Next Article in Journal
A Proposed Method of Converting Gait Speed and TUG Test in Older Subjects
Previous Article in Journal
How Stress Is Related to Age, Education, Physical Activity, Body Mass Index, and Body Fat Percentage in Adult Polish Men?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Efficacy of Mouth Rinses and Nasal Spray in the Inactivation of SARS-CoV-2: A Systematic Review and Meta-Analysis of In Vitro and In Vivo Studies

1
UWA Dental School, The University of Western Australia, Nedlands, WA 6009, Australia
2
Private Dental Practice, Darwin, NT 0810, Australia
3
College of Medicine, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2022, 19(19), 12148; https://doi.org/10.3390/ijerph191912148
Submission received: 16 August 2022 / Revised: 19 September 2022 / Accepted: 20 September 2022 / Published: 25 September 2022
(This article belongs to the Section Oral Health)

Abstract

:
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) is a global and evolving pandemic associated with heavy health and financial burdens. Considering the oral cavity as the major reservoir for SARS-CoV-2, a systematic review and meta-analysis were conducted to assess the efficacy of mouth rinses and nasal sprays in reducing the salivary viral load of SARS-CoV-2. All in vivo and in vitro studies that assessed the virucidal efficacy of mouth rinses and nasal sprays against SARS-CoV-2 and were published in the English language from December 2019 to April 2022 were considered for analyses. Special Medical Subject Headings terms were used to search Pubmed, Scopus, Embase Ovid, and Web of Science databases. The toxicological data reliability assessment tool (ToxRToool) was used to assess the quality of the included studies. Thirty-three studies (11 in vivo and 22 in vitro) were deemed eligible for inclusion in this analysis. Results of the pooled data showed that povidone-iodine is the most efficacious intervention in vivo in terms of reducing the SARS-CoV-2 salivary viral load, followed by chlorhexidine. The mean difference in the viral load was 86% and 72%, respectively. Similarly, povidone-iodine was associated with the highest log10 reduction value (LRV) in vitro, followed by cetylpyridinium chloride, (LRV = 2.938 (p < 0.0005) and LRV = 2.907 (p = 0.009), respectively). Povidone-iodine-based oral and nasal preparations showed favourable results in terms of reducing SARS-CoV-2 viral loads both in vivo and in vitro. Considering the limited number of patients in vivo, further studies among larger cohorts are recommended.

1. Introduction

The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), instigated a global health emergency since its discovery in Wuhan, Hubei Provence, China, in 2019 [1]. This virus is a betacoronavirus associated with symptoms ranging from mild respiratory symptoms to severe pneumonia in the lungs, requiring supplementary oxygen or ventilation [1].
The main modes of transmission include human-to-human transmission through droplets, contact with an infected individual, or direct or indirect contact with contaminated surfaces [2]. SARS-CoV-2 can also be transmitted via aerosols, which are defined as air-borne suspended particles with the potential to contain salivary components and microorganisms [3]. Studies have demonstrated a high viral load in the saliva and oropharynx of symptomatic and asymptomatic patients with COVID-19 [4].
Considering that the oral cavity represents the major reservoir for SARS-CoV-2 [4], dental professionals are at high risk of exposure to pathogens through exposure to secretions, inhalation during aerosol-generating procedures, and mucosal contact with infected particles [5]. SARS-CoV-2 has demonstrated the potential for aerosolization for up to 3–16 h in vitro studies [6,7]. Thus, many dental regulatory authorities released recommendations to support using pre-procedural mouth rinse in dental settings to reduce SARS-CoV-2 transmission [8,9]. The premise for this is based on the principle of reducing oral microbial loads and mitigating the potential transmission of microbes via aerosol.
In this respect, several active compounds against bacteria and viruses in the oral cavity are currently being assessed for their potential virucidal efficacy against SARS-CoV-2. Studies have been conducted predominately in vitro and increasingly in vivo on the efficacy and efficiency of various active compounds in mouthwashes to reduce viral loads and, thus, the transmission of COVID-19. Several previous reviews attempted to address this topic; however, due to incomplete ongoing studies, the results were inconclusive [10,11,12,13].
This study aims to assess the evidence from studies that utilized mouth rinses or nasal sprays to reduce the salivary viral load of SARS-CoV-2 and provide evidence-based recommendations that can be employed by decision makers and regulatory authorities to aid in limiting the spread of this infection in the community.

2. Materials and Methods

2.1. Protocol and Registration

The proposed systematic review and meta-analysis were registered on the International Prospective Register of Systematic Reviews (PROSPERO) platform (CRD42022323586) and performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) [14]. PRISMA checklists are shown in Supplementary Tables S1 and S2.

2.2. Focused Questions

This review was designed to address the following questions: (a) What is the efficacy of various mouth rinse ingredients in reducing the viral load of SARS-CoV-2 in vitro? (b) Are mouth rinses and nasal sprays clinically efficacious in reducing the viral load of SARS-CoV-2? (c) What is the most efficacious mouth rinse for reducing the viral load of SARS-CoV-2 in the community?

2.3. Eligibility Criteria

Inclusion criteria: Observational studies, both in vivo and in vitro and published in the English language from December 2019 to April 2022 to assess the virucidal efficacy of mouth rinses and/or nasal sprays against SARS-CoV-2, were assessed for inclusion.
In vivo studies were included if they (1) recruited subjects with confirmed positive SARS-CoV-2 as per the standard quantitative polymerase chain reaction (qPCR) assays; (2) reported individual subject-level reports of SARS-CoV-2 viral load at two time frames: basal level (viral load just before using a mouth rinse or nasal spray) and experimental level (viral load after 1 to 3 h post using a mouth rinse or nasal spray); and (3) reported values of viral load as either copies/mL or cycle threshold values of PCR assay.
In vitro studies were included if they: (1) assessed the virucidal efficacy of preparations against a strain of SARS-CoV-2; (2) are studies that reported the Log10 reduction value (LRV) between control and experimental groups; (3) are studies that followed the European standards for chemical disinfectants and antiseptics (EN 14476:2013+A2:2019) [15].
Exclusion criteria: Letters to the editor, author comments, systematic reviews, and books or book chapters were excluded. In vivo studies that used assays other than qPCR to confirm SARS-CoV-2 positivity were excluded. In vitro studies that used virus surrogates instead of SARS-CoV-2 or did not provide detailed methodological designs were excluded.

2.4. Search Strategy and Data Extraction

Eligible studies were identified by using a developed search strategy for the following databases: MEDLINE by PubMed, Scopus, Embase Ovid, and Web of Science (Supplementary Table S3). Titles, abstracts, and keywords of retrieved studies were first screened blindly by two reviewers (MI and OK) to assess their relevance. Subsequently, studies that were considered potentially eligible were assessed by the reviewers by reading the entire text against the pre-defined inclusion and exclusion criteria. Only studies that were eligible for inclusion as agreed by the two reviewers were included. Any discrepancy in the assessment was resolved by discussion and consensus.
Two authors (MI and OK) extracted relevant data from the included studies. For in vivo studies, the following details were collected: (1) authors, country of the experiments, and year of publication; (2) active ingredients, concentrations, and route of administration; (3) number of participants; and (4) viral loads vs. time plots in log10. Where possible, numerical values were extracted from tables directly. Viral load values that were reported in figures or plots were extracted using a special online tool, WebPlotDigitizer, accessed from https://automeris.io/WebPlotDigitizer/ (accessed on 15 April 2022). Values that were reported as copies/mL or copies/µL were converted to log10 copies/µL. Values that were reported as cycle threshold of PCR assays were converted to log10 using the proposed model of Gentilini et al. to convert cycle threshold values to log10 copies/µL values [16].
For the in vitro studies, the following details were extracted: (1) authors and year of publication; (2) active ingredients and concentrations; (3) the strain of SARS-CoV-2 and the used cell line for propagation; (4) number of replicates and p values; and (4) the log10 reduction value (LRV) between control and experimental groups.

2.5. Risk of Bias Assessment

The quality of the included studies was assessed using the toxicological data reliability assessment tool (ToxRToool) [17]. ToxRTool includes a specific part for each in vivo and in vitro study. Slight modifications were made to meet the nature of the included studies. The included in vivo and in vitro studies were assessed against 15 and 17 items, respectively (Supplementary Tables S7 and S8). Any study that scored 12 or more was considered “reliable without restrictions” according to the systematic approach of Klimisch et al. for evaluating the quality of experimental toxicological and ecotoxicological data [18].

2.6. Data Analysis

The pooled difference in the mean and the 95% confidence interval (CI) of viral load for in vivo studies and the difference in the mean of LRV for in vitro studies were calculated under the random-effects model of the DerSimonian and Laird method. The extent of heterogeneity was measured according to the Higgins method and reported as I2. The value of I2 was interpreted as low (0–40%), moderate (30% to 60%), substantial (50% to 90%), and considerable (75% to 100%). A p-value of <0.05 was considered statistically significant. Review Manager 5.3 (Copenhagen, Denmark) and Comprehensive Meta-Analysis 3.3.070 (Englewood, NJ, USA) were used for meta-analysis purposes.

3. Results

3.1. Results of Database Searches

Three hundred and eighty-three studies were retrieved for abstract screening out of one thousand two hundred and twenty-two studies that were initially identified via the search platforms. Of these, 85 studies were assessed by full-text reading, whereby 52 studies were deemed ineligible. Of the included 33 studies, there were 11 in vivo studies and 22 in vitro studies (Figure 1). Descriptions of the included studies are shown in Supplementary Table S4 for in vivo studies and Supplementary Table S5 for in vitro studies. The excluded studies and reasons for exclusion are shown in Supplementary Table S6.

3.2. General Characteristics of the Included Studies

3.2.1. In Vivo Studies

Eleven in vivo studies assessed the virucidal efficacies of various preparations among 339 SARS-CoV-2 positive patients [19,20,21,22,23,24,25,26,27,28,29]. Five studies [21,22,24,26,29] conducted several experiments using various preparations, which totaled up to 21 experiments. Only one study assessed the efficacy of nasal sprays in reducing viral load [29] while others used mouth rinses for this purpose.
Chlorhexidine mouth rinse was assessed in six experiments among 105 patients [20,21,22,24,26,28]. Two experiments assessed chlorhexidine combined with hydrogen peroxide in 23 patients [21,24], while three experiments assessed hydrogen peroxide alone among 28 patients [23,24]. Povidone-iodine was used in five experiments among 55 patients [21,22,25,26,29], and only two experiments utilized cetylpyridinium-chloride mouth rinse in 11 patients [21,26]. Finally, three different preparations were assessed once in three different experiments. These preparations are (1) b-cyclodextrinecitrox mouthwash (CDCM) (76 patients) [19], (2) Linola Sept, Dr August Wolff mouthwash (Linola Sept) (29 patients) [27], and (3) sodium hypochlorite (NaCIO) (12 patients) [24].
Regarding the risk of bias assessment of the in vivo studies, all studies were considered reliable without restrictions. However, none of them scored 15 out of 15 as the strain of SARS-CoV-2 was not mentioned in any study (Supplementary Table S7). Seven studies were found to have an overall score of 13, while four studies had a score of 12.

3.2.2. In Vitro Studies

In total, the included in vitro studies (22 studies) conducted 79 experiments on different test products at different timescales [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. The contact time ranged from 15 s to 10 min. Povidone-iodine was the most widely assessed preparation (32 experiments), followed by chlorhexidine and sodium fluoride/chloride in 11 and 9 experiments, respectively (Supplementary Table S5).
In terms of the risk of bias assessment, all studies were reliable for inclusion without restrictions. Four studies had a score of 17 out of 17. Sixteen studies showed an overall score of either 16 or 15. Only one study had a score of 14 (Supplementary Table S8).
The strain of the assessed virus has varied according to the country of origin. The most widely assessed strain in 11 studies was USA-WA1/2020 [30,31,32,33,34,35,36,37,38,39,40]. The other assessed strains were hCoV19/Singapore/2/2020 [41], England 2 strain [42], SARS-CoV-2/MY/UM/6-3 [43], JPN/TY/WK-521 [44,45], FI-100 strain [46], hCoV-19/ Germany/BY-Bochum-1/2020 [47], and isolate 026 V-03883 [48]. Oropharyngeal swabs from COVID-19-infected patients were used to isolate the virus in two studies [49,50]. All studies propagated the virus into Vero cells (lineages Vero 76 and Vero E6).

3.3. Meta-Analysis for the Virucidal Efficacy of Different Preparations against SARS-CoV-2 In Vivo

Pooled data revealed that using a mouth rinse or nasal spray in vivo significantly reduced the salivary viral load for a period ranging from 5 min to 3 h by 67% (CI 95% −0.86, −0.47) (p < 0.0001) (Figure 2). The level of heterogeneity among the experiments was low (I2 = 30%); however, this was not statistically significant (p = 0.10) (Figure 2).
For sub-group analysis, pooled data showed that povidone-iodine was associated with the highest virucidal efficacy in comparison to other preparations, with a significant mean difference in the viral load (−0.86 [CI 95% −1.5, −0.23], p = 0.008). The second most efficacious preparation in vivo was chlorhexidine, which showed a statistically significant reduction in the viral load (−0.72 [CI 95% −1.09, −0.36], p = 0.0001) (Figure 2). However, further subgroup analyses for povidone-iodine and chlorhexidine based on their concentrations were not possible due to the limited number of patients in certain concentrations.
Using cetylpyridinium-chloride and hydrogen peroxide preparations was not associated with a significant reduction in viral load (p = 0.4 and p = 0.26, respectively) (Figure 2).

3.4. Meta-Analysis for the Virucidal Efficacy of Different Preparations against SARS-CoV-2 In Vitro

Pooled data for the entire set of 79 in vitro experiments showed an LRV of 0.886 (p < 0.0005) (Figure 3). However, there was substantial heterogeneity among the experiments (I2 = 69.192, p < 0.0005) (Supplementary Figure S1).
After applying subgroup analyses, the most efficacious intervention was povidone-iodine, regardless of its concentration (LRV = 2.938, p < 0.0005) (Figure 4A). Additional subgroup analysis among povidone-iodine experiments showed that the LRV increased to 3.836 (p < 0.0005) when using povidone-iodine in concentrations of more than 1% (Figure 4B).
The second most efficacious intervention in terms of in vitro virucidal activity was cetylpyridinium-chloride with an LRV of 2.907 (p = 0.009) (Figure 5A). The third most efficacious intervention was Listerine® and other essential oils (LRV = 2.244, p = 0.001) (Figure 5B).
A statistically significant difference in LRV was reported for hydrogen peroxide (LRV = 0.969, p = 0.033) (Figure 5C). Eleven experiments utilizing chlorhexidine and their related pool data revealed an LRV of 0.695 (p < 0.0005) (Figure 5D). Finally, the least efficacious intervention was related to preparations containing sodium fluoride or sodium chloride (LRV = 0.539, p = 0.003) (Figure 5E).

4. Discussion

Since the emergence of the pandemic, viral loads of SARS-CoV-2 have varied greatly among patients and played a significant role in infectivity and fatality. Although many occupational-related organizations recommended using mouth rinses to reduce the SARS-CoV-2 viral load to levels that could be non-infectious [8,9], these recommendations were anecdotal, speculative, and not evidence-based. This systematic review presents an evaluation of the available evidence about the efficacy of mouth rinses and nasal sprays in decreasing the SARS-CoV-2 viral load to potentially prevent the spread of COVID-19 in the community. To the best of our knowledge, this work represents the first meta-analysis for both in vivo and in vitro studies in this area.
SARS-CoV-2 infection is usually diagnosed qualitatively as positive or negative, although the gold standard for its diagnosis, the quantitative polymerase chain reaction, was originally intended to be quantitative. This greatly hampered the understanding of the viral load’s dynamics and may explain why some individuals are infectious without symptoms, while similarly infectious individuals do not transmit the infection to their intimate partner and household members.
In this review, the majority of in vivo studies reported their viral load outcomes as cycle threshold values for qPCR. However, neither methods of cycle threshold conversion to viral load nor measured errors were provided [16]. Such inconsistencies in reports complicate comparisons between studies. To overcome this obstacle, this review used the proposed model of Gentilini et al. in 2021 to convert cycle threshold values to SARS-CoV-2 viral load [16]. This model was validated on more than 6200 COVID-19-positive patients and showed a reliability of almost 92% [16]. The results of this review found that in vitro studies followed the European standards for chemical disinfectants and antiseptics (EN14476:2013+A2:2019) [15]. According to these standards, a substance can be considered virucidal efficacious if it reduces the virus titre by at least four decimal logarithms (LRV ≥ 4 log10) [15]. However, the analysis of the pooled data showed that all preparations were associated with LRV less than 4 log10. This may highlight the potential limitations of in vitro studies. SARS-CoV-2 primarily targets the human lung epithelium; nonetheless, the most widely used cell line in SARS-CoV-2-related projects is the Vero cell line (kidney of an African green monkey). This is because Vero cells express high levels of ACE 2, which is the cellular receptor for SARS-CoV-2 entry [52]. Noteworthily, the members of the World Health Organization (WHO) working on the SARS-CoV-2 virus analysed genetic sequencing data and found that the virus propagation in Vero cells causes genetic variants that may impact the interpretation of results from animal and clinical trials [53]. This may, to some extent, explain the differences between in vivo and in vitro results for some preparations.
One of the most widely studied preparations is povidone-iodine. Povidone-iodine is a broad-spectrum antimicrobial compound of a potent bactericidal agent, iodine, which is within the carrier molecule, povidone [41]. The results of this meta-analysis revealed that povidone-iodine is the most efficacious virucidal preparation against SARS-CoV-2 in both in vivo and in vitro studies. The mean difference in viral load was found to be up to 86% for up to 3 h post rinsing with povidone-iodine. While its efficacy increases with concentrations of more than 1%, the time for oral rinses ranges between 1 and 2 min. Considering that povidone-iodine can safely be used in the oral cavity at concentrations up to 2.5% for up to 5 months [22], this indicates the potential efficaciousness of povidone-iodine in controlling the spread of SARS-CoV-2.
Chlorhexidine is a cationic surfactant and synthetic biguanide with broad-spectrum antimicrobial activity [11]. Although it is well documented that chlorhexidine mouthwash is effective against herpes, influenza, parainfluenza, and hepatitis B [11], the available evidence about its efficacy against SARS-CoV-2 is limited. Our analysis of the in vivo studies demonstrated that chlorhexidine is the second most efficacious intervention against SARS-CoV-2 as it lowered the mean oral viral load by 72% (p = 0.0001). On the contrary, in vitro experiments showed a very limited therapeutic utility for chlorhexidine in reducing the viral load. This can be attributed to the fact that chlorhexidine provides a long-lasting effect in vivo due to its ability to retain on oral cavity surfaces for several hours because of its cationic feature, whereas it cannot possess the same antiseptic effect with a short contact time in vitro [11].
Hydrogen peroxide is an antiseptic solution that exerts its microbicidal action by producing hydroxyl free radicals that can attack membrane lipids and other essential cell components of pathogens. [24,30]. It has been suggested that using 1% hydrogen peroxide would be more appropriate for reducing the salivary load of SARS-CoV-2 as the virus is vulnerable to oxidation in the oral environment. However, the results of this review revealed that hydrogen peroxide oral rinse is not superior to other preparations in reducing the salivary load of SARS-CoV-2, both in vivo and in vitro (35% and LRV = 0.969, respectively).
Cetylpyridinium-chloride is a quaternary ammonium compound that exerts its antiviral effect through a physiochemical disruption of the viral lipid envelope, which is the same as the membrane surrounding SARS-CoV-2 [30]. Although our results revealed that cetylpyridinium-chloride did not show promising results in vivo, it was the second most efficacious preparation after povidone-iodine in vitro (LRV = 2.907). However, as the number of patients in vivo was limited (11 patients), a further validation of cetylpyridinium-chloride in larger cohorts is required to provide reliable recommendations.
The major limitation of this study was the limited number of patients for in vivo studies. This can be attributed to strict ethical requirements for in vivo studies in some countries. The second major limitation is the difference in the number of studies and participants among in vivo studies for some active ingredients, which may make comparisons between the efficacy of these ingredients non-conclusive. However, after more than two years of this pandemic, we anticipate the easing of mandatory restrictions, which will translate to increased quality and quantity of clinical studies. Moreover, the absence of standardized protocols for sample collection and result reporting among in vivo studies accounts for heterogeneity among the relevant studies. Finally, all in vitro studies utilised Vero cells for the virus propagation; however, based on the recent WHO report, there is sufficient reason to question the reliability of Vero cells in these experiments [53].

5. Conclusions

In conclusion, povidone-iodine and chlorhexidine mouth rinses, regardless of concentration, were clinically the most efficacious interventions for reducing the SARS-CoV-2 oral viral load. Emerging evidence from in vivo studies using hydrogen peroxide, cetylpyridinium-chloride, and various other active ingredients remains inconclusive. Despite povidone-iodine and chlorhexidine mouth rinses demonstrating favourable efficacy, their effectiveness in terms of virucidal activity does not currently meet recommended standards of the European Standards for chemical disinfectants and antiseptics (EN 14476). Given these results, governing organisations should revisit their COVID-19 pandemic guidelines and consider recommending specific preparations of mouth rinses (1–5% povidone-iodine or 0.12–0.2% chlorhexidine) for individuals infected by SARS-CoV-2 or at a high risk of being infected. However, prescribers should be aware of the side effects of these mouthwashes when they plan to use them routinely. Likewise, consideration for routine use of mouth rinses by asymptomatic or uninfected individuals during high community transmission may reduce the burden on strained health care systems. Nonetheless, new mouthwashes with little to no side effects that can significantly reduce the SARS-CoV-2 oral viral load with remarkable virucidal impact are warranted.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijerph191912148/s1, Figure S1: Funnel plot of the standard error by the difference in means for in vitro studies; Table S1: PRISMA 2020 main checklist; Table S2: PRIMSA 2020 Abstract Checklist; Table S3: MeSH terms used for searching through Pubmed, Scopus, Embase Ovid, and Web of Science databases; Table S4: General descriptions of the included in vivo studies and their primary findings; Table S5: General descriptions of the included in vitro studies and their primary findings (all preparations are mouth rinse unless otherwise specified); Table S6: Excluded studies and reasons of exclusion; Table S7: Risk of bias assessment of in vivo studies; Table S8: Risk of bias assessment of in vitro studies. References [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105] are cited in the supplementary materials.

Author Contributions

Conceptualization, R.B. and O.K.; methodology, M.I., B.M. and O.K.; validation, M.I. and O.K.; formal analysis, M.I.; investigation, M.I., B.M., A.F., A.A.A., R.B. and O.K.; data curation, M.I., B.M., A.A.A., R.B. and O.K.; writing—original draft preparation, M.I. and B.M.; writing—review and editing, M.I. and O.K.; project administration, O.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was registered on the International Prospective Register of Systematic Reviews (PROSPERO) platform (CRD42022323586) and performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and supplementary materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yuki, K.; Fujiogi, M.; Koutsogiannaki, S. COVID-19 pathophysiology: A review. Clin. Immunol. 2020, 215, 108427. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, M.Y.; Zhao, R.; Gao, L.J.; Gao, X.F.; Wang, D.P.; Cao, J.M. SARS-CoV-2: Structure, Biology, and Structure-Based Therapeutics Development. Front. Cell. Infect. Microbiol. 2020, 10, 587269. [Google Scholar] [CrossRef] [PubMed]
  3. Harrel, S.K.; Molinari, J. Aerosols and splatter in dentistry: A brief review of the literature and infection control implications. J. Am. Dent. Assoc. 2004, 135, 429–437. [Google Scholar] [CrossRef] [PubMed]
  4. Hasanoglu, I.; Korukluoglu, G.; Asilturk, D.; Cosgun, Y.; Kalem, A.K.; Altas, A.B.; Kayaaslan, B.; Eser, F.; Kuzucu, E.A.; Guner, R. Higher viral loads in asymptomatic COVID-19 patients might be the invisible part of the iceberg. Infection 2021, 49, 117–126. [Google Scholar] [CrossRef]
  5. Fallahi, H.R.; Keyhan, S.O.; Zandian, D.; Kim, S.G.; Cheshmi, B. Being a front-line dentist during the COVID-19 pandemic: A literature review. Maxillofac. Plast. Reconstr. Surg. 2020, 42, 12. [Google Scholar] [CrossRef]
  6. van Doremalen, N.; Bushmaker, T.; Morris, D.H.; Holbrook, M.G.; Gamble, A.; Williamson, B.N.; Tamin, A.; Harcourt, J.L.; Thornburg, N.J.; Gerber, S.I.; et al. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N. Engl. J. Med. 2020, 382, 1564–1567. [Google Scholar] [CrossRef]
  7. Fears, A.C.; Klimstra, W.B.; Duprex, P.; Hartman, A.; Weaver, S.C.; Plante, K.S.; Mirchandani, D.; Plante, J.A.; Aguilar, P.V.; Fernandez, D.; et al. Persistence of Severe Acute Respiratory Syndrome Coronavirus 2 in Aerosol Suspensions. Emerg. Infect. Dis. 2020, 26, 2168–2171. [Google Scholar] [CrossRef]
  8. Australian Dental Association: Risk Management Principles for Dentistry during the COVID-19 Pandemic. Available online: https://www.ada.org.au/getdoc/d3eecaba-d0aa-4803-a7ea-89facae6f274/Risk-Management-Principles-for-Dentistry-(1).aspx (accessed on 10 April 2022).
  9. American Dental Association: ADA Interim Guidance for Minimizing Risk of COVID-19 Transmission. Available online: https://snlg.iss.it/wp-content/uploads/2020/04/ADA_COVID_Int_Guidance_Treat_Pts.pdf (accessed on 10 April 2022).
  10. Burton, M.J.; Clarkson, J.E.; Goulao, B.; Glenny, A.M.; McBain, A.J.; Schilder, A.G.; Webster, K.E.; Worthington, H.V. Antimicrobial mouthwashes (gargling) and nasal sprays to protect healthcare workers when undertaking aerosol-generating procedures (AGPs) on patients without suspected or confirmed COVID-19 infection. Cochrane Database Syst. Rev. 2020, 9, CD013628. [Google Scholar]
  11. Amber, A.; Abhishek, P.; Nikita, R. Efficacy of Mouth Rinses against SARS-CoV-2: A Scoping Review. Front. Dent. Med. 2021, 2, 648547. [Google Scholar]
  12. Cavalcante-Leao, B.L.; de Araujo, C.M.; Basso, I.B.; Schroder, A.G.; Guariza-Filho, O.; Ravazzi, G.C.; Goncalves, F.M.; Zeigelboim, B.S.; Santos, R.S.; Stechman-Neto, J. Is there scientific evidence of the mouthwashes effectiveness in reducing viral load in COVID-19? A systematic review. J. Clin. Exp. Dent. 2021, 13, e179–e189. [Google Scholar] [CrossRef]
  13. Garcia-Sanchez, A.; Pena-Cardelles, J.F.; Ruiz, S.; Robles, F.; Ordonez-Fernandez, E.; Salgado-Peralvo, A.O.; Balloch, J.; Simon, J.C. Efficacy of Pre-Procedural Mouthwashes against SARS-CoV-2: A Systematic Review of Randomized Controlled Trials. J. Clin. Med. 2022, 11, 1692. [Google Scholar] [CrossRef] [PubMed]
  14. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  15. European Standard: EN14476:2013+A1:2015, Chemical Disinfectants and Antiseptics—Quantitative Suspension Test for the Evaluation of Virucidal Activity in the Medical Area—Test Method and Requirements (Phase 2/Step 1). Available online: https://standards.iteh.ai/catalog/standards/cen/5e78911a-aedf-4456-90b7-39e1649f8acf/en-14476-2013a1-2015 (accessed on 12 April 2022).
  16. Gentilini, F.; Turba, M.E.; Taddei, F.; Gritti, T.; Fantini, M.; Dirani, G.; Sambri, V. Modelling RT-qPCR cycle-threshold using digital PCR data for implementing SARS-CoV-2 viral load studies. PLoS ONE 2021, 16, e0260884. [Google Scholar] [CrossRef]
  17. Schneider, K.; Schwarz, M.; Burkholder, I.; Kopp-Schneider, A.; Edler, L.; Kinsner-Ovaskainen, A.; Hartung, T.; Hoffmann, S. “ToxRTool”, a new tool to assess the reliability of toxicological data. Toxicol. Lett. 2009, 189, 138–144. [Google Scholar] [CrossRef] [PubMed]
  18. Klimisch, H.J.; Andreae, M.; Tillmann, U. A systematic approach for evaluating the quality of experimental toxicological and ecotoxicological data. Regul. Toxicol. Pharmacol. 1997, 25, 1–5. [Google Scholar] [CrossRef] [PubMed]
  19. Carrouel, F.; Valette, M.; Gadea, E.; Esparcieux, A.; Illes, G.; Langlois, M.E.; Perrier, H.; Dussart, C.; Tramini, P.; Ribaud, M.; et al. Use of an antiviral mouthwash as a barrier measure in the SARS-CoV-2 transmission in adults with asymptomatic to mild COVID-19: A multicentre, randomized, double-blind controlled trial. Clin. Microbiol. Infect. 2021, 27, 1494–1501. [Google Scholar] [CrossRef]
  20. Costa, D.D.; Brites, C.; Vaz, S.N.; de Santana, D.S.; Dos Santos, J.N.; Cury, P.R. Chlorhexidine mouthwash reduces the salivary viral load of SARS-CoV-2: A randomized clinical trial. Oral Dis. 2021. [Google Scholar] [CrossRef]
  21. Eduardo, F.P.; Correa, L.; Heller, D.; Daep, C.A.; Benitez, C.; Malheiros, Z.; Stewart, B.; Ryan, M.; Machado, C.M.; Hamerschlak, N.; et al. Salivary SARS-CoV-2 load reduction with mouthwash use: A randomized pilot clinical trial. Heliyon 2021, 7, e07346. [Google Scholar] [CrossRef]
  22. Elzein, R.; Abdel-Sater, F.; Fakhreddine, S.; Hanna, P.A.; Feghali, R.; Hamad, H.; Ayoub, F. In vivo evaluation of the virucidal efficacy of chlorhexidine and povidone-iodine mouthwashes against salivary SARS-CoV-2. A randomized-controlled clinical trial. J. Evid. Based Dent. Pract. 2021, 21, 101584. [Google Scholar] [CrossRef]
  23. Gottsauner, M.J.; Michaelides, I.; Schmidt, B.; Scholz, K.J.; Buchalla, W.; Widbiller, M.; Hitzenbichler, F.; Ettl, T.; Reichert, T.E.; Bohr, C.; et al. A prospective clinical pilot study on the effects of a hydrogen peroxide mouthrinse on the intraoral viral load of SARS-CoV-2. Clin. Oral Investig. 2020, 24, 3707–3713. [Google Scholar] [CrossRef]
  24. Guimaraes, T.C.; Marques, B.B.F.; Castro, M.V.; Secco, D.A.; Porto, L.; Tinoco, J.M.M.; Tinoco, E.M.B.; Fletcher, P.; Fischer, R.G. Reducing the viral load of SARS-CoV-2 in the saliva of patients with COVID-19. Oral Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
  25. Martinez Lamas, L.; Diz Dios, P.; Perez Rodriguez, M.T.; Del Campo Perez, V.; Cabrera Alvargonzalez, J.J.; Lopez Dominguez, A.M.; Fernandez Feijoo, J.; Diniz Freitas, M.; Limeres Posse, J. Is povidone iodine mouthwash effective against SARS-CoV-2? First in vivo tests. Oral Dis. 2020, 28 (Suppl. 1), 908–911. [Google Scholar] [CrossRef] [PubMed]
  26. Seneviratne, C.J.; Balan, P.; Ko, K.K.K.; Udawatte, N.S.; Lai, D.; Ng, D.H.L.; Venkatachalam, I.; Lim, K.S.; Ling, M.L.; Oon, L.; et al. Efficacy of commercial mouth-rinses on SARS-CoV-2 viral load in saliva: Randomized control trial in Singapore. Infection 2021, 49, 305–311. [Google Scholar] [CrossRef] [PubMed]
  27. Schurmann, M.; Aljubeh, M.; Tiemann, C.; Sudhoff, H. Mouthrinses against SARS-CoV-2: Anti-inflammatory effectivity and a clinical pilot study. Eur. Arch. Otorhinolaryngol. 2021, 278, 5059–5067. [Google Scholar] [CrossRef]
  28. Yoon, J.G.; Yoon, J.; Song, J.Y.; Yoon, S.Y.; Lim, C.S.; Seong, H.; Noh, J.Y.; Cheong, H.J.; Kim, W.J. Clinical Significance of a High SARS-CoV-2 Viral Load in the Saliva. J. Korean Med. Sci. 2020, 35, e195. [Google Scholar] [CrossRef]
  29. Zarabanda, D.; Vukkadala, N.; Phillips, K.M.; Qian, Z.J.; Mfuh, K.O.; Hatter, M.J.; Lee, I.T.; Rao, V.K.; Hwang, P.H.; Domb, G.; et al. The Effect of Povidone-Iodine Nasal Spray on Nasopharyngeal SARS-CoV-2 Viral Load: A Randomized Control Trial. Laryngoscope 2021. [Google Scholar] [CrossRef]
  30. Anderson, E.R.; Patterson, E.I.; Richards, S.; Pitol, A.K.; Edwards, T.; Wooding, D.; Buist, K.; Green, A.; Mukherjee, S.; Hoptroff, M.; et al. CPC-containing oral rinses inactivate SARS-CoV-2 variants and are active in the presence of human saliva. J. Med. Microbiol. 2022, 71, 001508. [Google Scholar] [CrossRef]
  31. Bidra, A.S.; Pelletier, J.S.; Westover, J.B.; Frank, S.; Brown, S.M.; Tessema, B. Rapid In-Vitro Inactivation of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Using Povidone-Iodine Oral Antiseptic Rinse. J. Prosthodont. 2020, 29, 529–533. [Google Scholar] [CrossRef]
  32. Bidra, A.S.; Pelletier, J.S.; Westover, J.B.; Frank, S.; Brown, S.M.; Tessema, B. Comparison of In Vitro Inactivation of SARS CoV-2 with Hydrogen Peroxide and Povidone-Iodine Oral Antiseptic Rinses. J. Prosthodont. 2020, 29, 599–603. [Google Scholar] [CrossRef]
  33. Frank, S.; Brown, S.M.; Capriotti, J.A.; Westover, J.B.; Pelletier, J.S.; Tessema, B. In Vitro Efficacy of a Povidone-Iodine Nasal Antiseptic for Rapid Inactivation of SARS-CoV-2. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 1054–1058. [Google Scholar] [CrossRef]
  34. Gudmundsdottir, A.; Scheving, R.; Lindberg, F.; Stefansson, B. Inactivation of SARS-CoV-2 and HCoV-229E in vitro by ColdZyme(R) a medical device mouth spray against the common cold. J. Med. Virol. 2021, 93, 1792–1795. [Google Scholar] [CrossRef]
  35. Kontos, Z. Efficacy of “Essential Iodine Drops” against Severe Acute Respiratory Syndrome-Coronavirus 2 (SARS-CoV-2). PLoS ONE 2021, 16, e0254341. [Google Scholar] [CrossRef] [PubMed]
  36. Liang, B.; Yuan, X.; Wei, G.; Wang, W.; Zhang, M.; Peng, H.; Javer, A.; Mendenhall, M.; Julander, J.; Huang, S.; et al. In-Vivo Toxicity Studies and In-Vitro Inactivation of SARS-CoV-2 by Povidone-iodine In-situ Gel Forming Formulations. bioRxiv 2020. [Google Scholar] [CrossRef]
  37. Pelletier, J.S.; Tessema, B.; Frank, S.; Westover, J.B.; Brown, S.M.; Capriotti, J.A. Efficacy of Povidone-Iodine Nasal and Oral Antiseptic Preparations Against Severe Acute Respiratory Syndrome-Coronavirus 2 (SARS-CoV-2). Ear Nose Throat J. 2021, 100 (Suppl. 2), 192S–196S. [Google Scholar] [CrossRef]
  38. Shet, M.; Westover, J.; Hong, R.; Igo, D.; Cataldo, M.; Bhaskar, S. In vitro inactivation of SARS-CoV-2 using a povidone-iodine oral rinse. BMC Oral Health 2022, 22, 47. [Google Scholar] [CrossRef]
  39. Shewale, J.G.; Gelhaus, H.C.; Ratcliff, J.L.; Hernandez-Kapila, Y.L. In vitro antiviral activity of stabilized chlorine dioxide containing oral care products. Oral Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
  40. Teagle, V.; Clem, D.S.; Yoon, T. Virucidal Properties of Molecular Iodine Oral Rinse Against SARS-CoV-2. Compend. Contin. Educ. Dent. 2022, 43, e13–e16. [Google Scholar] [PubMed]
  41. Anderson, D.E.; Sivalingam, V.; Kang, A.E.Z.; Ananthanarayanan, A.; Arumugam, H.; Jenkins, T.M.; Hadjiat, Y.; Eggers, M. Povidone-Iodine Demonstrates Rapid In Vitro Virucidal Activity against SARS-CoV-2, the Virus Causing COVID-19 Disease. Infect. Dis. Ther. 2020, 9, 669–675. [Google Scholar] [CrossRef]
  42. Davies, K.; Buczkowski, H.; Welch, S.R.; Green, N.; Mawer, D.; Woodford, N.; Roberts, A.D.G.; Nixon, P.J.; Seymour, D.W.; Killip, M.J. Effective in vitro inactivation of SARS-CoV-2 by commercially available mouthwashes. J. Gen. Virol. 2021, 102, 001578. [Google Scholar] [CrossRef]
  43. Hassandarvish, P.; Tiong, V.; Mohamed, N.A.; Arumugam, H.; Ananthanarayanan, A.; Qasuri, M.; Hadjiat, Y.; Abubakar, S. In vitro virucidal activity of povidone iodine gargle and mouthwash against SARS-CoV-2: Implications for dental practice. Br. Dent. J. 2020. [Google Scholar] [CrossRef]
  44. Komine, A.; Yamaguchi, E.; Okamoto, N.; Yamamoto, K. Virucidal activity of oral care products against SARS-CoV-2 in vitro. J. Oral Maxillofac. Surg. Med. Pathol. 2021, 33, 475–477. [Google Scholar] [CrossRef] [PubMed]
  45. Kariwa, H.; Sawa, H.; Kobayashi, S. Inactivation of SARS-CoV-2 by povidone-iodine products: Implications for effective mouth rinsing and gargling. Jpn. J. Vet. Res. 2021, 69, 183–187. [Google Scholar]
  46. Koch-Heier, J.; Hoffmann, H.; Schindler, M.; Lussi, A.; Planz, O. Inactivation of SARS-CoV-2 through Treatment with the Mouth Rinsing Solutions ViruProX((R)) and BacterX((R)) Pro. Microorganisms 2021, 9, 521. [Google Scholar] [CrossRef] [PubMed]
  47. Meister, T.L.; Todt, D.; Bruggemann, Y.; Steinmann, J.; Banava, S.; Brill, F.H.H.; Steinmann, J.; Pfaender, S.; Steinmann, E. Virucidal activity of nasal sprays against severe acute respiratory syndrome coronavirus-2. J. Hosp. Infect. 2022, 120, 9–13. [Google Scholar] [CrossRef] [PubMed]
  48. Pyrc, K.; Milewska, A.; Duran, E.B.; Botwina, P.; Dabrowska, A.; Jedrysik, M.; Benedyk, M.; Lopes, R.; Arenas-Pinto, A.; Badr, M.; et al. SARS-CoV-2 inhibition using a mucoadhesive, amphiphilic chitosan that may serve as an anti-viral nasal spray. Sci. Rep. 2021, 11, 20012. [Google Scholar] [CrossRef]
  49. Santos, C.; da Fonseca Orcina, B.; Brito Reia, V.C.; Ribeiro, L.G.; Grotto, R.M.T.; Prudenciatti, A.; de Moraes, L.N.; Ragghianti Zangrando, M.; Vilhena, F.V.; da Silva Santos, P.S. Virucidal Activity of the Antiseptic Mouthwash and Dental Gel Containing Anionic Phthalocyanine Derivative: In vitro Study. Clin. Cosmet. Investig. Dent. 2021, 13, 269–274. [Google Scholar] [CrossRef]
  50. Tiong, V.; Hassandarvish, P.; Bakar, S.A.; Mohamed, N.A.; Wan Sulaiman, W.S.; Baharom, N.; Abdul Samad, F.N.; Isahak, I. The effectiveness of various gargle formulations and salt water against SARS-CoV-2. Sci. Rep. 2021, 11, 20502. [Google Scholar] [CrossRef]
  51. Steinhauer, K.; Meister, T.L.; Todt, D.; Krawczyk, A.; Passvogel, L.; Becker, B.; Paulmann, D.; Bischoff, B.; Pfaender, S.; Brill, F.H.H.; et al. Comparison of the in-vitro efficacy of different mouthwash solutions targeting SARS-CoV-2 based on the European Standard EN 14476. J. Hosp. Infect. 2021, 111, 180–183. [Google Scholar] [CrossRef]
  52. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280 e278. [Google Scholar] [CrossRef]
  53. Funnell, S.G.P.; Afrough, B.; Baczenas, J.J.; Berry, N.; Bewley, K.R.; Bradford, R.; Florence, C.; Duff, Y.L.; Lewis, M.; Moriarty, R.V.; et al. A cautionary perspective regarding the isolation and serial propagation of SARS-CoV-2 in Vero cells. NPJ Vaccines 2021, 6, 83. [Google Scholar] [CrossRef]
  54. Abdelalim, A.A.; Mohamady, A.A.; Elsayed, R.A.; Elawady, M.A.; Ghallab, A.F. Corticosteroid nasal spray for recovery of smell sensation in COVID-19 patients: A randomized controlled trial. Am. J. Otolaryngol. 2021, 42, 102884. [Google Scholar] [CrossRef] [PubMed]
  55. Chaudhary, P.; Melkonyan, A.; Meethil, A.; Saraswat, S.; Hall, D.L.; Cottle, J.; Wenzel, M.; Ayouty, N.; Bense, S.; Casanova, F.; et al. Estimating salivary carriage of severe acute respiratory syndrome coronavirus 2 in nonsymptomatic people and efficacy of mouthrinse in reducing viral load: A randomized controlled trial. J. Am. Dent. Assoc. 2021, 152, 903–908. [Google Scholar] [CrossRef] [PubMed]
  56. Burgos-Ramos, E.; Urbieta, I.R.; Rodriguez, D. Is hydrogen peroxide an effective mouthwash for reducing the viral load of SARS-CoV-2 in dental clinics? Saudi Dent. J. 2022, 34, 237–242. [Google Scholar] [CrossRef] [PubMed]
  57. da Fonseca Orcina, B.; Vilhena, F.V.; Cardoso de Oliveira, R.; Marques da Costa Alves, L.; Araki, K.; Toma, S.H.; Ragghianti Zangrando, M.S.; da Silva Santos, P.S. A Phthalocyanine Derivate Mouthwash to Gargling/Rinsing as an Option to Reduce Clinical Symptoms of COVID-19: Case Series. Clin. Cosmet. Investig. Dent. 2021, 13, 47–50. [Google Scholar] [CrossRef]
  58. Domenico, M.B.D.; Collares, K.; Santos, R.B.D.; Lenz, U.; Antunes, V.P.; Godinho, V.W.; Cesca, H.; Ponciano, T.H.J.; Corazza, P.H. Hydrogen peroxide as an auxiliary treatment for COVID-19 in Brazil: A randomized double-blind clinical trial. Epidemiol. Health 2021, 43, e2021051. [Google Scholar] [CrossRef] [PubMed]
  59. Figueroa, J.M.; Lombardo, M.E.; Dogliotti, A.; Flynn, L.P.; Giugliano, R.; Simonelli, G.; Valentini, R.; Ramos, A.; Romano, P.; Marcote, M.; et al. Efficacy of a Nasal Spray Containing Iota-Carrageenan in the Postexposure Prophylaxis of COVID-19 in Hospital Personnel Dedicated to Patients Care with COVID-19 Disease. Int. J. Gen. Med. 2021, 14, 6277–6286. [Google Scholar] [CrossRef]
  60. Guenezan, J.; Garcia, M.; Strasters, D.; Jousselin, C.; Leveque, N.; Frasca, D.; Mimoz, O. Povidone Iodine Mouthwash, Gargle, and Nasal Spray to Reduce Nasopharyngeal Viral Load in Patients With COVID-19: A Randomized Clinical Trial. JAMA Otolaryngol. Head Neck Surg. 2021, 147, 400–401. [Google Scholar] [CrossRef]
  61. Huang, Y.H.; Huang, J.T. Use of chlorhexidine to eradicate oropharyngeal SARS-CoV-2 in COVID-19 patients. J. Med. Virol. 2021, 93, 4370–4373. [Google Scholar] [CrossRef]
  62. Kasiri, H.; Rouhani, N.; Salehifar, E.; Ghazaeian, M.; Fallah, S. Mometasone furoate nasal spray in the treatment of patients with COVID-19 olfactory dysfunction: A randomized, double blind clinical trial. Int. Immunopharmacol. 2021, 98, 107871. [Google Scholar] [CrossRef]
  63. Arefin, M.K.; Rumi, S.; Uddin, A.; Banu, S.S.; Khan, M.; Kaiser, A.; Chowdhury, J.A.; Khan, M.A.S.; Hasan, M.J. Virucidal effect of povidone iodine on COVID-19 in the nasopharynx: An open-label randomized clinical trial. Indian J. Otolaryngol. Head Neck Surg. 2021, 1–5. [Google Scholar] [CrossRef]
  64. Khan, M.M.; Parab, S.R.; Paranjape, M. Repurposing 0.5% povidone iodine solution in otorhinolaryngology practice in COVID-19 pandemic. Am. J. Otolaryngol. 2020, 41, 102618. [Google Scholar] [CrossRef]
  65. Saud, Z.; Tyrrell, V.J.; Zaragkoulias, A.; Protty, M.B.; Statkute, E.; Rubina, A.; Bentley, K.; White, D.A.; Rodrigues, P.D.S.; Murphy, R.C.; et al. The SARS-CoV2 envelope differs from host cells, exposes procoagulant lipids, and is disrupted in vivo by oral rinses. J. Lipid Res. 2022, 63, 100208. [Google Scholar] [CrossRef] [PubMed]
  66. Avhad, S.K.; Bhanushali, M.; Sachdev, S.S.; Save, S.S.; Kalra, D.; Kamala, D.N. Comparison of effectiveness of chlorine dioxide mouthwash and chlorhexidine gluconate mouthwash in reduction of oral viral load in patients with COVID-19. Indian J. Public Health Res. Dev. 2020, 11, 27–32. [Google Scholar]
  67. Almanza-Reyes, H.; Moreno, S.; Plascencia-Lopez, I.; Alvarado-Vera, M.; Patron-Romero, L.; Borrego, B.; Reyes-Escamilla, A.; Valencia-Manzo, D.; Brun, A.; Pestryakov, A.; et al. Evaluation of silver nanoparticles for the prevention of SARS-CoV-2 infection in health workers: In vitro and in vivo. PLoS ONE 2021, 16, e0256401. [Google Scholar] [CrossRef] [PubMed]
  68. Aref, Z.F.; Bazeed, S.; Hassan, M.H.; Hassan, A.S.; Rashad, A.; Hassan, R.G.; Abdelmaksoud, A.A. Clinical, Biochemical and Molecular Evaluations of Ivermectin Mucoadhesive Nanosuspension Nasal Spray in Reducing Upper Respiratory Symptoms of Mild COVID-19. Int. J. Nanomed. 2021, 16, 4063–4072. [Google Scholar] [CrossRef]
  69. Laferl, H.; Seitz, T.; Baier-Grabner, S.; Kelani, H.; Scholz, E.; Heger, F.; Gotzinger, F.; Frischer, P.T.; Wenisch, C.; Allerberger, P.F. Evaluation of RT-qPCR of mouthwash and buccal swabs for detection of SARS-CoV-2 in children and adults. Am. J. Infect. Control 2022, 50, 176–181. [Google Scholar] [CrossRef]
  70. Michel, W.; Farber, J.; Dilas, M.; Heuft, H.G.; Tammer, I.; Baar, J.; Kaasch, A.J. A combined oro-nasopharyngeal swab is more sensitive than mouthwash in detecting SARS-CoV-2 by a high-throughput PCR assay. Infection 2021, 49, 527–531. [Google Scholar] [CrossRef]
  71. Mora-Aguilera, G.; Martinez-Bustamante, V.; Acevedo-Sanchez, G.; Coria-Contreras, J.J.; Guzman-Hernandez, E.; Flores-Colorado, O.E.; Mendoza-Ramos, C.; Hernandez-Nava, G.; Alvarez-Maya, I.; Gutierrez-Espinosa, M.A.; et al. Surveillance Web System and Mouthwash-Saliva qPCR for Labor Ambulatory SARS-CoV-2 Detection and Prevention. Int. J. Environ. Res. Public Health 2022, 19, 1271. [Google Scholar] [CrossRef]
  72. Paull, J.R.A.; Luscombe, C.A.; Castellarnau, A.; Heery, G.P.; Bobardt, M.D.; Gallay, P.A. Protective Effects of Astodrimer Sodium 1% Nasal Spray Formulation against SARS-CoV-2 Nasal Challenge in K18-hACE2 Mice. Viruses 2021, 13, 1656. [Google Scholar] [CrossRef]
  73. Errecalde, J.; Lifschitz, A.; Vecchioli, G.; Ceballos, L.; Errecalde, F.; Ballent, M.; Marin, G.; Daniele, M.; Turic, E.; Spitzer, E.; et al. Safety and Pharmacokinetic Assessments of a Novel Ivermectin Nasal Spray Formulation in a Pig Model. J. Pharm. Sci. 2021, 110, 2501–2507. [Google Scholar] [CrossRef]
  74. Jain, A.; Grover, V.; Singh, C.; Sharma, A.; Das, D.K.; Singh, P.; Thakur, K.G.; Ringe, R.P. Chlorhexidine: An effective anticovid mouth rinse. J. Indian Soc. Periodontol. 2021, 25, 86–88. [Google Scholar] [CrossRef] [PubMed]
  75. Bano-Polo, M.; Martinez-Gil, L.; Sanchez Del Pino, M.M.; Massoli, A.; Mingarro, I.; Leon, R.; Garcia-Murria, M.J. Cetylpyridinium chloride promotes disaggregation of SARS-CoV-2 virus-like particles. J. Oral Microbiol. 2022, 14, 2030094. [Google Scholar] [CrossRef] [PubMed]
  76. Bansal, S.; Jonsson, C.B.; Taylor, S.L.; Figueroa, J.M.; Dugour, A.V.; Palacios, C.; Vega, J.C. Iota-carrageenan and xylitol inhibit SARS-CoV-2 in Vero cell culture. PLoS ONE 2021, 16, e0259943. [Google Scholar] [CrossRef] [PubMed]
  77. Bentley, K.; Stanton, R.J. Hydroxypropyl Methylcellulose-Based Nasal Sprays Effectively Inhibit In Vitro SARS-CoV-2 Infection and Spread. Viruses 2021, 13, 2345. [Google Scholar] [CrossRef] [PubMed]
  78. Bovard, D.; van der Toorn, M.; Schlage, W.K.; Constant, S.; Renggli, K.; Peitsch, M.C.; Hoeng, J. Iota-carrageenan extracted from red algae is a potent inhibitor of SARS-CoV-2 infection in reconstituted human airway epithelia. Biochem. Biophys. Rep. 2022, 29, 101187. [Google Scholar] [CrossRef] [PubMed]
  79. Haridas, M.; Sasidhar, V.; Nath, P.; Abhithaj, J.; Sabu, A.; Rammanohar, P. Compounds of Citrus medica and Zingiber officinale for COVID-19 inhibition: In silico evidence for cues from Ayurveda. Future J. Pharm. Sci. 2021, 7, 13. [Google Scholar] [CrossRef]
  80. Moakes, R.J.A.; Davies, S.P.; Stamataki, Z.; Grover, L.M. Formulation of a Composite Nasal Spray Enabling Enhanced Surface Coverage and Prophylaxis of SARS-COV-2. Adv. Mater. 2021, 33, e2008304. [Google Scholar] [CrossRef]
  81. Morokutti-Kurz, M.; Froba, M.; Graf, P.; Grosse, M.; Grassauer, A.; Auth, J.; Schubert, U.; Prieschl-Grassauer, E. Iota-carrageenan neutralizes SARS-CoV-2 and inhibits viral replication in vitro. PLoS ONE 2021, 16, e0237480. [Google Scholar] [CrossRef]
  82. Munoz-Basagoiti, J.; Perez-Zsolt, D.; Leon, R.; Blanc, V.; Raich-Regue, D.; Cano-Sarabia, M.; Trinite, B.; Pradenas, E.; Blanco, J.; Gispert, J.; et al. Mouthwashes with CPC Reduce the Infectivity of SARS-CoV-2 Variants In Vitro. J. Dent. Res. 2021, 100, 1265–1272. [Google Scholar] [CrossRef]
  83. Paolacci, S.; Ergoren, M.C.; De Forni, D.; Manara, E.; Poddesu, B.; Cugia, G.; Dhuli, K.; Camilleri, G.; Tuncel, G.; Kaya Suer, H.; et al. In vitro and clinical studies on the efficacy of alpha-cyclodextrin and hydroxytyrosol against SARS-CoV-2 infection. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 81–89. [Google Scholar] [CrossRef]
  84. Rodriguez, K.; Saunier, F.; Rigaill, J.; Audoux, E.; Botelho-Nevers, E.; Prier, A.; Dickerscheit, Y.; Pillet, S.; Pozzetto, B.; Bourlet, T.; et al. Evaluation of in vitro activity of copper gluconate against SARS-CoV-2 using confocal microscopy-based high content screening. J. Trace Elem. Med. Biol. 2021, 68, 126818. [Google Scholar] [CrossRef] [PubMed]
  85. Sharad, S.; Kapur, S. Indian Herb-Derived Phytoconstituent-Based Antiviral, Antimicrobial and Antifungal Formulation: An Oral Rinse Candidate for Oral Hygiene and the Potential Prevention of COVID-19 Outbreaks. Pathogens 2021, 10, 1130. [Google Scholar] [CrossRef] [PubMed]
  86. Tateyama-Makino, R.; Abe-Yutori, M.; Iwamoto, T.; Tsutsumi, K.; Tsuji, M.; Morishita, S.; Kurita, K.; Yamamoto, Y.; Nishinaga, E.; Tsukinoki, K. The inhibitory effects of toothpaste and mouthwash ingredients on the interaction between the SARS-CoV-2 spike protein and ACE2, and the protease activity of TMPRSS2 in vitro. PLoS ONE 2021, 16, e0257705. [Google Scholar] [CrossRef] [PubMed]
  87. Yadalam, P.K.; Varatharajan, K.; Rajapandian, K.; Chopra, P.; Arumuganainar, D.; Nagarathnam, T.; Sohn, H.; Madhavan, T. Antiviral Essential Oil Components against SARS-CoV-2 in Pre-procedural Mouth Rinses for Dental Settings during COVID-19: A Computational Study. Front. Chem. 2021, 9, 642026. [Google Scholar] [CrossRef] [PubMed]
  88. Rodriguez-Casanovas, H.J.; la Rosa, M.; Bello-Lemus, Y.; Rasperini, G.; Acosta-Hoyos, A.J. Virucidal Activity of Different Mouthwashes Using a Novel Biochemical Assay. Healthcare 2021, 10, 63. [Google Scholar] [CrossRef] [PubMed]
  89. Robinson, T.E.; Moakes, R.J.A.; Grover, L.M. Low Acyl Gellan as an Excipient to Improve the Sprayability and Mucoadhesion of Iota Carrageenan in a Nasal Spray to Prevent Infection With SARS-CoV-2. Front. Med. Technol. 2021, 3, 687681. [Google Scholar] [CrossRef]
  90. Westover, J.B.; Ferrer, G.; Vazquez, H.; Bethencourt-Mirabal, A.; Go, C.C. In Vitro Virucidal Effect of Intranasally Delivered Chlorpheniramine Maleate Compound against Severe Acute Respiratory Syndrome Coronavirus 2. Cureus 2020, 12, e10501. [Google Scholar] [CrossRef]
  91. Cannon, M.L.; Westover, J.B.; Bleher, R.; Sanchez-Gonzalez, M.A.; Ferrer, G. In Vitro Analysis of the Anti-viral Potential of nasal spray constituents against SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
  92. Mohamed, N.A.; Baharom, N.; Sulaiman, W.S.W.; Rashid, Z.Z.; Ken, W.K.; Ali, U.K.; Othman, S.N.; Samat, M.N.; Kori, N.; Periyasamy, P.; et al. Early viral clearance among COVID-19 patients when gargling with povidone-iodine and essential oils—A clinical trial. medRvix 2020. [Google Scholar] [CrossRef]
  93. Statkute, E.; Rubina, A.; O’Donnell, V.B.; Thomas, D.W.; Stanton, R.J. Brief Report: The Virucidal Efficacy of Oral Rinse Components against SARS-CoV-2 In Vitro. bioRxiv 2020. [Google Scholar] [CrossRef]
  94. Xu, C.; Wang, A.; Hoskin, E.R.; Cugini, C.; Markowitz, K.; Chang, T.L.; Fine, D.H. Differential effects of antiseptic mouth rinses on SARS-CoV-2 infectivity in vitro. bioRxiv 2020, 10, 272. [Google Scholar] [CrossRef] [PubMed]
  95. Buonavoglia, A.; Camero, M.; Lanave, G.; Catella, C.; Trombetta, C.M.; Gandolfi, M.G.; Palazzo, G.; Martella, V.; Prati, C. Virucidal activity in vitro of mouthwashes against a feline coronavirus type II. Oral Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
  96. Green, A.; Roberts, G.; Tobery, T.; Vincent, C.; Barili, M.; Jones, C. In vitro assessment of the virucidal activity of four mouthwashes containing Cetylpyridinium Chloride, ethanol, zinc and a mix of enzyme and proteins against a human coronavirus. bioRxiv 2020. [Google Scholar] [CrossRef]
  97. Shet, M.; Hong, R.; Igo, D.; Cataldo, M.; Bhaskar, S. In Vitro Evaluation of the Virucidal Activity of Different Povidone-Iodine Formulations against Murine and Human Coronaviruses. Infect. Dis. Ther. 2021, 10, 2777–2790. [Google Scholar] [CrossRef] [PubMed]
  98. Balouch, B.; Vontela, S.; Yeakel, H.; Alnouri, G.; Sataloff, R.T. Role of Famotidine and Other Acid Reflux Medications for SARS-CoV-2: A Pilot Study. J. Voice 2021. [Google Scholar] [CrossRef]
  99. Frank, S.; Capriotti, J.; Brown, S.M.; Tessema, B. Povidone-Iodine Use in Sinonasal and Oral Cavities: A Review of Safety in the COVID-19 Era. Ear Nose Throat J. 2020, 99, 586–593. [Google Scholar] [CrossRef]
  100. Vergara-Buenaventura, A.; Castro-Ruiz, C. Use of mouthwashes against COVID-19 in dentistry. Br. J. Oral Maxillofac. Surg. 2020, 58, 924–927. [Google Scholar] [CrossRef]
  101. de Toledo Telles-Araujo, G.; Caminha, R.D.G.; Kallas, M.S.; Sipahi, A.M.; da Silva Santos, P.S. Potential mouth rinses and nasal sprays that reduce SARS-CoV-2 viral load: What we know so far? Clinics 2020, 75, e2328. [Google Scholar] [CrossRef]
  102. Gerlach, M.; Wolff, S.; Ludwig, S.; Schafer, W.; Keiner, B.; Roth, N.J.; Widmer, E. Rapid SARS-CoV-2 inactivation by commonly available chemicals on inanimate surfaces. J. Hosp. Infect. 2020, 106, 633–634. [Google Scholar] [CrossRef]
  103. Carrouel, F.; Conte, M.P.; Fisher, J.; Goncalves, L.S.; Dussart, C.; Llodra, J.C.; Bourgeois, D. COVID-19: A Recommendation to Examine the Effect of Mouthrinses with beta-Cyclodextrin Combined with Citrox in Preventing Infection and Progression. J. Clin. Med. 2020, 9, 1126. [Google Scholar] [CrossRef]
  104. Peng, X.; Xu, X.; Li, Y.; Cheng, L.; Zhou, X.; Ren, B. Transmission routes of 2019-nCoV and controls in dental practice. Int. J. Oral Sci. 2020, 12, 9. [Google Scholar] [CrossRef] [PubMed]
  105. Pattanshetty, S.; Narayana, A.; Radhakrishnan, R. Povidone-iodine gargle as a prophylactic intervention to interrupt the transmission of SARS-CoV-2. Oral Dis. 2021, 27 (Suppl. 3), 752–753. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA flow chart shows the number of screened, included, and excluded studies.
Figure 1. PRISMA flow chart shows the number of screened, included, and excluded studies.
Ijerph 19 12148 g001
Figure 2. Forest plot of virucidal efficacy of several preparations against SARS-CoV-2 in vivo and the associated level of heterogeneity [19,20,21,22,23,24,25,26,27,28,29].
Figure 2. Forest plot of virucidal efficacy of several preparations against SARS-CoV-2 in vivo and the associated level of heterogeneity [19,20,21,22,23,24,25,26,27,28,29].
Ijerph 19 12148 g002
Figure 3. Forest plot of virucidal efficacy of several preparations against SARS-CoV-2 in vitro [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51].
Figure 3. Forest plot of virucidal efficacy of several preparations against SARS-CoV-2 in vitro [30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51].
Ijerph 19 12148 g003
Figure 4. (A) Forrest plot of virucidal efficacy of povidone-iodine (PVP-I) (all concentrations) in vitro [31,32,33,34,35,36,37,38,41,42,43,45]. (B) Forrest plot of virucidal efficacy of povidone-iodine (PVP-I) > 1.0% in vitro [31,32,33,37,41,43].
Figure 4. (A) Forrest plot of virucidal efficacy of povidone-iodine (PVP-I) (all concentrations) in vitro [31,32,33,34,35,36,37,38,41,42,43,45]. (B) Forrest plot of virucidal efficacy of povidone-iodine (PVP-I) > 1.0% in vitro [31,32,33,37,41,43].
Ijerph 19 12148 g004aIjerph 19 12148 g004b
Figure 5. (A) Forrest plot of virucidal efficacy of cetylpyridinium-chloride (CPC) in vitro [30,44,46]. (B) Forrest plot of virucidal efficacy of Listerine® and other essential oils in vitro [42,43]. (C) Forrest plot of virucidal efficacy of hydrogen peroxide (HP) in vitro [32,42,46]. (D) Forrest plot of virucidal efficacy of chlorhexidine (CHX) in vitro [30,42,44,46,50,51]. (E) Forrest plot of virucidal efficacy of sodium fluoride, sodium chloride, and sodium fluoride (NaF) in vitro [39,47,50].
Figure 5. (A) Forrest plot of virucidal efficacy of cetylpyridinium-chloride (CPC) in vitro [30,44,46]. (B) Forrest plot of virucidal efficacy of Listerine® and other essential oils in vitro [42,43]. (C) Forrest plot of virucidal efficacy of hydrogen peroxide (HP) in vitro [32,42,46]. (D) Forrest plot of virucidal efficacy of chlorhexidine (CHX) in vitro [30,42,44,46,50,51]. (E) Forrest plot of virucidal efficacy of sodium fluoride, sodium chloride, and sodium fluoride (NaF) in vitro [39,47,50].
Ijerph 19 12148 g005aIjerph 19 12148 g005bIjerph 19 12148 g005c
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Idrees, M.; McGowan, B.; Fawzy, A.; Abuderman, A.A.; Balasubramaniam, R.; Kujan, O. Efficacy of Mouth Rinses and Nasal Spray in the Inactivation of SARS-CoV-2: A Systematic Review and Meta-Analysis of In Vitro and In Vivo Studies. Int. J. Environ. Res. Public Health 2022, 19, 12148. https://doi.org/10.3390/ijerph191912148

AMA Style

Idrees M, McGowan B, Fawzy A, Abuderman AA, Balasubramaniam R, Kujan O. Efficacy of Mouth Rinses and Nasal Spray in the Inactivation of SARS-CoV-2: A Systematic Review and Meta-Analysis of In Vitro and In Vivo Studies. International Journal of Environmental Research and Public Health. 2022; 19(19):12148. https://doi.org/10.3390/ijerph191912148

Chicago/Turabian Style

Idrees, Majdy, Bridget McGowan, Amr Fawzy, Abdulwahab Ali Abuderman, Ramesh Balasubramaniam, and Omar Kujan. 2022. "Efficacy of Mouth Rinses and Nasal Spray in the Inactivation of SARS-CoV-2: A Systematic Review and Meta-Analysis of In Vitro and In Vivo Studies" International Journal of Environmental Research and Public Health 19, no. 19: 12148. https://doi.org/10.3390/ijerph191912148

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop