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

Open Access 01.12.2020 | Research

Therapeutic effect of subcutaneous injection of low dose recombinant human granulocyte-macrophage colony-stimulating factor on pulmonary alveolar proteinosis

verfasst von: Fen Zhang, Dong Weng, Yiliang Su, Chengsheng Yin, Li Shen, Yuan Zhang, Ying Zhou, Qiuhong Li, Yang Hu, Huiping Li

Erschienen in: Respiratory Research | Ausgabe 1/2020

Abstract

Objective

To observe the efficacy of recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) for pulmonary alveolar proteinosis (PAP).

Materials and methods

A total of 55 patients with PAP were screened at Shanghai Pulmonary Hospital between May 2014 and May 2018. Among these, 42 were diagnosed with idiopathic PAP, 24 were included in this study, 20 were treated for 6 months, and 17 were followed up for additional 6 months. All patients received a subcutaneous injection of 75μg/d GM-CSF qd for 1 month. The therapeutic dose was adjusted according to the changes in the lesions of chest CT. If the lesions were absorbed, subcutaneous injections of 75μg/d GM- CSF qd and 75μg/d GM-CSF qod were given for 2 and 3 months, otherwise, the dose was increased to 150μg/d GM-CSF qd and 150μg/d qod for 2 and 3 months, respectively. All cases were treated once a day in the first 3 months and once every other day in the last 3 months. The total course of treatment was 6 months. After withdrawal, the patients were followed up for another 6 months. The deadline of follow up was September 30, 2019.

Results

Twenty patients completed the treatment and efficacy evaluation. One patient was completely cured, 16 cases improved, three cases were noneffective. After 1-month evaluation, 12 patients received an increased dose (150μg) from the second month of treatment. Seventeen patients completed the 12-month follow-up, among which fourteen improved. CT showed the lesions were slightly increased in three cases. Economic burden was the following: RMB 7324–15,190 Yuan were required for the 6-month treatment course, which is significantly lower compared to other treatment methods.

Conclusion

Subcutaneous injection of rhGM-CSF at low dose (75μg-150μg /d) is effective treatment for patients with idiopathic PAP.

Trial registration

NCT01983657. Registered 16 April 2013.
Hinweise
Fen Zhang, Dong Weng, Yiliang Su and Chengsheng Yin contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
DLCO
Carbon monoxide diffusion capacity
HRCT
High resolution computed tomography
Pa(A-O)2
Alveolar-arterial oxygen partial pressure difference
PaO2
Partial pressure difference
PAP
Pulmonary alveolar proteinosis
PAS
Periodic-acid-schiff
rhGM-CSF
Recombinant human granulocyte-macrophage colony-stimulating factor
WLL
Whole lavage

Background

Pulmonary alveolar proteinosis (PAP) is a rare, diffuse lung disease of unknown etiology, characterized by the filling of the alveolar cavity with phospholipid and protein-containing periodic acid- schiff (PAS) staining positive granular substances. PAP was discovered in 1958 by three pathologists, SH Rosen, Benjamin Castleman and AA Liebow, thus is also known as Rosen-Castleman-Liebow’s Triad Syndrome [1].
PAP consists of three types: congenital, secondary and idiopathic (also known as acquired or autoimmune) [24]. Idiopathic PAP is the main type of PAP, accounting for more than 90%, and is associated with autoimmune abnormalities [5, 6]. GM-CSF is a hematopoietic cell growth-stimulating factor that not only stimulates the proliferation and differentiation of bone marrow cells and their stem cells, but also regulates the phagocytic function of alveolar macrophages and their ability to degrade surfactants, having an important role in the balance of pulmonary surfactants. In 1994, Dranoff et al [7] have found an association between pulmonary alveolar proteinosis and GM-CSF; pulmonary alveolar proteinosis was found in mice with GM-CSF-encoding gene deletion, while the expression of GM-CSF in lung tissue by transgenic technology could make protein-like substances of alveolar deposition in gene knockout mice disappear. Its pulmonary pathological changes could be restored to normal through re-expression of GM-CSF in the lung tissue. Nevertheless, no deletion or abnormalities of GM-CSF coding gene have been found in patients with PAP [8]. Moreover, a number of studies [913] have indicated that autoantibodies are able to neutralize granulocyte-macrophage colony-stimulating factor in alveolar lavage fluid or serum samples from patients with idiopathic PAP; however, this effect was not found in secondary PAP, healthy controls and other patients with lung disease. The presence of this antibody impairs the function of granulocyte-macrophage colony-stimulating factor, resulting in increased number of alveolar lipoproteins that eventually lead to pulmonary alveolar proteinosis. Takaki and colleagues [13] have found that patients with pulmonary alveolar proteinosis relapse after double lung transplantation, further confirming the view that “the presence of autoantibodies neutralizes neutrophil-macrophage colony-stimulating factor in patients with idiopathic PAP”. Subsequently, some studies have extracted anti-GM-CSF antibodies from the blood of PAP patients and used it in healthy non-human primates, proving that the biochemical, cytological and pathological characteristics of pulmonary alveolar proteinosis could be observed via this approach [14]. Based on these studies, it is believed that the presence of anti-GM-CSF antibodies in the body is an important cause of idiopathic pulmonary alveolar proteinosis. Therefore, the ability of alveolar macrophages to degrade surfactants should be partially restored by supplementing GM-CSF, thus achieving the purpose of treating PAP.
Thus far, many studies have reported the treatment of PAP with GM-CSF. However, no uniform standard existed. The main methods followed were inhalation therapy and subcutaneous therapy. The time of therapy included daily medication and different time intervals. The total course of treatment varied from 3 to 36 months, and the doses were different between studies [1524] (Table 1). The dose of subcutaneous injection was from 3 to 20 μg/(kg · day) and the time course of the therapy was from 3 to 68 weeks, which brought huge financial burden and side effects. The minimum dose of inhalation therapy was 125 μg qd [22, 24], and the maximum dose was 1000 μg/day [21]. The use of atomizing device also brought the risk of secondary infection to patients. Although the efficacy was positive, these treatment approaches had some drawbacks, such as high dose, high cost, and increased risk of infection due to atomized devices. The clinical experience in the present study indicated that some patients with idiopathic PAP responded well to lower doses of GM-CSF (75 μg/day to 150 μg/day) by a subcutaneous injection and had lower financial burden. A clinical trial named “clinical observation of subcutaneous injection of low-dose rhGM–CSF on treatment of PAP” (ID number is NCT01983657) was registered to further verify the efficacy of low-dose recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) for PAP. The goal of this study was to explore the efficacy and safety of a lower dose of rhGM-CSF for idiopathic PAP, which should also be convenient to perform and help reduce the cost of therapy.
Table 1
Literature summary of GM-CSF treatment for PAP
Trials
Usage of GM-CSF
Maximum dose of GM-CSF
Medication timea
Effective rate
Yearb
Seymour et al. [15]
subcutaneous injection
5 μg/kg/day
300 days
100% (n = 1)
1996
Kavuru et al. [16]
subcutaneous injection
9μg/kg/day
12 weeks
75% (n = 4)
2000
Seymour et al [17]
subcutaneous injection
20μg/kg/d
6–12 weeks
43%(n = 14)
2001
Bonfield et al. [18]
subcutaneous injection
18 μg/kg/day
12–48 weeks
55% (n = 11)
2002
Tazawa et al. [19]
inhalation therapy
250 μg/day; every other week
24 weeks
100% (n = 3)
2005
Venkateshiah et al. [20]
subcutaneous injection
18 μg/kg/day
12–52 weeks
48% (n = 21)
2006
Wylam et al. [21]
inhalation therapy
500ugbid week-on, week-off
3–68 weeks
92%(n = 12)
2006
Tazawa et al. [22]
inhalation therap
250 μg/day d1-d8, d9-d14 no use of drugs × 6 cycles
24 weeks
62% (n = 39)
2010
Spyros et al. [23]
inhalation therapy
250μg qd 4 days-on, 4 days-off
14–65 weeks
100%(n = 6)
2014
Tazawa et al. [24]
inhalation therapy
(125 μg bid d1-d8, d9-d14 no use of drugs × 6 cycles) + (125 μg qd d1-d4,d5-d14 no use of drugs× 6 cycles)
24 weeks
66%(n = 35
2014
GM-CSF Granulocyte-macrophage colony-stimulating factor, PAP Pulmonary alveolar proteinosis
aMedication time:The time of administration was fixed in some studies, while varied in some studies. In this situation, the medication time was showed as the shortest time - the longest time in this table
byear: The publication time of the study

Materials and methods

Inclusion and exclusion criteria

A total of 55 patients with PAP who were admitted at the Shanghai Lung Hospital from May 2014 to May 2018 were included in this study. Diagnostic criteria for PAP [2527] were the following: 1) patients with or without cough and polypnea; 2) chest HRCT showing typical paving stone signs; 3) under electron microscope, oval granules were observed in the lavage solution, with bright and dark threaded or fingerprint-like stripes, which suggested the presence of lamellar bodies (Fig. 1); 4) secondary factors such as industrial dust exposure, various infectious diseases, malignant hematological diseases or immunodeficiency diseases were excluded. The inclusion criteria were: (1) diagnosis of idiopathic pulmonary alveolar proteinosis according to PAP diagnostic criteria; (2) 18–70 years old, both male and female; (3) Informed consent. Exclusion criteria were the following: (1) patients with secondary pulmonary alveolar proteinosis; (2) patients who received rhGM-CSF treatment within 6 months before enrollment; (3) those who received whole lung lavage treatment within 4 weeks before enrollment; (4) leukocyte count (> 12,000/ul); (5) fever > 38 °C; (6) severe edema: systemic diseases, such as cardiac insufficiency, liver and kidney insufficiency, etc.; (7) patients with severe diseases of other systems, including liver, kidney, lung, blood and cardiovascular diseases; (8) pregnant women or those planning the pregnancy, and lactating women; (9) patients with severe history of drug allergy or allergy constitution, or those allergic to Escherichia coli preparations; (10) unable to express subjective discomfort symptoms; (11) patients who are allergic to GM-CSF or not suitable for GM-CSF; (12) other conditions determined by the investigator that do not meet the inclusion criteria. Finally, 42 patients were diagnosed with PAP, 24 patients were included in the study. Twenty patients completed 6 months treatment and 6 months follow-up; 17 patients completed 12 months follow-up (Fig. 2), including 3 female patients and 17 male patients.

Drug administration and evaluation scheme

The drug administration and evaluation scheme are presented in Fig. 3. Briefly, enrolled patients were given subcutaneous injection of 75μg/d GM-CSF qd for 1 month. Consequently, all patients underwent chest CT. According to the observation of preclinical medication, the efficacy for 3 months has been relatively stable. Therefore, this study was designed as follows: once a day for the first 3 months and once every other day for the next 3 months after the evaluation of the efficacy. We hope to achieve the purpose of not only maintaining the treatment effect but also reducing the economic burden. The results of this study also confirm the rationality of this study design. If the lesions were absorbed, subcutaneous injection of 75μg/d GM-CSF qd for 2 months and subcutaneous injection of 75μg/d GM-CSF qod for 3 months were continued. After 1 month, if the chest CT showed no absorption or increase, the dose was increased to 150 μg/d GM-CSF qd for 2 months and 150 μg/d GM-CSF qod for 3 months. After that, all patients were followed up to 12 months.

Evaluation of efficacy and safety

Evaluation indexes of efficacy

Treatment efficacy was calculated based on chest CT paving stone sign proportion score (Table 2), lung function (FVC, DLCO%), blood gas analysis (oxygen partial pressure, alveolar-arterial oxygen partial pressure difference), six-minute walking test, symptom score (score of cough and polypnea) (Table 3).
Table 2
Imaging examinations (scoring method according to the proportion of paving stone signs displayed on CT)
0 point
no paving stone signs
1 point
paving stone signs less than 5%
2 points
5–24% of paving stone signs
3 points
25–49% of paving stone signs
4 points
50–74% of paving stone signs
5 points
paving stone signs more than≥75%
Table 3
Clinical symptom score
Polypnea Or dyspnea
□1 point Dyspnea only when exerting themselves.
□2 points Shortness of breath when walking fast on flat ground or climbing small slopes on foot.
□3 points Because of shortness of breath, walking on flat ground is slower than that of their peers or requires stopping to rest.
□4 points Stop to catch breath after walking on flat ground for about 100 m or a few minutes
□5 points Severe dyspnea prevents the patient from leaving his home or dyspnea while dressing or undressing.
cough
□0 point No cough during the day; no cough at night.
□1 point There are occasional short coughs in the daytime; temporary coughs or occasional coughs when sleeping at night.
□2 points Frequent cough during the day slightly affects daily activities; cough slightly affects sleep at night.
□3 points Frequent cough during the day seriously affects daily activities; cough seriously affects sleep at night.
 
Other clinical symptoms:

Efficacy evaluation

Curative effect was calculated based on clinical symptoms and signs, imaging data, arterial blood gas, pulmonary function, etc.. According to the study protocol, all patients included in the analysis were evaluated three times at 1 month of treatment, completion of 6-month medication (6 months) and 6 months of follow-up (12 months) after withdrawal. Three grades were used: (1) Cure: all clinical symptoms and abnormal signs disappeared; arterial blood gas and pulmonary function returned to normal levels and no abnormalities were visible on chest CT scan; (2) Improvement: chest CT showed improved images, and pulmonary function, arterial blood gas, clinical symptoms and signs all improved; (3) inefficacy or deterioration: no improvement or deterioration of imaging, pulmonary function, arterial blood gas, clinical symptoms and signs were observed. Cases with cure and improvement were effective ones.

Evaluation indexes of safety

Evaluation indexes of safety were based on blood routine, liver function, renal function, electrolyte, myocardial zymogram, electrocardiogram; and eventual adverse events.

Drug cost estimation

According to the therapeutic regimen and the current drug market price, the costs of each study treatment were calculated based on the following formula: time of administration × the dose of the drug × the unit price of the drug. In addition, the minimum cost (the shortest time × the minimum dose × the unit price of the drug) and the maximum cost (the longest time × the maximum dose × the unit price of the drug) were also calculated.

Statistical analysis

Related data of all eligible PAP patients were statistically analyzed. Pulmonary function (FVC, DLCO%), blood gas analysis (oxygen partial pressure, alveolar-arterial oxygen partial pressure difference), six-minute walking distance and other continuous numerical variables were analyzed by paired sample t test. The specific values were recorded by mean ± standard deviation. P < 0.05 was considered to be statistically significant. Orderly categorical variables such as Chest CT paving stone sign proportion score, symptom score (score of cough and polypnea) were analyzed by Wilcoxon symbolic rank test. Results of efficacy analysis were compared and described using percentages. During the clinical study, all patients were followed up to record the frequency, time and severity of adverse events, and the safety analysis was descriptive.

Results

Twenty patients completed 6 months of treatment, including 17 males and 3 females (average age of 46.65 ± 10.84 years old). Seventeen patients were followed up for 6 months after the treatment was completed.

Evaluation of efficacy after treatment (6 months)

Chest CT data

Chest CT examination indicated complete absorption of the lesion in one patient, partial absorption in 16 patients, and no changes in 3 patients (Fig. 4). The paving stone sign proportion scores before and after the treatment were compared with Wilcoxon symbolic rank test, and the results showed that the lesions were absorbed (z = 3.686, P = 0.000003; Table 5a).

Arterial blood gas

The mean ± standard deviation of oxygen partial pressure (Table 4, Fig. 5a) before and after treatment were 69.05 ± 11.14 and 88.16 ± 15 respectively (p = 0.000162) while the mean ± standard deviation of alveolar-arterial oxygen partial pressure (Table 4, Fig. 5b) before and after treatment were 35.77 ± 16 and 18.16 ± 13.69 respectively (p = 0.001).
Table 4
General information of patients and statistical summary of continuous numerical observation indexes
Indexes
Before treatment VS 6 months (n = 20)
Before treatment VS 12 months (n = 17)
Before treatment
6 months
P value
Before treatment
12 months
P value
Oxygen partial pressure(mmHg)
69.05 ± 1.14
88.16 ± 15
< 0.001
67.56 ± 11.42
81.25 ± 13.55
0.002
Alveolar-arterial oxygen partial pressure difference(mmHg)
35.77 ± 16.00
18.1 ± 13.69
0.001
35.84 ± 15.52
28.75 ± 12.21
0.220
FVC(L)
3.34 ± 0.88
3.54 ± 0.83
0.022
3.165 ± 0.952
3.365 ± 0.849
0.139
DLCO (%)
69.65 ± 22.49
84.49 ± 13.73
0.004
68.75 ± 26.02
89.24 ± 14.70
0.010
Six-minute walking distance(miles)
501.9 ± 133.22
645 ± 117.18
0.002
486.0 ± 134.0
624.0 ± 149.7
0.021
FVC Forced vital capacity, DLCO Carbon monoxide diffusion capacity
Pulmonary function, blood gas analysis and six-minute walking distance test results (mean ± standard deviation)

Pulmonary function

The mean ± standard deviation of FVC (Table 4, Fig. 6a) before and after treatment were 3.34 ± 0.88 and 3.54 ± 0.83, respectively (p = 0.022), while the mean ± standard deviation of DLCO% (Table 4, Fig. 6b) before and after treatment were 69.65 ± 22.49 and 84.49 ± 13.73, respectively (p = 0.004).

Symptom score

The results showed that the cough and polypnea improved after treatment, and the difference was statistically significant (cough: z = 3.419, P = 0.001, Table 6; polypnea: z = 3.562, P = 0.000011, Table 7).

Six-minute walking test

Six-minute walking test is shown in Table 4 and Fig. 7. Briefly, 6 months after treatment, the walking distance of patients was prolonged, and the results were statistically significant. The mean ± standard deviation before and after treatment was 501.9 ± 133.2 and 645 ± 117.18, respectively (p = 0.002).

Judgment of efficacy

Among the 20 patients, 1 patient was cured, 16 patients improved, and 3 patients showed no changes after 6 months treatment, with an improvement rate of 17/20 (85%) (Table 8).

Evaluation of efficacy after 12 months fellow up

A total of 17 of the 20 patients completed the 12-months follow-up. The comparison of Chest CT paving stone sign proportion scores before and after the treatment (Table 5b) was statistically significant (z = 4.103, P = 0.000041). In addition, the comparison of polypnea scores before and after treatment was statistically significant (Table 6b) (z = 3.830, P = 0.000128). Moreover, compared with those before treatment, arterial blood gas (Table 4 and Fig. 5), pulmonary function (Table 4 and Fig. 6), cough score (Table 6b) and six-minute walking distance test (Table 4 and Fig. 7) showed improving trend, nevertheless the statistical difference could not be reflected due to small number of cases.
Table 5
Comparisons of CT paving stone sign score before and after treatment
(A) Changes of CT paving stone sign score before treatment and after 6 months of treatment (n = 20)
Scores before treatment
Scores after 6 months of treatment
Number of cases
2
1
4
2
2
2
3
1
4
3
2
4
3
3
1
4
1
1
4
2
1
5
0
1
5
1
2
(B) Changes of CT paving stone signs score before treatment and after 12 months of treatment (n = 17)
Scores before treatment
Scores after 12 months of treatment
Number of cases
2
1
3
2
2
1
3
1
5
3
2
1
3
3
2
4
3
1
4
1
1
5
1
2
5
2
1
Compared with that before treatment, the proportion scores of chest CT paving stone sign improved significantly after 6 months of treatment, with significant difference (z = 3.686, P = 0.000003). Seventeen patients were followed up for 6 months (12 months) after treatment, and their CT scores improved significantly compared with that before treatment, with statistical significance (z = 4.103, P = 0.00004)
Table 6
Comparisons of cough scores before and after treatment
(A) Changes of cough scores before treatment and after 6 months of treatment (n = 20)
Scores before treatment
Scores after 6 months of treatment
Number of cases
0
0
2
1
0
1
1
1
5
2
1
10
3
1
2
(B) Changes of cough scores before treatment and after 12 months of treatment (n = 17)
Scores before treatment
Scores after 12 months of treatment
Number of cases
0
0
1
1
0
1
1
1
3
1
2
1
2
1
8
2
2
1
3
1
2
The cough score was improved before and after 6 months of treatment, with significant difference (z = 3.419, P = 0.001); Seventeen patients were followed up after 12 months and the cough after 12 months of treatment improved when compared with that before treatment, with significant difference (z = 2.578, P = 0.010)
Table 7
Comparison of polypnea scores before and after treatment
(A): Changes of polypnea scores before treatment and after 6 months of treatment (n = 20)
Scores before treatment
Scores after 6 months of treatment
Number of cases
1
1
4
2
1
4
3
1
4
3
2
3
4
1
4
5
1
1
(B): Changes of polypnea scores before treatment and after 12 months of treatment (n = 17)
Scores before treatment
Scores after 12 months of treatment
Number of cases
1
0
1
1
1
2
2
0
1
2
1
1
2
3
1
3
0
2
3
1
3
3
2
1
3
3
1
4
1
3
5
1
1
The score of polypnea was also improved before and after 6 months of treatment, with significant difference (z = 3.562, P = 0.000011). Seventeen patients were followed up after 12 months, and compared with that before treatment, the polypnea after 12 months of treatment was also improved, with statistically significant difference (z = 3.830, P = 0.000128)
Table 8
Evaluation of efficacy
 
Cure
Improved
Invalidity or deterioration
Effective cases (Effective rate)
End of treatment(6 months)
1
16
3
17/20(85%)
End of follow-up (12 months)
0
14
3
14/17(82.35%)
Table 9
Estimates of funds
Medication method
Minimum Dose x Minimum Time
RMB (yuan)
Maximum Dose x Maximum Time
RMB (yuan)
Inhalation
250μg qd × 14 weeks, 4 days-on, 4 days-off
15,190
500μg bid*68 weeks
344,306
Subcutaneous injection
9μg/kg/day*12 weeks
32,811
18μg/kg/d*52 weeks
284,357
This method
75μg qd*6 months
7324
150μg qd*6 months
13,020

Cost

Comparing the cost of each study (calculated by the time of administration × the dose of the drug) we discovered that the cost of this study was significantly lower compared to other studies (Table 9). The minimum cost of 6 months was RMB 7324, which was only half of the minimum cost of RMB 15,190 [23] for the study of PAP treatment by inhaling this drug, and only a quarter of the minimum cost of RMB 32,811 Yuan [16] for the study of PAP treatment by subcutaneous injection of this drug. The maximum cost of this study was RMB 13,020, which was 1/26 of the maximum cost of RMB 344,306 [21] for the study of PAP treatment by inhaling this drug and 1/21 of the maximum cost of RMB 284,357 [20] for the study of PAP treatment by subcutaneous injection of this drug.

Safety analysis

Mild skin stiffness at the subcutaneous injection site (arm) appeared in two patients, and it almost disappeared after 2 weeks of the replacement of the injection site. No other adverse events occurred, and the blood routine, biochemical and other safety indicators were normal.

Discussion

Whole lung lavage (WLL) treatment has been considered the most effective and preferred treatment approach for pulmonary alveolar proteinosis (PAP). WLL has applied in clinical practice for nearly 50 years [28]. However, some patients show poor or ineffective response to the therapy, or require repeated treatment. Moreover, WLL is an invasive treatment method that needs to be carried out in the operating room under general anesthesia. The risks of anesthesia, washing operation and corresponding economic costs are relatively high. With the deep understanding of the pathogenesis of PAP, subcutaneous injection or inhalation of granulocyte-macrophage colony-stimulating factor (GM-CSF) has become an important treatment technique for idiopathic PAP. Although Cormac McCarthy et al. [29] suggested that statins could be used for PAP treatment by reducing the cholesterol level in alveolar macrophages, the actual application in clinical practice still needs to be confirmed by data of a large number of studies. Therefore, GM-CSF is still considered the main therapeutic drug for PAP.
At present, some studies reported on the treatment of PAP by GM-CSF [1524]. Of the five studies [1518, 20] describing the treatment of PAP by GM-CSF subcutaneous injection, a single dose recommended by Seymour [15] was the smallest, which was 5 μg/(kg · day). The study by Seymour included only one patient, whose therapy duration was 300 days and the effective rate was 100%. The single dose of another subcutaneous injection was 9 μg/(kg · day) [16]; a total of four patients were included, and the duration of therapy was 12 weeks. The effective rate of this study was 75%. In another study [18], the single dose of subcutaneous injection was 18 μg/(kg · day), and the duration of therapy ranged from 12 to 48 weeks. Eleven patients were included, and the effective rate was 55%. In another study [20], the single dose of subcutaneous injection was 18 μg/(kg · day), and the duration of medication ranged from 12 to 52 weeks, in which 21 patients were included in the study. In another study [17], the single dose of subcutaneous injection was also 20 μg/ (kg · day). A total of 14 patients were included. The duration of medication was 6–12 weeks, and the effective rate was 43%. Five studies reported on the GM-CSF inhalation treatment for PAP [19, 2124] of which the minimum single dose of inhalation treatment was 125 μg/kg [24] (twice/day inhalation, 1 week to stop, and therapy time was 24 weeks). A total of 35 patients were included, and the effective rate was 66%. The minimum single dose of another inhalation treatment study [19] was 250 μg/ (kg · day). The medication time was 24 weeks (1 week to stop). Three patients were included, and the effective rate was 100%. In another inhalation treatment study [21], the minimum single dose was 500 μg/kg (twice/day, 1 week to stop), and the duration of medication varied from 3 to 68 weeks. Twelve patients were included, and the effective rate was 92%. In one inhalation treatment study [22], the minimum single dose was 250 μg/(kg · day) (1 week to stop), and the duration of therapy was 24 weeks. Thirty-nine patients were included, and the effective rate was 62%. The minimum single dose of the inhalation treatment study [23] was 250 μg/(kg · day). The duration of therapy was 14–65 weeks, and the treatment was stopped for 4 days after every 4 days. A total of 35 patients were included, and the effective rate was 66%.
In this study, idiopathic PAP was treated by subcutaneous injection of low-dose GM-CSF (75 μg/d or 150 μg/d, ie 1.25 μg/kg/d or 2.5 μg/kg/d, calculated according to 60 kg body weight), which was far lower than the dose reported by other studies. The treatment time was 6 months, and the overall effective rate reached 85%. No obvious adverse reactions were found, and the economic burden of patients was significantly reduced compared with other studies reported so far. This type of therapy is especially suitable for patients with mild disease. In this study, seventeen of the 20 patients were followed up for 6 months after treatment (a total of 12 months), and three of them showed slightly increased lesions after withdrawal, indicating that the disease might relapse after withdrawal. In addition, eight of the 20 patients received a single dose of 75μg for 6 months, and no improvement was observed in 12 patients after 1-month evaluation. The single dose was increased to 150μg from the second month. After the increase of the dose, the efficacy of 9 patients among the 12 patients was evaluated as improved, indicating that appropriate increase of the dose can increase the efficacy. Nevertheless, to what extent can the dose ensure good efficacy, low side effects and the lowest economic burden, these problems need to be confirmed by further research by increasing the sample size.

Conclusion

Subcutaneous injection of rhGM-CSF at low dose (75μg-150μg /d) is effective treatment for patients with idiopathic PAP.
In conclusion, the method of subcutaneous injection of GM-CSF low dose designed in this study has the advantages of good clinical efficacy, low economic burden and light side effects compared with the methods reported by other studies and is suitable for clinical promotion and application.

Acknowledgements

Very appreciate the work on statistical analysis by Professor Ai-Hong Zhang who works for Tongji University School of Medicine.

Conflict of interest

The authors confirm that there are no conflicts of interest.
The Institutional Ethics Committee of Shanghai Pulmonary Hospital approved the study (No. K13–105).
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.
Literatur
1.
Zurück zum Zitat Rosen SH, Castleman B, Liebow AA. Pulmonary alveolar proteinosis. N Engl J Med. 1958;258:1123–42.CrossRef Rosen SH, Castleman B, Liebow AA. Pulmonary alveolar proteinosis. N Engl J Med. 1958;258:1123–42.CrossRef
2.
Zurück zum Zitat Saber H, Khaled H, Khaled L, Samir H, Chebil BM, Néji GM. Congenital pulmonary alveolar proteinosis. Case Rep Pediatr. 2013;2013:764216. Saber H, Khaled H, Khaled L, Samir H, Chebil BM, Néji GM. Congenital pulmonary alveolar proteinosis. Case Rep Pediatr. 2013;2013:764216.
3.
Zurück zum Zitat Haruyuki I, Seymour John F, Ryushi T, Yoshikazu I, Naoyuki U, Aya N, et al. Secondary pulmonary alveolar proteinosis complicating myelodysplastic syndrome results in worsening of prognosis: a retrospective cohort study in Japan. BMC Pulm Med. 2014;14(37):1471–80. Haruyuki I, Seymour John F, Ryushi T, Yoshikazu I, Naoyuki U, Aya N, et al. Secondary pulmonary alveolar proteinosis complicating myelodysplastic syndrome results in worsening of prognosis: a retrospective cohort study in Japan. BMC Pulm Med. 2014;14(37):1471–80.
4.
Zurück zum Zitat Nogee LM, deMello DE, Dehner LP, Colten HR. Deficiency of pulmonary surfactant protein B in congenital alveolar proteinosis. N Engl J Med. 1993;328:406–10.CrossRef Nogee LM, deMello DE, Dehner LP, Colten HR. Deficiency of pulmonary surfactant protein B in congenital alveolar proteinosis. N Engl J Med. 1993;328:406–10.CrossRef
5.
Zurück zum Zitat Kanji U, Koh N, Trapnell Bruce C, Takahiro T, Emi H, Ayako M, et al. High-affinity autoantibodies specifically eliminate granulocyte-macrophage colony-stimulating factor activity in the lungs of patients with idiopathic pulmonary alveolar proteinosis. Blood. 2004;103:1089–98. Kanji U, Koh N, Trapnell Bruce C, Takahiro T, Emi H, Ayako M, et al. High-affinity autoantibodies specifically eliminate granulocyte-macrophage colony-stimulating factor activity in the lungs of patients with idiopathic pulmonary alveolar proteinosis. Blood. 2004;103:1089–98.
6.
Zurück zum Zitat Seymour John F, Presneill JJ. Pulmonary alveolar proteinosis: progress in the first 44 years. Am J Respir Crit Care Med. 2002;166:215–35.CrossRef Seymour John F, Presneill JJ. Pulmonary alveolar proteinosis: progress in the first 44 years. Am J Respir Crit Care Med. 2002;166:215–35.CrossRef
7.
Zurück zum Zitat Dranoff G, Crawford AD, Sadelain M, Ream B, Rashid A, Bronson RT, et al. Involvement of granulocyte-macrophage colony-stimulating factor in pulmonary homeostasis. Science. 1994;264:713–6.CrossRef Dranoff G, Crawford AD, Sadelain M, Ream B, Rashid A, Bronson RT, et al. Involvement of granulocyte-macrophage colony-stimulating factor in pulmonary homeostasis. Science. 1994;264:713–6.CrossRef
8.
Zurück zum Zitat Carraway MS, Ghio AJ, Carter JD, Piantadosi CA. Detection of granulocyte-macrophage colony-stimulating factor in patients with pulmonary alveolar proteinosis. Am. J. Respir. Crit. Care Med. 2000;161:1294–9. Carraway MS, Ghio AJ, Carter JD, Piantadosi CA. Detection of granulocyte-macrophage colony-stimulating factor in patients with pulmonary alveolar proteinosis. Am. J. Respir. Crit. Care Med. 2000;161:1294–9.
9.
Zurück zum Zitat Kitamura T, Uchida K, Tanaka N, Tsuchiya T, Watanabe J, Yamada Y, et al. Serological diagnosis of idiopathic pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2000;162:658–62.CrossRef Kitamura T, Uchida K, Tanaka N, Tsuchiya T, Watanabe J, Yamada Y, et al. Serological diagnosis of idiopathic pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2000;162:658–62.CrossRef
10.
Zurück zum Zitat Bonfield Tracey L, Debra R, Sujata B, Anagha M, Kavuru Mani S, Jane TM. Autoantibodies against granulocyte macrophage colony-stimulating factor are diagnostic for pulmonary alveolar proteinosis. Am J Respir Cell Mol Biol. 2002;27:481–6.CrossRef Bonfield Tracey L, Debra R, Sujata B, Anagha M, Kavuru Mani S, Jane TM. Autoantibodies against granulocyte macrophage colony-stimulating factor are diagnostic for pulmonary alveolar proteinosis. Am J Respir Cell Mol Biol. 2002;27:481–6.CrossRef
11.
Zurück zum Zitat Kitamura T, Tanaka N, Watanabe J, Uchida K, Kanegasaki S, Yamada Y, Nakata K. Idiopathicpulmonary alveolar proteinosis as an autoimmune disease with neutralizing antibody against granulocyte macrophage colony-stimulating factor. Exp Med. 1999;190(6):875–80.CrossRef Kitamura T, Tanaka N, Watanabe J, Uchida K, Kanegasaki S, Yamada Y, Nakata K. Idiopathicpulmonary alveolar proteinosis as an autoimmune disease with neutralizing antibody against granulocyte macrophage colony-stimulating factor. Exp Med. 1999;190(6):875–80.CrossRef
12.
Zurück zum Zitat Venkateshiah Saiprakash B, Jane TM, Kavuru Mani S. Pulmonary alveolar proteinosis. Clinical manifestations and optimal treatment strategies. Treat Respir Med. 2004;3:217–27.CrossRef Venkateshiah Saiprakash B, Jane TM, Kavuru Mani S. Pulmonary alveolar proteinosis. Clinical manifestations and optimal treatment strategies. Treat Respir Med. 2004;3:217–27.CrossRef
13.
Zurück zum Zitat Takaki M, Tanaka T, Komohara Y, Tsuchihashi Y, Mori D, Hayashi K, Fukuoka J, et al. Recurrence of pulmonary alveolar proteinosis after bilateral lung transplantation in a patient with a nonsense mutation in CSF2RB. Respir Med Case Rep. 2016;19:89–93.PubMedPubMedCentral Takaki M, Tanaka T, Komohara Y, Tsuchihashi Y, Mori D, Hayashi K, Fukuoka J, et al. Recurrence of pulmonary alveolar proteinosis after bilateral lung transplantation in a patient with a nonsense mutation in CSF2RB. Respir Med Case Rep. 2016;19:89–93.PubMedPubMedCentral
14.
Zurück zum Zitat Takuro S, David B, Kanji U, Takuji S, Carey Brenna C, Koh N, et al. Patient-derived granulocyte/macrophage colony-stimulating factor autoantibodies reproduce pulmonary alveolar proteinosis in nonhuman primates. Am J Respir Crit Care Med. 2010;182:49–61.CrossRef Takuro S, David B, Kanji U, Takuji S, Carey Brenna C, Koh N, et al. Patient-derived granulocyte/macrophage colony-stimulating factor autoantibodies reproduce pulmonary alveolar proteinosis in nonhuman primates. Am J Respir Crit Care Med. 2010;182:49–61.CrossRef
15.
Zurück zum Zitat Seymour JF, Dunn AR, Vincent JM, Presneill JJ, Pain MC. Efficacy of granulocyte-macrophage colony-stimulating factor in acquired alveolar proteinosis. N Engl J Med. 1996;335:1924–5.CrossRef Seymour JF, Dunn AR, Vincent JM, Presneill JJ, Pain MC. Efficacy of granulocyte-macrophage colony-stimulating factor in acquired alveolar proteinosis. N Engl J Med. 1996;335:1924–5.CrossRef
16.
Zurück zum Zitat Kavuru MS, Sullivan EJ, Piccin R, Thomassen MJ, Stoller JK. Exogenous granulocyte-macrophage colony-stimulating factor administration for pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2000;161:1143–8.CrossRef Kavuru MS, Sullivan EJ, Piccin R, Thomassen MJ, Stoller JK. Exogenous granulocyte-macrophage colony-stimulating factor administration for pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2000;161:1143–8.CrossRef
17.
Zurück zum Zitat Seymour JF, Presneill JJ, Schoch OD, Downie GH, Moore PE, Doyle IR, et al. Therapeutic efficacy of granulocyte-macrophage colony-stimulating factor in patients with idiopathic acquired alveolar proteinosis. Am J Respir Crit Care Med. 2001;163:524–31.CrossRef Seymour JF, Presneill JJ, Schoch OD, Downie GH, Moore PE, Doyle IR, et al. Therapeutic efficacy of granulocyte-macrophage colony-stimulating factor in patients with idiopathic acquired alveolar proteinosis. Am J Respir Crit Care Med. 2001;163:524–31.CrossRef
18.
Zurück zum Zitat Bonfield Tracey L, Kavuru Mani S, Jane TM. Anti-GM-CSF titer predicts response to GM-CSF therapy in pulmonary alveolar proteinosis. Clin Immunol. 2002;105:342–50.CrossRef Bonfield Tracey L, Kavuru Mani S, Jane TM. Anti-GM-CSF titer predicts response to GM-CSF therapy in pulmonary alveolar proteinosis. Clin Immunol. 2002;105:342–50.CrossRef
19.
Zurück zum Zitat Ryushi T, Emi H, Toru A, Hiromitsu O, Osamu I, Kanji U, et al. Granulocyte-macrophage colony-stimulating factor and lung immunity in pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2005;171:1142–9.CrossRef Ryushi T, Emi H, Toru A, Hiromitsu O, Osamu I, Kanji U, et al. Granulocyte-macrophage colony-stimulating factor and lung immunity in pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2005;171:1142–9.CrossRef
20.
Zurück zum Zitat Venkateshiah Saiprakash B, Yan Tom D, Bonfield Tracey L, Jane TM, Moulay M, Carmen C, et al. An open-label trial of granulocyte macrophage colony stimulating factor therapy for moderate symptomatic pulmonary alveolar proteinosis. Chest. 2006;130:227–37.CrossRef Venkateshiah Saiprakash B, Yan Tom D, Bonfield Tracey L, Jane TM, Moulay M, Carmen C, et al. An open-label trial of granulocyte macrophage colony stimulating factor therapy for moderate symptomatic pulmonary alveolar proteinosis. Chest. 2006;130:227–37.CrossRef
21.
Zurück zum Zitat Wylan ME, Ten R, Prakash UBS, Nadrous HF, Clawson ML, Anderson PM. Aerosol granulocyte-macrophage colonystimulating factor for pulmonary alveolar proteinosis. Eur Respir J. 2006;27(3):585–93.CrossRef Wylan ME, Ten R, Prakash UBS, Nadrous HF, Clawson ML, Anderson PM. Aerosol granulocyte-macrophage colonystimulating factor for pulmonary alveolar proteinosis. Eur Respir J. 2006;27(3):585–93.CrossRef
22.
Zurück zum Zitat Ryushi T, Trapnell Bruce C, Yoshikazu I, Toru A, Toshinori T, Yasuyuki N, et al. Inhaled granulocyte/macrophage-colony stimulating factor as therapy for pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2010;181:1345–54.CrossRef Ryushi T, Trapnell Bruce C, Yoshikazu I, Toru A, Toshinori T, Yasuyuki N, et al. Inhaled granulocyte/macrophage-colony stimulating factor as therapy for pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2010;181:1345–54.CrossRef
23.
Zurück zum Zitat Papiris Spyros A, Panagiotis T, Likurgos K, Georgia P, Papaioannou Andriana I, Christina T, et al. Long-term inhaled granulocyte macrophage-colony-stimulating factor in autoimmune pulmonary alveolar proteinosis: effectiveness, safety, and lowest effective dose. Clin Drug Investig. 2014;34:553–64.CrossRef Papiris Spyros A, Panagiotis T, Likurgos K, Georgia P, Papaioannou Andriana I, Christina T, et al. Long-term inhaled granulocyte macrophage-colony-stimulating factor in autoimmune pulmonary alveolar proteinosis: effectiveness, safety, and lowest effective dose. Clin Drug Investig. 2014;34:553–64.CrossRef
24.
Zurück zum Zitat Ryushi T, Yoshikazu I, Toru A, Toshinori T, Yasunori K, Masayuki H, et al. Duration of benefit in patients with autoimmune pulmonary alveolar proteinosis after inhaled granulocyte-macrophage colony-stimulating factor therapy. Chest. 2014;145:729–37.CrossRef Ryushi T, Yoshikazu I, Toru A, Toshinori T, Yasunori K, Masayuki H, et al. Duration of benefit in patients with autoimmune pulmonary alveolar proteinosis after inhaled granulocyte-macrophage colony-stimulating factor therapy. Chest. 2014;145:729–37.CrossRef
25.
Zurück zum Zitat Lee KN, Levin DL, Webb WR, Chen D, Storto ML, Golden JA. Pulmonary alveolar proteinosis: high-resolution CT, chest radiographic, and functional correlations. Chest. 1997;111:989–95.CrossRef Lee KN, Levin DL, Webb WR, Chen D, Storto ML, Golden JA. Pulmonary alveolar proteinosis: high-resolution CT, chest radiographic, and functional correlations. Chest. 1997;111:989–95.CrossRef
26.
Zurück zum Zitat Holbert JM, Costello P, Li W, Hoffman RM, Rogers RM. CT features of pulmonary alveolar proteinosis. AJR Am J Roentgenol. 2001;176:1287–94.CrossRef Holbert JM, Costello P, Li W, Hoffman RM, Rogers RM. CT features of pulmonary alveolar proteinosis. AJR Am J Roentgenol. 2001;176:1287–94.CrossRef
27.
Zurück zum Zitat Borie R, Danel C, Debray MP, Taille C, Dombret MC, Aubier M, et al. Pulmonary alveolar proteinosis. Eur Respir Rev. 2011;20:98–107.CrossRef Borie R, Danel C, Debray MP, Taille C, Dombret MC, Aubier M, et al. Pulmonary alveolar proteinosis. Eur Respir Rev. 2011;20:98–107.CrossRef
28.
Zurück zum Zitat Ramirez J, Kieffer RJ, Ball WJ. Bronchopumonary lavage in man. Ann Intem Med. 1965;63(5):819–28.CrossRef Ramirez J, Kieffer RJ, Ball WJ. Bronchopumonary lavage in man. Ann Intem Med. 1965;63(5):819–28.CrossRef
29.
Zurück zum Zitat Cormac MC, Lee E, Bridges James P, Anthony S, Takuji S, Woods Jason C, Tarling Elizabeth J, Trapnell Bruce C. Statin as a novel pharmacotherapy of pulmonary alveolar proteinosis. Nat Commun. 2018;9(1):3127–35.CrossRef Cormac MC, Lee E, Bridges James P, Anthony S, Takuji S, Woods Jason C, Tarling Elizabeth J, Trapnell Bruce C. Statin as a novel pharmacotherapy of pulmonary alveolar proteinosis. Nat Commun. 2018;9(1):3127–35.CrossRef
Metadaten
Titel
Therapeutic effect of subcutaneous injection of low dose recombinant human granulocyte-macrophage colony-stimulating factor on pulmonary alveolar proteinosis
verfasst von
Fen Zhang
Dong Weng
Yiliang Su
Chengsheng Yin
Li Shen
Yuan Zhang
Ying Zhou
Qiuhong Li
Yang Hu
Huiping Li
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2020
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-019-1261-1

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

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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