Skip to main content

Nebulized inhalation of plasma-activated water in the treatment of progressive moderate COVID-19 patients with antiviral treatment failure: a randomized controlled pilot trial

Abstract

Background

Antiviral drugs show significant efficacy in non-severe COVID-19 cases, yet there remains a subset of moderate COVID-19 patients whose pneumonia continues to progress post a complete course of treatment. Plasma-activated water (PAW) possesses anti-SARS-CoV-2 properties. To explore the potential of PAW in improving pneumonia in COVID-19 patients following antiviral treatment failure, we conducted this study.

Methods

This was a randomized, controlled trial. Moderate COVID-19 patients with antiviral treatment failure were randomly assigned to the experimental group or the control group. They inhaled nebulized PAW or saline respectively. This was done twice daily for four consecutive days. We assessed improvement in chest CT on day 5, the rate of symptom resolution within 10 days, and safety.

Results

A total of 23 participants were included, with 11 receiving PAW and 12 receiving saline. The baseline characteristics of both groups were comparable. The experimental group showed a higher improvement rate in chest CT on day 5 (81.8% vs. 33.3%, p = 0.036). The cumulative disappearance rate of cough within 10 days was higher in the experimental group. Within 28 days, 4 patients in each group progressed to severe illness, and no patients died. No adverse reactions were reported from inhaling nebulized PAW.

Conclusion

This pilot trial preliminarily confirmed that nebulized inhalation of PAW can alleviate pneumonia in moderate COVID-19 patients with antiviral treatment failure, with no adverse reactions observed. This still needs to be verified by large-scale studies.

Trial registration

Chinese Clinical Trial Registry; No.: ChiCTR2300078706 (retrospectively registered, 12/15/2023); URL: www.chictr.org.cn.

Peer Review reports

Introduction

Coronavirus disease 2019 (COVID-19) continues to be pandemic globally, and a variety of antiviral drugs have been widely used to treat COVID-19 pneumonia. Commonly used oral antiviral drugs such as Nirmatrelvir/Ritonavir and Molnupiravir are small molecules that suppress viral replication and propagation by acting on key enzymes in the replication process of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [1,2,3]. These drugs can reduce the hospitalization and mortality rates of non-severe COVID-19 patients [4, 5], but a portion of patients still progress to severe disease and death after treatment. For example, a retrospective study by Wai et al. found that among high-risk mild to moderate COVID-19 hospitalized patients in Hong Kong, the 28-day all-cause mortality rates for Nirmatrelvir/Ritonavir and Molnupiravir were 4.3% and 8.5%, respectively [6]. Additionally, the aforementioned drugs also have adverse reactions, such as disease recurrence, dysgeusia, diarrhea, and rash [7, 8]. Therefore, the search for new, more effective, and safer antiviral drugs is imperative.

Plasma-activated water (PAW) is a category of aqueous solution activated by discharge plasma, containing abundant reactive species with various biological effects such as anti-microbial, anti-cancer, and stopping bleeding [9, 10]. Generally, PAW is produced by the reaction between plasma-generated gaseous reactive species and aqueous solutions via diffusion, bubbling, or jetting to produce aqueous species [11]. Plasma can inactivate SARS-CoV-2 in the environment [12, 13] and degrade its RNA [14]. Guo et al. found that PAW can disrupt the receptor-binding domain (RBD) of SARS-CoV-2 spike protein, preventing its binding to human angiotensin-converting enzyme 2 (ACE2) [15]. Acute and chronic toxicity tests in animals confirmed the biosafety of PAW [16, 17]. In addition, we have observed no adverse events in C57 mice after nebulizing PAW for 1 week. Blood indicators and histopathological examinations of important organs were normal. C57 mice infected with influenza A virus H1N1 PR8 were given aerosolized PAW for 3 days, and we observed its effectiveness. The results showed that the experimental group was superior to the control group in terms of survival status, body weight, and decreased inflammatory factors. Two clinical studies have confirmed the safety of PAW in humans [18, 19]. Guo et al. found that gargling with PAW can accelerate the conversion of viral RNA from positive to negative in COVID-19 patients with persistently positive oropharyngeal swabs, improve sore throat, and without adverse reactions [20]. However, for COVID-19 patients with pneumonia, gargling treatments may be less effective. Inhalation administration has the advantages of rapid onset, high local drug concentration, low dosage, and fewer systemic adverse reactions [21]. Several studies suggest that inhaled antiviral drugs may be effective in treating COVID-19 [22,23,24,25]. Cortázar et al. found in vitro that nebulized PAW can reduce the infectivity of SARS-CoV-2 [26]. In December 2022, we provided remedial nebulized inhalation of PAW to 5 hospitalized COVID-19 patients whose chest computed tomography (CT) showed worsening pneumonia after 5 days of oral antiviral drug treatment. Worsening pneumonia refers to an increase in the number and/or size of pneumonia lesions on chest CT. Results showed significant improvement in chest CT on day 5 in 4 patients, and 1 patient with slight worsening on day 5 improved on day 12 after one week of standard treatment. Their symptoms gradually improved, with no adverse events occurring.

Based on the aforementioned findings, to further explore the effectiveness and safety of nebulized inhalation of PAW in COVID-19 patients, we selected moderate COVID-19 patients with high risk of severity and worsening pulmonary imaging post-oral antiviral treatment, and administered nebulized PAW. This is the first study of nebulized inhalation of PAW for the treatment of pulmonary infections and COVID-19.

Methods

Study design and randomization

This is a single-center, single-blind, randomized, controlled trial. All subjects were randomly assigned in a 1:1 ratio to the experimental and control groups by simple randomization. Random numbers were generated by a separate researcher using computer software (SPSS software version 27), and then secured in sequentially numbered opaque sealed envelopes to conceal allocation. The researcher in charge of enrollment contacted this independent researcher to obtain random numbers and corresponding groups when the subjects were enrolled.

Ethical review

The study follows international scientific research standards and the Declaration of Helsinki, approved by the Medical Ethics Committee of the First Affiliated Hospital of the Air Force Medical University (Approval No.: KY20232004-C-1), registered with the Chinese Clinical Trial Registry (Registration No.: ChiCTR2300078706), and written informed consent was obtained from all participants.

Subjects

The eligible population consists of moderate COVID-19 patients hospitalized in Xijing Hospital from January to July 2023. The COVID-19 classification is based on the “Diagnosis and Treatment Protocol for Novel Coronavirus Pneumonia (Trial Version 9)” issued by China’s National Health Commission. The criteria for moderate COVID-19 are as follows: presence of fever and/or respiratory symptoms and other clinical manifestations related to COVID-19, with radiological evidence of pneumonia. Additionally, the following criteria must be met: aged 18–85 years; presence of high-risk factors for severe disease; positive SARS-CoV-2 polymerase chain reaction or rapid antigen test within the past week; chest CT after 5 days of oral Nirmatrelvir/Ritonavir treatment showing increased number and/or size of pneumonia lesions. Patients with severe or critical COVID-19, allergic constitution, bacterial or fungal co-infection of the lungs, pregnant or lactating women, and those planning to conceive were excluded. Detailed COVID-19 classification criteria and high-risk factors for severe disease are provided in the Supplementary Materials.

Preparation of PAW

The equipment and preparation method used in the study of PAW gargling treatment for COVID-19 were used [20]. A hybrid plasma discharge configuration using ambient air was used for the PAW preparation, where a dielectric barrier discharge reactor and a gliding arc discharge reactor generated air plasmas in O3 mode and NOx mode, respectively. The dielectric barrier discharge reactor was powered by a sinusoidal high-voltage power supply (Xiaozhong Environmental Ltd., XZ-10GKT) with an output voltage (peak-to-peak value) of about 12 kV and a frequency of about 10 kHz, while the gliding arc discharge reactor was driven by a high-voltage transformer (Hongba Electronics Ltd., MXP-12KV-40MA) with an output voltage (peak-to-peak value) of about 15 kV and a frequency of about 50 Hz. In brief, gas plasma was introduced into 200 ml of saline (0.9%NaCl) and reacted for 10 min to prepare PAW.

Interventions and blinding

All subjects received standard treatment according to COVID-19 treatment guidelines. On this basis, the experimental group inhaled nebulized PAW twice daily (interval > 4 h). Using an oxygen-driven jet nebulizer (oxygen flow rate 6 L/min) to nebulize 5 ml of PAW for 10 min each time, for 4 consecutive days. PAW was nebulized immediately after preparation for treatment of the subjects. The control group inhaled nebulized 5 ml saline using the same method. Subjects were blinded in the study. There was no difference in appearance between PAW and saline, so the subjects did not know what they received. Subjects gargled and washed their faces with water after the nebulization treatment.

Efficacy

The primary efficacy indicator was the improvement rate of chest CT on day 5. Each chest CT was evaluated by two radiologists with over 15 years of experience in chest imaging diagnosis and a respiratory physician with over 20 years of pulmonary medicine experience. All three evaluators were blinded. This qualitative assessment was classified as improvement, stability, or aggravation, with the following specific criteria: improvement: lesion range decreased; lesion range unchanged, density reduced. Stability: lesion range and density basically unchanged. Aggravation: lesion range increased; lesion range unchanged, density increased; new lesions appeared. Stability and aggravation were classified as non-improvement. Scans were performed using a 64-slice spiral CT (GE, LightSpeed VCT). In addition, we conducted quantitative analysis of chest CT using a computerized quantitative analysis method that was utilized in the study by Shen et al. [27]. The volume and density of lung lesions were automatically measured in the Digital Lung software (DEXIN, China). The results of lesion segmentation obtained by the software were inspected by another radiologist. If there were any inaccuracies, they were manually rectified by the radiologist.

Secondary efficacy indicators included the rate of symptom disappearance within 10 days, the rate of symptom relief within 10 days, hospitalization duration, levels of inflammatory factors on day 5 (procalcitonin [PCT], interleukin 6 [IL-6]), the proportion of moderate to severe cases within 28 days, and all-cause death within 28 days. Symptom scores were assessed and reported by patients according to the severity of their symptoms. We collected three symptoms: cough, sputum production, and shortness of breath. The scoring criteria from the U.S. FDA COVID-19-related symptom questionnaire were used [28]. Specific scoring criteria are as follows: none = 0 points, mild = 1 point, moderate = 2 points, severe = 3 points. The aforementioned symptoms were recorded at the same time every day, for 10 consecutive days, starting from enrollment. Face-to-face interviews during hospitalization and follow-up calls after discharge. The changes of each symptom were recorded in detail. Symptom disappearance was defined as the symptom being absent for two consecutive days, with the first day of absence recorded as the time of symptom disappearance. Symptom relief was defined as a decrease of 1 point or more in the patient’s symptom score compared to baseline, maintained for at least two days, with the first day of relief recorded as the time of symptom relief. Symptom disappearance and symptom relief were both for each symptom. Venous blood from patients was collected at the study baseline and at the end of the trial, with PCT and IL-6 levels measured using a fluorescence immunoassay analyzer (Getein1600, GeteinBiotech).

Safety

Potential adverse reactions such as dry mouth, nausea, palpitations, cough, chest tightness, dyspnea, difficulty breathing, and skin/mucosal damage were monitored in patients during the study, as well as vital signs before and after the intervention. Venous blood was collected from patients at the study baseline and at the end of the trial, and a complete blood count was performed using a hematology analyzer (XN-3000, SYSMEX). Biochemical markers (alanine aminotransferase, aspartate aminotransferase, blood urea nitrogen, and creatinine) were measured using a dry chemistry analyzer (FS 5.1, Johnson & Johnson MedTech).

Statistical analysis

Data were analyzed in the per-protocol analysis population. The Shapiro-Wilke test was used to check whether the continuous data were normally distributed. Continuous data adhering to a normal distribution were expressed as mean ± standard deviation and compared between groups using the independent samples t-test. Continuous data following a skewed distribution were expressed as median (interquartile range) and compared between groups using the Mann-Whitney U test. Categorical data were expressed as frequency (percentage) and compared between groups using Fisher’s exact test. Ordinal data were expressed as frequency (percentage) and compared between groups using the Mann-Whitney U test. The rate of symptom disappearance and symptom relief within 10 days were displayed using Kaplan-Meier curves and compared using the Log-Rank test. The Kaplan-Meier curves were generated and the Log-Rank test was conducted using R software (version 4.3.2). All other statistical analyses were performed in SPSS software version 27. A P-value < 0.05 (two-sided) was considered statistically significant.

Sample size

Before this study was conducted, no literature reported the improvement rate of chest CT after Nirmatrelvir/Ritonavir treatment in moderate COVID-19. Therefore, we preliminarily analyzed the data of hospitalized COVID-19 patients in our hospital before the study. Among 152 moderate COVID-19 patients treated with Nirmatrelvir/Ritonavir, 16 cases showed worsening pneumonia on chest CT. Those with worsening pneumonia continued to receive standard treatment according to COVID-19 treatment guidelines. Of the 11 patients who received only standard treatment, 3 showed improvement in chest CT, an improvement rate of approximately 27%. Among the 5 patients who received both standard treatment and PAW, 4 showed improvement in chest CT, an improvement rate of approximately 80%. Conservatively estimating the improvement rate of PAW at 60%, with a two-sided alpha of 0.05, a power of 0.8, and a dropout rate of 20%, the sample size was calculated to be 80 cases, with 40 cases in each group.

Results

Baseline characteristics

From January to July 2023, a total of 335 subjects were screened. 311 individuals were excluded, of which 301 patients did not meet the inclusion criteria and 10 eligible patients refused to participate. Subjects who withdrew consent after randomization were excluded. Due to slow recruitment, patient enrollment was stopped on August 31, 2023, as recommended by the independent data and safety monitoring board. A total of 24 subjects were involved in the randomization, with 11 in the experimental group and 13 in the control group. One patient in the control group withdrew from the study due to an acute myocardial infarction before the first intervention on the day of randomization. Ultimately, 23 subjects received the intervention and completed follow-up (Fig. 1). According to data from the Chinese Center for Disease Control and Prevention, the prevalent strains of SARS-CoV-2 in China during this period were all Omicron variants, mainly BA.5.2, BF.7, XBB, and EG.5. There were no significant differences between the experimental and control groups in terms of age, sex, body mass index, smoking history, comorbidities, disease duration, and vaccination status (Table 1).

Fig. 1
figure 1

Randomization, treatment assignments, and follow-up

Table 1 Demographic and clinical characteristics of the subjects

Primary outcomes

After 4 days of intervention, qualitative assessment of follow-up chest CT showed that the experimental group was superior to the control group. The improvement rate of chest CT in the experimental group was significantly higher than that in the control group (81.8% vs. 33.3%, estimate difference [95%CI], 48.48% [13.40–83.57%], p = 0.036) (Table 2). The images from the quantitative analysis of chest CT are shown in Fig. 2. Quantitative analysis showed similar results: the experimental group’s DLeV% (percentage of lung volume occupied by lesions before treatment - percentage of lung volume occupied by lesions after treatment) and DLW (mass of lesions before treatment - mass of lesions after treatment) were significantly higher than those of the control group (p = 0.010, p = 0.006). The experimental group’s DMleD (mean density of lesions before treatment - mean density of lesions after treatment) was also higher than that of the control group (p = 0.055).

Table 2 Primary outcomes: qualitative and quantitative analysis of chest CT on day 5
Fig. 2
figure 2

Illustration of lesion segmentation and dynamic changes. As shown in A and C, all lesions were segmented and marked by red lines. B and D represent the three-dimensional reconstruction of lesions. A and B are pre-intervention images, while C and D are post-intervention images

Secondary outcomes

During the 10-day observation period, the cumulative disappearance rate of cough in the experimental group was higher than in the control group (p = 0.029) (Fig. 3-A). No significant differences were observed between groups in the cumulative disappearance rates and cumulative relief rates of other symptoms (Fig. 3-B, C, D, E, F).

Fig. 3
figure 3

The cumulative disappearance rate and cumulative relief rate of symptoms within 10 days. A: The cumulative disappearance rate of cough within 10 days. B: The cumulative disappearance rate of sputum production within 10 days. C: The cumulative disappearance rate of shortness of breath within 10 days. D: The cumulative relief rate of cough within 10 days. E: The cumulative relief rate of sputum production within 10 days. F: The cumulative relief rate of shortness of breath within 10 days

The average hospital stay for the experimental group was approximately 5 days shorter than the control group, and the median time from the first intervention to discharge was 3 days shorter for the experimental group, although these differences were not statistically significant (Table 3). There were no significant differences in inflammatory markers (PCT, IL-6) between the two groups on day 5. Within a 28-day follow-up period, 4 cases in each group progressed to severe COVID-19, with no significant difference in the proportion of moderate-to-severe cases between groups. No patients died, and there was no difference in 28-day all-cause mortality.

Table 3 Secondary outcomes

Safety

Throughout the study, no patients reported discomfort or adverse reactions. No new instances of dry mouth, nausea, palpitations, cough, chest tightness, dyspnea, difficulty breathing, or skin/mucosal damage were observed after the intervention, nor did any existing symptoms worsen. There were no significant differences in complete blood count or hepatic and renal function indicators between the groups after the intervention (Table 4).

Table 4 Safety of plasma-activated water

Discussion

In moderate COVID-19 patients with high risk of severity and progression of pneumonia on radiological imaging following oral antiviral medication, we administered nebulized PAW or saline. The results showed that the PAW group had significantly higher rates of improvement in chest CT and cough disappearance compared to the saline group, with a trend toward shorter hospital stays in the PAW group. No patient died within 28 days. No adverse reactions occurred.

Chest imaging is a commonly used and objective indicator for clinicians to evaluate whether pneumonia is controlled by medication. Chest CT allows for a direct visual assessment of the severity of pneumonia lesions in COVID-19 patients [29,30,31]. This study found that PAW could halt or even reverse the deterioration of chest CT in moderate COVID-19 patients post-antiviral treatment. Possible reasons for PAW facilitating radiological improvement in COVID-19 patients include: on one hand, PAW can disrupt the structure of SARS-CoV-2 and inhibit its replication. PAW contains a large number of reactive oxygen species and reactive nitrogen species, such as O2•–, O3, •OH, ONOOH, H2O2, NO•, etc [10, 11, 32]. Guo et al. found that the short-lived reactive species such as ONOO − in PAW play a significant role in damaging the RBD of the SARS-CoV-2 spike protein [15]. Wang et al. confirmed that •OH play a dominant role in triggering ACE2 nucleus translocation [33]. Khanikar et al. demonstrated that H2O2 can inactivate the SARS-CoV-2 spike protein and reduce its gene abundance [34]. Guo et al. have verified that gargling with PAW can accelerate the viral RNA clearance in COVID-19 patients with prolonged positive oropharyngeal swabs [20]. On the other hand, we postulate that PAW has anti-inflammatory and immunosuppressive effects on pulmonary inflammatory lesions. Pulmonary pathology in COVID-19 patients reveals substantial immune cell accumulation [35, 36]. Immune cells, cytokines, and chemokines are significantly elevated in bronchoalveolar lavage fluid [37]. Numerous animal experiments and human studies have confirmed that plasma has local immunosuppressive and anti-inflammatory effects [38,39,40,41]. Therefore, we speculate that PAW might reduce the infiltration of immune cells and the levels of inflammatory cytokines in the lungs of COVID-19 patients, thus avoiding an excessive immune response and promoting the improvement of pneumonia. There was no significant difference between the two groups in terms of improvement in serum IL-6. This may be because all patients included in this study were moderate COVID-19 cases, with overall low and comparable baseline serum IL-6 levels. Additionally, the samples used for IL-6 measurement were venous blood, not from the lungs or bronchoalveolar lavage fluid. Cytokine levels in the blood may not match those in the lungs [42]. Whether PAW can reduce pulmonary IL-6 and other cytokine levels remains to be explored.

Common clinical manifestations in COVID-19 patients include fever, fatigue, myalgia, cough, and sputum production [43, 44]. Our study results indicate that nebulized inhalation of PAW can accelerate the resolution of cough in COVID-19 patients. According to the literature, the potential mechanism of COVID-19-related cough may involve accumulation of inflammatory cells and release of inflammatory mediators in the respiratory tract, epithelial cell damage, and increased secretions following infection with SARS-CoV-2. These stimuli can induce a cough hypersensitivity state through neuroinflammatory or neuroimmune processes [45,46,47]. This is a complex process that may involve factors such as viral invasion, inflammation, immunity, neuroreflex, and hypersensitivity. The improvement in cough observed in COVID-19 patients treated with PAW may result from the combined effects of multiple pathways. First, PAW can block the binding of SARS-CoV-2 Spike protein RBD to the respiratory tract ACE2, preventing viral invasion. Second, PAW can suppress the infiltration of immune cells and reduce the levels of inflammatory mediators in the respiratory tract. Finally, PAW may have anti-allergic effects. In vitro studies have found that liquid-type plasma inhibits mast cell activation [48]. Animal experiments suggest that plasma can suppress the recruitment of mast cells and eosinophils in the skin lesions of mice with atopic dermatitis and reduce IgE levels in the lesions [48, 49]. Interestingly, an improvement in cough was also observed in another randomized controlled study where we used nebulized inhalation of PAW to treat acute upper respiratory infections caused by the influenza A virus.

There was no difference in the rate of progression to severe disease between the two groups in this study, which may be related to the small number of cases. This study suggests a trend toward reduced hospital stay duration with PAW treatment, which could help save medical costs and alleviate medical congestion during epidemic outbreaks.

During the study, no local or systemic adverse reactions occurred in patients in the PAW group. There were no significant differences in routine blood tests, liver function, or kidney function laboratory indicators between the groups. A large number of animal studies have confirmed the safety of PAW [16, 17]. Studies by Hwang et al. and Jang et al. confirmed that PAW spray treatment for bacterial vaginosis did not cause adverse reactions [18, 19]. PAW gargle treatment for COVID-19 patients did not cause adverse reactions [20].

This study is a preliminary investigation into the efficacy and safety of nebulized inhalation of PAW for the treatment of COVID-19 and has some limitations. First, the sample size of this study was small. The main reason is the significant reduction in the incidence of COVID-19 in China, especially the scarcity of moderate COVID-19 patients with oral antiviral treatment failure at the hospital where the researchers are located. Second, this trial was a pilot study and used a single-blind design. Finally, studies of this nature often use mortality as a primary endpoint. However, due to the small sample size and the low mortality rate among moderate COVID-19 patients, it was not appropriate to use mortality as the primary endpoint.

Currently, there is a lack of effective treatment methods for COVID-19 pneumonia that fails to respond to antiviral medication. We innovatively used nebulized inhalation of PAW to treat such COVID-19 patients and preliminarily confirmed its efficacy and safety. Further multicenter, large-sample studies are needed for validation. We are researching the use of nebulized inhalation of PAW for the initial treatment of COVID-19 and the treatment of acute upper respiratory infections caused by the influenza A virus. Additionally, we will explore the therapeutic effects of PAW on other pathogen infections.

Conclusion

This pilot trial preliminarily confirmed that for moderate COVID-19 patients with worsening pneumonia radiologically after antiviral treatment and high-risk factors for severe illness, nebulized inhalation of PAW can alleviate pneumonia, with no adverse reactions observed. The clinical efficacy of nebulized inhalation of PAW in the treatment of COVID-19 still needs to be confirmed by large-scale, multicenter clinical studies.

Data availability

Data supporting the findings of this study are available from the corresponding author upon request.

Abbreviations

ACE2:

Angiotensin-converting enzyme 2

COVID-19:

Coronavirus disease 2019

CT:

Computed tomography

DLeV% :

LeV% at baseline minus LeV% on day 5

DLW :

LW at baseline minus LW on day 5

DMLeD :

MLeD at baseline minus MLeD on day 5

IL-6:

Interleukin 6

LeV%:

The ratio of lesion volume to lung volume

LeV:

Lesion volume

LV:

Lung volume

LW:

Lesion weight

MLeD:

Mean lesion density

PAW:

Plasma-activated water

PCT:

Procalcitonin

RBD:

Receptor-binding domain

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

References

  1. Ng TI, Correia I, Seagal J, DeGoey DA, Schrimpf MR, Hardee DJ, Noey EL, Kati WM. Antiviral drug discovery for the treatment of COVID-19 infections. Viruses-Basel. 2022;14(5):961.

    Article  CAS  Google Scholar 

  2. Owen DR, Allerton CMN, Anderson AS, Aschenbrenner L, Avery M, Berritt S, Boras B, Cardin RD, Carlo A, Coffman KJ et al. An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science. 2021;374(6575):1586–1593.

  3. Kabinger F, Stiller C, Schmitzová J, Dienemann C, Kokic G, Hillen HS, Höbartner C, Cramer P. Mechanism of molnupiravir-induced SARS-CoV-2 mutagenesis. Nat Struct Mol Biol. 2021;28(9):740–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hammond J, Leister-Tebbe H, Gardner A, Abreu P, Bao W, Wisemandle W, Baniecki M, Hendrick VM, Damle B, Simón-Campos A, et al. Oral nirmatrelvir for high-risk, nonhospitalized adults with Covid-19. New Engl J Med. 2022;386(15):1397–408.

    Article  CAS  PubMed  Google Scholar 

  5. Jayk Bernal A, Gomes Da Silva MM, Musungaie DB, Kovalchuk E, Gonzalez A, Delos Reyes V, Martín-Quirós A, Caraco Y, Williams-Diaz A, Brown ML, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. New Engl J Med. 2022;386(6):509–20.

    Article  PubMed  Google Scholar 

  6. Wai AK, Chan CY, Cheung AW, Wang K, Chan SC, Lee TT, Luk LY, Yip ET, Ho JW, Tsui OW, et al. Association of Molnupiravir and Nirmatrelvir-Ritonavir with preventable mortality, hospital admissions and related avoidable healthcare system cost among high-risk patients with mild to moderate COVID-19. Lancet Reg Health - Western Pac. 2023;30:100602.

    Article  Google Scholar 

  7. Zhuang W, Xu J, Wu Y, Yang J, Lin X, Liao Y, Wan J, Weng L, Lin W. Post-marketing safety concerns with nirmatrelvir: a disproportionality analysis of spontaneous reports submitted to the FDA adverse event reporting system. Brit J Clin Pharmaco. 2023;89(9):2830–42.

    Article  CAS  Google Scholar 

  8. Santi Laurini G, Montanaro N, Motola D. Safety profile of Molnupiravir in the treatment of COVID-19: a descriptive study based on FAERS data. J Clin Med. 2023;12(1):34.

    Article  Google Scholar 

  9. Kaushik NK, Ghimire B, Li Y, Adhikari M, Veerana M, Kaushik N, Jha N, Adhikari B, Lee S, Masur K, et al. Biological and medical applications of plasma-activated media, water and solutions. Biol Chem. 2019;400(1):39.

    Article  CAS  Google Scholar 

  10. Kim S, Kim C. Applications of plasma-activated liquid in the medical field. Biomedicines. 2021;9(11):1700.

  11. Zhou R, Zhou R, Wang P, Xian Y, Mai-Prochnow A, Lu X, Cullen PJ, Ostrikov KK, Bazaka K. Plasma-activated water: generation, origin of reactive species and biological applications. J Phys D. 2020;53(30):303001.

    Article  CAS  Google Scholar 

  12. Bisag A, Isabelli P, Laghi G, Laurita R, Dirani G, Taddei F, Bucci C, Capelli F, Gherardi M, Paglianti A et al. Cold atmospheric plasma decontamination of SARS-CoV‐2 bioaerosols. Plasma Process Polym. 2022;19(3).

  13. Loizou C, Kniazeva V, Apostolou T, Kornev A, Kostevitch S, Roslyakov E, Constantinou C, Hadjihannas L. Effect of cold atmospheric plasma on SARS-CoV-2 inactivation: a pilot study in the hospital environment. COVID. 2022;2(10):1396–404.

    Article  Google Scholar 

  14. Bisag A, Isabelli P, Laurita R, Bucci C, Capelli F, Dirani G, Gherardi M, Laghi G, Paglianti A, Sambri V et al. Cold atmospheric plasma inactivation of aerosolized microdroplets containing bacteria and purified SARS-CoV‐2 RNA to contrast airborne indoor transmission. Plasma Process Polym. 2020;17(10).

  15. Guo L, Yao Z, Yang L, Zhang H, Qi Y, Gou L, Xi W, Liu D, Zhang L, Cheng Y, et al. Plasma-activated water: an alternative disinfectant for S protein inactivation to prevent SARS-CoV-2 infection. Chem Eng J. 2021;421:127742.

    Article  CAS  PubMed  Google Scholar 

  16. XU D, CUI Q, XU Y, WANG B, TIAN M, LI Q, LIU Z, LIU D, CHEN H, KONG MG. Systemic study on the safety of immuno-deficient nude mice treated by atmospheric plasma-activated water. Plasma Sci Technol. 2018;20(4):44003.

    Article  Google Scholar 

  17. Nastasa V, Pasca A, Malancus R, Bostanaru A, Ailincai L, Ursu E, Vasiliu A, Minea B, Hnatiuc E, Mares M. Toxicity assessment of long-term exposure to non-thermal plasma activated water in mice. Int J Mol Sci. 2021;22(21):11534.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hwang Y, Jeon H, Wang GY, Kim HK, Kim J, Ahn DK, Choi JS, Jang Y. Design and medical effects of a vaginal cleaning device generating plasma-activated water with antimicrobial activity on bacterial vaginosis. Plasma. 2020;3(4):204–13.

    Article  Google Scholar 

  19. Jang Y, Bok J, Ahn DK, Kim C, Kang J. Human trial for the effect of plasma-activated water spray on vaginal cleaning in patients with bacterial vaginosis. Med Sci. 2022;10(2):33.

    CAS  Google Scholar 

  20. Guo X, Lv X, Wu Y, Wang M, Wu S, Guo L, Wang Z, Shi Z, Huang R, Zhang H et al. Safety and efficacy of plasma-activated water on prolonged viral shedding of COVID‐19 patients: a randomized controlled trial. Plasma Process Polym. 2023.

  21. Anderson S, Atkins P, Bäckman P, Cipolla D, Clark A, Daviskas E, Disse B, Entcheva-Dimitrov P, Fuller R, Gonda I, et al. Inhaled medicines: past, present, and future. Pharmacol Rev. 2022;74(1):48–118.

    Article  CAS  PubMed  Google Scholar 

  22. Gül F, Gonen ZB, Jones OY, Taşlı NP, Zararsız G, Ünal E, Özdarendeli A, Şahin F, Eken A, Yılmaz S et al. A pilot study for treatment of severe COVID-19 pneumonia by aerosolized formulation of convalescent human immune plasma exosomes (ChipEXO™). Front Immunol. 2022;13.

  23. Li C, Zhan W, Yang Z, Tu C, Hu G, Zhang X, Song W, Du S, Zhu Y, Huang K, et al. Broad neutralization of SARS-CoV-2 variants by an inhalable bispecific single-domain antibody. Cell. 2022;185(8):1389–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Poulakou G, Barakat M, Israel RJ, Bacci MR. Ribavirin aerosol in hospitalized adults with respiratory distress and COVID-19: an open‐label trial. Clin Transl Sci. 2023;16(1):165–74.

    Article  CAS  PubMed  Google Scholar 

  25. Zheng F, Zhou Y, Zhou Z, Ye F, Huang B, Huang Y, Ma J, Zuo Q, Tan X, Xie J, et al. SARS-CoV-2 clearance in COVID-19 patients with Novaferon treatment: a randomized, open-label, parallel-group trial. Int J Infect Dis. 2020;99:84–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cortázar OD, Megía-Macías A, Moreno S, Brun A, Gómez-Casado E. Vulnerability of SARS-CoV-2 and PR8 H1N1 virus to cold atmospheric plasma activated media. Sci Rep-UK. 2022;12(1).

  27. Shen C, Yu N, Cai S, Zhou J, Sheng J, Liu K, Zhou H, Guo Y, Niu G. Quantitative computed tomography analysis for stratifying the severity of Coronavirus disease 2019. J Pharm Anal. 2020;10(2):123–9.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Assessing COVID-19-related symptoms in outpatient adult and adolescent subjects in clinical trials of drugs and biological products for COVID-19 prevention or treatment. https://www.fda.gov/media/142143/download. Accessed 13 August 2024.

  29. Azizmohammadi S, Azizmohammadi S, Dahmardeh S, Azargashb H, Shokouh SJH, Mohajeri-Iravani M, Mobasher M, Soleiman-Meigooni S, Zabihi M. Analysis of 239 ordinary and severe cases of COVID-19: clinical features and treatment. Eur J Transl Myol. 2021;31(3).

  30. Li D, Liu C, Liu J, Hu J, Yang Y, Zhou Y. Analysis of risk factors for 24 patients with COVID-19 developing from moderate to severe condition. Front Cell Infect Mi. 2020;10.

  31. Zhou Z, Guo D, Li C, Fang Z, Chen L, Yang R, Li X, Zeng W. Coronavirus disease 2019: initial chest CT findings. Eur Radiol. 2020;30(8):4398–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guo L, Xu R, Gou L, Liu Z, Zhao Y, Liu D, Zhang L, Chen H, Kong MG. Mechanism of virus inactivation by cold atmospheric-pressure plasma and plasma-activated water. Appl Environ Microb. 2018;84(17).

  33. Wang P, Zhou R, Zhou R, Li W, Weerasinghe J, Chen S, Rehm BHA, Zhao L, Frentiu FD, Zhang Z, et al. Cold atmospheric plasma for preventing infection of viruses that use ACE2 for entry. Theranostics. 2022;12(6):2811–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Khanikar RR, Kalita M, Kalita P, Kashyap B, Das S, Khan MR, Bailung H, Sankaranarayanan K. Cold atmospheric pressure plasma for attenuation of SARS-CoV-2 spike protein binding to ACE2 protein and the RNA deactivation. Rsc Adv. 2022;12(15):9466–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Luo W, Yu H, Gou J, Li X, Sun Y, Li J, He J, Liu L. Histopathologic findings in the explant lungs of a patient with COVID-19 treated with bilateral orthotopic lung transplant. Transplantation. 2020;104(11):e329–31.

    Article  CAS  PubMed  Google Scholar 

  36. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respiratory Med. 2020;8(4):420–2.

    Article  CAS  Google Scholar 

  37. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, Cheng L, Li J, Wang X, Wang F, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. 2020;26(6):842–4.

    Article  CAS  PubMed  Google Scholar 

  38. Amini MR, Sheikh Hosseini M, Fatollah S, Mirpour S, Ghoranneviss M, Larijani B, Mohajeri-Tehrani MR, Khorramizadeh MR. Beneficial effects of cold atmospheric plasma on inflammatory phase of diabetic foot ulcers; a randomized clinical trial. J Diabetes Metabolic Disorders. 2020;19(2):895–905.

    Article  CAS  Google Scholar 

  39. Lee YS, Lee M, Kim H, Won H, Kim C. Non-thermal atmospheric plasma ameliorates imiquimod-induced psoriasis-like skin inflammation in mice through inhibition of immune responses and up-regulation of PD-L1 expression. Sci Rep-UK. 2017;7(1):15564.

    Article  Google Scholar 

  40. Seebauer C, Freund E, Hasse S, Miller V, Segebarth M, Lucas C, Kindler S, Dieke T, Metelmann HR, Daeschlein G, et al. Effects of cold physical plasma on oral lichen planus: an in vitro study (effects of CAP on OLP). Oral Dis. 2021;27(7):1728–37.

    Article  PubMed  Google Scholar 

  41. Zhai S, Xu M, Li Q, Guo K, Chen H, Kong MG, Xia Y. Successful treatment of vitiligo with cold atmospheric plasma–activated hydrogel. J Invest Dermatol. 2021;141(11):2710–9.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang W, Chua B, Selva K, Kedzierski L, Ashhurst T, Haycroft E, Shoffner S, Hensen L, Boyd D, James F et al. Immune responses in COVID-19 respiratory tract and blood reveal mechanisms of disease severity. Res Sq. 2021.

  43. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, Liu L, Shan H, Lei C, Hui DSC, et al. Clinical characteristics of Coronavirus disease 2019 in China. New Engl J Med. 2020;382(18):1708–20.

    Article  CAS  PubMed  Google Scholar 

  44. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Braman SS. Postinfectious cough. Chest. 2006;129(1):S138–46.

    Article  Google Scholar 

  46. Song W, Hui CKM, Hull JH, Birring SS, McGarvey L, Mazzone SB, Chung KF. Confronting COVID-19-associated cough and the post-COVID syndrome: role of viral neurotropism, neuroinflammation, and neuroimmune responses. Lancet Respiratory Med. 2021;9(5):533–44.

    Article  CAS  Google Scholar 

  47. Tamasauskiene L, Sitkauskiene B. Immune system in the pathogenesis of chronic cough. Immunol Lett. 2020;218:40–3.

    Article  CAS  PubMed  Google Scholar 

  48. Lee M, Lee YS, Kim HJ, Han CH, Kang SU, Kim C. Non-thermal plasma inhibits mast cell activation and ameliorates allergic skin inflammatory diseases in NC/Nga mice. Sci Rep-UK. 2019;9(1).

  49. Choi J, Song Y, Lee H, Hong J, Kim G. Inhibition of inflammatory reactions in 2,4-dinitrochlorobenzene induced Nc/Nga atopic dermatitis mice by non-thermal plasma. Sci Rep-UK. 2016;6(1).

Download references

Acknowledgements

We thank our hospital colleagues for their help in recruiting patients.

Funding

This work was supported by the Key projects of basic research of natural science in Shaanxi Province (No. 2022JZ-58), the Discipline Promotion Plan of Xijing Hospital (No. XJZT21CZ06), the Clinical Research Program of Air Force Military Medical University (No. 2021JSTS36), the Shaanxi Provincial Health Scientific Research Fund Project (No. 2021A006).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, L.S. and D.L.; Methodology, L.S., X.L. and H.Z.; Software, C.S. and H.Z.; Validation, L.S., H.Z. and W.M.; Formal Analysis, L.S. and H.Z.; Investigation, X.L. and L.S.; Resources, D.L., Z.W., L.G., M.R., H.Z., W.M., J.L. and C.S.; Data Curation, H.Z and W.M.; Writing – Original Draft Preparation, H.Z. and W.M.; Writing – Review & Editing, L.S., D.L. and X.L.; Visualization, H.Z.; Supervision, J.L., X.G. and Z.C.; Project Administration, L.S. and X.L.; Funding Acquisition, L.S. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Dingxin Liu or Liqiang Song.

Ethics declarations

Ethics approval and consent to participate

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Medical Ethics Committee of the First Affiliated Hospital of the Air Force Medical University (Approval No.: KY20232004-C-1, January 5, 2023), registered with the Chinese Clinical Trial Registry (Registration No.: ChiCTR2300078706). Informed consent was obtained from all subjects involved in the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

: COVID-19 classification criteria and high-risk factors for severe disease. Table S1 Detailed qualitative assessment of chest CT on day 5

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, H., Meng, W., Lv, X. et al. Nebulized inhalation of plasma-activated water in the treatment of progressive moderate COVID-19 patients with antiviral treatment failure: a randomized controlled pilot trial. BMC Infect Dis 24, 960 (2024). https://doi.org/10.1186/s12879-024-09886-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-024-09886-w

Keywords