Abstract
Idiopathic granulomatous mastitis is a benign non-specific inflammatory disease that occurs in women during non-lactation. Main symptoms in the early stages of disease include red breast lumps, swelling, heat and pain, whereas in the later stages, breast abscesses, sinus tracts, and even ulcers may develop. At present, the etiology and pathogenesis of IGM remains unclear, and there are no standardized guidelines for IGM treatment, which leads to treatment resistance and frequent recurrence, causing serious distress to women’s physical and mental health. Based on the existing literature evidence, this paper proposes the “immunity-pathogen imbalance pathogenesis” model, the disease is caused by the imbalance between pathogens and immune system in the breast, resulting in granuloma as the characteristic immune damage response, and Corynebacterium may be the driving factor of the cascade reaction. This article summarizes etiology, diagnostic methods, treatment plans, and recurrence factors, and aims to translate existing mechanisms into a practical clinical framework.
Keywords: idiopathic granulomatous mastitis, etiology, pathogenic bacteria, therapy, antibiotics, methylprednisolone
Introduction
Idiopathic Granulomatous mastitis (IGM) was first described in 1972 by Kessler and Wolloch.1 IGM is a benign and non-specific inflammatory disease that affects non-lactating females. The etiology of IGM remains unclear. However, several risk factors have been associated with it, including autoimmunity, infections, high prolactin levels, and use of oral contraceptives and psychoactive drugs. Main symptoms in the early stages of disease include red breast lumps, swelling, heat and pain, whereas in the later stages, breast abscesses, sinus tracts, and even ulcers may develop. There are currently no standardized guidelines for IGM treatment. Treatment methods such as hormones, antibiotics, immunosuppressive agents and surgery have been reported. However, these methods either require a long treatment time, prolong course of the disease, or lead to easy recurrence. Consequently, patients may develop scar hyperplasia, hyperpigmentation of the breast skin, and breast deformity, causing significant physical and mental distress.
Epidemiology
Idiopathic granulomatous mastitis, a type of non-lactating mastitis, lacks clear data on its incidence rate, which varies by region. Studies conducted in Germany have shown that mastitis accounts for 3% of benign breast diseases2 with an annual prevalence of 2.4 per 100,000 women, and an incidence of 0.37%.3–6 The disease has been frequently reported in China, Turkey, Iran, Spain,7–13 but infrequently in Europe and in the US. Moreover, the disease primarily affects women who are approximately 30 years old after childbirth,14 often within five years of ceasing breastfeeding.15 Idiopathic granulomatous mastitis is rarely reported in men or nulliparous women.16,17 Most patients have high body-mass index (BMI) and increased prolactin, smoke, and use oral contraceptives. The literature reports that BMI value were ≥30, 25–30, and < 25 for 10%, 45%, and 45% of patients, respectively.14,18 A childbearing history was reported for 93% of patients, whereas 7% had no children.9,10,15–19 The time from last lactation to disease onset was > 5 years for 12%, and ≤5 years for 88% of patients.9,14,19–23 93% of cases present with unilateral breast disease, while 7% present with bilateral breast disease. Fifteen percent of patients had a smoking history, whereas 85% did not smoke.9,18,19,21,24,25 A history of oral contraceptives was reported for 23% whereas 77% has not used oral contraceptives.9,18,21,24 Thirteen percent has elevated prolactin levels and 87% had normal levels14,23 (Figure 1).
Figure 1.
Clinical characteristics of patients with granulomatous mastitis.
Etiology
Autoimmunity
Immune Cells and IGM
Innate immune cells, including macrophages and NK cells, exhibit altered functions in IGM. Macrophage differentiate into M1 and M2 subtypes.26 M1 macrophages kill microorganisms and promote inflammation,27 while M2 macrophages secrete cytokines that inhibit inflammation and enable tissue repair.28 M1 and M2 macrophages can transform into each other and maintain a balance between pro-inflammatory and anti-inflammatory responses to ensure homeostasis.29 NK regulate immunity via cytokines or cytotoxins that are crucial for immune protection.30,31 Immunohistochemical studies showed higher expression of M2 (CD68+ and CD163+) macrophages and CD57+ NK cells in IGM versus breast cystic hyperplasia (Figure 2A) IgG4+ plasma cell expression was significantly higher in IGM patients without nipple retraction.29
Figure 2.
Changes of immune system in patients with granulomatous mastitis: (A) The number of CD163-positive M2 macrophages, CD68-positive M2 macrophages and CD57-positive NK cells increased. (B) The secretion of IL-10 by Th2 cells increased. The secretion of IL-22, IL-21 by Th17 cells increased. (C) The secretion of IL-6, IL-23, IL-17, IL-8, IL-33 increased. (D) C3, C5 and MAC numbers increased.
Related studies found that T lymphocytes were predominant in IGM, and a T-cell-mediated reactive inflammation in IGM may be caused by ductal epithelial cell injury and form a lobular-centered granuloma lesion.10 Subsequent studies further confirmed the dominance of T lymphocytes in the inflammatory microenvironment in IMG, highlighting their importance in IGM pathogenesis.32 Based on their function, T cells can be divided into T helper (Ths) and regulatory T cells (Treg).33,34 Treg cells were reported to play a suppressive role in the immune system, and any disruption in the function of Treg cells may cause immune imbalance, and result in immune injury to the body,35 as in giant cell arteritis (GCA) and other autoimmune vasculitis.36 Studying exploring differences in Tregs and B cells across IGM subgroups found variation in Treg expression. Specifically, Treg levels differed between the and remission stages of IGM patients as well as between IGM patients and healthy controls. However, there was no significant difference in the regulatory B cell subpopulations.37 Hence, changes in regulatory T cell subpopulations and quantities appear integral to the immune response of the mammary glands. Another study found that the percentage of lymphocytes, percentage of T helper lymphocytes, and the ratio of CD4/CD8 in the IGM group were lower than those in the healthy control group. The counts and percentages of white blood cells and neutrophils, absolute values of monocytes and cytotoxic T lymphocytes, and the percentages and absolute values of natural killer cells (NK cells) and natural killer T cells (NKT cells) were also found to be higher than those in the healthy control group.32
TREM-1 is a triggering receptor expressed in myeloid cells. In autoimmune disorders, the expression of TREM-1 in myeloid cells increases,38–40 promoting the secretion of monocyte chemoattractant protein-1, monocyte chemoattractant protein-3, interleukin-6, interleukin-8, and other pro-inflammatory factors.38 The level of TREM-1 in IGM patients was found to be higher than that in the normal control group in both the active and remission stages.38
Cytokines
Cytokines constitute an integral part of immune regulation, produced by immune and non-immune cells, with activated T cells as the main source.41–44 Cytokines mediate immunity, inflammation,45,46 and immune injury. In previous studies, levels of inflammatory cytokines IL-22 (Figure 2B) and IL-23 (Figure 2C) were found to be significantly increased in IGM patients.47 Moreover, serum interleukin (IL)-6 level in IGM patients was found to be significantly higher than that in healthy controls, and the serum IL-6 and C-reactive protein (CRP) levels in patients with severe IGM were significantly higher than those in patients with mild to moderate IGM.48 Similarly, the IL-33 and sST2 levels were significantly higher in the IGM group than in the healthy control group.49
In another study, IL-10 (Figure 2B), IL-17 (Figure 2C) and IL-8 (Figure 2C), levels in IGM patients were found to be higher than those in healthy controls.50 IL-17 induces neutrophilia by increasing the expression of granulocyte colony-stimulating factor (GCSF) and responding to neutrophil chemokines.51 IL-17 also induces chemokine production in lymphocytes, dendritic cells and monocytes.52 IL-8 is produced by a variety of cells, but is released only under inflammatory conditions.53 IL-8 also plays a pivotal role in attracting neutrophils to sites of inflammation.54 Interestingly, IL-10 is mainly produced by Th2 cells yet has a significant negative immunoregulatory effect.55 HLA-A*10, HLA-A*24:03, HLA-B*18, and HLA-DR*17 frequencies were also found to be high in IGM patients. In addition, the frequencies of HLA-A*10, HLA-A*24, HLA-A*32, HLA-B*18, HLA-B*103, and HLADR*17 were found to be higher in patients with recurrent IGM than in those without recurrence.56 High frequencies of HLA-A*24:03 and HLA-B*18 may in particular suggesting an association of IGM with humoral and auto immunity.56
Complement Activation Mediates Immune Injury
The membrane attack complex (MAC) is the terminal component of the complement activation pathway,57 which triggers inflammatory response.58,59 Elevated MAC levels lead to apoptosis and death.60,61 MAC was reported at elevated levels in the peripheral blood and tissues of IGM patient (Figure 2D), and was considered a key substance causing injury to mammary ductal epithelial cells in IGM.62 IGM pathology involves the loss of the acinar structure and ductal injury.63,64 This study confirmed that the abnormally activated complement pathway in patients with IGM increases MAC, which activates other immune mechanisms and causes mammary ductal epithelial cell injury. Another study reported elevated levels of C3/C3a-C3aR and C5/C5a-C5aR1 in IGM tissues65 (Figure 2D). The increased expression levels of the MAC, C3, and C5 indicate that complement-mediated activation contributes to cell damage in IGM, further evidencing complement-mediated immune injury (Figure 2).
Pathogen Presence
In 1985, a study66 found that patients with nipple discharge caused by mammary duct ectasia had a higher rates of secretion than those without nipple discharge. Therefore, certain pathogens are considered to be closely-associated with this disease. Corynebacterium was considered the main bacterial flora that causes IGM, as it was detected in 41–60% of patients in previous studies.67,68 In another study found Corynebacterium in all patients, with 57.89% of whom having Corynebacterium kroppenstedtii.69 In similar studies, Corynebacterium also had the highest detection rate among other pathogens.70–72 Corynebacterium is a recognized cause of livestock diseases.67,73 However, improvements in detection technology revealed the presence of not only Corynebacterium, but also other pathogenic microorganisms and viruses as well. In accordance with a previous report,19 24 genera, including Ochrobactrum, Delftia, Streptococcus, and Peptoniphilus (Table 1) were detected in samples. Epstein-Barr virus has been detected in IGM samples68 (Table 1). Thus, IGM pathogens usually occur as mixed infections. It remains unclear if the diversity of bacterial genera underlies antibiotic resistance and recurrence in IGM.
Table 1.
Pathogenic Factors of Idiopathic Granulomatous Mastitis
| Factors | Type | Detection Method | Ref |
|---|---|---|---|
| Pathogen | Propionibacterium | 16S rRNA sequencing | [19] |
| Staphylococcus | |||
| Corynebacterium | |||
| Delftia | |||
| Pseudomonas | |||
| Streptococcus | |||
| Finegoldia magna | |||
| Serratia | |||
| Fusobacterium | |||
| Ochrobactrum | |||
| Peptoniphilus | |||
| Acinetobacter | |||
| Prevotella | |||
| Enhydrobacter | |||
| Anaerococcus | |||
| Pathogen | Epstein-Barr virus | Metagenomic Next-Generation Sequencing | [68] |
| Corynebacterium | |||
| Acinetobacter baumannii | |||
| Thermophilic bacteria | |||
| Streptococcus pneumoniae | |||
| Pseudomonas oleovorans | |||
| Klebsiella | |||
| Staphylococcus |
The role of infections in IGM remains unclear. Owing to the anatomical structure and function of the mammary gland, certain bacterial flora can be detected in the breast tissue under normal conditions,74 such as Lactobacillus, Bacteroides, Anaerococcus, Staphylococcus. However the pathogenic role of isolated bacterial species is also not yet clear, complicating distinctions between infection, colonization, and contamination.75–79 While studies find microorganisms in breast tissue, Corynebacteria – dominant in IGM – constitute a low proportion of normal tissue flora.
Genes and Signal Pathways in Idiopathic Granulomatous Mastitis
Previous studies examined miRNAs expression in tissues IGM patients, finding significantly higher serum miR451a and miR55713p levels than those in healthy individuals.80 Another study analyzed 12,115 mRNAs, with 207 differentially-expressed (136 upregulated and 71 downregulated) between GM and normal tissues.81 Overexpressed GM genes also correlated with breast cancer, suggesting that chronic inflammatory stimuli are risk factors for breast cancer. Another study also found that the C677T polymorphism in the methylenetetrahydrofolategene was closely related to granulomatous lobular mastitis.82
In some studies,51 human plasma cell mastitis tissue was converted into an oil emulsion and injected into mice to confer pathological features that were highly similar to those of human plasma cell mastitis. Thus, a mouse model of plasma cell mastitis was established. Later studies injected IL-6 into this model, and found that the IL-6/JAK2/STAT3 signaling pathway was activated. Mice treated with the signaling pathway inhibitor (AG-490) also showed significantly fewer mammary plasma cells and controlled inflammation.83
Exosomes are involved in in the regulation of inflammatory responses and immune balance.84,85 In IGM cells, the expression of exosomes and the activity of the signaling pathways mediated by them are increased. A previous study86 found that the secretion of exosomes in the PCM was increased, and the PI3K-Akt-mTOR signaling pathway was activated. An injection of an exosome inhibitor (GW4869) to into the PCM mouse model revealed that the structure of the mammary ducts in mice was improved, and the infiltration of plasma cells and lymphocytes was decreased.87 Another study demonstrated that exosome MiR-221 derived from mammary epithelial cells mediated the polarization of M1 macrophages through SOCS1/STATs, and promoted the development of local mammary inflammation.88 These findings on exosomes and signaling pathways provide further evidence regarding the immune injury mechanism in IGM. The inhibition of exosome secretion can significantly inhibit the infiltration of inflammatory cells, and may provide with a viable therapeutic treatment strategy in the future.
Other Factors
Elevated serum prolactin levels, and use of oral contraceptives and psychoactive drugs are considered risk factors for IGM. However, these associations are based on relatively few studies, and the underlying mechanisms are still unclear.89–91 Prolactin (PRL) is a neuroendocrine polypeptide hormone that stimulates DNA synthesis and epithelial cell proliferation, increases milk yield in the mammary glands, and thus plays a role in the pathogenesis of mastitis.92 A retrospective study also reported elevated serum prolactin levels in patients with IGM.68 Two studies93,94 reported development of IGM in nonpregnant patients due to drug-induced hyperprolactinemia. Previous studies also indicated the rate of oral contraceptive use in IGM patients can reach up to 54.79%.95 Additionally, the inhibitory effect of antipsychotic drugs on dopamine increases prolactin secretion of prolactin.96 A study reported97 that 19 cases of IGM were caused by long-term oral psychoactive drugs, and other studies have also suggested that hyperprolactinemia induced by psychoactive drugs is related to IGM.98,99 Elevated prolactin levels are caused by disease and drug-related factors. Diseases include pituitary adenoma, craniopharyngioma, spontaneous elevation, and drug factors, including antidepressants and antipsychotics (Table 2). Therefore, imbalances in the secretion of estrogen, progestogen, or prolactin, either inherent or drug-induced, can alter the microenvironment of the mammary gland. This may induce immune and bacterial dysregulation, thereby promoting mammary duct injury and inflammation100 (Table 1).
Table 2.
Risk Factors of Hyperdense Prolactinemia
| Factors | Type | Number | Ref |
|---|---|---|---|
| Disease Factors | Pituitary adenoma | 2 | [89] |
| Craniopharyngioma | 1 | ||
| Spontaneous elevation | 1 | ||
| Drug Factors | Prednisolone, Azathioprine, alfacalcidol, Sulpiride | 1 | [93] |
| Fluphenazine, Zolpidem, Duloxetine | 1 | ||
| Risperidone | 1 | [94] | |
| Flufenazine, Zolpidem, Duloxetine, Risperidone, Risperidone | 6 | [97] | |
| Risperidone+Clozapine | 4 | ||
| Aripiprazole+Fluvoxamine | 2 | ||
| Clozapine | 1 | ||
| Olanzapine | 2 | ||
| Olanzapine+ Ziprasidone+Quetiapine | 2 | ||
| Aripiprazole+Sulpiride | 2 | ||
| Risperidone | 1 | [101] |
Diagnosis
Imaging diagnosis
Imaging examinations included breast ultrasound, mammography and breast magnetic resonance imaging. However, the imaging findings of IGM are highly similar to breast cancer, and it is often difficult to distinguish it from breast cancer.102
Breast ultrasound is cost-effective and convenient to perform, making it the preferred choice for imaging examinations. Under ultrasound, the IGM demonstrated solitary or multiple hypoechoic regions with irregular configurations and indistinct boundaries, which were tubular extensions or cavities containing foam debris and separation.103–106 In advanced cases, due to the liquefaction of necrotic tissue, the echo patterns become diverse and may form abscess cavities or fistulas.107,108 IGM is similar to breast cancer under ultrasound, but its diameter is often larger than that of breast cancer.108,109
IGM can be manifested as single or multiple irregular masses with increased density and blurred boundaries in mammography, which is characterized by focal asymmetry.103,110,111 Related clinical manifestations also include skin thickening and axillary lymph node enlargement.103 Mammography is superior to breast ultrasound in the identification of calcification, but it is necessary to compress the breast during the examination to increase the pain of the affected breast. Patients may discontinue the examination due to severe pain. In the process of strong extrusion, it even increases the risk of inflammation diffusion.
Magnetic resonance imaging (MRI) can show the range of the mass and its relationship with the surrounding tissue more clearly. Heterogeneous enhancement mass or edge enhancement lesion is the most common imaging manifestation of IGM, which may also be accompanied by segmental or regional non-mass enhancement.109–112 Imaging examination is often considered to lack specificity.113 Finally, further pathological biopsy is needed to confirm the diagnosis.
Pathological Diagnosis
Pathological diagnosis is still the gold standard for the diagnosis of immune granuloma (IGM), which can make up for the deficiency of imaging examination. Correct pathological diagnosis depends on high-quality samples. Fine needle aspiration and biopsy needle aspiration are two commonly used pathological diagnosis methods. Studies have reported that the sensitivity of tissue samples for fine needle aspiration is 21.1%, and the sensitivity of tissue samples for biopsy needle aspiration is 96%.114,115 The reason for the difference may be the difference in tissue volume.114,115 Compared with fine needle aspiration, the samples obtained by biopsy needle puncture are more ideal in quality and quantity. In addition, the space between the inflammatory tissues can be opened while the biopsy needle is punctured, which promotes the discharge of pus.
Pathological features lobular-centered, non-necrotizing granulomas with multinucleated giant cells, epithelioid histiocytes, lymphocytes, plasma cells and neutrophils.116 The parenchyma usually manifests as a loss of acinar structure and ductal injury.63–65 Recent studies have also discovered a special pathological type characterized by a well-defined vacuole in the center of the granuloma surrounded by neutrophils. Corynebacterium can be seen under Gram stain, and IGMwith this pathological feature is thus named “cystic neutrophilic granulomatous mastitis”.73 However, whether this form of IGM is a subtype of idiopathic granulomatous mastitis or a separate type of mastitis remains controversial.117,118
The pathological diagnosis of IGM should be distinguished from breast tuberculosis. Lacambra et alshowed that IGM is characterized by a significant increasein plasma cells, while tuberculosis changes are characterized by fibrosis, eosinophilia, and necrosis.118
Pathogen Detection
Current pathogen detection methods include traditional culturing, first-generationsequencing (Sanger sequencing), next-generation sequencing (NGS), and nanopore-targeted sequencing (NTS). Conventional culture method is commonly used to detect pathogenic bacteria in clinic. However, the positive detection rate remains low for unknown pathogens or those with highly stringent growth requirements. For example, Corynebacterium kroppenstedtii, which needs at least 72 hours ofculture time, grows poorly on ordinary medium,67,119 but prolongs the culture time and grows well in the same medium supplemented with 1% Tween 80.67,120,121
NTS being the latest gene sequencing technology.113,122 Compared to traditional next-generation sequencing, NTS has the advantages of ultra-long reads, high sensitivity/accuracy, simultaneous detection of DNA and RNA, and the ability to report within 24 h.117 NTS is also capable of meeting rapid, comprehensive, and accurate clinical needs. Previous studies have shown that the early detection rates of nanopore-targeted sequencing are 91.7% and 94.4%, respectively.20,118 In clinical settings, NTS is currently used to assist the diagnosis of endophthalmitis, respiratory tract infections, meningitis, among other diseases.118 A recent study using NTS to detect pathogenic bacteria in IGM showed that this method achieves a higher bacterial detection rate than that of the culture method, with the dominant bacteria being Corynebacterium species (64%), especially Corynebacterium kroppenstedtii.20
Treatment
There is currently no standard treatment for IGM. Methods used in IGM treatment can be divided into conservative and surgical treatment categories. Conservative treatment options include hormone therapy, antibiotics, immunosuppressants. Surgical treatments included drainage and extended resection. Debate surrounds optimal treatment for IGM, including the appropriateness of conservative versus surgical approaches, treatment effects, and recurrence rates. An overview of treatment methods and outcomes is presented below.
Hormone Treatment
Drug therapy is the first choice for initial treatment of IGM, and a large number of reports have reported the use of hormones as the first-line treatment.119 Hormone therapy includes administration of systemic oral hormones, local injections, and steroids. No standard procedure for the dosage and duration of treatment exists to date. A clinical trial120 reported the oral administration of prednisone 32 mg (16 mg bid) daily, with the dose being tapered until the medication was stopped after 2 months. All patients were found to enter remission, and a recurrence rate of 11.9% was observed. Another clinical trial121 reported that 20 mg of methylprednisolone was administered orally per day, and the dose was gradually reduced until the condition of the patients stabilized. A total of 80.7% of patients responded well to steroid therapy, and 33% experienced recurrence thereafter. However, the efficacy of glucocorticoids such as methylprednisolone varies between studies. This compound can completely revert the symptoms and prevent recurrence; however, it can also lead to poor treatment response. During treatment, a dose reduction can lead to disease progression or recurrence after the end of treatment. Several studies also showed that, upon reducing the dosage levels of hormones, an approximately 50% recurrence rate was obtained123 (Table 3).
Table 3.
Treatment of Idiopathic Granulomatous Mastitis
| Treatment Method | Number | Medication | Remission | No Remission | Recurrence | No Recurrence | Side Effect | Ref |
|---|---|---|---|---|---|---|---|---|
| Bromocriptine+ drainage | 3 | NA | 3 | 0 | 3 | 16 | NA | [97] |
| Bromocriptine+ corticosteroid+ surgery | 4 | Methylprednisolone 20 mg/day | 4 | 0 | NA | |||
| Bromocriptine+ corticosteroid | 6 | Methylprednisolone 20 mg/day | 6 | 0 | NA | |||
| Bromocriptine+ glucocorticoids+antibacterials | 3 | Rifampicin 0.45g, Isoniazid 0.3g, Pyrazinamide 0.75g/day+Methylprednisolone 20mg/day | 3 | 0 | NA | |||
| Bromocriptine+ surgery | 2 | NA | 2 | 0 | NA | |||
| Bromocriptine+ corticosteroid+ antibacterials+ surgery | 1 | Rifampicin 0.45g, Isoniazid 0.3g, Pyrazinamide 0.75g/day+Methylprednisolone 20mg/day | 1 | 0 | NA | |||
| Hormones+ antibiotics | 21 | Methylprednisolone 16mg, twice a day, dose reduced after 2 weeks for 2 months+unknown antibiotics | 21 | 0 | 4 | 17 | NA | [120] |
| Hormones | 36 | Methylprednisolone 16mg, twice a day, dose reduced after 2 weeks for 2 months | 36 | 0 | 8 | 28 | NA | |
| Hormones+ surgery | 9 | Methylprednisolone 16mg, twice a day, dose reduced after 2 weeks for 2 months | 9 | 0 | 1 | 8 | NA | |
| Hormones+ immunosuppressive drugs | 2 | Methylprednisolone 16mg, twice a day, dose reduced after 2 weeks for 2 months+Azathioprine | 2 | 0 | 0 | 0 | NA | |
| No treatment | 50 | NA | 50 | 0 | 1 | 49 | NA | |
| Hormones | 72 | Methylprednisolone 20g per day. After achieving complete remission (CR) or stable disease (SD), the dose of methylprednisolone reduced by 4mg every 2–4 weeks. During steroid treatment, patients undergo regular outpatient clinical visits every 2–4 weeks. | 30 | 9 | 21 | 42 | Cushing’s syndrome+obesity | [121] |
| Low-dose hormones | 15 | 5mg per day for two months | 8 | 7 | 3 | 12 | NA | [124] |
| High-dose hormones | 15 | 50mg for 3 consecutive days, 25mg for 3 consecutive days, then 12.5mg for 3 consecutive days, followed by 5mg daily for 2 months. | 14 | 1 | 0 | 0 | NA | |
| Surgery | 17 | Surgical removal of inflamed breast tissue at healthy edges | NA | NA | 7 | 10 | NA | [125] |
| High-dose hormones | 23 | High-dose Prednisolone (50mg), twice a day for two consecutive weeks, gradually reduced to 50mg, 25mg, 10mg and 5mg per day according to the treatment response and clinical examination. | NA | NA | 9 | 14 | 7 cases of hyperglycemia, 8 cases of headache, 6 cases of hair loss, 2 cases of mild blurred vision | |
| Low-dose hormones+ drainage | 47 | Steroids 30mg/day for 2 weeks, tapered to 20mg/day for 4 weeks, 10mg/day for 2 weeks, and then 5mg/day for 2 weeks. | NA | NA | 9 | 38 | 4 cases of mild hypoglycemia, and 5 cases of gastrointestinal problems such as stomach pain. | |
| Observation | 28 | 28 | 0 | 28 | 0 | NA | [126] | |
| Local injection | 12 | 40 (Triamcinolone acetonide 40mg/mL), ranging from 2mL (80mg) to 4mL (160mg) | 12 | 0 | 12 | 0 | NA | |
| Surgery | 9 | 9 | 0 | 9 | 0 | NA | ||
| Local injection of steroids+skin application of steroid cream | 46 | Triamcinolone acetonide (40mg/mL), 20mg/cm3, once a month+local administration of 0.1% triamcinolone acetonide, twice a day to treat the skin of breast lesions, every other day, for one month | 37 | 9 | 4 | 42 | 1 case of local skin thinning | [24] |
| Oral hormones | 32 | Methylprednisolone 32mg/day for one month | 23 | 9 | 15 | 17 | 3 cases of hirsutism, 1 case of weight gain | |
| Skin application of steroid cream | 41 | Prednisolone 0.125% cream, twice a day until complete remission | 34 | 7 | 5 | 36 | 10 patients with local side effects, 1 patient with skin cellulitis, 9 patients with thinning/dry skin and telangiectasia, 1 patient with systemic side effects, and 1 patient with weight gain±hirsutism | [127] |
| Oral hormones | 34 | 0.8 mg/kg once after meals | 29 | 5 | 6 | 28 | 13 patients with systemic side effects, 11 patients with weight gain±hirsutism, and 2 patients with iatrogenic treatment | |
| Oral hormones+local skin application of hormone cream | 33 | Prednisolone 0.125% cream, twice a day | 27 | 6 | 6 | 27 | 7 patients with local side effects, 2 patients with skin cellulitis, 5 patients with skin thinning/dryness and telangiectasia, 10 patients with systemic side effects, 9 patients with weight gain±hirsutism, 1 patient with iatrogenic treatment | |
| Antibiotics | 19 | Amoxicillin plus Clavulanic acid | 29 | NA | 2 | 29 | NA | [21] |
| 5 | Sodium fusidate | |||||||
| 1 | Azithromycin | |||||||
| 1 | Ciprofloxacin+ Ornidazole | |||||||
| 5 | Others | |||||||
| Antibiotics | 30 | Rifampicin at a dose of 300mg, twice a day | 30 | 0 | 0 | 30 | NA | [128] |
| Antibiotics | 22 | Doxycycline at a dose of 100 mg, twice a day | 16 | 6 | NA | NA | NA | [23] |
| Immunosuppressive drugs | 60 | Take methotrexate monotherapy 15mg/week for 24 consecutive weeks, and folate 10mg/week as a supplement for all patients. | 52 | 8 | 52 | 8 | NA | [129] |
| Immunosuppressive drugs+ hormones | 41 | The median dose of methotrexate is 15mg/week, respectively | 41 | 0 | NA | NA | Gastrointestinal symptoms | [18] |
| 3 | Azathioprine 125 mg/day | 2 | 1 | NA | NA | |||
| 3 | Steroid hormone 40 mg/day | 3 | 0 | NA | NA | |||
| Immunosuppressive drugs+ surgery | 4 | NA | 4 | 0 | NA | NA | ||
| Immunosuppressive drugs | 19 | Methotrexate is administered at a dose of 10–15mg/week, increasing to 20–25mg/week per oral (PO) or subcutaneous (SC) based on clinical response. | 12 | 5 | 3 | 14 | Two cases (10.5%) of nausea, and one case (5.2%) of elevated liver function test | [130] |
| Immunosuppressive drugs+ hormones | 17 | Methotrexate 5mg/week+prednisone 8mg/day for 2–3 months | 13 | 4 | 0 | 17 | NA | [131] |
| Hormones+minimally invasive rotary resection | 30 | The initial dose of methylprednisolone is 20mg/day, reduced to 16, 12, 8 and 4mg/day every 1–2 weeks until discontinuation. | 26 | 4 | 3 | 27 | One case of postoperative bleeding | [132] |
| Vacuum sealing drainage (VSD) | 30 | NA | 30 | 0 | 2 | 28 | NA | [133] |
| Ultrasound-guided precise debridement | 29 | NA | 29 | 0 | 2 | 27 | NA | |
| Minimally invasive comprehensive treatment | 69 | Perform pus drainage, lesion resection and drainage tube placement after minimally invasive surgery, and iodophor, metronidazole, and dexamethasone are used to flush the lesion after the operation | 69 | 0 | 7 | 0 | NA | [134] |
| Wide resection+ skin flap transplantation technology | 68 | NA | 68 | 0 | 3 | 65 | NA | [25] |
| Surgery+Yanghe Decoction Scheme | 28 | Yanghe Decoction is a traditional Chinese medicine made from degelatined antler, licorice, cinnamon, processed ginger root, white mustard seeds, rehmannia glutinosa and ephedra herb (honey-fried). | 27 | 1 | NA | NA | NA | [135] |
| IEPED+surgery | 60 | IEPED is made from 12 grams of atractylodes macrocephala, 12 grams of pangolin, 12 grams of ginseng, 12 grams of angelica sinensis, 9 grams of cohosh, 9 grams of licorice, 15 grams of angelica sinensis, 15 grams of astragalus membranaceus, 10 grams of licorice, and 10 grams of dried green orange peel. | 54 | 6 | 0 | 60 | NA | [136] |
| Bromocriptine | 1 | Bromocriptine 2.5mL, twice a day | 1 | 0 | 0 | 1 | NA | [101] |
A previous study has compared the efficacy and recurrence rates of different doses. The remission rate with 5 mg of oral prednisolone daily was 53.3% and the recurrence rate was 37.5%. However, after oral prednisolone 50 mg on Day 1–3, 25 mg on Day 4–6, 12.5 mg on Day 7–9, and then maintained at 5 mg, the remission rate was 93.3% without recurrence.124 However, another study found that higher doses were ineffective.125 The study reported a 39.1% recurrence rate with a daily tapering from 100 mg to 5 mg, whereas 30 mg tapered daily to 5 mg with drainage had a recurrence rate of 19.1%. Therefore, the choice of dosage remains key to treatment. According to previous studies, patients treated with high doses have high remission and low recurrence rates. However, excessively high doses not only do not reduce the recurrence rate, but also increase the risk of recurrence and are likely to cause serious steroid-related complications such as Cushing’s syndrome, weight gain, dyspepsia, hyperglycemia, hypertension, and blurred vision.111,125,137 These complications lead to poor patient compliance and self-discontinuation of medications, resulting in treatment failure. However, extremely low doses have a poor remission effect and may lead to disease progression and recurrence.124 Therefore, excessively high and low doses were not conducive to disease control (Table 3).
In addition to systemic oral steroid therapy, local steroid injections and topical hormone creams are also effective in IGM treatment. Local treatment with injectable steroids is based on the well-established injection-based procedure to treat diseases such as arthritis.126 According to previous reports, local injection of 80–160 mg triamcinolone acetonide (40 mg/mL) achieved remission after an average of 2 months.126 Another study reported that local injection of triamcinolone acetonide (20 mg/cm3) in addition to skin application of 0.1% triamcinolone acetonide cream resulted in a remission rate of 93.5% and a recurrence rate of 8.7% after three months.24 Moreover, 0.125% prednisolone cream was applied topically twice daily until clinical remission, with a remission rate of 82.9% and a recurrence rate of 14.7%.127 Nevertheless, local injection and skin application of steroids have certain side effects, mainly manifested as local skin thinning, dryness, local hematoma, skin telangiectasia, and secondary skin infection cellulitis.24,127,138 Hence, compared to systemic treatment, local treatment can achieve higher effectiveness, with side effects mostly localized and lighter than those of systemic treatment and higher patient compliance (Table 3).
Antibiotic Therapy
Antibiotics are also commonly-used in IGM treatment, yet there are no standard procedures for medication and dosage adjustment. A total of 206 patients in Iran were treated with cloxacillin, cefalexin, ciprofloxacin, or clindamycin for 20 days. Only 6 (3%) patients entered remission.139 In another study, 31 patients were treated with a combination of amoxicillin and clavulanic acid, sodium fusidate, azithromycin, and a combination of ciprofloxacin ornidazole, of which 29 achieved complete remission, and two patients experienced recurrence. The average remission time was 2.5 months.21 In another study, 17 of 21 patients achieved complete remission after antibiotic treatment, four recurred in subsequent follow-up. No time of remission was not reported.120 In an 11-year single-center study, only nine of 17 patients treated with antibiotics alone reported remission, with an addition four subsequently entering remission.22 Antibiotic therapy thus remains controversial due to its variable therapeutic effects. The reasons behind this may include the complexity of pathogens, gradual resistance of pathogens to antibiotics, and the length of treatment time (Table 3).
Corynebacterium has been reported to show resistance to several drugs.140 Corynebacterium is a lipophilic bacterium that relies on lipids for its growth and reproduction. The bacterium is resistant to hydrophilic antibiotics, such as B-type amide (penicillin) and quinolone penicillin (levofloxacin).71,141 The resistance rate of IGM infected with Corynebacterium to penicillin can be as high as 70%.22 Most Corynebacterium strains were found to be sensitive to rifampicin, co-trimoxazole, doxycycline, linezolid, and vancomycin.1,13,140,142,143 In a study that used rifampicin as the first-line treatment, 18 and 12 out of 30 patients showed remission at 6 and 9 months, respectively. The median follow-up time was 15.5 months with no disease recurrence.128 Another study used doxycycline as the first-line drug for IGM, and 50% of the patients achieved complete remission after treatment.23 In general, Corynebacterium species were associated with a longer duration of treatment and a higher risk of recurrence.22,144,145 Corynebacterium has been reported to require a shorter treatment duration in patients receiving anti-Corynebacterium therapy than in those who did not receive treatment.144 Therefore, anti-Corynebacterial treatment is a key step that should not be ignored. Other studies have also shown that the recurrence rate of antibiotics plus hormone therapy is lower than that of hormone therapy alone, indicating that antibiotic therapy plays an important role in reducing the duration of treatment and recurrence rate142 (Table 3).
Immunosuppressive Agent Therapy
The most commonly used immunosuppressive drug used in IGM treatment is methotrexate (MTX), followed by azathioprine.18,146 The main mechanism of action of methotrexate is the inhibition of the transformation of folate into tetrahydrofolic acid, which participates in cell proliferation. Methotrexate also inhibit other folate-dependent enzymes and exert anti-inflammatory effects.147 It is also used to treat psoriasis, rheumatoid arthritis, and other diseases.147 However, methotrexate usage is less common than that of antibiotics or corticosteroids in IGM treatment, and is usually considered an alternative in case of cortisol resistance or recurrence.148 The largest MTX monotherapy studies to date have shown that after 6 months of MTX treatment in 64 patients, 81.25% achieved complete remission.129 In a study by Postolova et al,130 patients for whom hormones, antibiotics, and surgery failed, were treated with methotrexate alone for a median treatment time of 15 months. A total of 75% of the patients achieved remission, 94% showed improvement, and 15.8% showed improvement after switching to subcutaneous administration (Table 3).
Immunosuppressive drug treatment combined with steroid hormones can achieve certain level of effectiveness with lower steroid hormone doses and less side effects. In such a study, 41, 3, 3, and 4 out of 51 patients received methotrexate, steroid hormones, azathioprine and steroid hormones, and only steroid hormones, respectively. Among them, 50 patients achieved complete or partial remission, and one patient developed thrombocytopenia and Sjögren’s syndrome after using azathioprine. No recurrence was observed in any of the patients.18 Another study reported that 80 patients who failed initial treatment with methylprednisolone were treated with methotrexate (7.5 mg/week) and low-dose methylprednisolone (4–8 mg/day). Among them, 44 patients achieved complete remission, 17 achieved partial remission, and subsequent surgical treatment achieved complete remission. Only five patients experienced mild hair loss, and no other side effects were observed.148 In addition, 17 patients were treated with 5 mg/week methotrexate and 8 mg/day low-dose prednisone. After 3 months of treatment, 10 patients achieved complete remission, 3 achieved partial remission and subsequent complete remission after surgery, 4 showed no response to treatment, and no side effects occurred131 (Table 3).
Surgical Treatment
Surgical treatment is an option for patients for whom conservative treatment fails, recurrence is observed within a short period, or for those who refuse treatment with steroid hormones. In patients with pus in the lesion, traditional incision and drainage, needle aspiration, or needle or catheter drainage can be performed. However, the use of drainage alone has been reported in only a few cases, which may explain the higher recurrence rates associated with traditional drainage.21 Lesions that cannot be cured by drainage or appear as masses can be surgically removed. The surgical methods used for this purpose include minimally invasive rotary resection,134 breast lesion resection,149 extended lumpectomy,149 lumpectomy plus skin flap transplantation,25 and nipple-areola complex-sparing subcutaneous mastectomy and prosthesis implantation150(Table 3).
Minimally invasive surgery is feasible when drug treatment results in no acute lesions or when the lesions shrink. In one study, the lesions of 30 patients were divided into four types under ultrasonography: 0 cases of diffuse type, 17 cases of patchy hypoechoic type, 9 cases of localized abscess type, and 4 cases of localized hypoechoic mass type. A minimally invasive rotary resection was performed after glucocorticoid treatment, which cured 26 patients (86.7%), whereas 1 patient had postoperative bleeding, and 3 patients had recurrence (10.00%). All 3 recurrent cases were hypoechoic.132 In addition, another study reported that patients treated with vacuum sealing drainage (VSD) and ultrasound-guided precise debridement were cured, with an overall recurrence rate of 6.8%, an overall new incidence rate of 8.5%, and an average disease duration of 5.3 months.133 Another study proposed a minimally-invasive comprehensive treatment. In this study, 69 patients underwent pus drainage, lesion resection, and drainage tube placement after minimally invasive surgery. Iodophor, metronidazole, and dexamethasone were used to flush the lesion after the operation. All patients were cured, and seven (10.17%) patients experienced recurrence after a year of follow-up134 (Table 3).
If there are multiple fistulas and large ulcers on the surface of a patient’s breast, skin flap transplantation technology can be used to repair the wound after wide resection. In such a surgical study, a random breast dermo-glandular flap (BDGF) was used to correct tissue defects, with only three patients experiencing recurrence after surgery (3/68, 4.4%).25 For patients with large lesions that cannot be completely relieved or even progress with other treatments, to avoid scarring caused by multiple breast surgeries, subcutaneous mastectomy and prosthesis implantation can be performed to avoid scarring caused by multiple breast surgeries, and to obtain an aesthetic effect150 (Table 3).
However, the effectiveness of the surgical treatment remains controversial. According to statistics, the recurrence rate after surgical treatment ranges between 5% and 50%,3,151 and the highest reported incidence rate is 72.7%.151 However, other studies reported that patients who do not respond to conservative treatment can achieve complete remission without recurrence after surgery.139 A meta-analysis noted no significant difference in recurrence rates between patients treated nonoperatively and surgically.152 In general, surgical treatment can save patients who fail to respond to conservative treatment to a certain extent, preventing further development of skin ulceration or even extensive necrosis. Early surgery can prevent unnecessary mastectomy as well (Table 3).
Traditional Chinese Medicine
Traditional Chinese medicine has a history of thousands of years. Traditional Chinese medicine aims to regulate the balance of Yin and Yang in the body through different combinations of animal and plant products as well as minerals to cure diseases. Traditional Chinese medicine has also been used to treat IGM. Chuang Ling Ye (CLY) is a traditional Chinese medicine compound composed of rhubarb, safflower, A. manihot, and T. chebula. A study included 40 patients who needed to change dressings. Twenty patients in the experimental group used 30 mL CLY local flushing when changing dressings, while the control group did not use CLY. After 4 weeks of follow-up, the range of inflammation in the experimental group was significantly smaller than that in the control group, and the difference was statistically significant.153
Yanghe decoction is another traditional Chinese medicine obtained from degelatine antler, licorice, cinnamon, processed ginger root, white mustard seeds, Rehmannia glutinosa, and ephedra herb (honey-fried). The results showed that Compared with patients who underwent surgery only, those who received adjuvant Yanghe decoction following surgery showed a shorter remission time with a higher rate (96.4% vs 76.0%) as well as a lower recurrence rate (0% vs 16.0%)135 (Table 3).
Internal expulsion pus-expelling decoction (IEPED) was prepared from 12 g of Atractylodes macrocephala, 12 g of pangolin, 12 g of ginseng, 12 g of Angelica sinensis, 9 g of cohosh, 9 g of licorice, 15 g of Angelica sinensis, 15 g of Astragalus membranaceus, 10 g of licorice, and 10 g of dried green orange peel. In several studies,136 IEPED was soaked in sterile gauze, which was stuffed into the lesion after debridement. The total effective rate of the control group without IEPED was 68%, and the total effective rate of the experimental group with IEPED was 90%. During the 1-year follow-up, 1 patient in the control group relapsed, and there was no recurrence in the experimental group. In addition, it has been reported that traditional Chinese medicine combined with surgery leads to a higher recovery rate and shorter disease duration.154 The efficacy of traditional Chinese medicine in IGM is thus promising (Table 3).
Targeted Prolactin-Lowering Therapy
Treatment of IGM caused by anti psychoactive drug-induced prolactin elevation has also been reported. In a study on mastitis caused by elevated prolactin levels induced by psychoactive drugs, anti psychoactive drugs were first reduced or replaced, and bromocriptine was used to reduce prolactin, followed by surgery, steroid hormones, and antibiotics. A total of 16 of 19 patients were completely cured, and 3 patients experienced recurrence.96 Similar studies have suggested that a targeted reduction in prolactin levels could effectively control inflammation and avoid unnecessary surgery as well101 (Table 3).
Observation
Several studies have suggested that IGM is a self-limiting disease, and mere monitoring of the patient is also a viable option for disease management.8 This is based on the observation that 15.1% of patients achieve remission within nine months of diagnosis without any treatment.155 Another report also stated that 42.4% of the patients recovered under monitoring without any treatment120 (Table 3).
Recurrence Factor
Patients with idiopathic granulomatous mastitis are prone to recurrent attacks that pose a significant challenge to clinicians. Therefore, the recurrence rate should be reduced during the course of treatment. Univariate analysis of related studies showed that differences in body mass index (BMI) and follicle-stimulating hormone (FSH)/luteinizing hormone (LH) ratios were positively correlated with IGM recurrence. In particular, the difference in PRL levels before and after treatment is an independent risk factor for recurrence, and patients with higher PRL levels after treatment have a higher risk of recurrence156 (Table 4). A Multivariate analysis identified low The serum albumin to globulin ratio and smoking as independent risk factors for IGM recurrence157 (Table 4). Another Retrospective analysis revealed that patients with relapsing IGM exhibited high BI-RADS-defined background parenchymal enhancement (moderate to intense) and low mean apparent diffusion coefficient values (less than 1.00×10−3 mm2 /s) on MRI. The authors accordingly proposed that early-stage MRI evaluation should be considered, as high BPE and low mean ADC values might serve as potential markers for future recurrence.158 A 2.16-fold to 2.64-fold increase in the recurrence rate of Corynebacterium infections has been reported22,144,159 (Table 4). A systematic analysis explored the relationship between treatment methods and recurrence. Statistics found that the recurrence rate of the antibiotic treatment group was the lowest, and the difference was statistically significant.160 Further studies are required to predict recurrence factors, and the risk of recurrence should be considered during the initial treatment to reduce the recurrence rate and avoid secondary lesions in patients.
Table 4.
List of Recurrence Risk Factors
Discussion
The etiology of IGM remains unclear, associated with many factors.62,68,80,93,94 Whether IGM is an autoimmune disease or an infectious disease is controversial. Some authors believe that it is an autoimmune disease, because the study found that the immune indexes of IGM patients have different degrees of abnormalities, and methylprednisolone has a control effect on it. Another part of the authors believe that it is an infectious disease. Although pathogens are detected in IGM tissues, there is no conclusive evidence that the detected pathogens are pathogenic bacteria or colonized bacteria. Literature suggests that the occurrence and development of IGM is not determined by a single factor but rather by a combination of multiple factors, including hormone imbalance, flora disorder, and immune imbalance.68,76,86
Based on the existing literature, we believe that IGM is a damage response induced by the imbalance between the immune system and pathogens. In the microenvironment of the mammary ducts and acini, immune cells and pathogenic bacteria in mammary ducts and acini are in a dynamic balance (Figure 3A). However, changes such as hormone imbalance, increased endotoxins, cytokines, and complements released by the body,161 disrupts the local microenvironment.37 Alternatively, local breast impact injury and the increase in the type or number of local pathogenic bacteria may also disrupt the balance between the immune system and pathogens in the mammary ducts and acini162 (Figure 3B).
Figure 3.
The “immunity-pathogen imbalance pathogenesis” model. (A) (I) In normal breast, the immune system is in balance with pathogens, and (II) the ductal epithelium is lined up. (B) (I) immune system imbalance, abnormal recognition ability enhancement. (II) breast impact injury directly leads to breast duct injury and exposes pathogens (III) hormone imbalance causes breast duct deformation to expose pathogens. (C) The above reasons lead to imbalance between pathogens and the immune system, immune cells attack exposed pathogens. (D) Long-term chronic inflammatory stimulation leads to granuloma formation. (E) (I) painful mass formation (II) galactapostema (III) breast sinus. (IV) breast ulceration with pus outflow. The C arrow in the figure shows that the immune cells attack the pathogens.
Once imbalance occurs, immune cells attack the exposed pathogen. On one hand, due to the non-specific killing of some immune cells, and the pathogens are mostly located in epithelial cells, the immune cells directly damage the mammary epithelial cells in the process of killing pathogens. On the other hand, due to the release of granzymes, perforin, and other substances by activated immune cells, mammary epithelial cells are indirectly injured.163–165 The destruction of mammary epithelial cells indirectly leads to pathogen invasion, which further activates the immune system. This cycle aggravates local inflammation, and expands the scope of the lesions (Figure 3C). To prevent expansion of the lesion, the body forms granulomas in the early stages of the lesion to prevent the development of pathogens (Figure 3D), which clinically manifest as a painful mass. As the disease progresses, immune cells kill pathogens, breast tissue increases, and a cavity with a large amount of pus is formed locally in the breast. If the process continues and the scope of the lesions gradually expands, breast sinus tracts and even breast skin ulcers form (Figure 3E).
The validity of this reasoning can be substantiated by multiple perspectives. Based on the pathology of the IGM lesions, granulomas are nodular foci formed by the proliferation of local inflammatory macrophages and their derivative cells. This is a special type of chronic inflammation166,167 accompanied by delayed-type hypersensitivity during the entire process.168
Pathological results of IGM reveal not only a large amount of immune cell infiltration in the lesion114 but also neutrophils surrounding Corynebacterium and other pathogens in the granuloma vacuoles,72 in line with the characteristics of chronic inflammation.
From the perspective of pathogens, pus cavities and large amounts of pus can form in the IGM lesions. Various genera of pathogens and viruses have been identified in the IGM tissues and pus.166,167 These studies have shown that pathogenic infections may be an important initiating factor for IGM,20 especially Corynebacterium. A study infected rat models with Corynebacterium bacteria isolated from patient tissues. Researchers observed typical IGM pathological features in the mammary tissue of infected rats, as well as upregulation of the inflammatory factor IL-6 in rat blood and upregulation of IL-6 and TNF-αin mammary tissue.169 The research team speculated that Corynebacterium is attracted by fatty acids in the breast to colonize in the breast, and secretes Corynekropbactin during colonization, which may play a role in chelating iron, destroying breast cells, inhibiting the growth of other colonized bacteria, and causing the up-regulation of inflammatory factor IL-6, which in turn leads to the formation and development of IGM.169 Our speculation is consistent with this study, that is, external factors break the balance between pathogens and the immune system in the breast, causing the immune system to attack pathogens, thereby forming an immune injury response characterized by granuloma, in which Corynebacterium may be the main driver of this cascade.
In addition, the increase in white blood cells and CRP levels in blood indicators also proves the existence of a pathogenic infection.48 Not all patients have standardized clinical manifestations and indicator changes; however, most patients undergo changes at a certain stage.
During immune regulation in IGM, the cell types and cytokines whose expression changes include NK cells, macrophages, T cells, B cells, CD4+ cells, CD8+ cells,29,32 IL-23, IL-22, IL-6, IL-33, IL-17, IL-8, IL-10.47–50 After recognizing pathogens, macrophages induce Th0 cells to differentiate into Th1, Th2, Th17, and Treg cells by producing cytokines, presenting antigens, and expressing co-stimulatory molecule.170–172 NK cells release INF-y after activation, and DC release IL-12 and IL-23 after activation. IL-12 and INF-y act together on TH1 cells to activate TH1 cells,173,174 and activated Th1 cells promote the macrophage secretion of INF-α, and INF-αpromotes macrophages to become epithelioid cells and form granulomas.175 IL-12 promotes the release of IL-17, IL-6, IL-8, G-CSF, IL-17, IL-6, and IL-8 from CD4+T cells, whereas G-CSF promotes the activation of neutrophils, B lymphocytes, and Th0 cells. Tregs release IL-10 to inhibit TH1 cells, thereby suppressing the inflammatory response.176 TH17 cells release IL-21 and IL-22; IL-22 inhibits the production of Th2 cells, and IL-12 combines with IL-17 to form antimicrobial peptides.177,178 Th2 cells release IL-4 to promote neutrophil and B lymphocytes.179–181 During activation, CD8+ T cells release granzymes, perforin, and express FasL, resulting in epithelial cell injury182,183 (Figure 4).
Figure 4.
Immune mechanism of granulomatous mastitis.
Currently, no standardized treatment protocols are available for IGM. Methods using hormone therapy, antibiotics, surgery, immunosuppressive drugs, probiotics, and traditional Chinese medicine have been reported. However, the absence of a standard treatment protocols lead to application of different treatment strategies by doctors with different levels of experience or using the same strategy for all patients. Hormone therapy is the first-line treatment used by most clinicians.119
Studies have shown that hormone therapy is indeed effective for IGM; however, the hormone dosage that maximizes the benefits and minimizes the side effects is still not clear, and some patients may still experience recurrence after hormone dose reduction or discontinuation.123
There are differences in the therapeutic effect of antibiotics, which may be related to the following factors: blind use of antibiotics without pathogen identification; there may be a mixture of multiple bacteria, and a single antibiotic or empirical antibiotic cannot cover all bacterial flora;19 The presence of drug-resistant bacteria or bacterial resistance to empirical antibiotics may also lead to ineffective antibiotic treatment.184,185 The identification of pathogens and the selection of antibiotics are difficult problems in treatment. In the past, due to the difficulty of pathogen identification, the results of pathogen culture were mostly negative in many cases, thus ignoring the use of antibiotics or blindly using antibiotics.
Long-term benefits of antibiotic therapy.144 Therefore, the use of antimicrobial agents should be done before the pathogen detection, targeted drug strategy, to avoid the use of empirical antibiotics and frequent replacement of antibiotic regimens.162,185,186 The sequencing method is preferred for the identification method, and the culture conditions conducive to the growth of Corynebacterium should be provided when using the culture method. After obtaining clear pathogen detection results, sensitive antibiotics were selected according to drug sensitivity. At present, the difficulty is the treatment of Corynebacterium krusei. It is reported that Corynebacterium is sensitive to rifampicin and gentamicin. Rifampicin shows good therapeutic effect and safety in the treatment of IGM, and rifampicin can be given priority.187
Immunosuppressive agents are often used as an alternative to hormone therapy. Although they are effective, they cannot be used as a first-line regimen. Methotrexate is a folic acid reductase inhibitor, which has dual effects of anti-tumor and immune regulation. It is mainly used in anti-tumor therapy and immunotherapy.188 From the curative effect analysis, glucocorticoids can more quickly and effectively control the inflammatory response and relieve acute symptoms, while methotrexate has a slower onset and is not suitable for acute patients who need to quickly control symptoms. From the safety analysis, the side effects of methotrexate mainly include bone marrow suppression, liver function injury, renal function injury, gastrointestinal discomfort, fatal skin diseases.189,190 Compared with the side effects of steroid hormones, such as edema, weight gain, hair removal, osteoporosis. The side effects of methotrexate are more serious. In contrast, in the case of obtaining the same therapeutic effect, the smaller the side effect, the greater the patient ‘s benefit. There is currently no evidence of methotrexate as a first-line treatment drug. In clinical practice, methotrexate is rarely used as a first-line drug. Further clinical trials are expected to evaluate the efficacy of methotrexate in the future.
The goal of IGM treatment is to improve the recovery rate and reduce recurrence. Studies have found that BMI, hormonal imbalance, and Corynebacterium infection are important factors that lead to an increased recurrence rate.22,144 This highlights that mastitis cannot be treated using a single method, and individualized comprehensive treatment options should be adopted for different patients. Therefore, a comprehensive assessment should be made before deciding on treatment, strategy by considering possible systemic and local factors, such as the immune system, lesion condition (size, location, and presence of pus), pathogen detection results, sex hormone and prolactin levels, and the presence of oral contraceptives or psychoactive drugs. Only by considering all possible recurrence factors through comprehensive treatment can a good outcome and prognosis be obtained with fewer recurrences. It is expected that with the deepening of research, the etiology of IGM can be clarified, and effective and standardized treatment methods can be found.
Conclusion
Idiopathic granulomatous mastitis is an immune injury reaction caused by the imbalance between pathogenic bacteria and immune system under the action of multiple factors. Corynebacterium may be the initiating factor of immune cascade reaction.
Funding Statement
This work was supported by the National Natural Science Foundation of China (82360557, 82260537) and the Biomedical Project of the Major Science and Technology Program of Yunnan Province (202302AA310046).
Disclosure
Siyi Wang and Rui Liu are co-first authors for this study. The authors report no conflicts of interest in this work.
References
- 1.Kessler E, Wolloch Y. Granulomatous mastitis: a lesion clinically simulating carcinoma. Am J Clin Pathol. 1972;58(6):642–646. doi: 10.1093/ajcp/58.6.642 [DOI] [PubMed] [Google Scholar]
- 2.Bässler R. [Mastitis. Classification, histopathology and clinical aspects]. Pathologe. 1997;18(1):27–36. German. doi: 10.1007/s002920050193 [DOI] [PubMed] [Google Scholar]
- 3.Uysal E, Soran A, Sezgin E, Granulomatous Mastitis Study Group. Factors related to recurrence of idiopathic granulomatous mastitis: what do we learn from a multicentre study. ANZ J Surg. 2018;88(6):635–639. doi: 10.1111/ans.14115 [DOI] [PubMed] [Google Scholar]
- 4.Atak T, Sagiroglu J, Eren T, Özemir I A, Alimoglu O. Strategies to treat idiopathic granulomatous mastitis: retrospective analysis of 40 patients. Breast Dis. 2015;35(1):19–24. doi: 10.3233/BD-140373 [DOI] [PubMed] [Google Scholar]
- 5.Ahmed R, Sultan F. Granulomatous mastitis: a review of 14 cases. J Ayub Med Coll Abbottabad. 2006;18(1):52–54. [PubMed] [Google Scholar]
- 6.Centers for Disease Control and Prevention (CDC). Idiopathic granulomatous mastitis in Hispanic women - Indiana, 2006-2008. MMWR Morb Mortal Wkly Rep. 2009;58(47):1317–1321. [PubMed] [Google Scholar]
- 7.Zuo X, Shi X, Gao X, Lai R, Liu P, Zhao Z. Treatment effect of mammary duct exploration combined with focal resection on granulomatous lobular mastitis. J Inflamm Res. 2021;14:2641–2646. doi: 10.2147/JIR.S309101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Davis J, Cocco D, Matz S, et al. Re-evaluating if observation continues to be the best management of idiopathic granulomatous mastitis. Surgery. 2019;166(6):1176–1180. doi: 10.1016/j.surg.2019.06.030 [DOI] [PubMed] [Google Scholar]
- 9.Barreto DS, Sedgwick EL, Nagi CS, Benveniste AP. Granulomatous mastitis: etiology, imaging, pathology, treatment, and clinical findings. Breast Cancer Res Treat. 2018;171(3):527–534. doi: 10.1007/s10549-018-4870-3 [DOI] [PubMed] [Google Scholar]
- 10.Erhan Y, Veral A, Kara E, et al. A clinicopthologic study of a rare clinical entity mimicking breast carcinoma: idiopathic granulomatous mastitis. Breast. 2000;9(1):52–56. doi: 10.1054/brst.1999.0072 [DOI] [PubMed] [Google Scholar]
- 11.Asoglu O, Ozmen V, Karanlik H, et al. Feasibility of surgical management in patients with granulomatous mastitis. Breast J. 2005;11(2):108–114. doi: 10.1111/j.1075-122X.2005.21576.x [DOI] [PubMed] [Google Scholar]
- 12.Velidedeoglu M, Umman V, Kilic F, et al. Idiopathic granulomatous mastitis: introducing a diagnostic algorithm based on 5 years of follow-up of 152 cases from Turkey and a review of the literature. Surg Today. 2022;52(4):668–680. doi: 10.1007/s00595-021-02367-6 [DOI] [PubMed] [Google Scholar]
- 13.Memis A, Bilgen I, Ustun EE, Ozdemir N, Erhan Y, Kapkac M. Granulomatous mastitis: imaging findings with histopathologic correlation. Clin Radiol. 2002;57(11):1001–1006. doi: 10.1053/crad.2002.1056 [DOI] [PubMed] [Google Scholar]
- 14.Deng Y, Xiong Y, Ning P, et al. A case management model for patients with granulomatous mastitis: a prospective study. BMC Womens Health. 2022;22(1):143. doi: 10.1186/s12905-022-01726-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Goulabchand R, Hafidi A, Van de Perre P, et al. Mastitis in autoimmune diseases: review of the literature, diagnostic pathway, and pathophysiological key players. J Clin Med. 2020;9(4):958. doi: 10.3390/jcm9040958 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Al Manasra AR, Al-Hurani MF. Granulomatous mastitis: a rare cause of male breast lump. Case Rep Oncol. 2016;9(2):516–519. doi: 10.1159/000448990 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yaghan RJ, Ayoub NM, Shenawi HM, Yaghan LR. Idiopathic granulomatous mastitis in the male population: a clinical analysis of 13 reported cases. Breast J. 2020;26(7):1481–1482. doi: 10.1111/tbj.13778 [DOI] [PubMed] [Google Scholar]
- 18.Tekgöz E, Çolak S, Çinar M, Yilmaz S. Treatment of idiopathic granulomatous mastitis and factors related with disease recurrence. Turk J Med Sci. 2020;50(5):1380–1386. doi: 10.3906/sag-2003-93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ong SS, Xu J, Sim CK, et al. Profiling microbial communities in idiopathic granulomatous mastitis. Int J Mol Sci. 2023;24(2):1042. doi: 10.3390/ijms24021042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Li XQ, Yuan JP, Fu AS, et al. New insights of corynebacterium kroppenstedtii in granulomatous lobular mastitis based on nanopore sequencing. J Invest Surg. 2022;35(3):639–646. doi: 10.1080/08941939.2021.1921082 [DOI] [PubMed] [Google Scholar]
- 21.Koksal H. What are the new findings with regard to the mysterious disease idiopathic granulomatous mastitis. Surg Today. 2021;51(7):1158–1168. doi: 10.1007/s00595-020-02204-2 [DOI] [PubMed] [Google Scholar]
- 22.Tan QT, Tay SP, Gudi MA, Nadkarni NV, Lim SH, Chuwa E. Granulomatous mastitis and factors associated with recurrence: an 11-year single-centre study of 113 patients in Singapore. World J Surg. 2019;43(7):1737–1745. doi: 10.1007/s00268-019-05014-x [DOI] [PubMed] [Google Scholar]
- 23.Steuer AB, Stern MJ, Cobos G, et al. Clinical characteristics and medical management of idiopathic granulomatous mastitis. JAMA Dermatol. 2020;156(4):460–464. doi: 10.1001/jamadermatol.2019.4516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Toktas O, Konca C, Trabulus DC, et al. A novel first-line treatment alternative for noncomplicated idiopathic granulomatous mastitis: combined intralesional steroid injection with topical steroid administration. Breast Care. 2021;16(2):181–187. doi: 10.1159/000507951 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhang X, Li Y, Zhou Y, et al. A systematic surgical approach for the treatment of idiopathic granulomatous mastitis: a case series. Gland Surg. 2020;9(2):261–270. doi: 10.21037/gs.2020.02.06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gordon S. Macrophage heterogeneity and tissue lipids. J Clin Invest. 2007;117(1):89–93. doi: 10.1172/JCI30992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cutolo M, Campitiello R, Gotelli E, Soldano S. The role of M1/M2 macrophage polarization in rheumatoid arthritis synovitis. Front Immunol. 2022;13:867260. doi: 10.3389/fimmu.2022.867260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mamilos A, Winter L, Schmitt VH, et al. Macrophages: from simple phagocyte to an integrative regulatory cell for inflammation and tissue regeneration-A review of the literature. Cells. 2023;12(2):276. doi: 10.3390/cells12020276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kong C, Zhang C, Wu Y, et al. The expression and meaning of CD68, CD163, CD57, and IgG4 in granulomatous lobular mastitis. Gland Surg. 2020;9(4):936–949. doi: 10.21037/gs-20-419 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Stokic-Trtica V, Diefenbach A, Klose C. NK cell development in times of innate lymphoid cell diversity. Front Immunol. 2020;11:813. doi: 10.3389/fimmu.2020.00813 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Stary V, Stary G. NK cell-mediated recall responses: memory-like, adaptive, or antigen-specific. Front Cell Infect Microbiol. 2020;10:208. doi: 10.3389/fcimb.2020.00208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Emsen A, Köksal H, Uçaryılmaz H, Kadoglou N, Artaç H. The alteration of lymphocyte subsets in idiopathic granulomatous mastitis. Turk J Med Sci. 2021;51(4):1905–1911. doi: 10.3906/sag-2012-192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Reinherz EL, Schlossman SF. The characterization and function of human immunoregulatory T lymphocyte subsets. Pharmacol Rev. 1982;34(1):17–22. doi: 10.1016/S0031-6997(25)06741-9 [DOI] [PubMed] [Google Scholar]
- 34.Kronenberg M, Rudensky A. Regulation of immunity by self-reactive T cells. Nature. 2005;435(7042):598–604. doi: 10.1038/nature03725 [DOI] [PubMed] [Google Scholar]
- 35.Rezaei Kahmini F, Shahgaldi S, Azimi M, Mansourabadi AH. Emerging therapeutic potential of regulatory T (Treg) cells for rheumatoid arthritis: new insights and challenges. Int Immunopharmacol. 2022;108:108858. doi: 10.1016/j.intimp.2022.108858 [DOI] [PubMed] [Google Scholar]
- 36.Jin K, Parreau S, Warrington KJ, et al. Regulatory T cells in autoimmune vasculitis. Front Immunol. 2022;13:844300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ucaryilmaz H, Koksal H, Emsen A, Kadoglou N, Dixon JM, Artac H. The role of regulatory T and B cells in the etiopathogenesis of idiopathic granulomatous mastitis. Immunol Invest. 2022;51(2):357–367. doi: 10.1080/08820139.2020.1832114 [DOI] [PubMed] [Google Scholar]
- 38.Ates D, Doner HC, Kurban S, Koksal H. The effect of soluble TREM-1 in idiopathic granulomatous mastitis. Immunol Invest. 2022;51(4):839–850. doi: 10.1080/08820139.2021.1879846 [DOI] [PubMed] [Google Scholar]
- 39.de Oliveira Matos A, Dos Santos Dantas PH, Figueira marques silva-sales M, Sales-Campos H. The role of the triggering receptor expressed on myeloid cells-1 (TREM-1) in non-bacterial infections. Crit Rev Microbiol. 2020;46(3):237–252. doi: 10.1080/1040841X.2020.1751060 [DOI] [PubMed] [Google Scholar]
- 40.Dantas P, Matos AO, da Silva Filho E, Silva-Sales M, Sales-Campos H. Triggering receptor expressed on myeloid cells-1 (TREM-1) as a therapeutic target in infectious and noninfectious disease: a critical review. Int Rev Immunol. 2020;39(4):188–202. doi: 10.1080/08830185.2020.1762597 [DOI] [PubMed] [Google Scholar]
- 41.Mills K. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol. 2023;23(1):38–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang T, Mei Y, Li Z. Research progress on regulatory B cells in systemic lupus erythematosus. Biomed Res Int. 2019;2019:7948687. doi: 10.1155/2019/7948687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kiełbasa A, Gadzała-Kopciuch R, Buszewski B. Cytokines-biogenesis and their role in human breast milk and determination. Int J Mol Sci. 2021;22(12):6238. doi: 10.3390/ijms22126238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Romani N, Kämpgen E, Koch F, Heufler C, Schuler G. Dendritic cell production of cytokines and responses to cytokines. Int Rev Immunol. 1990;6(2–3):151–161. doi: 10.3109/08830189009056626 [DOI] [PubMed] [Google Scholar]
- 45.Komlósi ZI, van de Veen W, Kovács N, et al. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med. 2022;85:100995. doi: 10.1016/j.mam.2021.100995 [DOI] [PubMed] [Google Scholar]
- 46.Saraiva M, Vieira P, O’Garra A. Biology and therapeutic potential of interleukin-10. J Exp Med. 2020;217(1). doi: 10.1084/jem.20190418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Saydam M, Yilmaz KB, Sahin M, et al. New findings on autoimmune etiology of idiopathic granulomatous mastitis: serum IL-17, IL-22 and IL-23 levels of patients. J Invest Surg. 2021;34(9):993–997. doi: 10.1080/08941939.2020.1725190 [DOI] [PubMed] [Google Scholar]
- 48.Huang YM, Lo C, Cheng CF, Lu CH, Hsieh SC, Li KJ. Serum C-reactive protein and interleukin-6 levels as biomarkers for disease severity and clinical outcomes in patients with idiopathic granulomatous mastitis. J Clin Med. 2021;10(10):2077. doi: 10.3390/jcm10102077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yigitbasi MR, Guntas G, Atak T, Sonmez C, Yalman H, Uzun H. The role of interleukin-33 as an inflammatory marker in differential diagnosis of idiopathic granulomatous mastitis and breast cancer. J Invest Surg. 2017;30(4):272–276. doi: 10.1080/08941939.2016.1240270 [DOI] [PubMed] [Google Scholar]
- 50.Koksal H, Vatansev H, Artac H, Kadoglou N. The clinical value of interleukins-8, −10, and −17 in idiopathic granulomatous mastitis. Clin Rheumatol. 2020;39(5):1671–1677. doi: 10.1007/s10067-020-04925-8 [DOI] [PubMed] [Google Scholar]
- 51.Yu JJ, Bao SL, Yu SL, et al. Mouse model of plasma cell mastitis. J Transl Med. 2012;10(Suppl 1):S11. doi: 10.1186/1479-5876-10-S1-S11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fossiez F, Djossou O, Chomarat P, et al. T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med. 1996;183(6):2593–2603. doi: 10.1084/jem.183.6.2593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Balkwill FR, Burke F. The cytokine network. Immunol Today. 1989;10(9):299–304. doi: 10.1016/0167-5699(89)90085-6 [DOI] [PubMed] [Google Scholar]
- 54.Matsushima K, Yang D, Oppenheim JJ. Interleukin-8: an evolving chemokine. Cytokine. 2022;153:155828. doi: 10.1016/j.cyto.2022.155828 [DOI] [PubMed] [Google Scholar]
- 55.Kawamoto M, Matsui E, Kaneko H, et al. IL-10 plays an important role as an immune-modulator in the pathogenesis of atopic diseases. Mol Med Rep. 2008;1(6):837–842. doi: 10.3892/mmr_00000037 [DOI] [PubMed] [Google Scholar]
- 56.Koksal H. Human leukocyte antigens class I and II in patients with idiopathic granulomatous mastitis. Am J Surg. 2019;218(3):605–608. doi: 10.1016/j.amjsurg.2019.01.038 [DOI] [PubMed] [Google Scholar]
- 57.Morgan BP. The membrane attack complex as an inflammatory trigger. Immunobiology. 2016;221(6):747–751. doi: 10.1016/j.imbio.2015.04.006 [DOI] [PubMed] [Google Scholar]
- 58.Bloch EF, Knight EM, Carmon T, et al. C5b-7 and C5b-8 precursors of the membrane attack complex (C5b-9) are effective killers of E. coli J5 during serum incubation. Immunol Invest. 1997;26(4):409–419. doi: 10.3109/08820139709022698 [DOI] [PubMed] [Google Scholar]
- 59.Tomlinson S, Taylor PW, Morgan BP, Luzio JP. Killing of gram-negative bacteria by complement. Fractionation of cell membranes after complement C5b-9 deposition on to the surface of salmonella minnesota Re595. Biochem J. 1989;263(2):505–511. doi: 10.1042/bj2630505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bayly-Jones C, Bubeck D, Dunstone MA. The mystery behind membrane insertion: a review of the complement membrane attack complex. Philos Trans R Soc Lond B Biol Sci. 2017;372:1726. doi: 10.1098/rstb.2016.0221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Takano T, Elimam H, Cybulsky AV. Complement-mediated cellular injury. Semin Nephrol. 2013;33(6):586–601. doi: 10.1016/j.semnephrol.2013.08.009 [DOI] [PubMed] [Google Scholar]
- 62.Zhang HJ, Ding PP, Zhang XS, et al. MAC mediates mammary duct epithelial cell injury in plasma cell mastitis and granulomatous mastitis. Int Immunopharmacol. 2022;113(Pt A):109303. doi: 10.1016/j.intimp.2022.109303 [DOI] [PubMed] [Google Scholar]
- 63.Benson JR, Dumitru D. Idiopathic granulomatous mastitis: presentation, investigation and management. Future Oncol. 2016;12(11):1381–1394. doi: 10.2217/fon-2015-0038 [DOI] [PubMed] [Google Scholar]
- 64.Illman JE, Terra SB, Clapp AJ, et al. Granulomatous diseases of the breast and axilla: radiological findings with pathological correlation. Insights Imaging. 2018;9(1):59–71. doi: 10.1007/s13244-017-0587-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Li XQ, Sun HG, Wang XH, et al. Activation of C3 and C5 may be involved in the inflammatory progression of PCM and GM. Inflammation. 2022;45(2):739–752. doi: 10.1007/s10753-021-01580-2 [DOI] [PubMed] [Google Scholar]
- 66.Bundred NJ, Dixon JM, Lumsden AB, et al. Are the lesions of duct ectasia sterile. Br J Surg. 1985;72(10):844–845. doi: 10.1002/bjs.1800721023 [DOI] [PubMed] [Google Scholar]
- 67.Taylor GB, Paviour SD, Musaad S, Jones WO, Holland DJ. A clinicopathological review of 34 cases of inflammatory breast disease showing an association between corynebacteria infection and granulomatous mastitis. Pathology. 2003;35(2):109–119. [PubMed] [Google Scholar]
- 68.Bi J, Li Z, Lin X, et al. Etiology of granulomatous lobular mastitis based on metagenomic next-generation sequencing. Int J Infect Dis. 2021;113:243–250. doi: 10.1016/j.ijid.2021.10.019 [DOI] [PubMed] [Google Scholar]
- 69.Yu HJ, Deng H, Ma J, et al. Clinical metagenomic analysis of bacterial communities in breast abscesses of granulomatous mastitis. Int J Infect Dis. 2016;53:30–33. doi: 10.1016/j.ijid.2016.10.015 [DOI] [PubMed] [Google Scholar]
- 70.Tauch A, Fernández-Natal I, Soriano F. A microbiological and clinical review on Corynebacterium kroppenstedtii. Int J Infect Dis. 2016;48:33–39. doi: 10.1016/j.ijid.2016.04.023 [DOI] [PubMed] [Google Scholar]
- 71.Johnson MG, Leal S, Plongla R, Leone PA, Gilligan PH. Closing the brief case: recurrent granulomatous mastitis due to corynebacterium kroppenstedtii. J Clin Microbiol. 2016;54(8):2212. doi: 10.1128/JCM.03132-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Johnstone KJ, Robson J, Cherian SG, Wan Sai Cheong J, Kerr K, Bligh JF. Cystic neutrophilic granulomatous mastitis associated with corynebacterium including corynebacterium kroppenstedtii. Pathology. 2017;49(4):405–412. doi: 10.1016/j.pathol.2017.01.006 [DOI] [PubMed] [Google Scholar]
- 73.Stary CM, Lee YS, Balfour J. Idiopathic granulomatous mastitis associated with corynebacterium sp. Infection Hawaii Med J. 2011;70(5):99–101. [PMC free article] [PubMed] [Google Scholar]
- 74.Fernández L, Pannaraj PS, Rautava S, Rodríguez JM. The microbiota of the human mammary ecosystem. Front Cell Infect Microbiol. 2020;10:586667. doi: 10.3389/fcimb.2020.586667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Costantini L, Magno S, Albanese D, et al. Characterization of human breast tissue microbiota from core needle biopsies through the analysis of multi hypervariable 16S-rRNA gene regions. Sci Rep. 2018;8(1):16893. doi: 10.1038/s41598-018-35329-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hieken TJ, Chen J, Hoskin TL, et al. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci Rep. 2016;6:30751. doi: 10.1038/srep30751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Urbaniak C, Gloor GB, Brackstone M, Scott L, Tangney M, Reid G. The microbiota of breast tissue and its association with breast cancer. Appl Environ Microbiol. 2016;82(16):5039–5048. doi: 10.1128/AEM.01235-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Urbaniak C, Cummins J, Brackstone M, et al. Microbiota of human breast tissue. Appl Environ Microbiol. 2014;80(10):3007–3014. doi: 10.1128/AEM.00242-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ransjö U, Asplund OA, Gylbert L, Jurell G. Bacteria in the female breast. Scand J Plast Reconstr Surg. 1985;19(1):87–89. doi: 10.3109/02844318509052869 [DOI] [PubMed] [Google Scholar]
- 80.Ling J, Xie X, Wang Y, et al. Differential expression profiles of miRNA in granulomatous lobular mastitis and identification of possible biomarkers. Exp Ther Med. 2022;24(2):500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhu Q, Wang L, Wang P. The identification of gene expression profiles associated with granulomatous mastitis. Breast Care. 2021;16(4):319–327. doi: 10.1159/000507474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lei QR, Yang X, Miao CM, Wang JC, Yang Y. Relationship between granulomatous lobular mastitis and methylene tetrahydrofolate reductase gene polymorphism. World J Clin Cases. 2020;8(18):4017–4021. doi: 10.12998/wjcc.v8.i18.4017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Liu Y, Zhang J, Zhou YH, et al. Activation of the IL-6/JAK2/STAT3 pathway induces plasma cell mastitis in mice. Cytokine. 2018;110:150–158. doi: 10.1016/j.cyto.2018.05.002 [DOI] [PubMed] [Google Scholar]
- 84.Console L, Scalise M, Indiveri C. Exosomes in inflammation and role as biomarkers. Clin Chim Acta. 2019;488:165–171. doi: 10.1016/j.cca.2018.11.009 [DOI] [PubMed] [Google Scholar]
- 85.Scherer PE. The many secret lives of adipocytes: implications for diabetes. Diabetologia. 2019;62(2):223–232. doi: 10.1007/s00125-018-4777-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang X, Han Y, Liu J, et al. Exosomes play an important role in the progression of plasma cell mastitis via the PI3K-Akt-mTOR signaling pathway. Mediators Inflamm. 2019;2019:4312016. doi: 10.1155/2019/4312016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244–1247. doi: 10.1126/science.1153124 [DOI] [PubMed] [Google Scholar]
- 88.Cai M, Shi Y, Zheng T, et al. Mammary epithelial cell derived exosomal MiR-221 mediates M1 macrophage polarization via SOCS1/STATs to promote inflammatory response. Int Immunopharmacol. 2020;83:106493. doi: 10.1016/j.intimp.2020.106493 [DOI] [PubMed] [Google Scholar]
- 89.Nikolaev A, Blake CN, Carlson DL. Association between hyperprolactinemia and granulomatous mastitis. Breast J. 2016;22(2):224–231. doi: 10.1111/tbj.12552 [DOI] [PubMed] [Google Scholar]
- 90.Bani-Hani KE, Yaghan RJ, Matalka II, Shatnawi NJ. Idiopathic granulomatous mastitis: time to avoid unnecessary mastectomies. Breast J. 2004;10(4):318–322. doi: 10.1111/j.1075-122X.2004.21336.x [DOI] [PubMed] [Google Scholar]
- 91.Parperis K, Achilleos S, Costi E, Vardas M. Granulomatous mastitis, erythema nodosum and arthritis syndrome: case-based review. Rheumatol Int. 2021;41(6):1175–1181. doi: 10.1007/s00296-021-04820-8 [DOI] [PubMed] [Google Scholar]
- 92.Boutet P, Sulon J, Closset R, et al. Prolactin-induced activation of nuclear factor κB in bovine mammary epithelial cells: role in chronic mastitis. J Dairy Sci. 2007;90(1):155–164. doi: 10.3168/jds.S0022-0302(07)72617-6 [DOI] [PubMed] [Google Scholar]
- 93.Kutsuna S, Mezaki K, Nagamatsu M, et al. Two cases of granulomatous mastitis caused by corynebacterium kroppenstedtii infection in nulliparous young women with hyperprolactinemia. Intern Med. 2015;54(14):1815–1818. doi: 10.2169/internalmedicine.54.4254 [DOI] [PubMed] [Google Scholar]
- 94.Cai R, Zhao J, Qiao Z, Li Y. Idiopathic granulomatous mastitis with normal prolactin level caused by risperidone. Asian J Surg. 2021;44(5):763–764. doi: 10.1016/j.asjsur.2021.02.026 [DOI] [PubMed] [Google Scholar]
- 95.Prasad S, Jaiprakash P, Dave A, Pai D. Idiopathic granulomatous mastitis: an institutional experience. Turk J Surg. 2017;33(2):100–103. doi: 10.5152/turkjsurg.2017.3439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jiao Y, Chang K, Jiang Y, Zhang J. Identification of periductal mastitis and granulomatous lobular mastitis: a literature review. Ann Transl Med. 2023;11(3):158. doi: 10.21037/atm-22-6473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Tian C, Wang H, Liu Z, Han X, Ning P. Characteristics and management of granulomatous lobular mastitis associated with antipsychotics-induced hyperprolactinemia. Breastfeed Med. 2022;17(7):599–604. doi: 10.1089/bfm.2021.0341 [DOI] [PubMed] [Google Scholar]
- 98.Tang MW, Reedquist KA, Garcia S, et al. The prolactin receptor is expressed in rheumatoid arthritis and psoriatic arthritis synovial tissue and contributes to macrophage activation. Rheumatology. 2016;55(12):2248–2259. doi: 10.1093/rheumatology/kew316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Song GG, Lee YH. Circulating prolactin level in systemic lupus erythematosus and its correlation with disease activity: a meta-analysis. Lupus. 2017;26(12):1260–1268. doi: 10.1177/0961203317693094 [DOI] [PubMed] [Google Scholar]
- 100.Barra F, Centurioni MG, Gustavino C, Alessandri F, Ferrero S. Idiopathic granulomatous mastitis: the importance of summarizing the heterogenous evidence of the current literature. J Invest Surg. 2022;35(3):721–722. doi: 10.1080/08941939.2021.1901162 [DOI] [PubMed] [Google Scholar]
- 101.Li J, McGregor HP. Idiopathic granulomatous mastitis associated with hyperprolactinemia: a nonoperative approach. Breast J. 2017;23(6):742–744. doi: 10.1111/tbj.12914 [DOI] [PubMed] [Google Scholar]
- 102.Yuan QQ, Xiao SY, Farouk O, et al. Management of granulomatous lobular mastitis: an international multidisciplinary consensus (2021 edition). Mil Med Res. 2022;9(1):20. doi: 10.1186/s40779-022-00380-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Fazzio RT, Shah SS, Sandhu NP, Glazebrook KN. Idiopathic granulomatous mastitis: imaging update and review. Insights Imaging. 2016;7(4):531–539. doi: 10.1007/s13244-016-0499-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Alikhassi A, Azizi F, Ensani F. Imaging features of granulomatous mastitis in 36 patients with new sonographic signs. J Ultrasound. 2020;23(1):61–68. doi: 10.1007/s40477-019-00392-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Zheng Y, Bai L, Sun J, et al. Diagnostic value of radiomics model based on gray-scale and contrast-enhanced ultrasound for inflammatory mass stage periductal mastitis/duct ectasia. Front Oncol. 2022;12:981106. doi: 10.3389/fonc.2022.981106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Yin L, Agyekum EA, Zhang Q, et al. Differentiation between granulomatous lobular mastitis and breast cancer using quantitative parameters on contrast-enhanced ultrasound. Front Oncol. 2022;12:876487. doi: 10.3389/fonc.2022.876487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Arslan S, Öncü F, Eryılmaz MA, Durmaz MS, Altunkeser A, Ünlü Y. Advantages of b-mode ultrasound combined with strain elastography in differentiation of idiopathic granulomatous mastitis from malignant breast lesions. Turk J Med Sci. 2018;48(1):16–23. doi: 10.3906/sag-1708-34 [DOI] [PubMed] [Google Scholar]
- 108.Sripathi S, Ayachit A, Bala A, Kadavigere R, Kumar S. Idiopathic granulomatous mastitis: a diagnostic dilemma for the breast radiologist. Insights Imaging. 2016;7(4):523–529. doi: 10.1007/s13244-016-0497-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Imoto S, Kitaya T, Kodama T, Hasebe T, Mukai K. Idiopathic granulomatous mastitis: case report and review of the literature. Jpn J Clin Oncol. 1997;27(4):274–277. doi: 10.1093/jjco/27.4.274 [DOI] [PubMed] [Google Scholar]
- 110.Dursun M, Yilmaz S, Yahyayev A, et al. Multimodality imaging features of idiopathic granulomatous mastitis: outcome of 12 years of experience. Radiol Med. 2012;117(4):529–538. doi: 10.1007/s11547-011-0733-2 [DOI] [PubMed] [Google Scholar]
- 111.Pluguez-Turull CW, Nanyes JE, Quintero CJ, et al. Idiopathic granulomatous mastitis: manifestations at multimodality imaging and pitfalls. Radiographics. 2018;38(2):330–356. doi: 10.1148/rg.2018170095 [DOI] [PubMed] [Google Scholar]
- 112.Oztekin PS, Durhan G, Nercis Kosar P, Erel S, Hucumenoglu S. Imaging findings in patients with granulomatous mastitis. Iran J Radiol. 2016;13(3):e33900. doi: 10.5812/iranjradiol.33900 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tyler AD, Mataseje L, Urfano CJ, et al. Evaluation of oxford nanopore’s MinION sequencing device for microbial whole genome sequencing applications. Sci Rep. 2018;8(1):10931. doi: 10.1038/s41598-018-29334-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Wolfrum A, Kümmel S, Theuerkauf I, Pelz E, Reinisch M. Granulomatous mastitis: a therapeutic and diagnostic challenge. Breast Care. 2018;13(6):413–418. doi: 10.1159/000495146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hovanessian Larsen LJ, Peyvandi B, Klipfel N, Grant E, Iyengar G. Granulomatous lobular mastitis: imaging, diagnosis, and treatment. AJR Am J Roentgenol. 2009;193(2):574–581. doi: 10.2214/AJR.08.1528 [DOI] [PubMed] [Google Scholar]
- 116.Cui L, Sun C, Guo J, Zhang X, Liu S. Pathological manifestations of granulomatous lobular mastitis. Front Med. 2024;11:1326587. doi: 10.3389/fmed.2024.1326587 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Fu Y, Chen Q, Xiong M, et al. Clinical performance of nanopore targeted sequencing for diagnosing infectious diseases. Microbiol Spectr. 2022;10(2):e0027022. doi: 10.1128/spectrum.00270-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Huang Q, Fu A, Wang Y, Zhang J, Zhao W, Cheng Y. Microbiological diagnosis of endophthalmitis using nanopore targeted sequencing. Clin Exp Ophthalmol. 2021;49(9):1060–1068. doi: 10.1111/ceo.13992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Godazandeh G, Shojaee L, Alizadeh-Navaei R, Hessami A. Corticosteroids in idiopathic granulomatous mastitis: a systematic review and meta-analysis. Surg Today. 2021;51(12):1897–1905. doi: 10.1007/s00595-021-02234-4 [DOI] [PubMed] [Google Scholar]
- 120.Çetinkaya G, Kozan R, Emral AC, Tezel E. Granulomatous mastitis, watch and wait is a good option. Ir J Med Sci. 2021;190(3):1117–1122. doi: 10.1007/s11845-020-02406-0 [DOI] [PubMed] [Google Scholar]
- 121.Tan QW, Zhang YN, Jia YP, Gou J, Lv Q, Yang XQ. Methylprednisolone for idiopathic granulomatous mastitis: a prospective observational cohort study. Gland Surg. 2022;11(9):1538–1545. doi: 10.21037/gs-22-484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang Y, Yang Q, Wang Z. The evolution of nanopore sequencing. Front Genet. 2014;5:449. doi: 10.3389/fgene.2014.00449 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Azlina AF, Ariza Z, Arni T, Hisham AN. Chronic granulomatous mastitis: diagnostic and therapeutic considerations. World J Surg. 2003;27(5):515–518. doi: 10.1007/s00268-003-6806-1 [DOI] [PubMed] [Google Scholar]
- 124.Montazer M, Dadashzadeh M, Moosavi Toomatari SE. Comparison of the outcome of low dose and high-dose corticosteroid in the treatment of idiopathic granulomatous mastitis. Asian Pac J Cancer Prev. 2020;21(4):993–996. doi: 10.31557/APJCP.2020.21.4.993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Shojaee L, Rahmani N, Moradi S, Motamedi A, Godazandeh G. Idiopathic granulomatous mastitis: challenges of treatment in iranian women. BMC Surg. 2021;21(1):206. doi: 10.1186/s12893-021-01210-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tang A, Dominguez DA, Edquilang JK, Green AJ, Khoury AL, Godfrey RS. Granulomatous mastitis: comparison of novel treatment of steroid injection and current management. J Surg Res. 2020;254:300–305. doi: 10.1016/j.jss.2020.04.018 [DOI] [PubMed] [Google Scholar]
- 127.Çetin K, Sıkar HE, Göret NE, et al. Comparison of topical, systemic, and combined therapy with steroids on idiopathic granulomatous mastitis: a prospective randomized study. World J Surg. 2019;43(11):2865–2873. doi: 10.1007/s00268-019-05084-x [DOI] [PubMed] [Google Scholar]
- 128.Farouk O, Abdelkhalek M, Abdallah A, et al. Rifampicin for idiopathic granulomatous lobular mastitis: a promising alternative for treatment. World J Surg. 2017;41(5):1313–1321. doi: 10.1007/s00268-016-3857-7 [DOI] [PubMed] [Google Scholar]
- 129.Papila Kundaktepe B, Velidedeoğlu M, Mete B. The effect of methotrexate monotherapy on treatment-resistant idiopathic granulomatous mastitis patients. Surgeon. 2022;20(3):e13–e19. doi: 10.1016/j.surge.2021.03.001 [DOI] [PubMed] [Google Scholar]
- 130.Postolova A, Troxell ML, Wapnir IL, Genovese MC. Methotrexate in the treatment of idiopathic granulomatous mastitis. J Rheumatol. 2020;47(6):924–927. doi: 10.3899/jrheum.181205 [DOI] [PubMed] [Google Scholar]
- 131.Kafadar MT, Bahadır MV, Girgin S. Low-dose methotrexate use in idiopathic granulomatous mastitis: an alternative treatment method. Breast Care. 2021;16(4):402–407. doi: 10.1159/000513879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Liao H, Guo J, Chen X, Hua Z, Lin J, Weng Y. Ultrasound classification-guided minimally invasive rotary cutting in granulomatous lobular mastitis. BMC Womens Health. 2020;20(1):252. doi: 10.1186/s12905-020-01118-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Chen R, Chen J, Peng A, Yang L, Zhou R. Clinical therapeutic evaluation of vacuum sealing drainage and precise ultrasound-guided debridement in the treatment of non-lactational mastitis. Exp Ther Med. 2021;21(5):480. doi: 10.3892/etm.2021.9911 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Yaohuai W, Junlong S, Yi T, Chuang C, Shengrong S. Minimally invasive comprehensive treatment for granulomatous lobular mastitis. BMC Surgery. 2020;20(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zhang X, Li J, Hu XJ. Postoperative Yanghe decoction regimen improves outcomes for idiopathic granulomatous mastitis: a retrospective cohort study. Medicine. 2020;99(45):e23136. doi: 10.1097/MD.0000000000023136 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Liu PZ, Shi XG, Zuo XM, et al. A clinical study on the treatment of granulomatous lobular mastitis by the external application of the internal pus-expelling decoction and operation. Ann Palliat Med. 2020;9(5):2631–2641. doi: 10.21037/apm-19-684 [DOI] [PubMed] [Google Scholar]
- 137.Mathew Thomas V, Alexander SA, Bindal P, Vredenburgh J. Idiopathic granulomatous mastitis-a mystery yet to be unraveled: a case series and review of literature. Cureus. 2020;12(2):e6895. doi: 10.7759/cureus.6895 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ertürk TF, Çakır Ö, Yaprak Bayrak B, Güneş A, Aydemir S, Utkan NZ. Local steroid treatment: an effective procedure for idiopathic granulomatous mastitis, including complicated cases. J Invest Surg. 2022;35(4):745–751. doi: 10.1080/08941939.2021.1933272 [DOI] [PubMed] [Google Scholar]
- 139.Aghajanzadeh M, Hassanzadeh R, Alizadeh Sefat S, et al. Granulomatous mastitis: presentations, diagnosis, treatment and outcome in 206 patients from the north of Iran. Breast. 2015;24(4):456–460. doi: 10.1016/j.breast.2015.04.003 [DOI] [PubMed] [Google Scholar]
- 140.Saraiya N, Corpuz M. Corynebacterium kroppenstedtii: a challenging culprit in breast abscesses and granulomatous mastitis. Curr Opin Obstet Gynecol. 2019;31(5):325–332. doi: 10.1097/GCO.0000000000000541 [DOI] [PubMed] [Google Scholar]
- 141.Dobinson HC, Anderson TP, Chambers ST, Doogue MP, Seaward L, Werno AM. Antimicrobial treatment options for granulomatous mastitis caused by corynebacterium species. J Clin Microbiol. 2015;53(9):2895–2899. doi: 10.1128/JCM.00760-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zeng W, Lao S, Jia W, et al. Clinical features and recurrence of Corynebacterium kroppenstedtii infection in patients with mastitis. BMC Womens Health. 2022;22(1):276. doi: 10.1186/s12905-022-01859-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Wu JM, Turashvili G. Cystic neutrophilic granulomatous mastitis: an update. J Clin Pathol. 2020;73(8):445–453. doi: 10.1136/jclinpath-2019-206180 [DOI] [PubMed] [Google Scholar]
- 144.Tsai MJ, Huang WC, Wang JT, et al. Factors associated with treatment duration and recurrence rate of complicated mastitis. J Microbiol Immunol Infect. 2020;53(6):875–881. doi: 10.1016/j.jmii.2020.03.028 [DOI] [PubMed] [Google Scholar]
- 145.Co M, Cheng V, Wei J, et al. Idiopathic granulomatous mastitis: a 10-year study from a multicentre clinical database. Pathology. 2018;50(7):742–747. doi: 10.1016/j.pathol.2018.08.010 [DOI] [PubMed] [Google Scholar]
- 146.Kaya MN, Tekgöz E, Çolak S, Kılıç Ö, Çınar M, Yılmaz S. Drug-free remission is an achievable target with immunosuppressive treatment in idiopathic granulomatous mastitis. Ir J Med Sci. 2023;192(6):2815–2819. doi: 10.1007/s11845-023-03338-1 [DOI] [PubMed] [Google Scholar]
- 147.Baran W, Batycka-Baran A, Zychowska M, Bieniek A, Szepietowski JC. Folate supplementation reduces the side effects of methotrexate therapy for psoriasis. Expert Opin Drug Saf. 2014;13(8):1015–1021. doi: 10.1517/14740338.2014.933805 [DOI] [PubMed] [Google Scholar]
- 148.Tian C, Han X, Liu Z, Lv X, Ning P. Methotrexate and low-dose corticosteroid: an effective alternate against corticosteroid-resistant granulomatous lobular mastitis. J Obstet Gynaecol Res. 2022;48(11):2956–2963. doi: 10.1111/jog.15396 [DOI] [PubMed] [Google Scholar]
- 149.Li J. Diagnosis and treatment of 75 patients with idiopathic lobular granulomatous mastitis. J Invest Surg. 2019;32(5):414–420. doi: 10.1080/08941939.2018.1424270 [DOI] [PubMed] [Google Scholar]
- 150.Zhang C, Wu Y, Wang H, et al. A clinical observation of stage I implant breast reconstruction for mass-like granulomatous lobular mastitis. Gland Surg. 2021;10(9):2663–2672. doi: 10.21037/gs-21-417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Yau FM, Macadam SA, Kuusk U, Nimmo M, Van Laeken N. The surgical management of granulomatous mastitis. Ann Plast Surg. 2010;64(1):9–16. doi: 10.1097/SAP.0b013e3181a20cae [DOI] [PubMed] [Google Scholar]
- 152.Zhou F, Liu L, Liu L, et al. Comparison of conservative versus surgical treatment protocols in treating idiopathic granulomatous mastitis: a meta-analysis. Breast Care. 2020;15(4):415–420. doi: 10.1159/000503602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Xue JX, Ye B, Liu S, Cao SH, Bian WH, Yao C. Treatment efficacy of chuang ling ye, a traditional chinese herbal medicine compound, on idiopathic granulomatous mastitis: a randomized controlled trial. Evid Based Complement Alternat Med. 2020;2020:6964801. doi: 10.1155/2020/6964801 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Sawuer R, Wu C, Sun Z, Liu S. The effectiveness of traditional chinese medicine combined with surgery to treat granulomatous mastitis: a propensity-matched analysis. Front Oncol. 2022;12:833742. doi: 10.3389/fonc.2022.833742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Azizi A, Prasath V, Canner J, et al. Idiopathic granulomatous mastitis: management and predictors of recurrence in 474 patients. Breast J. 2020;26(7):1358–1362. doi: 10.1111/tbj.13822 [DOI] [PubMed] [Google Scholar]
- 156.Huang Y, Wu H. A retrospective analysis of recurrence risk factors for granulomatous lobular mastitis in 130 patients: more attention should be paied to prolactin level. Ann Palliat Med. 2021;10(3):2824–2831. doi: 10.21037/apm-20-1972 [DOI] [PubMed] [Google Scholar]
- 157.Ciftci AB, Bük ÖF, Yemez K, Polat S, Yazıcıoğlu İM. Risk factors and the role of the albumin-to-globulin ratio in predicting recurrence among patients with idiopathic granulomatous mastitis. J Inflamm Res. 2022;15:5401–5412. doi: 10.2147/JIR.S377804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Kayadibi Y, Ucar N, Akan YN, et al. Magnetic resonance imaging findings associated with recurrence in idiopathic granulomatous mastitis. Clin Imaging. 2022;84:47–53. doi: 10.1016/j.clinimag.2022.01.010 [DOI] [PubMed] [Google Scholar]
- 159.Li S, Huang Q, Song P, et al. Clinical characteristics and therapeutic strategy of granulomatous mastitis accompanied by Corynebacterium kroppenstedtii: a retrospective cohort study. BMC Womens Health. 2023;23(1):388. doi: 10.1186/s12905-023-02509-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Fattahi AS, Amini G, Sajedi F, Mehrad-Majd H. Factors affecting recurrence of idiopathic granulomatous mastitis: a systematic review. Breast J. 2023;2023:9947797. doi: 10.1155/2023/9947797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Zhao C, Hu X, Bao L, et al. Gut dysbiosis induces the development of mastitis through a reduction in host anti-inflammatory enzyme activity by endotoxemia. Microbiome. 2022;10(1):205. doi: 10.1186/s40168-022-01402-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Li XQ, Wu HL, Yuan JP, Liu TG, Sun SR, Chen C. Bacteria associated with granulomatous lobular mastitis and the potential for personalized therapy. J Invest Surg. 2022;35(1):164–170. doi: 10.1080/08941939.2020.1833262 [DOI] [PubMed] [Google Scholar]
- 163.Shivakumar P, Mourya R, Bezerra JA. Perforin and granzymes work in synergy to mediate cholangiocyte injury in experimental biliary atresia. J Hepatol. 2014;60(2):370–376. doi: 10.1016/j.jhep.2013.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Kiselevsky DB. Granzymes and mitochondria. Biochemistry. 2020;85(2):131–139. doi: 10.1134/S0006297920020017 [DOI] [PubMed] [Google Scholar]
- 165.Welz M, Eickhoff S, Abdullah Z, et al. Perforin inhibition protects from lethal endothelial damage during fulminant viral hepatitis. Nat Commun. 2018;9(1):4805. doi: 10.1038/s41467-018-07213-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Klapproth H, Huerta Arana M, Fabri M. Macrophage biology in human granulomatous skin inflammation. Int J Mol Sci. 2023;24(5):4624. doi: 10.3390/ijms24054624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Sacco KA, Gazzin A, Notarangelo LD, Delmonte OM. Granulomatous inflammation in inborn errors of immunity. Front Pediatr. 2023;11:1110115. doi: 10.3389/fped.2023.1110115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Maione C, Palumbo VD, Maffongelli A, et al. Diagnostic techniques and multidisciplinary approach in idiopathic granulomatous mastitis: a revision of the literature. Acta Biomed. 2019;90(1):11–15. doi: 10.23750/abm.v90i1.6607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Liu R, Luo Z, Dai C, et al. Corynebacterium parakroppenstedtii secretes a novel glycolipid to promote the development of granulomatous lobular mastitis. Signal Transduct Target Ther. 2024;9(1):292. doi: 10.1038/s41392-024-01984-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Vinuesa CG, Linterman MA, Yu D, MacLennan IC. Follicular Helper T Cells. Annu Rev Immunol. 2016;34:335–368. doi: 10.1146/annurev-immunol-041015-055605 [DOI] [PubMed] [Google Scholar]
- 171.Alonso MN, Wong MT, Zhang AL, et al. T(H)1, T(H)2, and T(H)17 cells instruct monocytes to differentiate into specialized dendritic cell subsets. Blood. 2011;118(12):3311–3320. doi: 10.1182/blood-2011-03-341065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Maldonado-López R, Moser M. Dendritic cell subsets and the regulation of Th1/Th2 responses. Semin Immunol. 2001;13(5):275–282. doi: 10.1006/smim.2001.0323 [DOI] [PubMed] [Google Scholar]
- 173.Athie-Morales V, Smits HH, Cantrell DA, Hilkens CM. Sustained IL-12 signaling is required for Th1 development. J Immunol. 2004;172(1):61–69. doi: 10.4049/jimmunol.172.1.61 [DOI] [PubMed] [Google Scholar]
- 174.Ma N, Xing C, Xiao H, et al. C5a regulates IL-12+ DC migration to induce pathogenic Th1 and Th17 cells in sepsis. PLoS One. 2013;8(7):e69779. doi: 10.1371/journal.pone.0069779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Dang B, Gao Q, Zhang L, et al. The glycolysis/HIF-1α axis defines the inflammatory role of IL-4-primed macrophages. Cell Rep. 2023;42(5):112471. doi: 10.1016/j.celrep.2023.112471 [DOI] [PubMed] [Google Scholar]
- 176.Wang S, Gao X, Shen G, et al. Interleukin-10 deficiency impairs regulatory T cell-derived neuropilin-1 functions and promotes Th1 and Th17 immunity. Sci Rep. 2016;6:24249. doi: 10.1038/srep24249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Kyttaris VC, Zhang Z, Kuchroo VK, Oukka M, Tsokos GC. Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice. J Immunol. 2010;184(9):4605–4609. doi: 10.4049/jimmunol.0903595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Beringer A, Noack M, Miossec P. IL-17 in chronic inflammation: from discovery to targeting. Trends Mol Med. 2016;22(3):230–241. doi: 10.1016/j.molmed.2016.01.001 [DOI] [PubMed] [Google Scholar]
- 179.Singh V, Agrewala JN. Regulatory role of pro-Th1 and pro-Th2 cytokines in modulating the activity of Th1 and Th2 cells when B cell and macrophages are used as antigen presenting cells. BMC Immunol. 2006;7:17. doi: 10.1186/1471-2172-7-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Sun L, Yu G, Yang S, Zhang L. Effects of hydrocortisone on the differentiation of human T helper 2 cells. Scand J Immunol. 2011;73(3):208–214. doi: 10.1111/j.1365-3083.2010.02500.x [DOI] [PubMed] [Google Scholar]
- 181.Zhang H, Yang X, Yang H, et al. Modulation of mast cell proteinase-activated receptor expression and IL-4 release by IL-12. Immunol Cell Biol. 2007;85(7):558–566. doi: 10.1038/sj.icb.7100085 [DOI] [PubMed] [Google Scholar]
- 182.Salti SM, Hammelev EM, Grewal JL, et al. Granzyme B regulates antiviral CD8+ T cell responses. J Immunol. 2011;187(12):6301–6309. doi: 10.4049/jimmunol.1100891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Morissette MC, Parent J, Milot J. Perforin, granzyme B, and FasL expression by peripheral blood T lymphocytes in emphysema. Respir Res. 2007;8(1):62. doi: 10.1186/1465-9921-8-62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Wang J, Xu H, Li Z, et al. Pathogens in patients with granulomatous lobular mastitis. Int J Infect Dis. 2019;81:123–127. doi: 10.1016/j.ijid.2019.01.034 [DOI] [PubMed] [Google Scholar]
- 185.Russell SP, Neary C, Abd Elwahab S, et al. Breast infections - microbiology and treatment in an era of antibiotic resistance. Surgeon. 2020;18(1):1–7. doi: 10.1016/j.surge.2019.03.008 [DOI] [PubMed] [Google Scholar]
- 186.Shoyele O, Vidhun R, Dodge J, et al. Cystic neutrophilic granulomatous mastitis: a clinicopathologic study of a distinct entity with supporting evidence of a role for Corynebacterium-targeted therapy. Ann Diagn Pathol. 2018;37:51–56. doi: 10.1016/j.anndiagpath.2018.08.005 [DOI] [PubMed] [Google Scholar]
- 187.Zhou F, Li H, Wang F, et al. Efficacy and safety of rifampicin-based triple therapy for non-puerperal mastitis: a single-arm, open-label, prospective clinical trial. Int J Infect Dis. 2024;140:25–30. doi: 10.1016/j.ijid.2023.12.008 [DOI] [PubMed] [Google Scholar]
- 188.Krijbolder DI, Verstappen M, van Dijk BT, et al. Intervention with methotrexate in patients with arthralgia at risk of rheumatoid arthritis to reduce the development of persistent arthritis and its disease burden (TREAT EARLIER): a randomised, double-blind, placebo-controlled, proof-of-concept trial. Lancet. 2022;400(10348):283–294. doi: 10.1016/S0140-6736(22)01193-X [DOI] [PubMed] [Google Scholar]
- 189.Nakafero G, Grainge MJ, Williams HC, et al. Risk stratified monitoring for methotrexate toxicity in immune mediated inflammatory diseases: prognostic model development and validation using primary care data from the UK. BMJ. 2023;381:e074678. doi: 10.1136/bmj-2022-074678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Garneau AP, Riopel J, Isenring P. Acute methotrexate-induced crystal nephropathy. N Engl J Med. 2015;373(27):2691–2693. doi: 10.1056/NEJMc1507547 [DOI] [PubMed] [Google Scholar]




