Skip to main content
Erschienen in: Respiratory Research 1/2020

Open Access 01.12.2020 | Research

The efficacy and safety of pharmacological treatments for lymphangioleiomyomatosis

verfasst von: Qi Wang, Mengqi Luo, Bo Xiang, Siyuan Chen, Yi Ji

Erschienen in: Respiratory Research | Ausgabe 1/2020

Abstract

Background

Lymphangioleiomyomatosis (LAM) is a rare, low-grade multisystem neoplastic disease. Most LAM patients are at a high risk of losing lung function at an accelerated rate and developing progressive dyspnea. Recently, several studies have reported their experience with pharmacological treatments for LAM. Therefore, we conducted a systematic review and meta-analysis to assess the efficacy and safety of these therapies.

Methods

PubMed (Medline), EMBASE, Cochrane Library, Web of Science and EBSCO Host were searched (until March 31, 2019) for eligible prospective studies regarding LAM patients treated with pharmacological treatments. Random effect models were used for quantitative analysis.

Results

Fourteen prospective studies regarding five pharmacological treatments (including sirolimus, everolimus, doxycycline, triptorelin, and a combination therapy of sirolimus and hydroxychloroquine) were enrolled in our systematic review, and ten of them were used for the meta-analysis. Seven prospective studies reported that sirolimus was effective at improving or stabilizing lung function and alleviating renal angiomyolipoma (AML) in LAM patients. Subsequent quantitative analyses showed that during sirolimus treatment, the pooled values of lung function and 6-min walk distance (6MWD) were not significantly changed (P > 0.05), with the pooled response rate of AML being 0.62 (95% confidence intervals [CIs]: 0.43 to 0.82, I2 = 65%). Regarding everolimus, three prospective studies reported similar effects to those of sirolimus with regard to preserving lung function and reducing AMLs. The meta-analysis showed that the changes in lung function during everolimus treatment were not statistically significant (P > 0.05), while the pooled response rate of AML was 0.78 (95% CI: 0.68 to 0.88, I2 = 8%). Neither the qualitative nor the quantitative results confirmed the benefits of doxycycline or triptorelin treatment, and the effects of the combination therapy were unclear in LAM patients. Most of the adverse events during pharmacological treatments were low or moderate grade and tolerable.

Conclusions

Overall, sirolimus and everolimus were recommended for the treatment of LAM because they could stabilize lung function and alleviate renal AML. Doxycycline and triptorelin were not recommended for the treatment of LAM because no beneficial outcomes were consistently observed. The efficacy and safety of combination therapy remain to be further explored.
Begleitmaterial
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-1316-3.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
6MWD
6-min walk distance
AEs
Adverse events
AML
Angiomyolipoma
CI
Confidence interval
DLco
Diffusion coefficient for carbon monoxide
FEV1
Forced expiratory volume in 1 s
FPI
Functional performance inventory
FVC
Forced vital capacity
LAM
Lymphangioleiomyomatosis
MMP
Matrix metalloproteinase
mTOR
Mammalian target of rapamycin
QOL
Quality of life
RCT
Randomized controlled trial
SGRQ
St George’s Respiratory Questionnaire
TSC
Tuberous sclerosis complex
VAS
Visual analogue scale
VEGF-D
Vascular endothelial growth factor D
WMD
Weighted mean difference

Background

Lymphangioleiomyomatosis (LAM) is a rare, low-grade multisystem neoplastic disease characterized by cystic lung destruction, chylous fluid accumulation (pleural or ascitic), angiomyolipomas (AMLs) and lymphangioleiomyomas [1, 2]. LAM occurs sporadically (sLAM) or is associated with tuberous sclerosis complex (TSC). It affects adult females almost exclusively, with a prevalence of approximately five per million, although LAM has also been reported in adult males and children [35]. In LAM, the progressive cystic destruction of the lung can lead to recurrent pneumothorax and dyspnea, resulting in lung function declines at rates of 2–4 times or more compared with the typical age-related decline rate [6, 7]. In many patients, dyspnea with daily activities and hypoxia requiring oxygen support can also occur within 10 years of symptom onset [8]. For LAM patients in the end stage, lung transplantation is the only treatment option [6, 7]. Although AMLs are benign and usually asymptomatic, they may enlarge and bleed, which can lead to chronic kidney disease or require urgent treatment [9]. However, LAM may recur in transplanted lungs [10, 11], and patients with AML often develop new lesions and relapse after treatment [12].
Genetic studies have revealed that patients with LAM have biallelic inactivating mutations in the TSC-1 or TSC-2 gene [13]. Loss of TSC gene function activates the mammalian target of rapamycin (mTOR) signaling pathway, which is a key regulator of cell growth, motility, and survival, resulting in the proliferation of LAM cells [14]. The lungs and lymphatics of LAM patients are infiltrated with LAM cells [15]. Although LAM has a benign histological appearance, LAM cells can circulate in the blood and lymphatic fluids [16]. Moreover, LAM cells can express vascular endothelial growth factor D (VEGF-D), which may facilitate the migration of LAM cells to lymphatic vessels and promote metastatic spread [17, 18].
In addition to lung transplantation for LAM, hormonal therapies (e.g., progestins, gonadotrophin-releasing hormone analogs, and antiestrogen therapies) have previously been the main initial treatment options [1921]. Doxycycline, a tetracycline antibiotic and matrix metalloproteinase (MMP) inhibitor, has also been reported to be effective in the improvement of lung function in LAM patients. Clinical trials have been performed to evaluate the exact effect of doxycycline [22, 23]. Interestingly, mTOR inhibitors (sirolimus and everolimus) have been reported to be effective at reducing AML volume and improving or stabilizing lung function in LAM patients [2426]. In addition, clinical practice guideline have recommended sirolimus for LAM patients with declining lung function and problematic chylous effusions, although this recommendation is based on moderate-quality to very low-quality evidence [1]. Because LAM is an orphan disease, it is inevitable that the sample size and data collection will be limited. To date, there is still a lack of studies evaluating the efficacy and safety of all pharmacological treatments for LAM.
Therefore, we performed this literature review to present updated insights into the pharmacological treatment of LAM and to conduct a meta-analysis that assesses the efficacy and safety of these therapies.

Methods

Search strategy

Studies were identified by searching PubMed (Medline), EMBASE, Cochrane Library, Web of Science and EBSCO Host until March 31, 2019. OpenThesis and OpenGrey were also searched for gray literature. Medical Subject Headings (MeSH) and free text words were applied during the search, including (‘Lymphangioleiomyomatosis’ OR ‘Lymphangioleiomyomatoses’ OR ‘Lymphangiomyomatosis’ OR ‘Lymphangiomyomatoses’) AND (‘Therapeutics’ OR ‘Therapeutic’ OR ‘Therapy’ OR ‘Therapies’ OR ‘Treatment’ OR ‘Treatments’). Publications were restricted to research on humans and studies written in English or Chinese. The reference lists were also searched for additional relevant studies. This systematic review was registered in the s database with the registration number CRD42019122366.

Inclusion criteria and exclusion criteria

Studies were included if they met the following inclusion criteria: (1) any pharmacological treatment for sLAM or TSC-LAM, with or without a control group, and (2) a clear description of outcomes, adverse events (AEs) and side effects. The exclusion criteria were set as follows: (1) duplicate publications; (2) research presenting interim or extended findings regarding the same group of patients; (3) studies with a sample size of fewer than five patients; (4) publications with no detailed original data (such as abstracts, posters, conference reports, letters, case reports, reviews and meta-analyses); and (5) studies with a retrospective design.

Data extraction

Two reviewers (WQ and LMQ) independently assessed the eligibility and risk of bias of the studies and extracted the data. Disagreements were resolved by discussion with the third investigator (JY). For each suitable study, the following data were collected using a standard Excel form: (1) general information: name of first author, publication year, study location, study design, number and diagnosis of participants, study duration, treatment name and regimen, and study outcomes; (2) baseline and follow-up efficacy data: forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), diffusing capacity for carbon monoxide (DLco), 6-min walk distance (6MWD), quality of life (QOL) scores, serum level of VEGF-D and response rate of AML; and (3) AEs (mentioned in at least 2 studies): types of AEs and numbers of patients with AEs. First author and corresponding author were contacted to obtain missing data when possible.

Quality assessment

The risk of bias of the randomized controlled trials (RCTs) was assessed by the Cochrane criteria with regard to the following items: random sequence generation, allocation concealment, blinding of participants, personnel and outcome assessments, incomplete outcome data, selective reporting and other bias. The quality of single-arm trials was assessed according to the methodological index for nonrandomized studies (MINORS) [27], and the items included a clear stated aim, inclusion of consecutive patients, prospective collection of data, endpoints appropriate to the aim of the study, unbiased assessment of the study endpoint, follow-up period appropriate to the aim of the study, loss to follow-up less than 5% and prospective calculation of the sample size.

Data synthesis and statistical analysis

In this study, the protocol was performed based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [28]. Review Manager (version 5.3) and R software were used to perform the meta-analysis. For continuous data including the 6MWD, serum level of VEGF-D, and absolute values of FEV1, FVC and DLco at baseline and the endpoint, we extracted the means and standard deviations (SDs) from the included articles. The means and SDs were estimated by the methods described in the Cochrane handbook [29] or were estimated from the sample size, median, range and/or interquartile range if they were not directly reported [3032]. Moreover, we converted some data to ensure that they had the same unit of measure. The effect sizes were analyzed using a random effect model and are reported as the weighted mean difference (WMD). For dichotomous data, the pooled proportions were analyzed by R software with a meta-package in a random effect model. In addition, 95% confidence intervals (CIs) were reported for each measure. Heterogeneity among the included studies was evaluated by I2 statistics (low heterogeneity: I2 ≤ 25%; moderate: 25–50%; high> 75%). A sensitivity analysis omitting each included study in turn was also performed to evaluate the effects of individual studies on the statistical results. Probability values < 0.05 were considered to indicate statistical significance.
As the number of included articles about each pharmacological treatment was fewer than 10, publication bias could not be explored in this study.

Results

Search results and characteristics of the included studies

The search and screening process for the studies is presented in Fig. 1. A total of 2081 records were identified through the initial database search. After removing duplicates and reviewing titles and abstracts, the eligibility of the remaining 118 articles was carefully assessed by a full-text review. Finally, only 14 studies met the inclusion criteria for the systematic review, including 3 RCTs [23, 25, 33] and 11 single-arm trials [9, 20, 22, 24, 3440], of which ten studies were included in the meta-analysis [2225, 3336, 39, 40]. Bissler et al. reported accurate data on lung function in LAM patients treated with everolimus in an article published in 2017 [41]; these data were not available in the initial article published in 2013 [25]. Therefore, we cited these two articles as the same RCT. Because there was only one article each describing triptorelin monotherapy [20] and a combination therapy of sirolimus plus hydroxychloroquine [38], these two articles were only used for qualitative analyses. Moreover, the endpoints and/or baseline lung function values were not available from the studies by Takada et al. [37] and Bee et al. [9]. Therefore, these two articles were not included in the meta-analysis.
The characteristics of the included studies are summarized in Table 1. The included prospective clinical trials and RCTs reported five pharmacological treatments for 330 LAM patients. Among them, 46 patients in 1 RCT treatment group [33] and 164 patients in 6 single-arm trials [9, 24, 34, 35, 37, 40] were treated with sirolimus. Twenty-three patients in 1 RCT treatment group [25] and 30 patients in 2 single-arm trials [36, 39] were treated with everolimus. Twelve patients in 1 RCT treatment group [23] and 31 patients in a single-arm trial [22] were treated with doxycycline. In addition, 11 patients were reported to be treated with triptorelin [20], and 13 patients were reported to be treated with a combination of sirolimus and hydroxychloroquine [38] in single-arm trials. The quality assessments of the RCTs and single-arm trials are presented in Additional file Table A1 and Table A2.
Table 1
Characteristics of the included studies
Author, Publication Year, Country
Journal
Study design
Diagnosis,
No. of patients
Study duration
Intervention and sample size
Treatment regimen
Study outcomes
Bissler [24] (2008) USA
N Engl J Med
Single-center, nonrandomized open-label phase 1–2 trial
LAM (n = 6),
TSC-LAM (n = 12),
TSC (n = 7),
1 year of treatment,
1 year of observation
Sirolimus (n = 25)
0.25 mg/m2 body-surface area
Renal AMLs, lung function, lung cyst volume, 6MWD, AEs, neurologic assessment.
Harari [20] (2008) Italy
Chest
Single-center, prospective phase 1 trial
LAM (n = 11)
3 years of treatment
Triptorelin (n = 11)
11.25 mg IM
Hormonal assays, pulmonary function tests, 6MWD, bone mineral density.
Dabora [34] (2011) USA
PLoS ONE
Multicenter open-label, phase 2 trial, single-arm
TSC-LAM
(n = 21),
TSC (n = 15)
1 year of treatment
Sirolimus (n = 36)
2 mg/day
AMLs, kidney cysts, skin lesions, pulmonary function, VEGF-D.
McCormack [33] (2011) USA
N Engl J Med
Multicenter, randomized, placebo-controlled study
LAM (n = 81),
TSC-LAM (n = 8)
1 year of treatment,
1 year of observation
Sirolimus group
(n = 46);
Placebo group
(n = 43)
2 mg/day
Lung function, 6MWD, VEGF-D, QOL scores, AEs.
Davies [35] (2011) UK
Clin Cancer Res
Multicenter nonrandomized open label phase 2 trial
LAM (n = 6),
TSC-LAM (n = 3),
TSC (n = 7),
2 years of treatment
Sirolimus (n = 16)
0.5 mg/m2 body-surface area
Renal AMLs, lung function, AEs and neurocognitive function.
Bissler [25] (2013) USA
Lancet
Multicenter, randomized, double-blind, placebo-controlled phase 3 trial
LAM (n = 5),
TSC-LAM (n = 24),
TSC (n = 89)
Median 38 weeks for everolimus;
median 34 weeks for placebo.
Everolimus group (n = 79);
Placebo group (n = 39)
10 mg/day
AMLs, skin lesion, pharmacokinetics of everolimus, pulmonary function, AEs, neuropsychological assessments, VEGF-D.
Piment a[22] (2013) Brazil
J Bras Pneumol
Single-center open-label, single-arm, interventional clinical trial
LAM (n = 31)
1 year of treatment
Doxycycline (n = 31)
100 mg/day
Pulmonary function, 6MWD, QOL, MMP-2, MMP-9, VEGF-D
Chang [23] (2014) UK
Eur Respir J
Single-center randomized placebo-controlled trial
LAM (n = 23)
2 years of treatment
Doxycycline group (n = 12);
Placebo group (n = 11)
100 mg/day for 3 months followed by 200 mg/day for 21 months
Lung function, exercise capacity, QOL, shuttle walk distance, MMP levels, VEGF-D.
Goldberg [36] (2015) USA
Eur Respir J
Multicenter, open-label, nonrandomized, phase 2 trial
LAM (n = 19),
TSC-LAM
(n = 5),
Treatment for 26 weeks
Everolimus (n = 24)
2.5 mg/day for 4 weeks, 5 mg/day for 4 weeks, 10 mg/day for 18 weeks
Lung function, VEGF-D, 6MWD, everolimus pharmacokinetics, AEs.
Takada [37] (2016) Japan
Ann Am Thorac Soc
Multicenter, single-arm, open-label trial
LAM and TSC-LAM (n = 63)
2 years of treatment
Sirolimus (n = 63)
2 mg/day
Lung function, QOL scores, AEs
EI-Chemaly [38] (2017) USA
Chest
Two-center phase 1 trial
LAM (n = 13)
24 weeks of treatment,
24 weeks of observation
Hydroxychloroquine and sirolimus (n = 13)
Hydroxychloroquine (200 mg or 400 mg)
Sirolimus (2 mg/day)
Lung function, 6MWD, QOL, AMLs, VEGF-D, AEs.
Author, Publication Year, Country
Journal
Study design
Diagnosis,
No of patients
Study duration
Intervention and sample size
Treatment regimen
Study outcomes
Bee [9] (2018)
UK
Thorax
A prospective national cohort, single-arm study,
LAM (n = 38),
TSC-LAM
(n = 9)
35.8 ± 18 months of treatment
Sirolimus (n = 47)
1–2 mg/day
Lung function, VEGF-D, AEs.
Cai [39] (2018) China
Orphanet J Rare Dis
Single-center, nonrandomized, open-label phase 2 trial
TSC-LAM (n = 6),
TSC (n = 12)
1 year of treatment
1 year of observation
Everolimus (n = 18)
10 mg/day
AMLs, skin lesions, lung function, AEs.
Aghaeimeybodi [40] (2019) Iran
Caspian J Intern Med
A prospective phase 1 trial
LAM (n = 2),
TSC-LAM (n = 4)
1 year of treatment
Sirolimus (n = 6)
2 mg/day
Lung function, 6MWD, AEs.
The general information of the included studies, including: name of the first author, publication year, study location, name of the journal, study design, number and diagnosis of patients, study duration, intervention and sample size of it, treatment regimen, and outcome indicator type for each study
LAM lymphangioleiomyomatosis, TSC tuberous sclerosis complex, AML angiomyolipomas, 6MWD 6-min walk distance, AEs adverse events, IM Intramuscular injection, VEGF-D vascular endothelial growth factor D, QOL quality of life, MMP matrix metalloproteinase

Effects on FEV1, FVC, DLco, 6MWD and QOL scores

In LAM patients treated with sirolimus, the results of this meta-analysis showed that the changes in the FEV1, FVC, DLco and 6MWD values from baseline to the endpoint were not statistically significant. The WMD values of FEV1, FVC, DLco and 6MWD were 0.03 L (95% CI: − 0.13 to 0.18, P = 0.74, I2 = 0%, Fig. 2), 0.14 L (95% CI: − 0.10 to 0.37, P = 0.25, I2 = 0%, Fig. 2), − 0.17 ml/min/mmHg (95% CI: − 1.58 to 1.24, P = 0.81, I2 = 0%, Fig. 2) and 23.76 m (95% CI: − 12.96 to 60.47, P = 0.20, I2 = 0%, Figure A1), respectively. The endpoint values for lung function and the 6MWD were measured after 1 year [24, 33, 34, 40] or 2 years [35] of sirolimus treatment. In addition, two studies reported the changes in the Visual Analogue Scale (VAS) scores and total Functional Performance Inventory (FPI) scores for LAM patients treated with sirolimus. Compared with the placebo group, the sirolimus group had significant improvements in the VAS and FPI scores [33]. However, Takada et al. [37] did not detect significant changes in the VAS and FPI scores in LAM patients after 2 years of sirolimus treatment. In addition, the dosages of sirolimus treatment were variable: 0.25 mg/m2 body surface area [24], 0.5 mg/m2 body surface area [35] and 1–2 mg/day [33, 34, 37, 38, 40].
In LAM patients treated with everolimus, the results also showed that the changes in the FEV1, FVC and DLco values from baseline to the endpoint were not statistically significant. The WMD values of FEV1, FVC and DLco were FEV1: 0.05 L (95% CI: − 0.18 to 0.27, P = 0.69, I2 = 0%), FVC: 0.16 L (95% CI: − 0.14 to 0.47, P = 0.30, I2 = 0%) and DLco: − 0.72 ml/min/mmHg (95% CI: − 2.77 to 1.32, P = 0.49, I2 = 0%), respectively (Fig. 3). Only Goldberg et al. [36] reported that the mean increase in the 6MWD was 47 m (95% CI: − 0.30 to 97.0) after 26 weeks of everolimus treatment, although the P value of this change was not provided. The everolimus treatment regimen in the included studies was 10 mg/day with a duration from 26 weeks to 1 year [25, 36, 39]. No prospective study has reported the effect of everolimus treatment on QOL scores.
In LAM patients treated with doxycycline, neither FVC nor DLco values changed significantly according to the results of meta-analysis, with WMD values of 0.02 L (95% CI: − 0.26 to 0.30, P = 0.91, I2 = 0%) and − 1.09 ml/min/mmHg (95% CI: − 3.52 to 1.34, P = 0.38, I2 = 0%), respectively (Fig. 4). Additionally, Pimenta et al. [22] reported that after 12 months of doxycycline treatment (100 mg/day), there was a significant mean decrease in the FEV1 (− 70 ml, P = 0.034) and a significant median increase in the mental health scores (18, P = 0.006) on the Medical Outcomes Study 36-item Short-form Health Survey (SF-36). No significant variation in the 6MWD or other domains of the SF-36 was identified. In an RCT by Chang et al. [23], the authors reported that there was no significant difference in the changes in the St. George’s Respiratory Questionnaire (SGRQ) scores between the doxycycline treatment group (200 mg/day) and the placebo group after 24 months of treatment.
Apart from the treatment mentioned above, triptorelin, a gonadotrophin-releasing hormone analog, was reported to have no significant effect on preventing the decline in lung function in LAM patients in a prospective single-arm study [20]. In addition, the use of triptorelin might be associated with the loss of bone mineral density. EI-Chemaly et al. [38] reported that after 24 weeks of combination therapy (sirolimus 2 mg/day and hydroxychloroquine 200 mg–400 mg), the FEV1 and 6MWD values increased significantly (P < 0.05) but subsequently declined toward the baseline values and were significantly different at the end of treatment (P < 0.05). However, the FVC and DLco values and the SGRQ score were not significantly changed throughout the combination treatment period.

Effects on AML and VEGF-D levels

The response rate of AML was available in 5 studies [24, 25, 34, 35, 39]. Among them, 4 studies provided the numbers of patients for whom the AML volume was reduced by at least 30% or more [24, 25, 34, 35], and 1 study provided the proportion of patients who achieved a ≥ 50% reduction in the AML volume [39]. In the random effect model, the pooled response rate of AML for LAM patients treated with sirolimus or everolimus was 0.62 (95% CI: 0.43 to 0.82, I2 = 65%) and 0.78 (95% CI: 0.68 to 0.88, I2 = 8%), respectively (Fig. 5).
Three articles provided the exact values of the VEGF-D levels in LAM patients before and after pharmacological treatment, including 2 studies describing doxycycline treatment [22, 23] and 1 study describing sirolimus treatment [33]. The meta-analysis showed no significant difference in VEGF-D levels before and after doxycycline treatment, with a WMD of − 2.65 pg/ml (95% CI: − 596.89 to 591.59, P = 0.99, I2 = 0%, Figure A2). However, McCormack et al. [33] found that the VEGF-D levels in 46 LAM patients decreased from 1848 ± 1514 pg/ml to 862 ± 540 pg/ml (P = 0.001) after 1 year of sirolimus treatment. Although no detailed VEGF-D values in LAM patients were provided, Dabora et al. [34] also reported a decrease in VEGF-D levels in TSC-LAM and TSC patients treated with sirolimus. The same trend was also reported in 2 articles describing everolimus treatment in LAM, TSC-LAM and TSC patients [25, 36]. Similar findings have also been documented in LAM patients treated with a combination of sirolimus and hydroxychloroquine [38].

Safety events associated with pharmacological treatments for LAM patients

With regard to sirolimus treatment, the exact number of patients experiencing AEs was available in 4 studies [24, 34, 35, 40]. The common AEs during treatment were oral mucositis (60%), hyperlipidemia (43%), upper respiratory infection (29%), proteinuria (26%), urinary system infection (18%), headache (18%), peripheral edema (18%) and diarrhea (16%) (Figure A3).
Data regarding the number of patients experiencing AEs were provided in 3 studies describing everolimus treatment for LAM [25, 36, 39]. The most common AE was oral mucositis (75%) (Figure A4). Other common AEs included headache (40%), abdominal or flank pain (39%), hyperlipidemia (36%), diarrhea (31%), hypercholesterolemia (30%), acne (24%), upper respiratory infection (23%), nasopharyngitis (23%), cough (22%) and fatigue (20%) (Figure A4).
With regard to AEs in patients treated with doxycycline, only 2 studies provided detailed data [22, 23]. Headache (25%) and diarrhea (14%) were the two most common AEs reported in both of the studies (Figure A5). In addition, nausea (19%) and upper respiratory infection (83%) were also common AEs reported in the included studies.
Regarding LAM patients treated with triptorelin, flushing (36%), arthralgias and paresthesias (18%), and fatigue and dizziness (18%) were the common AEs reported by Harari et al. [20]. In LAM patients treated with a combination of sirolimus and hydroxychloroquine, oral mucositis (62%), headache (62%) and diarrhea (62%) were the most commonly reported AEs [38].
Notably, most of the AEs mentioned in the included studies were reported to be low or moderate grade and were tolerable.

Sensitivity analysis

Sensitivity analysis confirmed that the exclusion of each study in turn did not change the meta-analysis results regarding lung function, the 6MWD, and VEGF-D levels. For sirolimus treatment, the data from Bissler et al. [24] were the main source of heterogeneity in the analyses of the AML response rate and AEs. Omitting that study [24], the I2 index of the AML response rate and the oral mucositis, proteinuria and diarrhea rates decreased to 0, 1.7, 0 and 0%, respectively. As for everolimus, the data by Cai et al. [39] were the main source of heterogeneity in the analyses of the acne and upper respiratory infection rates. After omitting these results, the I2 index decreased to 0 and 0%, respectively.

Discussion

The impairment of lung function in LAM patients has been reported to be associated with the aberrant infiltration of LAM cells or MMPs released from LAM cells [42, 43]. The abnormal proliferation of LAM cells is due to the activation of the mTOR signaling pathway, which is induced by inactivating mutations in the TSC1/2 gene [13, 14, 44, 45]. Therefore, mTOR inhibitors may be effective in the treatment of LAM.
Consistent with the speculation mentioned above, the prospective trials included in our study have reported the efficacy of sirolimus in the improvement or stabilization of lung function, amelioration of QOL scores, and reduction in AML volume [9, 24, 3335, 37, 38, 40]. Furthermore, 1 observational study [46] and 7 case reports [4753] also addressed the efficacy of sirolimus in the management of chylous effusions. In the present study, quantitative analyses of lung function in patients treated with sirolimus revealed that the changes in lung function and 6MWD values were not significant. These results suggest that sirolimus is effective at stabilizing the lung function in LAM patients. However, there is not enough strong evidence to support the effect of this drug on improving lung function. Other reported benefits of sirolimus therapy were the reduction in AML volume and decrease in VEGF-D levels. The pooled response rate of AML (reduced by at least 30%) was 0.62 (95% CI: 0.43 to 0.82), but there were not enough raw data regarding the changes in VEGF-D levels for a meta-analysis. Therefore, this study could not assess the role of VEGF-D in measuring treatment efficacy.
Everolimus is another type of mTOR inhibitor that has also been used in the treatment of LAM. Compared with sirolimus, everolimus has a shorter half-life and better bioavailability [54]. In addition, exposure to everolimus can be terminated faster than that to sirolimus when a transplant procedure is planned [36]. Therefore, everolimus is an attractive potential option for LAM patients on the list for lung transplantation. The quantitative analyses in our study also indicated similar changes observed during sirolimus therapy in FEV1, FVC and DLco values during the application of everolimus. Among the included trials, only Cai et al. [39] reported the effects of everolimus with respect to improvements in both the FEV1 and FVC values, but the sample size was small: only 6 LAM patients. Goldberg et al. [36] also reported the improvement of FEV1 values during everolimus treatment, but the P value was not available. Therefore, it is still difficult to confirm the actual effects of everolimus on lung function in LAM patients. Nonetheless, 2 prospective studies [25, 39] reported the effect of everolimus in reducing in the volume of AMLs, and the pooled response rate was 0.78 (95% CI: 0.68 to 0.88).
Doxycycline is a tetracycline antibiotic that inhibits the production and activity of MMPs, which are overexpressed in serum and lung tissue in LAM patients near the cyst area and may contribute to cyst formation. However, doxycycline therapy was only demonstrated to be effective in improving lung function and QOL scores in a subgroup of 13 LAM patients with less severe disease. Notably, the effects of doxycycline were more likely to be due to the better baseline lung function of these patients [22, 55]. In contrast, the results from an RCT [23] and our quantitative analyses showed that doxycycline did not affect any outcome in LAM patients.
Only 1 prospective study reported triptorelin, a gonadotrophin-releasing hormone analog, in the treatment of LAM. The results showed no beneficial outcomes during triptorelin therapy and indicated that the application of triptorelin may be associated with some AEs [20]. Regarding the combination therapy, no strong evidence has been found to support its superiority over monotherapy in the treatment of LAM [38].
Generally, the prospect is still not optimistic in the treatment of LAM. Only sirolimus and everolimus have been reported to be effective for LAM in prospective studies. In addition, the results of the current meta-analysis support only the opinion that these drugs are effective at stabilizing the condition. Although sirolimus has been recommended for the treatment of LAM [1], several problems still exist in its application. Questions about when and how much sirolimus should be used to treat which kind of LAM still needs to be answered.
With respect to patient selection and the timing of treatment initiation, Bee et al. [9] found that LAM patients with better pretreatment lung function and shorter disease duration have a better response to sirolimus. However, the MILES study [56] suggested that severe LAM patients with higher VEGF-D levels may benefit more from sirolimus treatment. Furthermore, although sirolimus was suggested for application in LAM patients with an annual FEV1 loss of 90 ml/year (which is threefold higher than the normal FEV1 loss) in one guideline [1], whether it should be applied in asymptomatic patients with normal or mildly impaired lung function is still unclear [57, 58]. Regarding the treatment dosage, Bee et al. [9] also found that different serum levels of sirolimus produced similar effects, and lower doses had fewer side effects. However, these results should be interpreted with caution because the sample size in each dosage group was quite small. The dose-response curve for sirolimus remains unclear, and the optimal dose with the fewest AEs and sufficient effects remains uncertain. With respect to treatment duration, two prospective studies have reported that the effects of sirolimus were only present during its administration; the decline in lung function and growth of AMLs resumed when treatment was stopped [24, 33]. Although Takada et al. [37] and Yao et al. [59] have reported the durable safety and efficacy of sirolimus (the mean durations of sirolimus treatment were 2 years and 4.6 years, respectively), the optimal treatment duration and whether the long-term application of sirolimus could reduce the need for lung transplantation or even replace it are still unknown. Moreover, similar questions also existed during the use of everolimus in LAM patients. It is hoped that two ongoing multicenter international trials will help us better understand the above issues (ClinicalTrials.gov identifiers NCT03150914 and NCT02432560).
Our study had several limitations. First, because LAM is a rare disease, the number and sample sizes of the included studies were limited. Only 3 RCTs were available, and the other studies were single-arm designed without a placebo control group. These factors could have added bias to this meta-analysis. Second, five studies that included TSC patients cofounded the evaluations of AEs and the response rate of AML. Third, the data of the included studies were unavailable for subgroup analyses to assess the impacts of certain variables. Finally, only three pharmacological treatments were quantitatively analyzed due to the lack of raw data.

Conclusions

In conclusion, our systematic review and meta-analysis support the application of sirolimus and everolimus in LAM, as these medications may stabilize lung function and alleviate renal AML. Further studies are required to reveal the optimal timing, duration and dosage in the application of sirolimus or everolimus. Doxycycline and triptorelin were not recommended for LAM because no beneficial outcomes were consistently reported. The efficacy and safety of combination therapy still require further exploration.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-1316-3.

Acknowledgments

Not applicable

Declarations of interest

None
Not applicable
Not applicable

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary information

Literatur
1.
Zurück zum Zitat McCormack FX, Gupta N, Finlay GR, Young LR, Taveira-DaSilva AM, Glasgow CG, Steagall WK, Johnson SR, Sahn SA, Ryu JH, et al. Official American Thoracic Society/Japanese respiratory society clinical practice guidelines: Lymphangioleiomyomatosis diagnosis and management. Am J Respir Crit Care Med. 2016;194(6):748–61.PubMedPubMedCentralCrossRef McCormack FX, Gupta N, Finlay GR, Young LR, Taveira-DaSilva AM, Glasgow CG, Steagall WK, Johnson SR, Sahn SA, Ryu JH, et al. Official American Thoracic Society/Japanese respiratory society clinical practice guidelines: Lymphangioleiomyomatosis diagnosis and management. Am J Respir Crit Care Med. 2016;194(6):748–61.PubMedPubMedCentralCrossRef
2.
3.
Zurück zum Zitat Harknett EC, Chang WYC, Byrnes S, Johnson J, Lazor R, Cohen MM, Gray B, Geiling S, Telford H, Tattersfield AE, et al. Use of variability in national and regional data to estimate the prevalence of lymphangioleiomyomatosis. QJM : monthly journal of the Association of Physicians. 2011;104(11):971–9.PubMedCrossRef Harknett EC, Chang WYC, Byrnes S, Johnson J, Lazor R, Cohen MM, Gray B, Geiling S, Telford H, Tattersfield AE, et al. Use of variability in national and regional data to estimate the prevalence of lymphangioleiomyomatosis. QJM : monthly journal of the Association of Physicians. 2011;104(11):971–9.PubMedCrossRef
4.
Zurück zum Zitat Aubry MC, Myers JL, Ryu JH, Henske EP, Logginidou H, Jalal SM, Tazelaar HD. Pulmonary lymphangioleiomyomatosis in a man. Am J Respir Crit Care Med. 2000;162(2 Pt 1):749–52.PubMedCrossRef Aubry MC, Myers JL, Ryu JH, Henske EP, Logginidou H, Jalal SM, Tazelaar HD. Pulmonary lymphangioleiomyomatosis in a man. Am J Respir Crit Care Med. 2000;162(2 Pt 1):749–52.PubMedCrossRef
5.
Zurück zum Zitat Ciftci AO, Sanlialp I, Tanyel FC, Buyukpamukcu N. The association of pulmonary lymphangioleiomyomatosis with renal and hepatic angiomyolipomas in a prepubertal girl: a previously unreported entity. Respiration; international review of thoracic diseases. 2007;74(3):335–7.PubMedCrossRef Ciftci AO, Sanlialp I, Tanyel FC, Buyukpamukcu N. The association of pulmonary lymphangioleiomyomatosis with renal and hepatic angiomyolipomas in a prepubertal girl: a previously unreported entity. Respiration; international review of thoracic diseases. 2007;74(3):335–7.PubMedCrossRef
6.
Zurück zum Zitat Taveira-DaSilva AM, Stylianou MP, Hedin CJ, Hathaway A, Moss J. Decline in lung function in patients with lymphangioleiomyomatosis treated with or without progesterone. Chest. 2004;126(6):1867–74.PubMedCrossRef Taveira-DaSilva AM, Stylianou MP, Hedin CJ, Hathaway A, Moss J. Decline in lung function in patients with lymphangioleiomyomatosis treated with or without progesterone. Chest. 2004;126(6):1867–74.PubMedCrossRef
7.
Zurück zum Zitat Johnson SR, Tattersfield AE. Decline in lung function in lymphangioleiomyomatosis: relation to menopause and progesterone treatment. Am J Respir Crit Care Med. 1999;160(2):628–33.PubMedCrossRef Johnson SR, Tattersfield AE. Decline in lung function in lymphangioleiomyomatosis: relation to menopause and progesterone treatment. Am J Respir Crit Care Med. 1999;160(2):628–33.PubMedCrossRef
8.
Zurück zum Zitat Johnson SR, Whale CI, Hubbard RB, Lewis SA, Tattersfield AE. Survival and disease progression in UK patients with lymphangioleiomyomatosis. Thorax. 2004;59(9):800–3.PubMedPubMedCentralCrossRef Johnson SR, Whale CI, Hubbard RB, Lewis SA, Tattersfield AE. Survival and disease progression in UK patients with lymphangioleiomyomatosis. Thorax. 2004;59(9):800–3.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Bee J, Fuller S, Miller S, Johnson SR. Lung function response and side effects to rapamycin for lymphangioleiomyomatosis: a prospective national cohort study. Thorax. 2018;73(4):369–75.PubMedCrossRef Bee J, Fuller S, Miller S, Johnson SR. Lung function response and side effects to rapamycin for lymphangioleiomyomatosis: a prospective national cohort study. Thorax. 2018;73(4):369–75.PubMedCrossRef
10.
Zurück zum Zitat Karbowniczek M, Astrinidis A, Balsara BR, Testa JR, Lium JH, Colby TV, McCormack FX, Henske EP. Recurrent lymphangiomyomatosis after transplantation: genetic analyses reveal a metastatic mechanism. Am J Respir Crit Care Med. 2003;167(7):976–82.PubMedCrossRef Karbowniczek M, Astrinidis A, Balsara BR, Testa JR, Lium JH, Colby TV, McCormack FX, Henske EP. Recurrent lymphangiomyomatosis after transplantation: genetic analyses reveal a metastatic mechanism. Am J Respir Crit Care Med. 2003;167(7):976–82.PubMedCrossRef
11.
Zurück zum Zitat Chen F, Bando T, Fukuse T, Omasa M, Aoyama A, Hamakawa H, Fujinaga T, Shoji T, Sakai H, Hanaoka N, et al. Recurrent lymphangioleiomyomatosis after living-donor lobar lung transplantation. Transplant Proc. 2006;38(9):3151–3.PubMedCrossRef Chen F, Bando T, Fukuse T, Omasa M, Aoyama A, Hamakawa H, Fujinaga T, Shoji T, Sakai H, Hanaoka N, et al. Recurrent lymphangioleiomyomatosis after living-donor lobar lung transplantation. Transplant Proc. 2006;38(9):3151–3.PubMedCrossRef
12.
13.
Zurück zum Zitat Yu J, Astrinidis A, Henske EP. Chromosome 16 loss of heterozygosity in tuberous sclerosis and sporadic lymphangiomyomatosis. Am J Respir Crit Care Med. 2001;164(8 Pt 1):1537–40.PubMedCrossRef Yu J, Astrinidis A, Henske EP. Chromosome 16 loss of heterozygosity in tuberous sclerosis and sporadic lymphangiomyomatosis. Am J Respir Crit Care Med. 2001;164(8 Pt 1):1537–40.PubMedCrossRef
15.
Zurück zum Zitat Henske EP. Metastasis of benign tumor cells in tuberous sclerosis complex. Genes, chromosomes & cancer. 2003;38(4):376–81.CrossRef Henske EP. Metastasis of benign tumor cells in tuberous sclerosis complex. Genes, chromosomes & cancer. 2003;38(4):376–81.CrossRef
16.
Zurück zum Zitat Cai X, Pacheco-Rodriguez G, Fan QY, Haughey M, Samsel L, El-Chemaly S, Wu HP, McCoy JP, Steagall WK, Lin JP, et al. Phenotypic characterization of disseminated cells with TSC2 loss of heterozygosity in patients with lymphangioleiomyomatosis. Am J Respir Crit Care Med. 2010;182(11):1410–8.PubMedPubMedCentralCrossRef Cai X, Pacheco-Rodriguez G, Fan QY, Haughey M, Samsel L, El-Chemaly S, Wu HP, McCoy JP, Steagall WK, Lin JP, et al. Phenotypic characterization of disseminated cells with TSC2 loss of heterozygosity in patients with lymphangioleiomyomatosis. Am J Respir Crit Care Med. 2010;182(11):1410–8.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Kumasaka T, Seyama K, Mitani K, Souma S, Kashiwagi S, Hebisawa A, Sato T, Kubo H, Gomi K, Shibuya K, et al. Lymphangiogenesis-mediated shedding of LAM cell clusters as a mechanism for dissemination in lymphangioleiomyomatosis. Am J Surg Pathol. 2005;29(10):1356–66.PubMedCrossRef Kumasaka T, Seyama K, Mitani K, Souma S, Kashiwagi S, Hebisawa A, Sato T, Kubo H, Gomi K, Shibuya K, et al. Lymphangiogenesis-mediated shedding of LAM cell clusters as a mechanism for dissemination in lymphangioleiomyomatosis. Am J Surg Pathol. 2005;29(10):1356–66.PubMedCrossRef
18.
Zurück zum Zitat Kumasaka T, Seyama K, Mitani K, Sato T, Souma S, Kondo T, Hayashi S, Minami M, Uekusa T, Fukuchi Y, et al. Lymphangiogenesis in lymphangioleiomyomatosis: its implication in the progression of lymphangioleiomyomatosis. Am J Surg Pathol. 2004;28(8):1007–16.PubMedCrossRef Kumasaka T, Seyama K, Mitani K, Sato T, Souma S, Kondo T, Hayashi S, Minami M, Uekusa T, Fukuchi Y, et al. Lymphangiogenesis in lymphangioleiomyomatosis: its implication in the progression of lymphangioleiomyomatosis. Am J Surg Pathol. 2004;28(8):1007–16.PubMedCrossRef
19.
Zurück zum Zitat Denoo X, Hermans G, Degives R, Foidart JM. Successful treatment of pulmonary lymphangioleiomyomatosis with progestins: a case report. Chest. 1999;115(1):276–9.PubMedCrossRef Denoo X, Hermans G, Degives R, Foidart JM. Successful treatment of pulmonary lymphangioleiomyomatosis with progestins: a case report. Chest. 1999;115(1):276–9.PubMedCrossRef
20.
Zurück zum Zitat Harari S, Cassandro R, Chiodini I, Taveira-DaSilva AM, Moss J. Effect of a gonadotrophin-releasing hormone analogue on lung function in lymphangioleiomyomatosis. Chest. 2008;133(2):448–54.PubMedCrossRef Harari S, Cassandro R, Chiodini I, Taveira-DaSilva AM, Moss J. Effect of a gonadotrophin-releasing hormone analogue on lung function in lymphangioleiomyomatosis. Chest. 2008;133(2):448–54.PubMedCrossRef
21.
Zurück zum Zitat Tomasian A, Greenberg MS, Rumerman H. Tamoxifen for lymphangioleiomyomatosis. N Engl J Med. 1982;306(12):745–6.PubMedCrossRef Tomasian A, Greenberg MS, Rumerman H. Tamoxifen for lymphangioleiomyomatosis. N Engl J Med. 1982;306(12):745–6.PubMedCrossRef
22.
Zurück zum Zitat Pimenta SP, Baldi BG, Kairalla RA, Carvalho CR. Doxycycline use in patients with lymphangioleiomyomatosis: biomarkers and pulmonary function response. Jornal brasileiro de pneumologia : publicacao oficial da Sociedade Brasileira de Pneumologia e Tisilogia. 2013;39(1):5–15.CrossRef Pimenta SP, Baldi BG, Kairalla RA, Carvalho CR. Doxycycline use in patients with lymphangioleiomyomatosis: biomarkers and pulmonary function response. Jornal brasileiro de pneumologia : publicacao oficial da Sociedade Brasileira de Pneumologia e Tisilogia. 2013;39(1):5–15.CrossRef
23.
Zurück zum Zitat Chang WY, Cane JL, Kumaran M, Lewis S, Tattersfield AE, Johnson SR. A 2-year randomised placebo-controlled trial of doxycycline for lymphangioleiomyomatosis. Eur Respir J. 2014;43(4):1114–23.PubMedCrossRef Chang WY, Cane JL, Kumaran M, Lewis S, Tattersfield AE, Johnson SR. A 2-year randomised placebo-controlled trial of doxycycline for lymphangioleiomyomatosis. Eur Respir J. 2014;43(4):1114–23.PubMedCrossRef
24.
Zurück zum Zitat Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leonard JM, Schmithorst VJ, Laor T, Brody AS, Bean J, et al. Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N Engl J Med. 2008;358(2):140–51.PubMedPubMedCentralCrossRef Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leonard JM, Schmithorst VJ, Laor T, Brody AS, Bean J, et al. Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N Engl J Med. 2008;358(2):140–51.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Frost M, Belousova E, Sauter M, Nonomura N, Brakemeier S, de Vries PJ, et al. Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2013;381(9869):817–24.CrossRef Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Frost M, Belousova E, Sauter M, Nonomura N, Brakemeier S, de Vries PJ, et al. Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2013;381(9869):817–24.CrossRef
26.
Zurück zum Zitat Zhan Y, Shen L, Xu W, Wu X, Zhang W, Wang J, Li X, Yang Y, Tian X, Xu KF. Functional improvements in patients with lymphangioleiomyomatosis after sirolimus: an observational study. Orphanet J Rare Dis. 2018;13(1):34.PubMedPubMedCentralCrossRef Zhan Y, Shen L, Xu W, Wu X, Zhang W, Wang J, Li X, Yang Y, Tian X, Xu KF. Functional improvements in patients with lymphangioleiomyomatosis after sirolimus: an observational study. Orphanet J Rare Dis. 2018;13(1):34.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73(9):712–6.PubMedCrossRef Slim K, Nini E, Forestier D, Kwiatkowski F, Panis Y, Chipponi J. Methodological index for non-randomized studies (minors): development and validation of a new instrument. ANZ J Surg. 2003;73(9):712–6.PubMedCrossRef
28.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ (Clinical research ed). 2009;339:b2535.CrossRef Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ (Clinical research ed). 2009;339:b2535.CrossRef
29.
Zurück zum Zitat Higgins J, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration 2011, Available from www.cochrane-handbook.org. Higgins J, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration 2011, Available from www.​cochrane-handbook.​org.
30.
Zurück zum Zitat Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med Res Methodol. 2005;5:13.PubMedPubMedCentralCrossRef Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med Res Methodol. 2005;5:13.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.PubMedPubMedCentralCrossRef Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Stat Methods Med Res. 2018;27(6):1785–805.PubMedCrossRef Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Stat Methods Med Res. 2018;27(6):1785–805.PubMedCrossRef
33.
Zurück zum Zitat McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, Stocks JM, et al. Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N Engl J Med. 2011;364(17):1595–606.PubMedPubMedCentralCrossRef McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, Stocks JM, et al. Efficacy and safety of sirolimus in lymphangioleiomyomatosis. N Engl J Med. 2011;364(17):1595–606.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Davies DM, de Vries PJ, Johnson SR, McCartney DL, Cox JA, Serra AL, Watson PC, Howe CJ, Doyle T, Pointon K, et al. Sirolimus therapy for angiomyolipoma in tuberous sclerosis and sporadic lymphangioleiomyomatosis: a phase 2 trial. Clin Can Res. 2011;17(12):4071–81.CrossRef Davies DM, de Vries PJ, Johnson SR, McCartney DL, Cox JA, Serra AL, Watson PC, Howe CJ, Doyle T, Pointon K, et al. Sirolimus therapy for angiomyolipoma in tuberous sclerosis and sporadic lymphangioleiomyomatosis: a phase 2 trial. Clin Can Res. 2011;17(12):4071–81.CrossRef
36.
Zurück zum Zitat Goldberg HJ, Harari S, Cottin V, Rosas IO, Peters E, Biswal S, Cheng Y, Khindri S, Kovarik JM, Ma S, et al. Everolimus for the treatment of lymphangioleiomyomatosis: a phase II study. Eur Respir J. 2015;46(3):783–94.PubMedCrossRef Goldberg HJ, Harari S, Cottin V, Rosas IO, Peters E, Biswal S, Cheng Y, Khindri S, Kovarik JM, Ma S, et al. Everolimus for the treatment of lymphangioleiomyomatosis: a phase II study. Eur Respir J. 2015;46(3):783–94.PubMedCrossRef
37.
Zurück zum Zitat Takada T, Mikami A, Kitamura N, Seyama K, Inoue Y, Nagai K, Suzuki M, Moriyama H, Akasaka K, Tazawa R, et al. Efficacy and safety of long-term Sirolimus therapy for Asian patients with Lymphangioleiomyomatosis. Ann Am Thorac Soc. 2016;13(11):1912–22.PubMedCrossRef Takada T, Mikami A, Kitamura N, Seyama K, Inoue Y, Nagai K, Suzuki M, Moriyama H, Akasaka K, Tazawa R, et al. Efficacy and safety of long-term Sirolimus therapy for Asian patients with Lymphangioleiomyomatosis. Ann Am Thorac Soc. 2016;13(11):1912–22.PubMedCrossRef
38.
Zurück zum Zitat El-Chemaly S, Taveira-Dasilva A, Goldberg HJ, Peters E, Haughey M, Bienfang D, Jones AM, Julien-Williams P, Cui Y, Villalba JA, et al. Sirolimus and autophagy inhibition in Lymphangioleiomyomatosis: results of a phase I clinical trial. Chest. 2017;151(6):1302–10.PubMedPubMedCentralCrossRef El-Chemaly S, Taveira-Dasilva A, Goldberg HJ, Peters E, Haughey M, Bienfang D, Jones AM, Julien-Williams P, Cui Y, Villalba JA, et al. Sirolimus and autophagy inhibition in Lymphangioleiomyomatosis: results of a phase I clinical trial. Chest. 2017;151(6):1302–10.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Cai Y, Guo H, Wang W, Li H, Sun H, Shi B, Zhang Y. Assessing the outcomes of everolimus on renal angiomyolipoma associated with tuberous sclerosis complex in China: a two years trial. Orphanet J Rare Dis. 2018;13(1):43.PubMedPubMedCentralCrossRef Cai Y, Guo H, Wang W, Li H, Sun H, Shi B, Zhang Y. Assessing the outcomes of everolimus on renal angiomyolipoma associated with tuberous sclerosis complex in China: a two years trial. Orphanet J Rare Dis. 2018;13(1):43.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Aghaeimeybodi F, Najafizadeh K, Razavi-Ratki SK, Namiranian N. Effects of Sirolimus on lung function in patients with Lymphangioleiomyomatosis. Caspian J Intern Med. 2019;10(1):7–10.PubMedPubMedCentral Aghaeimeybodi F, Najafizadeh K, Razavi-Ratki SK, Namiranian N. Effects of Sirolimus on lung function in patients with Lymphangioleiomyomatosis. Caspian J Intern Med. 2019;10(1):7–10.PubMedPubMedCentral
41.
Zurück zum Zitat Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Belousova E, Frost MD, Sauter M, Brakemeier S, de Vries PJ, Berkowitz N, et al. Everolimus long-term use in patients with tuberous sclerosis complex: four-year update of the EXIST-2 study. PLoS One. 2017;12(8):e0180939.PubMedPubMedCentralCrossRef Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Belousova E, Frost MD, Sauter M, Brakemeier S, de Vries PJ, Berkowitz N, et al. Everolimus long-term use in patients with tuberous sclerosis complex: four-year update of the EXIST-2 study. PLoS One. 2017;12(8):e0180939.PubMedPubMedCentralCrossRef
42.
43.
Zurück zum Zitat Chilosi M, Pea M, Martignoni G, Brunelli M, Gobbo S, Poletti V, Bonetti F. Cathepsin-k expression in pulmonary lymphangioleiomyomatosis. Mod Pathol. 2009;22(2):161–6.PubMedCrossRef Chilosi M, Pea M, Martignoni G, Brunelli M, Gobbo S, Poletti V, Bonetti F. Cathepsin-k expression in pulmonary lymphangioleiomyomatosis. Mod Pathol. 2009;22(2):161–6.PubMedCrossRef
44.
Zurück zum Zitat Sato T, Seyama K, Fujii H, Maruyama H, Setoguchi Y, Iwakami S, Fukuchi Y, Hino O. Mutation analysis of the TSC1 and TSC2 genes in Japanese patients with pulmonary lymphangioleiomyomatosis. J Hum Genet. 2002;47(1):20–8.PubMedCrossRef Sato T, Seyama K, Fujii H, Maruyama H, Setoguchi Y, Iwakami S, Fukuchi Y, Hino O. Mutation analysis of the TSC1 and TSC2 genes in Japanese patients with pulmonary lymphangioleiomyomatosis. J Hum Genet. 2002;47(1):20–8.PubMedCrossRef
45.
Zurück zum Zitat Sengupta S, Peterson TR, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell. 2010;40(2):310–22.PubMedPubMedCentralCrossRef Sengupta S, Peterson TR, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell. 2010;40(2):310–22.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Taveira-DaSilva AM, Hathaway O, Stylianou M, Moss J. Changes in lung function and chylous effusions in patients with lymphangioleiomyomatosis treated with sirolimus. Ann Intern Med. 2011;154(12):797–805 w-292-793.PubMedPubMedCentralCrossRef Taveira-DaSilva AM, Hathaway O, Stylianou M, Moss J. Changes in lung function and chylous effusions in patients with lymphangioleiomyomatosis treated with sirolimus. Ann Intern Med. 2011;154(12):797–805 w-292-793.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Barrera P, Simons SO, Luijk B, Wessels MJ, Heijdra YF. Efficacy of sirolimus therapy for chylous effusions in lymphangioleiomyomatosis. Ann Am Thorac Soc. 2013;10(4):408–9.PubMedCrossRef Barrera P, Simons SO, Luijk B, Wessels MJ, Heijdra YF. Efficacy of sirolimus therapy for chylous effusions in lymphangioleiomyomatosis. Ann Am Thorac Soc. 2013;10(4):408–9.PubMedCrossRef
48.
Zurück zum Zitat Harari S, Elia D, Torre O, Bulgheroni E, Provasi E, Moss J. Sirolimus therapy for patients with Lymphangioleiomyomatosis leads to loss of Chylous ascites and circulating LAM cells. Chest. 2016;150(2):e29–32.PubMedPubMedCentralCrossRef Harari S, Elia D, Torre O, Bulgheroni E, Provasi E, Moss J. Sirolimus therapy for patients with Lymphangioleiomyomatosis leads to loss of Chylous ascites and circulating LAM cells. Chest. 2016;150(2):e29–32.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Ohara T, Oto T, Miyoshi K, Tao H, Yamane M, Toyooka S, Okazaki M, Date H, Sano Y. Sirolimus ameliorated post lung transplant chylothorax in lymphangioleiomyomatosis. Ann Thorac Surg. 2008;86(6):e7–8.PubMedCrossRef Ohara T, Oto T, Miyoshi K, Tao H, Yamane M, Toyooka S, Okazaki M, Date H, Sano Y. Sirolimus ameliorated post lung transplant chylothorax in lymphangioleiomyomatosis. Ann Thorac Surg. 2008;86(6):e7–8.PubMedCrossRef
50.
Zurück zum Zitat Chachaj A, Drozdz K, Chabowski M, Dziegiel P, Grzegorek I, Wojnar A, Jazwiec P, Szuba A. Chyloperitoneum, chylothorax and lower extremity lymphedema in woman with sporadic lymphangioleiomyomatosis successfully treated with sirolimus: a case report. Lymphology. 2012;45(2):53–7.PubMed Chachaj A, Drozdz K, Chabowski M, Dziegiel P, Grzegorek I, Wojnar A, Jazwiec P, Szuba A. Chyloperitoneum, chylothorax and lower extremity lymphedema in woman with sporadic lymphangioleiomyomatosis successfully treated with sirolimus: a case report. Lymphology. 2012;45(2):53–7.PubMed
51.
Zurück zum Zitat Ando K, Kurihara M, Kataoka H, Ueyama M, Togo S, Sato T, Doi T, Iwakami S, Takahashi K, Seyama K, et al. Efficacy and safety of low-dose sirolimus for treatment of lymphangioleiomyomatosis. Respir Investig. 2013;51(3):175–83.PubMedCrossRef Ando K, Kurihara M, Kataoka H, Ueyama M, Togo S, Sato T, Doi T, Iwakami S, Takahashi K, Seyama K, et al. Efficacy and safety of low-dose sirolimus for treatment of lymphangioleiomyomatosis. Respir Investig. 2013;51(3):175–83.PubMedCrossRef
53.
Zurück zum Zitat Ellender CM, Williams TJ, Gooi J, Snell GI, Whitford HM. Management of refractory chylothorax in pulmonary lymphangioleiomyomatosis. Respirology Case Rep. 2015;3(2):72–4.CrossRef Ellender CM, Williams TJ, Gooi J, Snell GI, Whitford HM. Management of refractory chylothorax in pulmonary lymphangioleiomyomatosis. Respirology Case Rep. 2015;3(2):72–4.CrossRef
54.
Zurück zum Zitat Dancey JE. Inhibitors of the mammalian target of rapamycin. Expert Opin Investig Drugs. 2005;14(3):313–28.PubMedCrossRef Dancey JE. Inhibitors of the mammalian target of rapamycin. Expert Opin Investig Drugs. 2005;14(3):313–28.PubMedCrossRef
55.
Zurück zum Zitat Johnson SR, Chang WY, Tattersfield AE, Lewis S, Kumaran M, Cane JL. Doxycycline in lymphangioleiomyomatosis: not all questions are answered. Eur Respir J. 2014;43(5):1538.PubMedCrossRef Johnson SR, Chang WY, Tattersfield AE, Lewis S, Kumaran M, Cane JL. Doxycycline in lymphangioleiomyomatosis: not all questions are answered. Eur Respir J. 2014;43(5):1538.PubMedCrossRef
56.
Zurück zum Zitat Young L, Lee HS, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, et al. Serum VEGF-D a concentration as a biomarker of lymphangioleiomyomatosis severity and treatment response: a prospective analysis of the multicenter international Lymphangioleiomyomatosis efficacy of Sirolimus (MILES) trial. Lancet Respir Med. 2013;1(6):445–52.PubMedPubMedCentralCrossRef Young L, Lee HS, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, et al. Serum VEGF-D a concentration as a biomarker of lymphangioleiomyomatosis severity and treatment response: a prospective analysis of the multicenter international Lymphangioleiomyomatosis efficacy of Sirolimus (MILES) trial. Lancet Respir Med. 2013;1(6):445–52.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Xu KF, Tian X, Yang Y, Zhang H. Rapamycin for lymphangioleiomyomatosis: optimal timing and optimal dosage. Thorax. 2018;73(4):308–10.PubMedCrossRef Xu KF, Tian X, Yang Y, Zhang H. Rapamycin for lymphangioleiomyomatosis: optimal timing and optimal dosage. Thorax. 2018;73(4):308–10.PubMedCrossRef
58.
Zurück zum Zitat Harari S, Spagnolo P, Cocconcelli E, Luisi F, Cottin V. Recent advances in the pathobiology and clinical management of lymphangioleiomyomatosis. Curr Opin Pulm Med. 2018;24(5):469–76.PubMedCrossRef Harari S, Spagnolo P, Cocconcelli E, Luisi F, Cottin V. Recent advances in the pathobiology and clinical management of lymphangioleiomyomatosis. Curr Opin Pulm Med. 2018;24(5):469–76.PubMedCrossRef
59.
Zurück zum Zitat Yao J, Taveira-DaSilva AM, Jones AM, Julien-Williams P, Stylianou M, Moss J. Sustained effects of sirolimus on lung function and cystic lung lesions in lymphangioleiomyomatosis. Am J Respir Crit Care Med. 2014;190(11):1273–82.PubMedPubMedCentralCrossRef Yao J, Taveira-DaSilva AM, Jones AM, Julien-Williams P, Stylianou M, Moss J. Sustained effects of sirolimus on lung function and cystic lung lesions in lymphangioleiomyomatosis. Am J Respir Crit Care Med. 2014;190(11):1273–82.PubMedPubMedCentralCrossRef
Metadaten
Titel
The efficacy and safety of pharmacological treatments for lymphangioleiomyomatosis
verfasst von
Qi Wang
Mengqi Luo
Bo Xiang
Siyuan Chen
Yi Ji
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2020
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-020-1316-3

Weitere Artikel der Ausgabe 1/2020

Respiratory Research 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.