. Obstet. Gynecol. 2005, 105, 921–928. [Google Scholar] [CrossRef]
  6. Parker, M.; Sneddon, A.; Arbon, P. The menstrual disorder of teenagers (MDOT) study: Determining typical menstrual patterns and menstrual disturbance in a large population-based study of Australian teenagers. BJOG Int. J. Obs. Gynaecol. 2010, 117, 185–192. [Google Scholar] [CrossRef] [PubMed]
  7. Harel, Z. Dysmenorrhea in adolescents and young adults: An update on pharmacological treatments and management strategies. Expert. Opin. Pharmacother. 2012, 13, 2157–2170. [Google Scholar] [CrossRef] [PubMed]
  8. Micangeli, G.; Paparella, R.; Tarani, F.; Menghi, M.; Ferraguti, G.; Carlomagno, F.; Spaziani, M.; Pucarelli, I.; Greco, A.; Fiore, M.; et al. Clinical Management and Therapy of Precocious Puberty in the Sapienza University Pediatrics Hospital of Rome, Italy. Children 2023, 10, 1672. [Google Scholar] [CrossRef] [PubMed]
  9. Bulun, S.E.; Yilmaz, B.D.; Sison, C.; Miyazaki, K.; Bernardi, L.; Liu, S.; Kohlmeier, A.; Yin, P.; Milad, M.; Wei, J. Endometriosis. Endocr. Rev. 2019, 40, 1048–1079. [Google Scholar] [CrossRef] [PubMed]
  10. Hudelist, G.; Fritzer, N.; Thomas, A.; Niehues, C.; Oppelt, P.; Haas, D.; Tammaa, A.; Salzer, H. Diagnostic delay for endometriosis in Austria and Germany: Causes and possible consequences. Hum. Reprod. 2012, 27, 3412–3416. [Google Scholar] [CrossRef] [PubMed]
  11. American College of Obstetricians and Gynecologists. ACOG Committee Opinion No. 760: Dysmenorrhea and Endometriosis in the Adolescent. Obs. Gynecol. 2018, 132, e249–e258. [Google Scholar] [CrossRef] [PubMed]
  12. Greene, R.; Stratton, P.; Cleary, S.D.; Ballweg, M.L.; Sinaii, N. Diagnostic experience among 4,334 women reporting surgically diagnosed endometriosis. Fertil. Steril. 2009, 91, 32–39. [Google Scholar] [CrossRef] [PubMed]
  13. Agarwal, S.K.; Chapron, C.; Giudice, L.C.; Laufer, M.R.; Leyland, N.; Missmer, S.A.; Singh, S.S.; Taylor, H.S. Clinical diagnosis of endometriosis: A call to action. Am. J. Obs. Gynecol. 2019, 220, 354.e1–354.e12. [Google Scholar] [CrossRef] [PubMed]
  14. Shim, J.Y.; Laufer, M.R.; King, C.R.; Lee, T.T.M.; Einarsson, J.I.; Tyson, N. Evaluation and Management of Endometriosis in the Adolescent. Obstet. Gynecol. 2023, 143, 44–51. [Google Scholar] [CrossRef] [PubMed]
  15. Pascoal, E.; Wessels, J.M.; Aas-Eng, M.K.; Abrao, M.S.; Condous, G.; Jurkovic, D.; Espada, M.; Exacoustos, C.; Ferrero, S.; Guerriero, S.; et al. Strengths and limitations of diagnostic tools for endometriosis and relevance in diagnostic test accuracy research. Ultrasound Obs. Gynecol. 2022, 60, 309–327. [Google Scholar] [CrossRef] [PubMed]
  16. Zannoni, L.; DEL Forno, S.; Raimondo, D.; Arena, A.; Giaquinto, I.; Paradisi, R.; Casadio, P.; Meriggiola, M.C.; Seracchioli, R. Adenomyosis and endometriosis in adolescents and young women with pelvic pain: Prevalence and risk factors. Minerva Pediatr. 2024, 76, 57–63. [Google Scholar] [CrossRef]
  17. Martire, F.G.; D’abate, C.; Schettini, G.; Cimino, G.; Ginetti, A.; Colombi, I.; Cannoni, A.; Centini, G.; Zupi, E.; Lazzeri, L. Adenomyosis and Adolescence: A Challenging Diagnosis and Complex Management. Diagnostics 2024, 14, 2344. [Google Scholar] [CrossRef] [PubMed]
  18. Martire, F.G.; Giorgi, M.; D’abate, C.; Colombi, I.; Ginetti, A.; Cannoni, A.; Fedele, F.; Exacoustos, C.; Centini, G.; Zupi, E.; et al. Deep Infiltrating Endometriosis in Adolescence: Early Diagnosis and Possible Prevention of Disease Progression. J. Clin. Med. 2024, 13, 550. [Google Scholar] [CrossRef] [PubMed]
  19. Kocas, H.D.; Rubin, L.R.; Lobel, M. Stigma and mental health in endometriosis. Eur. J. Obs. Gynecol. Reprod. Biol. X 2023, 19, 100228. [Google Scholar] [CrossRef] [PubMed]
  20. Giudice, L.C. Endometriosis. N. Engl. J. Med. 2010, 362, 2389–2398. [Google Scholar] [CrossRef] [PubMed]
  21. Abbott, J.; Hawe, J.; Hunter, D.; Holmes, M.; Finn, P.; Garry, R. Laparoscopic excision of endometriosis: A randomized, placebo-controlled trial. Fertil. Steril. 2004, 82, 878–884. [Google Scholar] [CrossRef] [PubMed]
  22. Gruber, T.M.; Mechsner, S. Pathogenesis of Endometriosis: The Origin of Pain and Subfertility. Cells 2021, 10, 1381. [Google Scholar] [CrossRef] [PubMed]
  23. Rossi, V.; Tripodi, F.; Simonelli, C.; Galizia, R.; Nimbi, F.M. Endometriosis-associated pain: A review of quality of life, sexual health and couple relationship. Minerva Obstet. Gynecol. 2021, 73, 536–552. [Google Scholar] [CrossRef]
  24. Laganà, A.S.; La Rosa, V.L.; Rapisarda, A.M.C.; Valenti, G.; Sapia, F.; Chiofalo, B.; Rossetti, D.; Frangez, H.B.; Bokal, E.V.; Vitale, S.G. Anxiety and depression in patients with endometriosis: Impact and management challenges. Int. J. Womens Health 2017, 9, 323–330. [Google Scholar] [CrossRef] [PubMed]
  25. La Rosa, V.L.; De Franciscis, P.; Barra, F.; Schiattarella, A.; Török, P.; Shah, M.; Karaman, E.; Cerentini, T.M.; Di Guardo, F.; Gullo, G.; et al. Quality of life in women with endometriosis: A narrative overview. Minerva Med. 2020, 111, 68–78. [Google Scholar] [CrossRef] [PubMed]
  26. Rush, G.; Misajon, R. Examining subjective wellbeing and health-related quality of life in women with endometriosis. Health Care Women Int. 2018, 39, 303–321. [Google Scholar] [CrossRef] [PubMed]
  27. Profeta, G.; Micangeli, G.; Tarani, F.; Paparella, R.; Ferraguti, G.; Spaziani, M.; Isidori, A.M.; Menghi, M.; Ceccanti, M.; Fiore, M.; et al. Sexual Developmental Disorders in Pediatrics. Clin. Ter. 2022, 173, 475–488. [Google Scholar] [CrossRef] [PubMed]
  28. Millischer, A.-E.; Santulli, P.; Da Costa, S.; Bordonne, C.; Cazaubon, E.; Marcellin, L.; Chapron, C. Adolescent endometriosis: Prevalence increases with age on magnetic resonance imaging scan. Fertil. Steril. 2023, 119, 626–633. [Google Scholar] [CrossRef] [PubMed]
  29. Martire, F.G.; Russo, C.; Selntigia, A.; Nocita, E.; Soreca, G.; Lazzeri, L.; Zupi, E.; Exacoustos, C. Early noninvasive diagnosis of endometriosis: Dysmenorrhea and specific ultrasound findings are important indicators in young women. Fertil. Steril. 2023, 119, 455–464. [Google Scholar] [CrossRef] [PubMed]
  30. Hirsch, M.; Dhillon-Smith, R.; Cutner, A.S.; Yap, M.; Creighton, S.M. The Prevalence of Endometriosis in Adolescents with Pelvic Pain: A Systematic Review. J. Pediatr. Adolesc. Gynecol. 2020, 33, 623–630. [Google Scholar] [CrossRef] [PubMed]
  31. Martire, F.G.; Lazzeri, L.; Conway, F.; Siciliano, T.; Pietropolli, A.; Piccione, E.; Solima, E.; Centini, G.; Zupi, E.; Exacoustos, C. Adolescence and endometriosis: Symptoms, ultrasound signs and early diagnosis. Fertil. Steril. 2020, 114, 1049–1057. [Google Scholar] [CrossRef] [PubMed]
  32. Yeung, P.; Gupta, S.; Gieg, S. Endometriosis in Adolescents: A Systematic Review. J. Endometr. Pelvic Pain. Disord. 2017, 9, 17–29. [Google Scholar] [CrossRef]
  33. Janssen, E.B.; Rijkers, A.C.M.; Hoppenbrouwers, K.; Meuleman, C. D’Hooghe TM. Prevalence of endometriosis diagnosed by laparoscopy in adolescents with dysmenorrhea or chronic pelvic pain: A systematic review. Hum. Reprod. Update 2013, 19, 570–582. [Google Scholar] [CrossRef] [PubMed]
  34. Opoku-Anane, J.; Laufer, M.R. Prevalence of Endometriosis in Adolescent Girls With Chronic Pelvic Pain Not Responding to Conventional Therapy. Have We Underestimated? J. Pediatr. Adolesc. Gynecol. 2012, 25, e50–e51. [Google Scholar] [CrossRef]
  35. Gubbels, A.; Spivack, L.; Lindheim, S.R.; Bhagavath, B. Adolescent Endometriosis. Obs. Gynecol. Surv. 2020, 75, 483–496. [Google Scholar] [CrossRef] [PubMed]
  36. Fedele, F. An Overview on the Psychological Impact of Endometriosis on Adolescent Girls’ Life. Women’s Health Bull. 2021, 8, 195–199. [Google Scholar] [CrossRef]
  37. Fong, Y.F.; Hon, S.K.; Low, L.L.; Lim Mei Xian, K. The clinical profile of young and adolescent women with laparoscopically diagnosed endometriosis in a Singapore tertiary hospital. Taiwan. J. Obs. Gynecol. 2017, 56, 181–183. [Google Scholar] [CrossRef]
  38. Zannoni, L.; Giorgi, M.; Spagnolo, E.; Montanari, G.; Villa, G.; Seracchioli, R. Dysmenorrhea, Absenteeism from School, and Symptoms Suspicious for Endometriosis in Adolescents. J. Pediatr. Adolesc. Gynecol. 2014, 27, 258–265. [Google Scholar] [CrossRef] [PubMed]
  39. Söderman, L.; Edlund, M.; Marions, L. Prevalence and impact of dysmenorrhea in Swedish adolescents. Acta Obs. Gynecol. Scand. 2019, 98, 215–221. [Google Scholar] [CrossRef]
  40. Banikarim, C.; Chacko, M.R.; Kelder, S.H. Prevalence and Impact of Dysmenorrhea on Hispanic Female Adolescents. Arch. Pediatr. Adolesc. Med. 2000, 154, 1226. [Google Scholar] [CrossRef] [PubMed]
  41. Baker, F.C.; Driver, H.S.; Rogers, G.G.; Paiker, J.; Mitchell, D. High nocturnal body temperatures and disturbed sleep in women with primary dysmenorrhea. Am. J. Physiol.-Endocrinol. Metab. 1999, 277, E1013–E1021. [Google Scholar] [CrossRef] [PubMed]
  42. Martire, F.G.; Piccione, E.; Exacoustos, C.; Zupi, E. Endometriosis and Adolescence: The Impact of Dysmenorrhea. J. Clin. Med. 2023, 12, 5624. [Google Scholar] [CrossRef] [PubMed]
  43. González-Echevarría, A.M.; Rosario, E.; Acevedo, S.; Flores, I. Impact of coping strategies on quality of life of adolescents and young women with endometriosis. J. Psychosom. Obs. Gynecol. 2019, 40, 138–145. [Google Scholar] [CrossRef] [PubMed]
  44. Vitale, S.G.; La Rosa, V.L.; Rapisarda, A.M.C.; Laganà, A.S. Impact of endometriosis on quality of life and psychological well-being. J. Psychosom. Obstet. Gynecol. 2017, 38, 317–319. [Google Scholar] [CrossRef] [PubMed]
  45. Vitale, S.G.; Petrosino, B.; La Rosa, V.L.; Rapisarda, A.M.C.; Laganà, A.S. A Systematic Review of the Association Between Psychiatric Disturbances and Endometriosis. J. Obs. Gynaecol. Can. 2016, 38, 1079–1080. [Google Scholar] [CrossRef] [PubMed]
  46. Vannuccini, S.; Lazzeri, L.; Orlandini, C.; Morgante, G.; Bifulco, G.; Fagiolini, A.; Petraglia, F. Mental health, pain symptoms and systemic comorbidities in women with endometriosis: A cross-sectional study. J. Psychosom. Obs. Gynecol. 2018, 39, 315–320. [Google Scholar] [CrossRef] [PubMed]
  47. Ballweg, M.L.; Laufer, M.R. Adolescent Endometriosis: Improving the Comfort Level of Health Care Providers Treating Adolescents with Endometriosis. J. Pediatr. Adolesc. Gynecol. 2011, 24, S1. [Google Scholar] [CrossRef]
  48. Ballweg, M.L. Treating Endometriosis in Adolescents: Does It Matter? J. Pediatr. Adolesc. Gynecol. 2011, 24, S2–S6. [Google Scholar] [CrossRef]
  49. Soliman, A.M.; Coyne, K.S.; Zaiser, E.; Castelli-Haley, J.; Fuldeore, M.J. The burden of endometriosis symptoms on health-related quality of life in women in the United States: A cross-sectional study. J. Psychosom. Obs. Gynecol. 2017, 38, 238–248. [Google Scholar] [CrossRef] [PubMed]
  50. Cox, H.; Henderson, L.; Wood, R.; Cagliarini, G. Learning to take charge: Women’s experiences of living with endometriosis. Complement. Ther. Nurs. Midwifery 2003, 9, 62–68. [Google Scholar] [CrossRef] [PubMed]
  51. Cox, H.; Henderson, L.; Andersen, N.; Cagliarini, G.; Ski, C. Focus group study of endometriosis: Struggle, loss and the medical merry-go-round. Int. J. Nurs. Pract. 2003, 9, 2–9. [Google Scholar] [CrossRef]
  52. Hållstam, A.; Stålnacke, B.; Svensén, C.; Löfgren, M. Living with painful endometriosis—A struggle for coherence. A Qual. Study. Sex. Reprod. Healthc. 2018, 17, 97–102. [Google Scholar] [CrossRef]
  53. Lövkvist, L.; Boström, P.; Edlund, M.; Olovsson, M. Age-Related Differences in Quality of Life in Swedish Women with Endometriosis. J. Womens Health 2016, 25, 646–653. [Google Scholar] [CrossRef] [PubMed]
  54. Roomaney, R.; Kagee, A. Salient aspects of quality of life among women diagnosed with endometriosis: A qualitative study. J. Health Psychol. 2018, 23, 905–916. [Google Scholar] [CrossRef] [PubMed]
  55. Ballard, K.; Lowton, K.; Wright, J. What’s the delay? A qualitative study of women’s experiences of reaching a diagnosis of endometriosis. Fertil. Steril. 2006, 86, 1296–1301. [Google Scholar] [CrossRef] [PubMed]
  56. Denny, E. “I Never Know From One Day to Another How I Will Feel”: Pain and Uncertainty in Women With Endometriosis. Qual. Health Res. 2009, 19, 985–995. [Google Scholar] [CrossRef] [PubMed]
  57. Facchin, F.; Barbara, G.; Dridi, D.; Alberico, D.; Buggio, L.; Somigliana, E.; Saita, E.; Vercellini, P. Mental health in women with endometriosis: Searching for predictors of psychological distress. Hum. Reprod. 2017, 32, 1855–1861. [Google Scholar] [CrossRef] [PubMed]
  58. Chauvet, P.; Guiguet-Auclair, C.; Comptour, A.; Denouël, A.; Gerbaud, L.; Canis, M.; Bourdel, N. Feelings and expectations in endometriosis: Analysis of open comments from a cohort of endometriosis patients. J. Gynecol. Obs. Hum. Reprod. 2018, 47, 281–287. [Google Scholar] [CrossRef] [PubMed]
  59. Nielsen, L.J.; Poulsen, K.; Funch, A.L.; Petersen, K.S. The lived experiences of endometriosis in adolescence—A critical hermeneutic perspective. Scand J. Caring Sci. 2023, 37, 1038–1047. [Google Scholar] [CrossRef] [PubMed]
  60. Bucci, F.; Pluchino, N.; Luisi, S.; Cela, V.; Artini, P.G.; Giannini, A.; Russo, M.; Russo, N.; Casarosa, E.; Luisi, M.; et al. Daily Variation of Plasma Brain-derived Neurotrophic Factor in Women with Endometriosis. J. Endometr. 2011, 3, 40–46. [Google Scholar] [CrossRef]
  61. Shen, H.-H.; Zhang, T.; Yang, H.-L.; Lai, Z.-Z.; Zhou, W.-J.; Mei, J.; Shi, J.-W.; Zhu, R.; Xu, F.-Y.; Li, D.-J.; et al. Ovarian hormones-autophagy-immunity axis in menstruation and endometriosis. Theranostics 2021, 11, 3512–3526. [Google Scholar] [CrossRef] [PubMed]
  62. Begliuomini, S.; Casarosa, E.; Pluchino, N.; Lenzi, E.; Centofanti, M.; Freschi, L.; Pieri, M.; Luisi, S.; Genazzani, A.R. Influence of endogenous and exogenous sex hormones on plasma brain-derived neurotrophic factor. Hum. Reprod. 2007, 22, 995–1002. [Google Scholar] [CrossRef] [PubMed]
  63. Ferraguti, G.; Terracina, S.; Micangeli, G.; Lucarelli, M.; Tarani, L.; Ceccanti, M.; Spaziani, M.; D’orazi, V.; Petrella, C.; Fiore, M. NGF and BDNF in pediatrics syndromes. Neurosci. Biobehav. Rev. 2023, 145, 105015. [Google Scholar] [CrossRef]
  64. Miguez, M.J.; Bueno, D.; Espinoza, L.; Chan, W.; Perez, C. Among Adolescents, BDNF and Pro-BDNF Lasting Changes with Alcohol Use Are Stage Specific. Neural Plast. 2020, 2020, 3937627. [Google Scholar] [CrossRef] [PubMed]
  65. Petrella, C.; Nenna, R.; Petrarca, L.; Tarani, F.; Paparella, R.; Mancino, E.; Di Mattia, G.; Conti, M.G.; Matera, L.; Bonci, E.; et al. Serum NGF and BDNF in Long-COVID-19 Adolescents: A Pilot Study. Diagnostics 2022, 12, 1162. [Google Scholar] [CrossRef] [PubMed]
  66. Sornelli, F.; Fiore, M.; Chaldakov, G.N.; Aloe, L. Brain-derived neurotrophic factor: A new adipokine. Biomed. Rev. 2007, 18, 85–88. [Google Scholar] [CrossRef]
  67. Liu, Y.; Wan, L.; Ngai, C.K.; Wang, Y.; Lam, S.L.; Guo, P. Structures and conformational dynamics of DNA minidumbbells in pyrimidine-rich repeats associated with neurodegenerative diseases. Comput. Struct. Biotechnol. J. 2023, 21, 1584–1592. [Google Scholar] [CrossRef] [PubMed]
  68. van Barneveld, E.; Manders, J.; van Osch, F.H.; van Poll, M.; Visser, L.; van Hanegem, N.; Lim, A.C.; Bongers, M.Y.; Leue, C. Depression, Anxiety, and Correlating Factors in Endometriosis: A Systematic Review and Meta-Analysis. J. Womens Health. 2022, 31, 219–230. [Google Scholar] [CrossRef]
  69. Browne, A.S.; Yu, J.; Huang, R.P.; Francisco, A.M.C.; Sidell, N.; Taylor, R.N. Proteomic identification of neurotrophins in the eutopic endometrium of women with endometriosis. Fertil. Steril. 2012, 98, 713–719. [Google Scholar] [CrossRef] [PubMed]
  70. Ding, S.; Zhu, T.; Tian, Y.; Xu, P.; Chen, Z.; Huang, X.; Zhang, X. Role of Brain-Derived Neurotrophic Factor in Endometriosis Pain. Reprod. Sci. 2018, 25, 1045–1057. [Google Scholar] [CrossRef] [PubMed]
  71. Jafarabady, K.; Shafiee, A.; Bahri, R.A.; Mohammadi, I.; Amini, M.J.; Rajai, S.; Akbarzadeh, D.; Abhari, F.S.; Movahed, E.; Parvari, S.; et al. Brain-derived neurotrophic factor (BDNF) as a potential marker of endometriosis: A systematic review and meta-analysis. BMC Womens Health 2024, 24, 39. [Google Scholar] [CrossRef] [PubMed]
  72. D’angelo, A.; Ceccanti, M.; Petrella, C.; Greco, A.; Tirassa, P.; Rosso, P.; Ralli, M.; Ferraguti, G.; Fiore, M.; Messina, M.P. Role of neurotrophins in pregnancy, delivery and postpartum. Eur. J. Obs. Gynecol. Reprod. Biol. 2020, 247, 32–41. [Google Scholar] [CrossRef] [PubMed]
  73. Cuffaro, F.; Russo, E.; Amedei, A. Endometriosis, Pain, and Related Psychological Disorders: Unveiling the Interplay among the Microbiome, Inflammation, and Oxidative Stress as a Common Thread. Int. J. Mol. Sci. 2024, 25, 6473. [Google Scholar] [CrossRef] [PubMed]
  74. Micangeli, G.; Menghi, M.; Profeta, G.; Tarani, F.; Mariani, A.; Petrella, C.; Barbato, C.; Ferraguti, G.; Ceccanti, M.; Tarani, L.; et al. The Impact of Oxidative Stress on Pediatrics Syndromes. Antioxidants 2022, 11, 1983. [Google Scholar] [CrossRef]
  75. Khashchenko, E.P.; Vysokikh, M.Y.; Marey, M.V.; Sidorova, K.O.; Manukhova, L.A.; Shkavro, N.N.; Uvarova, E.V.; Chuprynin, V.D.; Fatkhudinov, T.K.; Adamyan, L.V.; et al. Altered Glycolysis, Mitochondrial Biogenesis, Autophagy and Apoptosis in Peritoneal Endometriosis in Adolescents. Int. J. Mol. Sci. 2024, 25, 4238. [Google Scholar] [CrossRef]
  76. Leonardi, M.; Hicks, C.; El-Assaad, F.; El-Omar, E.; Condous, G. Endometriosis and the microbiome: A systematic review. BJOG Int. J. Obs. Gynaecol. 2020, 127, 239–249. [Google Scholar] [CrossRef] [PubMed]
  77. Salliss, M.E.; Farland, L.V.; Mahnert, N.D.; Herbst-Kralovetz, M.M. The role of gut and genital microbiota and the estrobolome in endometriosis, infertility and chronic pelvic pain. Hum. Reprod. Update 2021, 28, 92–131. [Google Scholar] [CrossRef] [PubMed]
  78. Jiang, I.; Yong, P.J.; Allaire, C.; Bedaiwy, M.A. Intricate Connections between the Microbiota and Endometriosis. Int. J. Mol. Sci. 2021, 22, 5644. [Google Scholar] [CrossRef] [PubMed]
  79. Zizolfi, B.; Foreste, V.; Gallo, A.; Martone, S.; Giampaolino, P.; Di Spiezio Sardo, A. Endometriosis and dysbiosis: State of art. Front. Endocrinol. 2023, 14, 1140774. [Google Scholar] [CrossRef] [PubMed]
  80. Uzuner, C.; Mak, J.; El-Assaad, F.; Condous, G. The bidirectional relationship between endometriosis and microbiome. Front. Endocrinol. 2023, 14, 1110824. [Google Scholar] [CrossRef] [PubMed]
  81. Sobstyl, A.; Chałupnik, A.; Mertowska, P.; Grywalska, E. How Do Microorganisms Influence the Development of Endometriosis? Participation of Genital, Intestinal and Oral Microbiota in Metabolic Regulation and Immunopathogenesis of Endometriosis. Int. J. Mol. Sci. 2023, 24, 10920. [Google Scholar] [CrossRef] [PubMed]
  82. Svensson, A.; Brunkwall, L.; Roth, B.; Orho-Melander, M.; Ohlsson, B. Associations Between Endometriosis and Gut Microbiota. Reprod. Sci. 2021, 28, 2367–2377. [Google Scholar] [CrossRef]
  83. Ata, B.; Yildiz, S.; Turkgeldi, E.; Brocal, V.P.; Dinleyici, E.C.; Moya, A.; Urman, B. The Endobiota Study: Comparison of Vaginal, Cervical and Gut Microbiota Between Women with Stage 3/4 Endometriosis and Healthy Controls. Sci. Rep. 2019, 9, 2204. [Google Scholar] [CrossRef] [PubMed]
  84. Shan, J.; Ni, Z.; Cheng, W.; Zhou, L.; Zhai, D.; Sun, S.; Yu, C. Gut microbiota imbalance and its correlations with hormone and inflammatory factors in patients with stage 3/4 endometriosis. Arch. Gynecol. Obstet. 2021, 304, 1363–1373. [Google Scholar] [CrossRef] [PubMed]
  85. Huang, L.; Liu, B.; Liu, Z.; Feng, W.; Liu, M.; Wang, Y.; Peng, D.; Fu, X.; Zhu, H.; Cui, Z.; et al. Gut Microbiota Exceeds Cervical Microbiota for Early Diagnosis of Endometriosis. Front. Cell Infect. Microbiol. 2021, 11, 788836. [Google Scholar] [CrossRef] [PubMed]
  86. Shaikh, S.D.; Sun, N.; Canakis, A.; Park, W.Y.; Weber, H.C. Irritable Bowel Syndrome and the Gut Microbiome: A Comprehensive Review. J. Clin. Med. 2023, 12, 2558. [Google Scholar] [CrossRef]
  87. Aloe, L.; Fiore, M.; Probert, L.; Turrini, P.; Tirassa, P. Overexpression of tumour necrosis factor alpha in the brain of transgenic mice differentially alters nerve growth factor levels and choline acetyltransferase activity. Cytokine 1999, 11, 45–54. [Google Scholar] [CrossRef] [PubMed]
  88. Ross, H.E.; Guo, Y.; Coleman, K.; Ousley, O.; Miller, A.H. Association of IL-12p70 and IL-6: IL-10 ratio with autism-related behaviors in 22q11.2 deletion syndrome: A preliminary report. Brain Behav. Immun. 2013, 31, 76–81. [Google Scholar] [CrossRef]
  89. Patterson, S.L. Immune dysregulation and cognitive vulnerability in the aging brain: Interactions of microglia, IL-1β, BDNF and synaptic plasticity. Neuropharmacology 2015, 96, 11–18. [Google Scholar] [CrossRef] [PubMed]
  90. Ustianowska, K.; Ustianowski, Ł.; Machaj, F.; Gorący, A.; Rosik, J.; Szostak, B.; Szostak, J.; Pawlik, A. The Role of the Human Microbiome in the Pathogenesis of Pain. Int. J. Mol. Sci. 2022, 23, 13267. [Google Scholar] [CrossRef] [PubMed]
  91. Matsuda, M.; Huh, Y.; Ji, R.R. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J. Anesth. 2019, 33, 131–139. [Google Scholar] [CrossRef] [PubMed]
  92. Bajaj, P. Endometriosis is associated with central sensitization: A psychophysical controlled study. J. Pain 2003, 4, 372–380. [Google Scholar] [CrossRef]
  93. Salmeri, N.; Sinagra, E.; Dolci, C.; Buzzaccarini, G.; Sozzi, G.; Sutera, M.; Candiani, M.; Ungaro, F.; Massimino, L.; Danese, S.; et al. Microbiota in Irritable Bowel Syndrome and Endometriosis: Birds of a Feather Flock Together—A Review. Microorganisms 2023, 11, 2089. [Google Scholar] [CrossRef]
  94. Simkin, D.R. Microbiome and Mental Health, Specifically as It Relates to Adolescents. Curr. Psychiatry Rep. 2019, 21, 93. [Google Scholar] [CrossRef] [PubMed]
  95. Nisenblat, V.; Bossuyt, P.M.; Shaikh, R.; Farquhar, C.; Jordan, V.; Scheffers, C.S.; Mol, B.W.J.; Johnson, N.; Hull, M.L. Blood biomarkers for the non-invasive diagnosis of endometriosis. Cochrane Database Syst. Rev. 2016, 2016, CD012179. [Google Scholar] [CrossRef] [PubMed]
  96. Kimber-Trojnar, Ż.; Pilszyk, A.; Niebrzydowska, M.; Pilszyk, Z.; Ruszała, M.; Leszczyńska-Gorzelak, B. The Potential of Non-Invasive Biomarkers for Early Diagnosis of Asymptomatic Patients with Endometriosis. J. Clin. Med. 2021, 10, 2762. [Google Scholar] [CrossRef] [PubMed]
  97. Leonardi, M.; Condous, G. How to perform an ultrasound to diagnose endometriosis. Australas. J. Ultrasound Med. 2018, 21, 61–69. [Google Scholar] [CrossRef]
  98. El-Ali, A.M.; Tong, A.; Smereka, P.; Lala, S.V. MRI for endometriosis in adolescent patients. Pediatr. Radiol. 2024, 55, 24–35. [Google Scholar] [CrossRef] [PubMed]
  99. Chen, Y.; Pan, M.; Zuo, Y.; Yang, B.; Wang, S. Research progress of CA125 in endometriosis: Teaching an old dog new tricks. Gynecol. Obs. Clin. Med. 2022, 2, 191–198. [Google Scholar] [CrossRef]
  100. Agrawal, S.; Tapmeier, T.T.; Rahmioglu, N.; Kirtley, S.; Zondervan, K.T.; Becker, C.M. The miRNA Mirage: How Close Are We to Finding a Non-Invasive Diagnostic Biomarker in Endometriosis? A Systematic Review. Int. J. Mol. Sci. 2018, 19, 599. [Google Scholar] [CrossRef] [PubMed]
  101. Moustafa, S.; Burn, M.; Mamillapalli, R.; Nematian, S.; Flores, V.; Taylor, H.S. Accurate Diagnosis of Endometriosis Using Serum MicroRNAs. Obs. Gynecol. Surv. 2020, 75, 404–406. [Google Scholar] [CrossRef]
  102. Senapati, S.; Clauw, D.; As-Sanie, S. The relationship between serum tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) with pelvic pain symptoms in women with endometriosis. Fertil. Steril. 2009, 92, S112. [Google Scholar] [CrossRef]
  103. Maignien, C.; Santulli, P.; Kateb, F.; Caradeuc, C.; Marcellin, L.; Pocate-Cheriet, K.; Bourdon, M.; Chouzenoux, S.; Batteux, F.; Bertho, G.; et al. Endometriosis phenotypes are associated with specific serum metabolic profiles determined by proton-nuclear magnetic resonance. Reprod. Biomed. Online 2020, 41, 640–652. [Google Scholar] [CrossRef] [PubMed]
  104. Vicente-Muñoz, S.; Morcillo, I.; Puchades-Carrasco, L.; Payá, V.; Pellicer, A.; Pineda-Lucena, A. Pathophysiologic processes have an impact on the plasma metabolomic signature of endometriosis patients. Fertil. Steril. 2016, 106, 1733–1741.e1. [Google Scholar] [CrossRef] [PubMed]
  105. Vicente-Muñoz, S.; Morcillo, I.; Puchades-Carrasco, L.; Payá, V.; Pellicer, A.; Pineda-Lucena, A. Nuclear magnetic resonance metabolomic profiling of urine provides a noninvasive alternative to the identification of biomarkers associated with endometriosis. Fertil. Steril. 2015, 104, 1202–1209. [Google Scholar] [CrossRef] [PubMed]
  106. Warren, L.A.; Shih, A.; Renteira, S.M.; Seckin, T.; Blau, B.; Simpfendorfer, K.; Lee, A.; Metz, C.N.; Gregersen, P.K. Analysis of menstrual effluent: Diagnostic potential for endometriosis. Mol. Med. 2018, 24, 1. [Google Scholar] [CrossRef] [PubMed]
  107. Tindal, K.; Filby, C.E.; Cousins, F.L.; Ellery, S.J.; Vollenhoven, B.; Palmer, K.; Gordon, A.; Gargett, C.E.; Davies-Tuck, M. The composition of menstrual fluid, its applications, and recent advances to understand the endometrial environment: A narrative review. FS Rev. 2024, 5, 100075. [Google Scholar] [CrossRef]
  108. Chiorean, D.M.; Mitranovici, M.-I.; Toru, H.S.; Cotoi, T.C.; Tomuț, A.N.; Turdean, S.G.; Cotoi, O.S. New Insights into Genetics of Endometriosis—A Comprehensive Literature Review. Diagnostics 2023, 13, 2265. [Google Scholar] [CrossRef]
  109. Wang, M.; Zheng, L.; Lin, R.; Ma, S.; Li, J.; Yang, S. A comprehensive overview of exosome lncRNAs: Emerging biomarkers and potential therapeutics in endometriosis. Front. Endocrinol. 2023, 14, 1199569. [Google Scholar] [CrossRef] [PubMed]
  110. Shomali, N.; Hemmatzadeh, M.; Yousefzadeh, Y.; Soltani-Zangbar, M.S.; Hamdi, K.; Mehdizadeh, A.; Yousefi, M. Exosomes: Emerging biomarkers and targets in folliculogenesis and endometriosis. J. Reprod. Immunol. 2020, 142, 103181. [Google Scholar] [CrossRef] [PubMed]
  111. Agic, A.; Djalali, S.; Wolfler, M.M.; Halis, G.; Diedrich, K.; Hornung, D. Combination of CCR1 mRNA, MCP1, and CA125 Measurements in Peripheral Blood as a Diagnostic Test for Endometriosis. Reprod. Sci. 2008, 15, 906–911. [Google Scholar] [CrossRef] [PubMed]
  112. Mihalyi, A.; Gevaert, O.; Kyama, C.M.; Simsa, P.; Pochet, N.; De Smet, F.; De Moor, B.; Meuleman, C.; Billen, J.; Blanckaert, N.; et al. Non-invasive diagnosis of endometriosis based on a combined analysis of six plasma biomarkers. Hum. Reprod. 2010, 25, 654–664. [Google Scholar] [CrossRef] [PubMed]
  113. Socolov, R.; Butureanu, S.; Angioni, S.; Sindilar, A.; Boiculese, L.; Cozma, L.; Socolov, D. The value of serological markers in the diagnosis and prognosis of endometriosis: A prospective case–control study. Eur. J. Obs. Gynecol. Reprod. Biol. 2011, 154, 215–217. [Google Scholar] [CrossRef] [PubMed]
  114. Vodolazkaia, A.; El-Aalamat, Y.; Popovic, D.; Mihalyi, A.; Bossuyt, X.; Kyama, C.M.; Fassbender, A.; Bokor, A.; Schols, D.; Huskens, D.; et al. Evaluation of a panel of 28 biomarkers for the non-invasive diagnosis of endometriosis. Hum. Reprod. 2012, 27, 2698–2711. [Google Scholar] [CrossRef] [PubMed]
  115. Ozhan, E.; Kokcu, A.; Yanik, K.; Gunaydin, M. Investigation of diagnostic potentials of nine different biomarkers in endometriosis. Eur. J. Obs. Gynecol. Reprod. Biol. 2014, 178, 128–133. [Google Scholar] [CrossRef] [PubMed]
  116. Choi, Y.S.; Kim, S.; Oh, Y.S.; Cho, S.; Hoon Kim, S. Elevated serum interleukin-32 levels in patients with endometriosis: A cross-sectional study. Am. J. Reprod. Immunol. 2019, 82, e13149. [Google Scholar] [CrossRef] [PubMed]
  117. Teague, E.M.C.O.; Print, C.G.; Hull, M.L. The role of microRNAs in endometriosis and associated reproductive conditions. Hum. Reprod. Update 2010, 16, 142–165. [Google Scholar] [CrossRef] [PubMed]
  118. Jia, S.-Z.; Yang, Y.; Lang, J.; Sun, P.; Leng, J. Plasma miR-17-5p, miR-20a and miR-22 are down-regulated in women with endometriosis. Hum. Reprod. 2013, 28, 322–330. [Google Scholar] [CrossRef]
  119. Suryawanshi, S.; Vlad, A.M.; Lin, H.-M.; Mantia-Smaldone, G.; Laskey, R.; Lee, M.; Lin, Y.; Donnellan, N.; Klein-Patel, M.; Lee, T.; et al. Plasma microRNAs as novel biomarkers for endometriosis and endometriosis-associated ovarian cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 1213–1224. [Google Scholar] [CrossRef]
  120. Wang, W.T.; Zhao, Y.N.; Han, B.W.; Hong, S.J.; Chen, Y.Q. Circulating MicroRNAs Identified in a Genome-Wide Serum MicroRNA Expression Analysis as Noninvasive Biomarkers for Endometriosis. J. Clin. Endocrinol. Metab. 2013, 98, 281–289. [Google Scholar] [CrossRef]
  121. Cosar, E.; Mamillapalli, R.; Ersoy, G.S.; Cho, S.; Seifer, B.; Taylor, H.S. Serum microRNAs as diagnostic markers of endometriosis: A comprehensive array-based analysis. Fertil. Steril. 2016, 106, 402–409. [Google Scholar] [CrossRef] [PubMed]
  122. Nisenblat, V.; Sharkey, D.J.; Wang, Z.; Evans, S.F.; Healey, M.; Teague, E.M.C.O.; Print, C.G.; A Robertson, S.; Hull, M.L. Plasma miRNAs Display Limited Potential as Diagnostic Tools for Endometriosis. J. Clin. Endocrinol. Metab. 2019, 104, 1999–2022. [Google Scholar] [CrossRef] [PubMed]
  123. Vanhie, A.O.D.; Peterse, D.; Beckers, A.; Cuéllar, A.; Fassbender, A.; Meuleman, C.; Mestdagh, P.; D’hooghe, T. Plasma miRNAs as biomarkers for endometriosis. Hum. Reprod. 2019, 34, 1650–1660. [Google Scholar] [CrossRef] [PubMed]
  124. Xavier, P.; Belo, L.; Beires, J.; Rebelo, I.; Martinez de Oliveira, J.; Lunet, N.; Barros, H. Serum levels of VEGF and TNF-α and their association with C-reactive protein in patients with endometriosis. Arch. Gynecol. Obstet. 2006, 273, 227–231. [Google Scholar] [CrossRef]
  125. Othman, E.E.D.R.; Hornung, D.; Salem, H.T.; Khalifa, E.A.; El-Metwally, T.H.; Al-Hendy, A. Serum cytokines as biomarkers for nonsurgical prediction of endometriosis. Eur. J. Obs. Gynecol. Reprod. Biol. 2008, 137, 240–246. [Google Scholar] [CrossRef] [PubMed]
  126. Seeber, B.; Sammel, M.D.; Fan, X.; Gerton, G.L.; Shaunik, A.; Chittams, J.; Barnhart, K.T. Panel of markers can accurately predict endometriosis in a subset of patients. Fertil. Steril. 2008, 89, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  127. Li, W.-N.; Hsiao, K.-Y.; Wang, C.-A.; Chang, N.; Hsu, P.-L.; Sun, C.-H.; Wu, S.-R.; Wu, M.-H.; Tsai, S.-J. Extracellular vesicle-associated VEGF-C promotes lymphangiogenesis and immune cells infiltration in endometriosis. Proc. Natl. Acad. Sci. USA 2020, 117, 25859–25868. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, L.; Li, H.; Yuan, M.; Li, D.; Sun, C.; Wang, G. Serum Exosomal MicroRNAs as Potential Circulating Biomarkers for Endometriosis. Dis. Markers 2020, 2020, 2456340. [Google Scholar] [CrossRef] [PubMed]
  129. Shan, S.; Yang, Y.; Jiang, J.; Yang, B.; Yang, Y.; Sun, F.; Zhang, J.; Lin, Y.; Xu, H. Extracellular vesicle-derived long non-coding RNA as circulating biomarkers for endometriosis. Reprod. Biomed. Online 2022, 44, 923–933. [Google Scholar] [CrossRef]
  130. Ji, S.; Liu, Y.; Yan, L.; Zhang, Y.; Li, Y.; Zhu, Q.; Xia, W.; Ge, S.; Zhang, J. DIA-based analysis of the menstrual blood proteome identifies association between CXCL5 and IL1RN and endometriosis. J. Proteom. 2023, 289, 104995. [Google Scholar] [CrossRef]
  131. Amanda, C.R.; Asmarinah; Hestiantoro, A.; Tulandi, T.; Febriyeni. Gene expression of aromatase, SF-1, and HSD17B2 in menstrual blood as noninvasive diagnostic biomarkers for endometriosis. Eur. J. Obs. Gynecol. Reprod. Biol. 2024, 301, 95–101. [Google Scholar] [CrossRef] [PubMed]
  132. Starodubtseva, N.; Chagovets, V.; Tokareva, A.; Dumanovskaya, M.; Kukaev, E.; Novoselova, A.; Frankevich, V.; Pavlovich, S.V.; Sukhikh, G. Diagnostic Value of Menstrual Blood Lipidomics in Endometriosis: A Pilot Study. Biomolecules 2024, 14, 899. [Google Scholar] [CrossRef] [PubMed]
  133. Potlog-Nahari, C.; Stratton, P.; Winkel, C.; Widra, E.; Sinaii, N.; Connors, S.; Nieman, L.K. Urine vascular endothelial growth factor-A is not a useful marker for endometriosis. Fertil. Steril. 2004, 81, 1507–1512. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, W.T.; Sun, Y.M.; Huang, W.; He, B.; Zhao, Y.N.; Chen, Y.Q. Genome-wide Long Non-coding RNA Analysis Identified Circulating LncRNAs as Novel Non-invasive Diagnostic Biomarkers for Gynecological Disease. Sci. Rep. 2016, 6, 23343. [Google Scholar] [CrossRef]
  135. Qiu, J.; Zhang, X.; Ding, Y.; Hua, K. 1856 Circulating Exosomal Long Noncoding RNA-TC0101441 as a Non-Invasive Biomarker for the Prediction of Endometriosis Severity and Recurrence. J Minim Invasive Gynecol. 2019, 26, S170–S171. [Google Scholar] [CrossRef]
  136. Nabeta, M.; Abe, Y.; Kagawa, L.; Haraguchi, R.; Kito, K.; Ueda, N.; Sugita, A.; Yokoyama, M.; Kusanagi, Y.; Ito, M. Identification of anti-α-enolase autoantibody as a novel serum marker for endometriosis. PROTEOMICS–Clin. Appl. 2009, 3, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  137. Gajbhiye, R.; Sonawani, A.; Khan, S.; Suryawanshi, A.; Kadam, S.; Warty, N.; Raut, V.; Khole, V. Identification and validation of novel serum markers for early diagnosis of endometriosis. Hum. Reprod. 2012, 27, 408–417. [Google Scholar] [CrossRef] [PubMed]
  138. Kaya, C.; Alay, I.; Guraslan, H.; Gedikbasi, A.; Ekin, M.; Kaya, S.E.; Oral, E.; Yasar, L. The Role of Serum Caspase 3 Levels in Prediction of Endometriosis Severity. Gynecol. Obs. Investig. 2018, 83, 576–585. [Google Scholar] [CrossRef] [PubMed]
  139. Tarani, L.; Fiore, M. Disclosing the Complexities of Childhood Neurodevelopmental Disorders. Children 2024, 12, 16. [Google Scholar] [CrossRef]
  140. Zugaj, M.R.; Germeyer, A.; Kranz, K.; Züger, A.; Keßler, J. Experiences of patients with endometriosis with a digital health application: A qualitative analysis. Arch. Gynecol. Obstet. 2024, 310, 2253–2263. [Google Scholar] [CrossRef]
  141. Edgley, K.; Horne, A.W.; Saunders, P.T.K.; Tsanas, A. Symptom tracking in endometriosis using digital technologies: Knowns, unknowns, and future prospects. Cell Rep. Med. 2023, 4, 101192. [Google Scholar] [CrossRef] [PubMed]
  142. De França Moreira, M.; Gamboa, O.L.; Pinho Oliveira, M.A. Association between severity of pain, perceived stress and vagally-mediated heart rate variability in women with endometriosis. Women Health 2021, 61, 937–946. [Google Scholar] [CrossRef] [PubMed]
  143. Balci, B.K. Is Endometriosis Telemedicine Friendly? Gynecol. Minim. Invasive Ther. 2022, 11, 224–230. [Google Scholar] [CrossRef] [PubMed]
  144. Perelmuter, S.; Shin, J.H. Enhancing endometriosis care with telehealth: Opportunities and challenges. J. Telemed. Telecare 2024, 1357633X241287969. [Google Scholar] [CrossRef] [PubMed]
  145. Shoebotham, A.; Coulson, N.S. Therapeutic Affordances of Online Support Group Use in Women With Endometriosis. J. Med. Internet Res. 2016, 18, e109. [Google Scholar] [CrossRef]
  146. Adler, H.; Lewis, M.; Ng, C.H.M.; Brooks, C.; Leonardi, M.; Mikocka-Walus, A.; Bush, D.; Semprini, A.; Wilkinson-Tomey, J.; Condous, G.; et al. Social Media, Endometriosis, and Evidence-Based Information: An Analysis of Instagram Content. Healthcare 2024, 12, 121. [Google Scholar] [CrossRef] [PubMed]
  147. Kiser, A.C.; Schliep, K.C.; Hernandez, E.J.; Peterson, C.M.; Yandell, M.; Eilbeck, K. An artificial intelligence approach for investigating multifactorial pain-related features of endometriosis. PLoS ONE 2024, 19, e0297998. [Google Scholar] [CrossRef] [PubMed]
  148. Dungate, B.; Tucker, D.R.; Goodwin, E.; Yong, P.J. Assessing the Utility of artificial intelligence in endometriosis: Promises and pitfalls. Womens Health 2024, 20, 17455057241248121. [Google Scholar] [CrossRef] [PubMed]
  149. Merlot, B.; Elie, V.; Périgord, A.; Husson, Z.; Jubert, A.; Chanavaz-Lacheray, I.; Dennis, T.; Cotty-Eslous, M.; Roman, H. Pain Reduction With an Immersive Digital Therapeutic in Women Living With Endometriosis-Related Pelvic Pain: At-Home Self-Administered Randomized Controlled Trial. J. Med. Internet Res. 2023, 25, e47869. [Google Scholar] [CrossRef] [PubMed]
  150. Brown, J.; Crawford, T.J.; Allen, C.; Hopewell, S.; Prentice, A. Nonsteroidal anti-inflammatory drugs for pain in women with endometriosis. Cochrane Database Syst. Rev. 2017, 1, CD004753. [Google Scholar] [CrossRef] [PubMed]
  151. Lazzeri, L.; Andersson, K.L.; Angioni, S.; Arena, A.; Arena, S.; Bartiromo, L.; Berlanda, N.; Bonin, C.; Candiani, M.; Centini, G.; et al. How to Manage Endometriosis in Adolescence: The Endometriosis Treatment Italian Club Approach. J. Minim. Invasive Gynecol. 2023, 30, 616–626. [Google Scholar] [CrossRef] [PubMed]
  152. Brown, J.; Crawford, T.J.; Datta, S.; Prentice, A. Oral contraceptives for pain associated with endometriosis. Cochrane Gynaecology and Fertility Group, ed. Cochrane Database Syst. Rev. 2018, 2018, CD001019. [Google Scholar] [CrossRef]
  153. Jensen, J.T.; Schlaff, W.; Gordon, K. Use of combined hormonal contraceptives for the treatment of endometriosis-related pain: A systematic review of the evidence. Fertil. Steril. 2018, 110, 137–152.e1. [Google Scholar] [CrossRef] [PubMed]
  154. Grandi, G.; Barra, F.; Ferrero, S.; Sileo, F.G.; Bertucci, E.; Napolitano, A.; Facchinetti, F. Hormonal contraception in women with endometriosis: A systematic review. Eur. J. Contracept. Reprod. Health Care Off. J. Eur. Soc. Contracept. 2019, 24, 61–70. [Google Scholar] [CrossRef]
  155. Brown, J.; Kives, S.; Akhtar, M. Progestagens and anti-progestagens for pain associated with endometriosis. Cochrane Database Syst. Rev. 2012, 2012, CD002122. [Google Scholar] [CrossRef] [PubMed]
  156. Petraglia, F.; Hornung, D.; Seitz, C.; Faustmann, T.; Gerlinger, C.; Luisi, S.; Lazzeri, L.; Strowitzki, T. Reduced pelvic pain in women with endometriosis: Efficacy of long-term dienogest treatment. Arch. Gynecol. Obstet. 2012, 285, 167–173. [Google Scholar] [CrossRef] [PubMed]
  157. Andres, M.d.P.; Lopes, L.A.; Baracat, E.C.; Podgaec, S. Dienogest in the treatment of endometriosis: Systematic review. Arch. Gynecol. Obstet. 2015, 292, 523–529. [Google Scholar] [CrossRef] [PubMed]
  158. Lan, S.; Ling, L.; Jianhong, Z.; Xijing, J.; Lihui, W. Analysis of the levonorgestrel-releasing intrauterine system in women with endometriosis. J. Int. Med. Res. 2013, 41, 548–558. [Google Scholar] [CrossRef] [PubMed]
  159. Yoost, J.; LaJoie, A.S.; Hertweck, P.; Loveless, M. Use of the levonorgestrel intrauterine system in adolescents with endometriosis. J. Pediatr. Adolesc. Gynecol. 2013, 26, 120–124. [Google Scholar] [CrossRef] [PubMed]
  160. Margatho, D.; Carvalho, N.M.; Bahamondes, L. Endometriosis-associated pain scores and biomarkers in users of the etonogestrel-releasing subdermal implant or the 52-mg levonorgestrel-releasing intrauterine system for up to 24 months. Eur. J. Contracept. Reprod. Health Care Off. J. Eur. Soc. Contracept. 2020, 25, 133–140. [Google Scholar] [CrossRef] [PubMed]
  161. Brown, J.; Pan, A.; Hart, R.J. Gonadotrophin-releasing hormone analogues for pain associated with endometriosis. Cochrane Database Syst. Rev. 2010, 2010, CD008475. [Google Scholar] [CrossRef] [PubMed]
  162. Tang, H.; Wu, R.; Li, X.; Zhou, Y.; Liu, Z.; Wang, C.; Chen, Y.; Zhang, F. Curative effect of 1.88-mg and 3.75-mg gonadotrophin-releasing hormone agonist on stage III-IV endometriosis: Randomized controlled study. J. Obs. Gynaecol. Res. 2017, 43, 1550–1554. [Google Scholar] [CrossRef] [PubMed]
  163. Veth, V.B.; van de Kar, M.M.; Duffy, J.M.; van Wely, M.; Mijatovic, V.; Maas, J.W. Gonadotropin-releasing hormone analogues for endometriosis. Cochrane Database Syst. Rev. 2023, 6, CD014788. [Google Scholar] [CrossRef] [PubMed]
  164. Gallagher, J.S.; Feldman, H.A.; Stokes, N.A.; Laufer, M.R.; Hornstein, M.D.; Gordon, C.M.; DiVasta, A.D. The Effects of Gonadotropin-Releasing Hormone Agonist Combined with Add-Back Therapy on Quality of Life for Adolescents with Endometriosis: A Randomized Controlled Trial. J. Pediatr. Adolesc. Gynecol. 2017, 30, 215–222. [Google Scholar] [CrossRef]
  165. DiVasta, A.D.M.; Feldman, H.A.; Gallagher, J.B.S.; Stokes, N.A.B.; Laufer, M.R.; Hornstein, M.D.; Gordon, C.M.M. Hormonal Add-Back Therapy for Females Treated With Gonadotropin-Releasing Hormone Agonist for Endometriosis: A Randomized Controlled Trial. Obs. Gynecol. 2015, 126, 617–627. [Google Scholar] [CrossRef] [PubMed]
  166. Taylor, H.S.; Giudice, L.C.; Lessey, B.A.; Abrao, M.S.; Kotarski, J.; Archer, D.F.; Diamond, M.P.; Surrey, E.; Johnson, N.P.; Watts, N.B.; et al. Treatment of Endometriosis-Associated Pain with Elagolix, an Oral GnRH Antagonist. N. Engl. J. Med. 2017, 377, 28–40. [Google Scholar] [CrossRef] [PubMed]
  167. Donnez, J.; Taylor, H.S.; Taylor, R.N.; Akin, M.D.; Tatarchuk, T.F.; Wilk, K.; Gotteland, J.-P.; Lecomte, V.; Bestel, E. Treatment of endometriosis-associated pain with linzagolix, an oral gonadotropin-releasing hormone-antagonist: A randomized clinical trial. Fertil. Steril. 2020, 114, 44–55. [Google Scholar] [CrossRef] [PubMed]
  168. Osuga, Y.; Seki, Y.; Tanimoto, M.; Kusumoto, T.; Kudou, K.; Terakawa, N. Relugolix, an oral gonadotropin-releasing hormone receptor antagonist, reduces endometriosis-associated pain in a dose-response manner: A randomized, double-blind, placebo-controlled study. Fertil. Steril. 2021, 115, 397–405. [Google Scholar] [CrossRef]
  169. Rzewuska, A.M.; Żybowska, M.; Sajkiewicz, I.; Spiechowicz, I.; Żak, K.; Abramiuk, M.; Kułak, K.; Tarkowski, R. Gonadotropin-Releasing Hormone Antagonists-A New Hope in Endometriosis Treatment? J. Clin. Med. 2023, 12, 1008. [Google Scholar] [CrossRef]
  170. Ferrero, S.; Gillott, D.J.; Venturini, P.L.; Remorgida, V. Use of aromatase inhibitors to treat endometriosis-related pain symptoms: A systematic review. Reprod. Biol. Endocrinol. RBE 2011, 9, 89. [Google Scholar] [CrossRef] [PubMed]
  171. Almassinokiani, F.; Almasi, A.; Akbari, P.; Saberifard, M. Effect of Letrozole on endometriosis-related pelvic pain. Med. J. Islam. Repub. Iran. 2014, 28, 107. [Google Scholar] [PubMed]
  172. Nawathe, A.; Patwardhan, S.; Yates, D.; Harrison, G.R.; Khan, K.S. Systematic review of the effects of aromatase inhibitors on pain associated with endometriosis. BJOG Int. J. Obs. Gynaecol. 2008, 115, 818–822. [Google Scholar] [CrossRef]
  173. Ferrero, S.; Barra, F. Hormonal Therapy in Endometriosis and Adenomyosis: Danazol, Aromatase Inhibitors. In Endometriosis and Adenomyosis; Oral, E., Ed.; Springer International Publishing: Berlin/Heidelberg, Germany, 2022; pp. 557–575. [Google Scholar] [CrossRef]
  174. Farquhar, C.; Prentice, A.; Singla, A.A.; Selak, V. Danazol for pelvic pain associated with endometriosis. Cochrane Gynaecology and Fertility Group, ed. Cochrane Database Syst. Rev. 2007, 4, CD000068. [Google Scholar] [CrossRef]
  175. Fu, J.; Song, H.; Zhou, M.; Zhu, H.; Wang, Y.; Chen, H.; Huang, W. Progesterone receptor modulators for endometriosis. Cochrane Database Syst. Rev. 2017, 7, CD009881. [Google Scholar] [CrossRef] [PubMed]
  176. Islam, M.S.; Afrin, S.; Jones, S.I.; Segars, J. Selective Progesterone Receptor Modulators-Mechanisms and Therapeutic Utility. Endocr. Rev. 2020, 41, bnaa012. [Google Scholar] [CrossRef] [PubMed]
  177. Tyson, N.; Shim, J.; Lee, T.; King, C.R.; Einarsson, J.; Hornstein, M.D.; Laufer, M.R. Surgical Considerations in the Management of Adolescent Endometriosis—An Expert Commentary. J. Minim. Invasive Gynecol. 2024, 31, 378–386. [Google Scholar] [CrossRef] [PubMed]
  178. Doyle, J.O.; Missmer, S.A.; Laufer, M.R. The Effect of Combined Surgical-Medical Intervention on the Progression of Endometriosis in an Adolescent and Young Adult Population. J. Pediatr. Adolesc. Gynecol. 2009, 22, 257–263. [Google Scholar] [CrossRef] [PubMed]
  179. Vannuccini, S.; Clemenza, S.; Rossi, M.; Petraglia, F. Hormonal treatments for endometriosis: The endocrine background. Rev. Endocr. Metab. Disord. 2022, 23, 333–355. [Google Scholar] [CrossRef] [PubMed]
  180. Dunselman, G.A.J.; Vermeulen, N.; Becker, C.; Calhaz-Jorge, C.; D’Hooghe, T.; De Bie, B.; Heikinheimo, O.; Horne, A.W.; Kiesel, L.; Nap, A.; et al. ESHRE guideline: Management of women with endometriosis. Hum. Reprod. 2014, 29, 400–412. [Google Scholar] [CrossRef] [PubMed]
  181. Nnoaham, K.E.; Hummelshoj, L.; Webster, P.; d’Hooghe, T.; de Cicco Nardone, F.; de Cicco Nardone, C.; Jenkinson, C.; Kennedy, S.H.; Zondervan, K.T. Impact of endometriosis on quality of life and work productivity: A multicenter study across ten countries. Fertil. Steril. 2011, 96, 366–373.e8. [Google Scholar] [CrossRef]
  182. Facchin, F.; Barbara, G.; Saita, E.; Mosconi, P.; Roberto, A.; Fedele, L.; Vercellini, P. Impact of endometriosis on quality of life and mental health: Pelvic pain makes the difference. J. Psychosom. Obs. Gynaecol. 2015, 36, 135–141. [Google Scholar] [CrossRef] [PubMed]
  183. Giudice, L.C.; Kao, L.C. Endometriosis. Lancet 2004, 364, 1789–1799. [Google Scholar] [CrossRef] [PubMed]
  184. Liakopoulou, M.K.; Tsarna, E.; Eleftheriades, A.; Arapaki, A.; Toutoudaki, K.; Christopoulos, P. Medical and Behavioral Aspects of Adolescent Endometriosis: A Review of the Literature. Children 2022, 9, 384. [Google Scholar] [CrossRef] [PubMed]
  185. Burney, R.O.; Giudice, L.C. Pathogenesis and pathophysiology of endometriosis. Fertil. Steril. 2012, 98, 511–519. [Google Scholar] [CrossRef] [PubMed]
  186. Bulun, S.E. Endometriosis. N. Engl. J. Med. 2009, 360, 268–279. [Google Scholar] [CrossRef] [PubMed]
  187. Linton, S.J.; Shaw, W.S. Impact of psychological factors in the experience of pain. Phys. Ther. 2011, 91, 700–711. [Google Scholar] [CrossRef] [PubMed]
  188. Ehde, D.M.; Dillworth, T.M.; Turner, J.A. Cognitive-behavioral therapy for individuals with chronic pain: Efficacy, innovations, and directions for research. Am. Psychol. 2014, 69, 153–166. [Google Scholar] [CrossRef] [PubMed]
  189. Anastasiu, C.V.; Moga, M.A.; Neculau, A.E.; Bălan, A.; Scârneciu, I.; Dragomir, R.M.; Dull, A.-M.; Chicea, L.-M. Biomarkers for the Noninvasive Diagnosis of Endometriosis: State of the Art and Future Perspectives. Int. J. Mol. Sci. 2020, 21, 1750. [Google Scholar] [CrossRef] [PubMed]
  190. Gupta, D.; Hull, M.L.; Fraser, I.; Miller, L.; Bossuyt, P.M.; Johnson, N.; Nisenblat, V. Endometrial biomarkers for the non-invasive diagnosis of endometriosis. Cochrane Database Syst. Rev. 2016, 2016, CD012165. [Google Scholar] [CrossRef] [PubMed]
  191. Gallagher, J.S.; DiVasta, A.D.; Vitonis, A.F.; Sarda, V.; Laufer, M.R.; Missmer, S.A. The Impact of Endometriosis on Quality of Life in Adolescents. J. Adolesc. Health 2018, 63, 766–772. [Google Scholar] [CrossRef]
  192. Evans, S.; Fernandez, S.; Olive, L.; Payne, L.A.; Mikocka-Walus, A. Psychological and mind-body interventions for endometriosis: A systematic review. J. Psychosom. Res. 2019, 124, 109756. [Google Scholar] [CrossRef] [PubMed]
  193. Thorn, B.E.; Eyer, J.C.; Van Dyke, B.P.; Torres, C.A.; Burns, J.W.; Kim, M.; Newman, A.K.; Campbell, L.C.; Anderson, B.; Block, P.R.; et al. Literacy-Adapted Cognitive Behavioral Therapy Versus Education for Chronic Pain at Low-Income Clinics: A Randomized Controlled Trial. Ann. Intern. Med. 2018, 168, 471. [Google Scholar] [CrossRef]
  194. Donatti, L.; Malvezzi, H.; Azevedo, B.C.D.; Baracat, E.C.; Podgaec, S. Cognitive Behavioral Therapy in Endometriosis, Psychological Based Intervention: A Systematic Review. Rev. Bras. Ginecol. E Obs. RBGO Gynecol. Obstet. 2022, 44, 295–303. [Google Scholar] [CrossRef] [PubMed]
  195. Moreira, M.D.F.; Gamboa, O.L.; Pinho Oliveira, M.A. A single-blind, randomized, pilot study of a brief mindfulness-based intervention for the endometriosis-related pain management. Eur. J. Pain 2022, 26, 1147–1162. [Google Scholar] [CrossRef] [PubMed]
  196. Moreira, M.D.F.; Gamboa, O.L.; Oliveira, M.A.P. Mindfulness intervention effect on endometriosis-related pain dimensions and its mediator role on stress and vitality: A path analysis approach. Arch. Womens Ment. Health. 2024, 27, 45–55. [Google Scholar] [CrossRef] [PubMed]
  197. Gonçalves, A.V.; Makuch, M.Y.; Setubal, M.S.; Barros, N.F.; Bahamondes, L. A Qualitative Study on the Practice of Yoga for Women with Pain-Associated Endometriosis. J. Altern. Complement. Med. 2016, 22, 977–982. [Google Scholar] [CrossRef] [PubMed]
  198. Nirgianakis, K.; Egger, K.; Kalaitzopoulos, D.R.; Lanz, S.; Bally, L.; Mueller, M.D. Effectiveness of Dietary Interventions in the Treatment of Endometriosis: A Systematic Review. Reprod. Sci. Thousand Oaks Calif. 2022, 29, 26–42. [Google Scholar] [CrossRef] [PubMed]
  199. Paparella, R.; Panvino, F.; Leonardi, L.; Pucarelli, I.; Menghi, M.; Micangeli, G.; Tarani, F.; Niceta, M.; Rasio, D.; Pancheva, R.; et al. Water-Soluble Vitamins: Hypo- and Hypervitaminosis in Pediatric Population. Pharmaceutics 2025, 17, 118. [Google Scholar] [CrossRef] [PubMed]
  200. Stanek, D.B.; Hestbjerg, I.; Hansen, K.E.; Tomlinson, M.K.; Kirk, U.B. Not “just a bad period”—The impact of a co-created endometriosis social media health campaign: A mixed methods study. Front. Commun. 2023, 8, 1154297. [Google Scholar] [CrossRef]
  201. Das, J.K.; Salam, R.A.; Lassi, Z.S.; Khan, M.N.; Mahmood, W.; Patel, V.; Bhutta, Z.A. Interventions for Adolescent Mental Health: An Overview of Systematic Reviews. J. Adolesc. Health 2016, 59, S49–S60. [Google Scholar] [CrossRef]
  202. Ott, J.; Nouri, K.; Hrebacka, D.; Gutschelhofer, S.; Huber, J.; Wenzi, R. Endometriosis and nutrition-recommending a Mediterranean diet decreases endometriosis-associated pain: An experimental observational study. J. Aging Res. Clin. Pract. 2012, 1, 162–166. [Google Scholar]
  203. Moore, J.S.; Gibson, P.R.; Perry, R.E.; Burgell, R.E. Endometriosis in patients with irritable bowel syndrome: Specific symptomatic and demographic profile, and response to the low FODMAP diet. Aust. N. Z. J. Obs. Gynaecol. 2017, 57, 201–205. [Google Scholar] [CrossRef] [PubMed]
  204. van Haaps, A.P.; Wijbers, J.V.; Schreurs, A.M.F.; Vlek, S.; Tuynman, J.; De Bie, B.; de Vogel, A.L.; van Wely, M.; Mijatovic, V. The effect of dietary interventions on pain and quality of life in women diagnosed with endometriosis: A prospective study with control group. Hum. Reprod. 2023, 38, 2433–2446. [Google Scholar] [CrossRef] [PubMed]
  205. Borghini, R.; Porpora, M.G.; Casale, R.; Marino, M.; Palmieri, E.; Greco, N.; Donato, G.; Picarelli, A. Irritable Bowel Syndrome-Like Disorders in Endometriosis: Prevalence of Nickel Sensitivity and Effects of a Low-Nickel Diet. An Open-Label Pilot Study. Nutrients 2020, 12, 341. [Google Scholar] [CrossRef] [PubMed]
  206. Marziali, M.; Venza, M.; Lazzaro, S.; Lazzaro, A.; Micossi, C.; Stolfi, V.M. Gluten-free diet: A new strategy for management of painful endometriosis related symptoms? Minerva Chir. 2012, 67, 499–504. [Google Scholar] [PubMed]
  207. Nodler, J.L.; DiVasta, A.D.; Vitonis, A.F.; Karevicius, S.; Malsch, M.; Sarda, V.; Fadayomi, A.; Harris, H.R.; A Missmer, S. 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, 229–236. [Google Scholar] [CrossRef] [PubMed]
  208. Mier-Cabrera, J.; Aburto-Soto, T.; Burrola-Méndez, S.; Jiménez-Zamudio, L.; Tolentino, M.C.; Casanueva, E.; Hernández-Guerrero, C. Women with endometriosis improved their peripheral antioxidant markers after the application of a high antioxidant diet. Reprod. Biol. Endocrinol. RBE 2009, 7, 54. [Google Scholar] [CrossRef]
  209. Santanam, N.; Kavtaradze, N.; Murphy, A.; Dominguez, C.; Parthasarathy, S. Antioxidant supplementation reduces endometriosis-related pelvic pain in humans. Transl. Res. J. Lab. Clin. Med. 2013, 161, 189–195. [Google Scholar] [CrossRef]
  210. Qiu, Y.; Yuan, S.; Wang, H. Vitamin D status in endometriosis: A systematic review and meta-analysis. Arch. Gynecol. Obstet. 2020, 302, 141–152. [Google Scholar] [CrossRef]
  211. Harris, H.R.; Chavarro, J.E.; Malspeis, S.; Willett, W.C.; Missmer, S.A. Dairy-Food, Calcium, Magnesium, and Vitamin D Intake and Endometriosis: A Prospective Cohort Study. Am. J. Epidemiol. 2013, 177, 420–430. [Google Scholar] [CrossRef] [PubMed]
  212. Nodler, J.L.; Harris, H.R.; Chavarro, J.E.; Frazier, A.L.; Missmer, S.A. Dairy consumption during adolescence and endometriosis risk. Am. J. Obs. Gynecol. 2020, 222, 257.e1–257.e16. [Google Scholar] [CrossRef] [PubMed]
  213. Qi, X.; Zhang, W.; Ge, M.; Sun, Q.; Peng, L.; Cheng, W.; Li, X. Relationship Between Dairy Products Intake and Risk of Endometriosis: A Systematic Review and Dose-Response Meta-Analysis. Front. Nutr. 2021, 8, 701860. [Google Scholar] [CrossRef] [PubMed]
  214. Parazzini, F.; Chiaffarino, F.; Surace, M.; Chatenoud, L.; Cipriani, S.; Chiantera, V.; Benzi, G.; Fedele, L. Selected food intake and risk of endometriosis. Hum. Reprod. 2004, 19, 1755–1759. [Google Scholar] [CrossRef] [PubMed]
  215. Yamamoto, A.; Harris, H.R.; Vitonis, A.F.; Chavarro, J.E.; Missmer, S.A. A prospective cohort study of meat and fish consumption and endometriosis risk. Am. J. Obs. Gynecol. 2018, 219, 178.e1–178.e10. [Google Scholar] [CrossRef] [PubMed]
  216. Tsuchiya, M.; Miura, T.; Hanaoka, T.; Iwasaki, M.; Sasaki, H.; Tanaka, T.; Nakao, H.; Katoh, T.; Ikenoue, T.; Kabuto, M.; et al. Effect of Soy Isoflavones on Endometriosis: Interaction With Estrogen Receptor 2 Gene Polymorphism. Epidemiology 2007, 18, 402–408. [Google Scholar] [CrossRef]
  217. Harris, H.R.; Eke, A.C.; Chavarro, J.E.; Missmer, S.A. Fruit and vegetable consumption and risk of endometriosis. Hum. Reprod. 2018, 33, 715–727. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A visual representation of the classical diagnostic and therapeutic approach to adolescent endometriosis, emphasizing the sequential steps and multidisciplinary care involved. Abbreviations: MRI, magnetic resonance imaging; NSAIDs, non-steroidal anti-inflammatory drugs.
Figure 1. A visual representation of the classical diagnostic and therapeutic approach to adolescent endometriosis, emphasizing the sequential steps and multidisciplinary care involved. Abbreviations: MRI, magnetic resonance imaging; NSAIDs, non-steroidal anti-inflammatory drugs.
Diagnostics 15 00548 g001
Table 1. Prevalence of endometriosis in adolescents across different articles.
Table 1. Prevalence of endometriosis in adolescents across different articles.
StudyPopulationDiagnostic
Method
Prevalence of Endometriosis
Zannoni et al., 2024 [16]Young women (n = 100, 14–24 years) with chronic pelvic pain (CPP)Ultrasound (US)25% (US-diagnosed endometriosis); higher prevalence in young women (20–24 years) compared to adolescents (14–19 years)
Millischer et al., 2023 [28]Adolescents (12–20 years) with severe dysmenorrheaMagnetic
resonance
imaging
39.3% (121/345) overall; 20.7% (25/121) with ovarian endometrioma (OMA), 88.4% (107/121) with deep infiltrating endometriosis (DIE)
Martire et al., 2023 [29]Young women (12–25 years) with severe dysmenorrheaUS35.3% (131/371) overall; 41.2% (54/131) with OMA, 16.8% (22/131) with isolated endometrioma, and 53.4% (70/131) with posterior DIE
Hirsch et al.,
2020 [30]
Adolescents with CPP undergoing laparoscopyLaparoscopy64% (648/1011; range 25–100%)
Martire et al., 2020 [31]Adolescents (12–20 years) referred for USUSAt least one US feature of endometriosis in 13.3% (36/270); higher detection in adolescents with dysmenorrhea (21%) and dyspareunia (33%)
Yeung et al.,
2017 [32]
Adolescents with laparoscopically proven endometriosisLaparoscopyHistologically confirmed in 39% of cases (448/1148) [Global prevalence: up to 80% of adolescents with CPP who fail to respond to medical treatment]
Janssen et al., 2013 [33]Adolescents with CPPLaparoscopy62% (543/880; range 25–100%)
Opoku-Anane and Laufer, 2012 [34]Young women (12–21 years) with CPP unresponsive to non-steroidal anti-inflammatory drugs and oral contraceptives Laparoscopy98% (115/117); all patients had either stage I or II endometriosis
Table 2. Non-invasive diagnostic biomarkers for endometriosis.
Table 2. Non-invasive diagnostic biomarkers for endometriosis.
Biomarker TypeSample TypeDiagnostic PotentialLimitationsReferences
CA125
(Cancer Antigen 125)
SerumUseful for detecting
advanced disease
in some cases
Low specificity
for early/mild cases
Agic et al., 2008 [111]; Mihalyi et al., 2010 [112]; Socolov et al., 2010 [113]; Vodolazkaia et al., 2012 [114]; Ozhan et al., 2014 [115]; Choi et al., 2019 [116]
MicroRNAs
(e.g., miR-200 family)
SerumEarly diagnostic
potential, stable in
blood, non-invasive
Need for further
validation and
standardization
Teague et al., 2010 [117]; Jia et al., 2013 [118]; Suryawanshi et al., 2013 [119]; Wang et al., 2013 [120]; Cosar et al., 2016 [121]; Nisenblat et al., 2019 [122]; Vanhie et al., 2019 [123];
Cytokines
(IL-6, TNF-α)
SerumCan indicate
inflammation and
the presence of endometriosis
Limited accuracy in differentiating from other diseasesXavier et al., 2006 [124]; Othman et al., 2008 [125]; Seeber et al., 2008 [126]; Socolov et al., 2010 [113]; Choi et al., 2019 [116]
Exosomes/extracellular vesiclesBlood, urine,
menstrual fluid
Contains proteins,
lipids, and RNA
markers for diagnosis
Still in the early stages of researchLi et al., 2020 [127]; Zhang et al., 2020 [128]; Shan et al., 2022 [129]
Menstrual fluid
proteins and genes
Menstrual fluidNon-invasive, specific to endometriosisRequires more
validation and
standardized protocols
Ji et al., 2023 [130]; Amanda et al., 2024 [131]; Starodubtseva et al., 2024 [132]
Vascular Endothelial Growth Factor (VEGF)Serum, urinePotential biomarker for early detection and stagingLimited sensitivity in early stages, affected by other conditionsPotlog-Nahari et al., 2004 [133]; Xavier et al., 2006 [124]; Vodolazkaia et al., 2012 [114]
Long non-coding RNAsSerumEmerging role in the diagnosis and prognosis of endometriosisLimited standardization and variability in findingsWang et al., 2016 [134]; Qiu et al., 2019 [135]
AutoantibodiesSerumCan help in the detection of autoimmune responses associated with endometriosisLack of specificity for endometriosisNabeta et al., 2009 [136]; Gajbhiye et al., 2012 [137]
Apoptotic markers (e.g., caspase-3)SerumProvides insight into endometrial tissue damage and disease progressionNot widely validated for clinical useKaya et al., 2018 [138]
Table 3. Medical and surgical management of symptomatic endometriosis.
Table 3. Medical and surgical management of symptomatic endometriosis.
TreatmentIndicationMechanism of ActionKey Findings
Non-steroidal anti-inflammatory drugsFor pain relief in all stages of treatment (Brown et al., 2017 [150])Temporarily suppress cyclooxygenase (COX)-1 and COX-2 activity, leading to a reduction in prostaglandin synthesisReduces pain effectively without long-term narcotic use [11,151]
Combined hormonal contraceptives (CHC) (oral, vaginal ring, or transdermal)First-line treatment to reduce endometriosis-associated dyspareunia, dysmenorrhea, and non-menstrual pain (Brown et al., 2018 [152]; Jensen et al., 2018 [153]; Grandi et al., 2019 [154])Suppress follicle-stimulating hormone (FSH) and luteinizing hormone (LH), curbing cell growth and promoting endometrial cell deathEffective in reducing symptoms and suppressing endometriosis [11,151]
Progestin-only agents (e.g., dienogest)First-line treatment alternative to CHC, effective in reducing symptoms (Brown et al., 2012 [155]; Petraglia et al., 2012 [156]; Andres et al., 2015 [157])Lower FSH and LH levels, inducing shrinkage or regression of endometrial tissueReduces pain and promotes endometrial suppression [11,151]
Levonorgestrel-releasing intrauterine systemAlternative to CHC, provides localized therapy for symptom management (Lan et al., 2013 [158]; Yoost et al., 2013 [159]; Margatho et al., 2020 [160])Similar to progestin-only agents, with minimal systemic hormone exposureEffective for long-term symptom relief, reduces pain [159]
Gonadotropin-releasing hormone (GnRH) agonistsFor refractory cases, often used with add-back therapy (Brown et al., 2010 [161]; Tang et al., 2017 [162]; Veth et al., 2023 [163])Prolonged use suppresses steroid hormone production by lowering LH and FSH levels, although initially causes a hormone surgeNeed add-back therapy (combination of low-dose hormones) to prevent bone loss and hypoestrogenic symptoms [164,165]
GnRH antagonistsA second-line treatment (e.g., if CHC or progestogens have been ineffective) owing to their side effect profile (Taylor et al., 2017 [166]; Donnez et al., 2020 [167]; Osuga et al., 2021 [168])Competitively binds to GnRH receptors in the pituitary gland, leading to immediate suppression of LH and FSH levels without the initial hormone flare seen with GnRH agonistsSimilar to GnRH agonists [169]
Aromatase inhibitorsFor severe cases resistant to other treatments (Ferrero et al., 2011 [170]; Almassinokiani et al., 2014 [171])Prevents the transformation of androgens into estrogens, diminishing endometrial cell proliferationEffective in combination with CHC, progestins, and GnRH agonists or antagonists [172]
Androgens (e.g., danazol)Second-line treatment for refractory cases or patients with contraindications to other treatments (Ferrero and Barra, 2022 [173])Act as antiestrogens by inhibiting enzymes responsible for steroid production and reducing gonadotropin secretionReduces endometriosis symptoms, but typically avoided in adolescents and young women due to long-term androgenic effects [174]
Selective progesterone receptor modulatorsEmerging therapy for pain and lesion reduction (Fu et al., 2017 [175])Antagonistic effects on progesterone receptors in the endometrium, leading to the suppression of endometrial proliferation and induction of atrophy, contributing to symptom reliefReduces endometrial growth with fewer side effects compared to GnRH agonists; long-term safety data are limited, particularly regarding potential endometrial effects, such as changes in endometrial histology [176]
Surgical managementFor refractory cases with severe pain, organ dysfunction, or infertility risk (Tyson et al., 2024 [177])Removes endometriotic lesions to relieve symptoms and prevent recurrenceEffective in selected cases but requires specialized surgical expertise; postoperative hormonal therapy reduces recurrence [178]
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

Panvino, F.; Paparella, R.; Pisani, F.; Tarani, F.; Ferraguti, G.; Fiore, M.; Ardizzone, I.; Tarani, L. Endometriosis in Adolescence: A Narrative Review of the Psychological and Clinical Implications. Diagnostics 2025, 15, 548. https://doi.org/10.3390/diagnostics15050548

AMA Style

Panvino F, Paparella R, Pisani F, Tarani F, Ferraguti G, Fiore M, Ardizzone I, Tarani L. Endometriosis in Adolescence: A Narrative Review of the Psychological and Clinical Implications. Diagnostics. 2025; 15(5):548. https://doi.org/10.3390/diagnostics15050548

Chicago/Turabian Style

Panvino, Fabiola, Roberto Paparella, Francesco Pisani, Francesca Tarani, Giampiero Ferraguti, Marco Fiore, Ignazio Ardizzone, and Luigi Tarani. 2025. "Endometriosis in Adolescence: A Narrative Review of the Psychological and Clinical Implications" Diagnostics 15, no. 5: 548. https://doi.org/10.3390/diagnostics15050548

APA Style

Panvino, F., Paparella, R., Pisani, F., Tarani, F., Ferraguti, G., Fiore, M., Ardizzone, I., & Tarani, L. (2025). Endometriosis in Adolescence: A Narrative Review of the Psychological and Clinical Implications. Diagnostics, 15(5), 548. https://doi.org/10.3390/diagnostics15050548

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