Advances in Dermatology and Allergology
eISSN: 2299-0046
ISSN: 1642-395X
Advances in Dermatology and Allergology/Postępy Dermatologii i Alergologii
Current Issue Archive Manuscripts accepted About the journal Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
5/2025
vol. 42
 
Share:
Share:
Original paper

Effect of sirolimus on connective tissue disease related refractory thrombocytopenia: clinical efficacy and immunomodulatory mechanism

Xuemei Li
1
,
Li Su
1
,
Xuejing Sun
2
,
Xia Li
1
,
Zhen Tian
1
,
Qiuju Liao
1
,
Wanling Sun
2
,
Yi Zhao
1

  1. Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University, Beijing, China
  2. Department of Haematology, Xuanwu Hospital, Capital Medical University, Beijing, China
Adv Dermatol Allergol 2025; XLII (5): 494–501
Online publish date: 2025/10/10
Article file
- Effect.pdf  [0.17 MB]
Get citation
 
 

Introduction

Connective tissue diseases (CTDs), which include systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjogren’s syndrome (SS), and so on, represent a group of autoimmune disorders that predominantly affect the body’s connective tissues. The pathogenesis of CTDs is complex and not entirely understood, but these diseases are known to potentially affect various organs and systems. A notable and frequent complication of CTDs is thrombocytopenia, which is defined by a platelet count below 100 × 109/l. Thrombocytopenia, particularly in its severe form (platelet count below 30 × 109/l), poses significant health burdens, including the potential for life-threatening gastrointestinal and intracranial bleeding. Consequently, the severity of thrombocytopenia is recognized as an independent risk factor for increased morbidity and mortality in patients with SLE [1].

A challenging aspect in the management of CTDs is the occurrence of refractory thrombocytopenia (RTP), which is characterized by an inadequate response to standard treatments such as glucocorticoids and immunosuppressive therapy. In such cases, alternative therapeutic strategies, including thrombopoietin and its receptor agonists, rituximab, and splenectomy, are often considered. Dysregulation in the balance of immune cells, including different subsets of T cells (Th1, Th17 and Tregs) and B cells is considered to be a driving factor in the development of RTP [2, 3]. This situation may be caused by a deficiency or dysfunction of Treg cells, which allow autoreactive T cells to become uncontrollable and mediate the production of autoantibodies against platelets [4]. The anti-platelet autoantibodies produced by B cells play a central role in immune thrombocytopenia (ITP), both in the Fc-dependent pathway-mediated increase in platelet destruction and in the non-Fc-dependent pathway-mediated inhibition of platelet production by megakaryocytes and the induction of desialylation that interferes with platelet production and impairs their lifespan [5]. Compared to non-refractory thrombocytopenia, RTP has a higher proportion of T cells involved in inflammation and immune activation [6]. Semple et al. have suggested that the elevated levels of IL-2, IFN-γ, and IL-10 in the serum of RTP patients are due to the accumulation of serum cytokines caused by abnormal activation of T cells [2]. The main function of Tregs is to block the pathogenic immune responses mediated by autoreactive cells, to establish and maintain immune homeostasis. It is reported that the population of Tregs is reduced in SS patients’ blood and salivary glands [7]. Studies have confirmed that the decreased Tregs levels are closely associated with SLE progression [8, 9]. The increase of Tregs results in the suppression of inflammatory response and the relief on pathological injury in SLE mice [10].

Recently, sirolimus, a macrolide antifungal antibiotic known for its immunosuppressive properties, has emerged as a promising treatment option for CTD-RTP [11, 12]. Sirolimus exerts its therapeutic effects primarily through the inhibition of the mammalian target of the rapamycin (mTOR) pathway, which plays a crucial role in the activation of immune cells involved in inflammation [13].

Currently, there is limited research on the effect of sirolimus in the immune mechanism of CTD-RTP.

Aim

The main purpose of this study is to further verify the clinical efficacy and safety of sirolimus in patients with CTD-RTP and to explore the impact on various immunological parameters, including T cell subsets, regulatory T cells (Tregs), B cells and cytokine profiles in the peripheral blood.

Material and methods

Study design

A retrospective, open-label, single-arm study was performed to evaluate the safety and efficacy of sirolimus in patients with CTD-RTP. The study was conducted following approval from the Ethics Committee of Xuanwu Hospital, Capital Medical University (Approval No. [2022]091). Informed consent was obtained from all participants. Subjects diagnosed with CTD-RTP were recruited from Xuanwu Hospital between November 2020 and December 2023.

Inclusion and exclusion criteria

In this study, eligible participants were individuals 18 years of age or older who met the diagnostic criteria for connective tissue disease as outlined by either the American College of Rheumatology or the European League Against Rheumatism [14, 15]. All patients had thrombocytopenia (platelet count less than 30 × 109/l at enrolment) and had previously not responded to at least one round of methylprednisolone pulse therapy (1 g/day for 3 consecutive days) and/or intravenous immunoglobulin G (20 g/day for 3–5 consecutive days) and/or had an inadequate response to glucocorticoids (prednisone 1 mg/kg/day or equivalent dose) or standard treatments, or had relapsed after glucocorticoid reduction. Patients in the study received treatments such as cyclophosphamide (1000 mg intravenous pulse monthly or 100 mg/day orally), mycophenolate mofetil (2–3 g/day), FK506 (tacrolimus; 2–3 mg/day), or MMF A (3–5 mg/kg/day). Oral prednisone was either continued or tapered to 5 mg in 8 weeks for maintenance. Previous immunosuppressants were maintained without introducing new ones during the study. Exclusion criteria comprised incomplete baseline data, secondary thrombocytopenia diseases, and severe infectious complications. The patient inclusion flow chart is detailed in Figure 1.

Figure 1

The flowchart of this study. CTD-RTP patients who met the inclusion criteria were administered sirolimus (SRL) 1 mg/day, after 6 months of SRL oral administration, an evaluation of SRL efficacy was conducted

/f/fulltexts/PDIA/56963/PDIA-42-5-56963-g001_min.jpg

Efficacy evaluation

The efficacy of therapy was categorized as follows: complete response (CR), partial response (PR), and non-response (NR). CR was defined as a post-treatment platelet count of ≥ 100 × 109/l without bleeding. PR was indicated by a platelet count of ≥ 30 × 109/l, at least double the baseline count, without bleeding. NR was characterized by a post-treatment platelet count of < 30 × 109/l, less than double the baseline count or the presence of bleeding. Sustained effectiveness was defined as the maintenance of treatment efficacy for 6 months or longer. Early response was characterized by meeting the effectiveness criteria within 1 week of starting treatment. The initial response was identified when the effectiveness criteria were achieved 1 month after initiating treatment. Complete remission was indicated by a platelet count of ≥ 100 × 109/l, observed 12 months following the commencement of treatment. To determine CR or response (R), platelet counts were examined at least twice with a minimum interval of 7 days. Recurrence was determined based on at least two measurements with a minimum 1-day interval [16].

Laboratory measurements

(1) Cytokine detection: Cytokine levels were measured using flow cytometry. Peripheral blood samples were processed to obtain plasma, and the levels of cytokines (including interleukin [IL]-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12P70, IL-17A, IFN-γ, TNF-α, and IFN-α) were quantified using a cytokine kit (Jiangxi Saiji Biotechnology) and the BD FACSAria II flow cytometer.

(2) T-cell and B-cell subset analysis: Immunophenotyping was performed on EDTA-anticoagulated peripheral blood samples. Monoclonal antibodies (including CD3-FITC, CD45-PerCP-Cy5.5, CD4-PE Cy7, CD56- PE, CD16-PE, CD19-APC, and CD8-APC-Cy) were used to characterize lymphocyte subpopulations. Percentages and absolute counts of each subset were determined using BD TrucountTM tubes (BD Biosciences).

Treg cell detection

Treg cells in peripheral blood were identified using 100 μl of EDTA anticoagulant blood, labelled with fluorescent antibodies including HLA-DR-FITC, CD4-PE-Cy7, CD45-PerCP-Cy5.5, CD25-PE, and CD127-PE-Cy7. The identification of CD3+CD4+CD25+CD127- cells as total Treg cells was performed, and their proportion in CD4+ T cells was quantified.

Statistical analysis

Continuous variables following a normal distribution were expressed as mean ± standard deviation (SD), while non-normally distributed data were presented as median ± interquartile range (25th–75th percentile). The comparison of baseline characteristics was conducted using the independent samples t-test (for parameters with normal distribution) and Mann-Whitney test (for parameters that did not follow a normal distribution). The paired t-test was used to compare the two groups before and after treatment for numerical variables with a normal distribution, while the Wilcoxon signed-rank test was used for data not meeting the normal distribution assumption. A p-value of less than 0.05 was considered statistically significant. All statistical analyses were conducted using SPSS software, version 26.0.

Results

Patient characteristics

A total of 13 patients (comprising 3 men and 10 women) diagnosed with CTD-RTP receiving sirolimus treatment were included in our study. Six patients met the diagnostic criteria for SLE, 7 patients met the criteria for primary Sjogren’s syndrome (pSS). The median age was 67 years old (ranging from 23 to 84 years). The median duration of CTD and thrombocytopenia was 3 years (range: 0.5 to 13 years) and 2 years (range: 0.5 to 7 years), respectively. All patients had platelet count below 30 × 109/l at the time of enrolment and had previously not responded to at least one round of methylprednisolone pulse therapy (1 g/day for 3 consecutive days) and/or intravenous immunoglobulin G (20 g/day for 3–5 consecutive days) and/or had an inadequate response to glucocorticoids (prednisone 1 mg/kg/day or equivalent dose) or standard treatments, or had relapsed after glucocorticoid reduction. Thus, they received the sirolimus therapy.

Clinical efficacy of sirolimus

All 13 patients had baseline platelet counts below 30 × 109/l. Specifically, 2 (15.4%) patients had counts below 10 × 109/l, 4 (30.8%) patients had counts between 10 × 109/l and 20 × 109/l, and 7 (53.8%) patients had counts between 20 × 109/l and 30 × 109/l. After 3 months of sirolimus treatment (1 mg/day), 7 (53.8%) patients achieved PR, 5 (38.5%) patients showed NR, and 1 (7.69%) patient achieved CR, culminating in a total response rate of 61.5%. At the 6-month follow-up, 4 (30.8%) patients achieved CR, 6 (46.2%) patients maintained PR, and 3 (23.1%) patients continued to show NR. The overall effective rate (CR + PR) was 76.9% (Table 1, Figure 2). In 6 SLE patients, the CR rate, PR rate, and NR rate after 3 months of sirolimus treatment were 16.6%, 33.6%, and 50%, respectively. The CR rate, PR rate, and NR rate after 6 months of sirolimus treatment were 16.6%, 50%, and 33.3%, respectively. In 7 pSS patients, the CR rate, PR rate, and NR rate after 3 months of sirolimus treatment were 0%, 71.4%, and 28.6%, respectively. The CR rate, PR rate, and NR rate after 6 months of sirolimus treatment were 42.9%, 42.9%, and 14.3%, respectively.

Table 1

Clinical profile of 13 CTD-RTP patients treated with sirolimus

CaseGendert0 [years]t1 [years]t2 [years]CTDPrevious GCsmaxPrevious ISsSevere bleedingComplicationsDrug combinationCE3 mCE6 mADRs
1Female2320.5SLE1 mg/kg/dayHCQNoThyroid nodulesPred 60 mg – 5 mg/day, HCQCRCRNo
2Male7134pSS1 mg/kg/dayMMFNoAPS, HTNPred 5 mg/day, MMFPRCROral ulcers. Hypercholesterolemia. Fungal infection
3Male7340.58SLE1 mg/kg/dayIVIG, CTX, HCQNoIPFPred 5 mg/ day, HCQPRPROral ulcers
4Female66102SLE1000 mg/day × 3IVIG, CsANoDyslipidaemiaMP 4 mg/ day
CsA
PRPRGastrointestinal reaction
5Male7422pSS1 mg/kg/dayFK506, TII
CsA
NoDyslipidaemia, Diabetes, CHD, Atherosclerosis, Intermuscular artery thrombosis, HTNPred 5 mg/day, Rivaroxaban
CsA
PRPRHypercholesterolemia
6Female41133pSS1 mg/kg/dyIVIG, HCQNoPleural effusionPred 5 mg
HCQ
PRCROral ulcers
7Female5852pSS1 mg/kg/dayFK506 IVIG
CsA
Intracranial bleedingCHD, AnemiaPred 60 mg – 5 mg/day, CsANRNRHypercholesteremia
8Female6732SLE1 mg/kg/dayIVIG, CsAGastrointestinal bleedingHTN, Lower extremities DVTPred 60 mg – MP 4 mg/day, CsANRNROral ulcers
9Female7037SLE1 mg/kg/dayIVIG, CTXNoHTNPred 60 mg – 5 mg, HCQNRPRHypercholesteremia
10Female690.50.83SLE500 mg × 3IVIG, CsA,
Romiplostim
Alveolar haemorrhageAIHAPred 60 mg – 5 mg, CsANRNRNo
11Female6154pSS1 mg/kg/dayCsA, HCQNoAIHAPred 5 mg
CsA
PRPROral ulcers
12Female4923pSS1 mg/kg/dayIVIG, CsANoNoPred 5 mg, CsANRPROral ulcers
13Female8430.5pSS1 mg/kg/dayIVIG, CsA, RTXNoDiabetes, AFPred 60 mg
– 5 mg, CsA
PRCRHypercholesteremia

[i] t0 – age at diagnosis of connective tissue disease, t1 – duration of CTD. t2 – duration of thrombocytopenia. GCsmax – maximum dose of previous glucocorticoids, CTD – connective tissue disease, SLE – systemic lupus erythematosus, pSS – primary Sjogren’s syndrome, APS – antiphospholipid syndrome, CE – curative effect, CR – complete remission, PR – partial remission, NR – no response, Pred – prednisone, HCQ – hydroxychloroquine, MTX – methotrexate, MMF – mycophenolate mofetil, IVIG – intravenous immunoglobulin, CTX – cyclophosphamide, CsA – cyclosporine A, FK506 – tacrolimus, TII – tripterygium glycosides, MP – methylprednisolone, RTX – rituximab, HTN – hypertension, IPF – interstitial pulmonary fibrosis, AIHA – autoimmune haemolytic anemia, AF – atrial fibrillation, CHD – coronary heart disease, DVT – deep vein thrombosis, ADRs – adverse drug reactions

Figure 2

The number of complete response (CR), partial response (PR), and no response (NR) patients at 3 and 6 months after sirolimus treatment

/f/fulltexts/PDIA/56963/PDIA-42-5-56963-g002_min.jpg

Safety

Regarding the safety of sirolimus treatment, no patients had experienced grade 2 or higher adverse events. Adverse reactions to sirolimus included oral ulcers (46.2%), hypercholesterolemia (38.5%), gastrointestinal reactions (7.7%), and fungal infections (7.7%). The adverse events observed after treatment with sirolimus are summarized in Table 1.

Immunological parameters

Flow cytometry analysis was performed to measure the lymphocyte subsets in patients before and after sirolimus treatment. We observed a notable alteration in the proportions of T subsets before and after sirolimus treatment. The results showed that, after sirolimus treatment, the ratio of Tregs (p < 0.05) was significantly increased. In contrast, after sirolimus treatment, the CD3+, CD8+, and CD4+ cell counts were significantly decreased (p < 0.05). However, the cell numbers of CD16+CD56+ NK cells and CD19+ B cells were not statistically significant before and after treatment (p > 0.05) (Table 2).

Table 2

Changes in platelet counts, Treg cell counts, and other lymphocyte subsets, before and after sirolimus treatment

ParameterBefore treatmentAfter treatmentP-value
Treg cells (%)7.79 ±1.3912.44 ±2.150.019*
Platelets (× 109/l)17.92 ±2.4970.31 ±10.70< 0.001*
CD3+ T cells/μl1318.23 ±113.14888.97 ±63.520.005**
CD8+ T cells/μl539.92 ±57.07310.15 ±31.370.004**
CD4+ T cells/μl741.31 ±86.80564.00 ±66.940.001**
CD16+CD56+ NK cells/μl213.01 ±34.05184.69 ±26.860.270
CD19+ B cells/μl279.00 (232.50, 340.00)234.00 (184.00, 269.50)0.687

* A significant increase post-treatment (p < 0.05),

** a significant decrease post-treatment (p < 0.01). Treg – percentage of regulatory T cells.

Cytokine levels

The concentrations of cytokines in patients before and after treatment were detected. The results showed that, after sirolimus treatment, the levels of IL-8 and IL-17A were significantly reduced (p < 0.05). However, the levels of IL-2, IL-4, IL-5, IL-6, IL-1B, IL-10, TNF-α, INF-α, IL-12 P70, and INF-γ were not statistically significant before and after treatment (p > 0.05) (Table 3).

Table 3

Cytokine levels before and after sirolimus treatment

CytokineBefore treatment [pg/ml]After treatment [pg/ml]P-value
IL-21.20 (0.15, 1.99)1.11 (0.84, 1.78)0.349
IL-40.60 (0.04, 2.36)1.24 (0.35, 1.91)0.863
IL-50.13 (0, 1.41)1.20 (0, 6.74)0.278
IL-67.98 (2.39, 17.95)6.98 (3.75, 11.79)0.228
IL-87.80 (6.80, 9.75)6.90 (6.00, 8.65)0.049*
IL-1B0.44 (0, 2.15)1.00 (0.61, 2.40)0.136
IL-17A11.52 ±5.218.92 ±4.940.039*
IL-101.61 (0.93, 3.34)1.88 (1.43, 4.60)0.337
TNF-α0.39 (0.93, 3.34)0.23 (0, 0.84)0.129
INF-α0.38 (0, 0.95)0.30 (0, 0.89)0.943
IL-12 P700 (0, 0.87)0.60 (0, 1.22)0.590
INF-γ0.43 (0, 1.67)0.11 (0, 0.53)0.063

* A significant decrease post-treatment (p < 0.05). IL – interleukin, TNF-α – tumor necrosis factor-α, INF – interferon.

Discussion

CTD-RTP is characterized by an inadequate response to standard treatments such as glucocorticoids and immunosuppressive therapy. The current treatment methods include thrombopoietin and its receptor agonists, rituximab, and splenectomy. Our study concentrated on evaluating the efficacy of sirolimus in treating patients with CTD-RTP. We discovered that sirolimus treatment resulted in overall response rate of 61.5% at the 3 months after treatment and 76.9% at 6 months following treatment. These findings demonstrate that sirolimus may be effective for CTD-RTP treatment.

Sirolimus (an mTOR inhibitor), known for its low toxicity, has emerged as a potent treatment for refractory thrombocytopenia, particularly in patients with CTDs such as SLE [17] and primary Sjogren’s syndrome (pSS) [18]. A pilot study by Du et al. demonstrated that 50% of patients achieved CR and 16.7% achieved PR when sirolimus was administered in patients with CTD-RTP [12]. Our present clinical trial demonstrated that sirolimus was effective for CTD-RTP as evidenced by the significant increase of platelet count. After 3 months of sirolimus treatment, the total overall effective rate (CR + PR) was 61.5%. At the 6-month follow-up, 4 (30.8%) patients achieved CR, 6 (46.2%) patients maintained PR, and 3 (23.1%) patients continued to show NR. The overall effective rate was 76.9%, surpassing the rates reported in previous literature. The higher efficacy rate of sirolimus in our study, compared to previous reports, could be attributed to the inclusion of more patients with pSS. This study highlights sirolimus’s potential as an alternative regimen for refractory CTD-RTP, including in SLE and pSS patients. Importantly, the overall incidence of adverse reactions in our study was mild, primarily involving self-healing oral ulcers and mild hypercholesterolemia, which normalized following statin therapy. This safety profile is similar to findings from a study on high-dose sirolimus in kidney transplant rejection patients [19]. These results suggest that sirolimus, unlike other immunosuppressants such as rituximab or cyclosporine, which can have severe drug side effects leading to significant immune deficiency and infections, is suitable as a second-line option for treating immune-mediated thrombocytopenia. However, caution is advised when combining sirolimus with other immunosuppressants as there have been reports of fungal infections and alveolar haemorrhage in kidney transplant patients treated with sirolimus and mycophenolate mofetil [20]. In our study, 1 patient who received sirolimus and mycophenolate mofetil displayed a pulmonary fungal infection and subsequently experienced a temporary reduction in platelet count, which normalized after antifungal treatment. This underscores the importance of vigilance regarding potential fungal infections when using sirolimus in combination with other immunosuppressive agents. Altogether, sirolimus may be a safe and effective therapeutic option for CTD-RTP patients.

The efficacy of sirolimus in treating immune-mediated thrombocytopenia is attributed to its role as an mTOR inhibitor, which modulates lymphocyte activity, a key factor in the pathogenesis of ITP [21]. Importantly, sirolimus has been shown to restore the balance of various immune cell types, including Tregs, Th2, and Th17 cells, which are often disrupted in autoimmune conditions like ITP [22]. In an open-label, prospective clinical trial involving 86 patients with RTP [23], 40% achieved CR by the third month of treatment. After 6 months, 41% of patients maintained CR, and the overall response rate remained at 65% at the 12-month follow-up. The study also observed a decrease in the proportion of Th2 and active Th17 cells, while the percentage of Treg cells increased. These findings suggest that sirolimus may help reestablish peripheral immune tolerance in ITP patients. Common occurrences in SLE include both numerical and functional deficiencies of Tregs [24] as well as the activation of the mTOR pathway. It is reported that T cells in SLE patients exhibit mitochondrial hyperpolarization (MHP) and adenosine triphosphatase (ATP) depletion, leading to a propensity for pro-inflammatory death [25]. The mTOR pathway, acting as a sensor for MHP, plays a crucial role in T cell activation in SLE. In our study, the increase in Tregs and platelet counts observed aligns with the known role of the mTOR pathway in immune thrombocytopenia [26], especially in SLE patients [27]. Additionally, our study observed a reduction in CD3+, CD8+, and CD4+ cells, as well as a decrease in cytokine levels of IL-17A and IL-8 following sirolimus treatment. This aligns with previous reports indicating that sirolimus can reduce the levels of IL-17 and other inflammatory factors in SLE patients, which is consistent with its known immunomodulatory effects [28]. In addition, levels of activated Th17 cells were reported to be elevated in ITP patients, and produce large amounts of IL-17, IL-21, and IL-22 [29]. These studies support our findings. However, in this study, we found no significant change in the number of B cells before and after treatment of sirolimus. Research indicates that sirolimus can stimulate B-cell activating factor (BAFF)-induced B-cell proliferation and reduce the production of autoantibodies [30]. In vitro studies have shown that sirolimus can block the proliferation of anti-Sjogren’s syndrome B antibodies (anti-SSB antibodies) and inhibit immunoglobulin production [18]. This suggests that sirolimus may have a broader application in various CTDs, where conventional therapies might not be as effective.

Interestingly, in our study, there was a case considered to be thrombocytopenia secondary to antiphospholipid syndrome (APS) associated with SLE, where sirolimus treatment was also effective. This aligns with reports suggesting that the mTOR pathway plays a role in the development of vasculopathy in APS [31]. In kidney transplant recipients with APS, treatment with sirolimus has been associated with a lack of recurrence of vascular lesions and decreased vascular proliferation, as compared to patients not receiving sirolimus [32]. These findings suggest that combining sirolimus with anticoagulants may not only improve the prognosis of APS but also prevent new thrombosis.

Limitations

Some limitations should deserve our attention. First, the single-centre, retrospective nature, small sample size, and short observation period may limit the generalizability of the findings. Future research should focus on multi-centre, large-scale clinical trials to validate or refute these findings. In addition, the lack of assessment of bleeding scores and health-related quality of life during follow-up was also a limitation of this study. Additionally, exploring the long-term effects of sirolimus and its combination with other immunosuppressants in a broader range of CTDs would be valuable. Investigating the specific mechanisms through which sirolimus affects different immune cell types could also provide deeper insights into its therapeutic potential and help tailor treatment strategies for individual patients.

Conclusions

This study contributes to the field of CTD and immunotherapy by demonstrating the effectiveness of sirolimus in a specific subset of CTD-RTP patients. It highlights the potential of sirolimus not only in reducing thrombocytopenia but also in modulating the immune system. This study finds that sirolimus increases Treg cells and platelet counts, but decreases CD3+, CD8+, and CD4+ cells, as well as suppresses the levels of cytokines IL-8 and IL-17A. These findings may provide a basis for the use of sirolimus in clinical practice, making it a promising another agent for refractory or relapsed CTD-RTP.

Ethical approval

The study was approved by the Ethics Committee of Xuanwu Hospital, Capital Medical University (Approval No. [2022]091).

Conflict of interest

The authors declare no conflict of interest.

References

1 

Yang F, Tian J, Peng L, et al. Thrombocytopenia is an independent risk factor for the prognosis of thrombotic microangiopathy in Chinese patients with systemic lupus erythematosus. Front Med 2021; 8: 772607.

2 

Semple JW, Milev Y, Cosgrave D, et al. Differences in serum cytokine levels in acute and chronic autoimmune thrombocytopenic purpura: relationship to platelet phenotype and antiplatelet T-cell reactivity. Blood 1996; 87: 4245-54.

3 

Zhang J, Ma D, Zhu X, et al. Elevated profile of Th17, Th1 and Tc1 cells in patients with immune thrombocytopenic purpura. Haematologica 2009; 94: 1326-9.

4 

Audia S, Mahevas M, Nivet M, et al. Immune thrombocytopenia: recent advances in pathogenesis and treatments. Hemasphere 2021; 5: e574.

5 

Roeser A, Lazarus AH, Mahévas M. B cells and antibodies in refractory immune thrombocytopenia. Br J Haematol 2023; 203: 43-53.

6 

Yazdanbakhsh K, Provan D, Semple JW. The role of T cells and myeloid-derived suppressor cells in refractory immune thrombocytopenia. Br J Haematol 2023; 203: 54-61.

7 

Liu MF, Lin LH, Weng CT, et al. Decreased CD4+CD25+bright T cells in peripheral blood of patients with primary Sjogren’s syndrome. Lupus 2008; 17: 34-9.

8 

Zhu Y, Huang Y, Ming B, et al. Regulatory T-cell levels in systemic lupus erythematosus patients: a meta-analysis. Lupus 2019; 28: 445-54.

9 

Ohl K, Tenbrock K. Regulatory T cells in systemic lupus erythematosus. Eur J Immunol 2015; 45: 344-55.

10 

Scalapino KJ, Daikh DI. Suppression of glomerulonephritis in NZB/NZW lupus prone mice by adoptive transfer of ex vivo expanded regulatory T cells. PLoS One 2009; 4: e6031.

11 

Wu C, Wang Q, Xu D, et al. Sirolimus for patients with connective tissue disease-related refractory thrombocytopenia: a single-arm, open-label clinical trial. Rheumatology (Oxford) 2021; 60: 2629-634.

12 

Du H, Su W, Su J, et al. Sirolimus for the treatment of patients with refractory connective tissue disease-related thrombocytopenia: a pilot study. Rheumatology (Oxford) 2024; 63: 79-84.

13 

Panwar V, Singh A, Bhatt M, et al. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct Target Ther 2023; 8: 375.

14 

Aringer M, Costenbader K, Daikh D, et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann Rheum Dis 2019; 78: 1151-9.

15 

Shiboski CH, Shiboski SC, Seror R, et al. 2016 American College of Rheumatology/European League Against Rheumatism classification criteria for primary Sjögren’s syndrome: a consensus and data-driven methodology involving three international patient cohorts. Arthritis Rheumatol 2017; 69: 35-45.

16 

Neunert C, Terrell DR, Arnold DM, et al. American Society of Hematology 2019 guidelines for immune thrombocytopenia. Blood Adv 2019; 3: 3829-66.

17 

Peng L, Wu C, Hong R, et al. Clinical efficacy and safety of sirolimus in systemic lupus erythematosus: a real-world study and meta-analysis. Ther Adv Musculoskelet Dis 2020; 12: 1759720x20953336.

18 

Blokland SLM, Hillen MR, Wichers CGK, et al. Increased mTORC1 activation in salivary gland B cells and T cells from patients with Sjögren’s syndrome: mTOR inhibition as a novel therapeutic strategy to halt immunopathology? RMD Open 2019; 5: e000701.

19 

Yu H, Nysak S, Garg N, et al. ODAE: ontology-based systematic representation and analysis of drug adverse events and its usage in study of adverse events given different patient age and disease conditions. BMC Bioinformatics 2019; 20 (Suppl 7): 199.

20 

Serifoglu I, Dedekarginoglu BE, Soy EHA, et al. Causes of hemoptysis in renal transplant patients. Exp Clin Transplant 2018; 16 (Suppl 1): 70-4.

21 

Suto T, Karonitsch T. The immunobiology of mTOR in autoimmunity. J Autoimmun 2020; 110: 102373.

22 

Kargar M, Torabizadeh M, Purrahman D, et al. Regulatory factors involved in Th17/Treg cell balance of immune thrombocytopenia. Curr Res Transl Med 2023; 71: 103389.

23 

Feng Y, Xiao Y, Yan H, et al. Sirolimus as rescue therapy for refractory/relapsed immune thrombocytopenia: results of a single-center, prospective, single-arm study. Front Med (Lausanne) 2020; 7: 110.

24 

Dao LTM, Vu TT, Nguyen QT, et al. Current cell therapies for systemic lupus erythematosus. Stem Cells Transl Med 2024; 13: 859-872.

25 

Sharabi A, Tsokos GC. T cell metabolism: new insights in systemic lupus erythematosus pathogenesis and therapy. Nat Rev Rheumatol 2020; 16: 100-12.

26 

Wang H, Yu T, An N, et al. Enhancing regulatory T-cell function via inhibition of high mobility group box 1 protein signaling in immune thrombocytopenia. Haematologica 2023; 108: 843-58.

27 

Shan J, Jin H, Xu Y. T cell metabolism: a new perspective on Th17/Treg cell imbalance in systemic lupus erythematosus. Front Immunol 2020; 11: 1027.

28 

Lai ZW, Kelly R, Winans T, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet 2018; 391: 1186-96.

29 

Chen YY, Zhou YQ, Zhao N, et al. Evaluation of IVIG response in relation to Th1/Th2 cytokines in pediatricm immune thrombocytopenia. Cytokine 2019; 120: 234-41.

30 

Zeng Q, Qin S, Zhang H, et al. Rapamycin attenuates BAFF-extended proliferation and survival via disruption of mTORC1/2 signaling in normal and neoplastic B-lymphoid cells. J Cell Physiol 2018; 233: 516-29.

31 

Salet DM, Bekkering S, Middeldorp S, et al. Targeting thromboinflammation in antiphospholipid syndrome. J Thromb Haemost 2023; 21: 744-57.

32 

Dufour I, Venot Q, Aydin S, et al. mTORC pathway activation and effect of sirolimus on native kidney antiphospholipid syndrome nephropathy: a case report. Am J Kidney Dis 2020; 76: 288-91.

Copyright: © 2025 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2025 Termedia Sp. z o.o.
Developed by Bentus.