Iota-carrageenan and xylitol inhibit SARS-CoV-2 in Vero cell culture

Shruti Bansal, Colleen B Jonsson, Shannon L Taylor, Juan Manuel Figueroa, Andrea Vanesa Dugour, Carlos Palacios, Julio César Vega, Shruti Bansal, Colleen B Jonsson, Shannon L Taylor, Juan Manuel Figueroa, Andrea Vanesa Dugour, Carlos Palacios, Julio César Vega

Abstract

Last year observed a global pandemic caused by SARS-CoV-2 (severe acute respiratory syndrome-coronavirus 2) infection affecting millions of individuals worldwide. There is an urgent unmet need to provide an easily producible and affordable medicine to prevent transmission and provide early treatment for this disease. Since the nasal cavity and the rhinopharynx are the sites of initial replication of SARS-CoV-2, a nasal spray may be an effective option to target SARS-CoV-2 infection. In this study, we tested the antiviral action of three candidate nasal spray formulations against SARS-CoV-2 in vitro. We determined that iota-carrageenan in concentrations as low as 6 μg/mL inhibits SARS-CoV-2 in vitro. The concentrations of iota-carrageenan with activity against SARS-CoV-2 in vitro may be easily achieved through the application of nasal sprays as commonly used in several countries. Recently a double-blind, placebo-controlled study showed that iota-carrageenan in isotonic sodium chloride reduces ca. five times the risk of infection by SARS-CoV-2 in health care personnel. Further, xylitol at a concentration of 50 mg/mL (ca. 329 mM) was found to exert some antiviral action, though this preliminary finding needs further confirmation.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: I have a commercial affiliation to Laboratorio Pablo Cassará S.R.L, who provided the formulations for the study and to Amcyte Pharma Inc.. This submission is related to US Patent No. 11.013.687 “PREVENTIVE AND THERAPEUTIC TREATMENT FOR COVID 19 AND ANY OTHER DISEASE CAUSED BY SARS COV 2”, whose inventor is Julio César Vega. This does not alter our adherence to PLOS ONE’s policies on sharing data and materials.

Figures

Fig 1. SARS-CoV-2 viral titer after treatment…
Fig 1. SARS-CoV-2 viral titer after treatment with Samples 1 and P1.
A) Infection assay. Sample 1 composition: 1.2mg/mL iota-carrageenan, 9 mg/mL sodium chloride, pH 6–7. Vero E6 were pre-treated for two hours with dilutions of Sample 1 with Sample P1 (diluent without iota-carrageenan) to obtain 600 μg/mL, 60 μg/mL, 6 μg/mL, and 0.6 μg/mL iota-carrageenan final concentration. After this pretreatment, cells were infected with SARS-CoV-2 and incubated for 48 hours in the presence of the same dilutions of Sample 1. Supernatants were harvested and virus yield was determined using an end point dilution assay (TCID50). Controls consisted of untreated infected cells or infected cells treated with P1 (no iota-carrageenan). Results were determined using the Reed and Muench formula and expressed as log TCID50/mL. The dotted line shows the limit of detection (LOD). Testing of samples was performed in triplicate, and the p-values are p≤0.00025 (****). B) Cellular viability assays. Vero-E6 cells were treated with iota-carrageenan or vehicle (600 μg/mL to 0 μg/mL) for 48 h at 37°C. After incubation, cellular viability was analyzed, and no statistically significant difference was found between the groups compared to the untreated control group (Group 600 μg/ml, p = 0.7464, Group 60 μg/ml, p = 0.0908, Group 6 μg/ml, p = 0.1208, and Group 0.6 μg/ml, p = 0.8938). Data are expressed as mean ± SD. Therefore, these compositions do not adversely affect cell viability. For this reason, cell lysis and death detected in the reported experiments after infection must be attributed to the action of the virus.
Fig 2. SARS-CoV-2 viral titer after treatment…
Fig 2. SARS-CoV-2 viral titer after treatment with Samples 2 and P2.
A) Infection assay. Sample 2 composition: 1.2mg/mL iota-carrageenan, 5 mg/mL sodium chloride, pH 6–7. Vero E6 were pre-treated with dilutions of Sample 2 with Sample P2 (diluent without iota-carrageenan) to get 600 μg/mL, 60 μg/mL, 6 μg/mL, and 0.6 μg/mL final iota-carrageenan concentration for two hours. After this pretreatment, cells were infected with SARS-CoV-2 and incubated for 48 hours in the presence of the same dilutions of Sample 2. Supernatants were harvested and virus yield was determined using an end point dilution assay (TCID50). Controls consisted of untreated infected cells or infected cells treated with P2 (no iota-carrageenan). Results were determined using the Reed and Muench formula and expressed as log TCID50/mL. The dotted line shows the limit of detection (LOD). Testing of samples was performed in triplicate, and the p-values are p≤0.00074 (***) and p≤0.00001. B) Cellular viability assays. Vero-E6 cells were treated with iota-carrageenan or vehicle (600 μg/mL to 0 μg/mL) for 48 h at 37°C. After incubation, cellular viability was analyzed, and no statistically significant difference was found between the groups compared to the untreated control group (Group 600 μg/ml, p = 0.9880, Group 60 μg/ml, p = 0.0683, Group 6 μg/ml, p = 0.9993, and Group 0.6 μg/ml, p = 0.1957). Data are expressed as mean ± SD.
Fig 3. SARS-CoV-2 viral titer after treatment…
Fig 3. SARS-CoV-2 viral titer after treatment with Samples 3 and P3.
A) Infection assay. Sample 3 composition: 1.2mg/mL iota-carrageenan, 5% m/V xylitol, pH 6–7. Vero E6 were pre-treated with dilutions of Sample 3 and Sample P3 (placebo without iota-carrageenan) to get 600 μg/mL, 60 μg/mL, 6 μg/mL, and 0.6 μg/mL final iota-carrageenan concentration for two hours. After this pretreatment, cells were infected with SARS-CoV-2 and incubated for 48 hours in the presence of the same dilutions of Sample 3. Supernatants were harvested, and virus yield was determined by an endpoint dilution assay (TCID50). Controls consisted of untreated infected cells or infected cells treated with P3 (no iota-carrageenan). Results were determined using the Reed and Muench formula and expressed as log TCID50/mL. The dotted line shows the limit of detection (LOD). Testing of samples was performed in triplicate and the p-value indicates that the groups are significantly different p = 0.0045, nevertheless no post-hoc analysis could be performed. B) Cellular viability assays. Vero-E6 cells were treated with iota-carrageenan or vehicle (600 μg/mL to 0 μg/mL) for 48 h at 37°C. After incubation, cellular viability was analyzed. No statistically significant difference was found between compositions at low iota-carrageenan concentrations, 6 μg/mL (p = 0.6904) and 0.6 μg/mL (p > 0.9999) compared to the untreated control group (Group 6 μg/ml, p = 0.6904, and Group 0.6 μg/ml, p > 0.9999). Compositions with higher concentrations of iota-carrageenan tend to show a significant difference increasing cell viability (Group 600 μg/ml, p = 0.0031 (***), Group 60 μg/ml, p = 0.0417 (*), They are certainly not toxic, but may exert some cytoprotective effect.). Data are expressed as mean ± SD.

References

    1. Dong E, Du H, Gardner L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect Dis. 2020;20: 533–534. doi: 10.1016/S1473-3099(20)30120-1
    1. Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China. Jama. 2020;323: 1239–1242. doi: 10.1001/jama.2020.2648
    1. Kelvin AA, Halperin S. COVID-19 in children: the link in the transmission chain. Lancet Infect Dis. 2020;20: 633–634. doi: 10.1016/S1473-3099(20)30236-X
    1. Mathieu E, Ritchie H, Ortiz-Ospina E, Roser M, Hasell J, Appel C, et al.. A global database of COVID-19 vaccinations. Nat Hum Behav. 2021;5: 947–953. doi: 10.1038/s41562-021-01122-8
    1. Liu Y, Liao W, Wan L, Xiang T, Zhang W. Correlation Between Relative Nasopharyngeal Virus RNA Load and Lymphocyte Count Disease Severity in Patients with COVID-19. Viral Immunol. 2021;34: 330–335. doi: 10.1089/vim.2020.0062
    1. Liu Y, Yan L-M, Wan L, Xiang T-X, Le A, Liu J-M, et al.. Viral dynamics in mild and severe cases of COVID-19. Lancet Infect Dis. 2020;20: 656–657. doi: 10.1016/S1473-3099(20)30232-2
    1. Zou L, Ruan F, Huang M, Liang L, Huang H, Hong Z, et al.. SARS-CoV-2 Viral Load in Upper Respiratory Specimens of Infected Patients. New Engl J Med. 2020;382: 1177–1179. doi: 10.1056/NEJMc2001737
    1. Callahan C, Lee RA, Lee GR, Zulauf K, Kirby JE, Arnaout R. Nasal-Swab Testing Misses Patients with Low SARS-CoV-2 Viral Loads. Medrxiv. 2020; 2020.06.12.20128736. doi: 10.1101/2020.06.12.20128736
    1. Eccles R, Meier C, Jawad M, Weinmüllner R, Grassauer A, Prieschl-Grassauer E. Efficacy and safety of an antiviral Iota-Carrageenan nasal spray: a randomized, double-blind, placebo-controlled exploratory study in volunteers with early symptoms of the common cold. Respir Res. 2010;11: 108. doi: 10.1186/1465-9921-11-108
    1. Koenighofer M, Lion T, Bodenteich A, Prieschl-Grassauer E, Grassauer A, Unger H, et al.. Carrageenan nasal spray in virus confirmed common cold: individual patient data analysis of two randomized controlled trials. Multidiscip Resp Med. 2014;9: 57. doi: 10.1186/2049-6958-9-57
    1. Ludwig M, Enzenhofer E, Schneider S, Rauch M, Bodenteich A, Neumann K, et al.. Efficacy of a Carrageenan nasal spray in patients with common cold: a randomized controlled trial. Respir Res. 2013;14: 124. doi: 10.1186/1465-9921-14-124
    1. Ahmadi A, Moghadamtousi SZ, Abubakar S, Zandi K. Antiviral Potential of Algae Polysaccharides Isolated from Marine Sources: A Review. Biomed Res Int. 2015;2015: 1–10. doi: 10.1155/2015/825203
    1. Buck CB, Thompson CD, Roberts JN, Müller M, Lowy DR, Schiller JT. Carrageenan Is a Potent Inhibitor of Papillomavirus Infection. Plos Pathog. 2006;2: e69. doi: 10.1371/journal.ppat.0020069
    1. Girond S, Crance JM, Cuyck-Gandre HV, Renaudet J, Deloince R. Antiviral activity of carrageenan on hepatitis A virus replication in cell culture. Res Virology. 1991;142: 261–270. doi: 10.1016/0923-2516(91)90011-q
    1. Grassauer A, Weinmuellner R, Meier C, Pretsch A, Prieschl-Grassauer E, Unger H. Iota-Carrageenan is a potent inhibitor of rhinovirus infection. Virol J. 2008;5: 107. doi: 10.1186/1743-422X-5-107
    1. Shao Q, Guo Q, Xu W ping, Li Z, Zhao T tong. Specific Inhibitory Effect of κ-Carrageenan Polysaccharide on Swine Pandemic 2009 H1N1 Influenza Virus. Plos One. 2015;10: e0126577. doi: 10.1371/journal.pone.0126577
    1. Talarico LB, Damonte EB. Interference in dengue virus adsorption and uncoating by carrageenans. Virology. 2007;363: 473–485. doi: 10.1016/j.virol.2007.01.043
    1. Leibbrandt A, Meier C, König-Schuster M, Weinmüllner R, Kalthoff D, Pflugfelder B, et al.. Iota-Carrageenan Is a Potent Inhibitor of Influenza A Virus Infection. Plos One. 2010;5: e14320. doi: 10.1371/journal.pone.0014320
    1. Fazekas T, Eickhoff P, Pruckner N, Vollnhofer G, Fischmeister G, Diakos C, et al.. Lessons learned from a double-blind randomised placebo-controlled study with a iota-carrageenan nasal spray as medical device in children with acute symptoms of common cold. Bmc Complem Altern M. 2012;12: 147–147. doi: 10.1186/1472-6882-12-147
    1. Graf C, Bernkop-Schnürch A, Egyed A, Koller C, Prieschl-Grassauer E, Morokutti-Kurz M. Development of a nasal spray containing xylometazoline hydrochloride and iota-carrageenan for the symptomatic relief of nasal congestion caused by rhinitis and sinusitis. Int J Gen Medicine. 2018;11: 275–283. doi: 10.2147/IJGM.S167123
    1. Morokutti-Kurz M, König-Schuster M, Koller C, Graf C, Graf P, Kirchoff N, et al.. The Intranasal Application of Zanamivir and Carrageenan Is Synergistically Active against Influenza A Virus in the Murine Model. Plos One. 2015;10: e0128794. doi: 10.1371/journal.pone.0128794
    1. Salli K, Lehtinen MJ, Tiihonen K, Ouwehand AC. Xylitol’s Health Benefits beyond Dental Health: A Comprehensive Review. Nutrients. 2019;11: 1813. doi: 10.3390/nu11081813
    1. Sakallioğlu Ö, Güvenç IA, Cingi C. Xylitol and its usage in ENT practice. J Laryngology X0026 Otology. 2014;128: 580–585. doi: 10.1017/S0022215114001340
    1. Lin L, Tang X, Wei J, Dai F, Sun G. Xylitol nasal irrigation in the treatment of chronic rhinosinusitis. Am J Otolaryng. 2017;38: 383–389. doi: 10.1016/j.amjoto.2017.03.006
    1. Xu ML, Wi GR, Kim HJ, Kim H-J. Ameliorating Effect of Dietary Xylitol on Human Respiratory Syncytial Virus (hRSV) Infection. Biological Pharm Bulletin. 2016;39: 540–546. doi: 10.1248/bpb.b15-00773
    1. Hebar A, Koller C, Seifert J-M, Chabicovsky M, Bodenteich A, Bernkop-Schnürch A, et al.. Non-Clinical Safety Evaluation of Intranasal Iota-Carrageenan. Plos One. 2015;10: e0122911. doi: 10.1371/journal.pone.0122911
    1. Weissman JD, Fernandez F, Hwang PH. Xylitol nasal irrigation in the management of chronic rhinosinusitis: A pilot study. Laryngoscope. 2011;121: 2468–2472. doi: 10.1002/lary.22176
    1. Durairaj L, Launspach J, Watt JL, Businga TR, Kline JN, Thorne PS, et al.. Safety assessment of inhaled xylitol in mice and healthy volunteers. Respir Res. 2004;5: 13–13. doi: 10.1186/1465-9921-5-13
    1. Eccles R. Iota-Carrageenan as an Antiviral Treatment for the Common Cold. Open Virology J. 2020;14: 9–15. doi: 10.2174/1874357902014010009
    1. Grassauer A, Prieschl-Grassauer E, Bodenteich A, Koller C, Morokutti-Kurz M. Stuffy nose deblocking composition having antiviral activity—Google Patents. US10660914B2, 2020. pp. 1–7. Available:
    1. Mendoza EJ, Manguiat K, Wood H, Drebot M. Two Detailed Plaque Assay Protocols for the Quantification of Infectious SARS-CoV-2. Curr Protoc Microbiol. 2020;57: ecpmc105. doi: 10.1002/cpmc.105
    1. Reed LJ, Muench H. A simple method of estimating fifty per cent endpoints. Am J Epidemiol. 1938;27: 493–497. doi: 10.1093/oxfordjournals.aje.a118408
    1. Morokutti-Kurz M, Fröba M, Graf P, Große M, Grassauer A, Auth J, et al.. Iota-carrageenan neutralizes SARS-CoV-2 and inhibits viral replication in vitro. Plos One. 2021;16: e0237480. doi: 10.1371/journal.pone.0237480
    1. Schütz D, Conzelmann C, Fois G, Groß R, Weil T, Wettstein L, et al.. Carrageenan-containing over-the-counter nasal and oral sprays inhibit SARS-CoV-2 infection of airway epithelial cultures. Am J Physiol-lung C. 2021;320: L750–L756. doi: 10.1152/ajplung.00552.2020
    1. Bitter C, Suter-Zimmermann K, Surber C. Nasal Drug Delivery in Humans. Curr Probl Dermatol. 2011;40: 20–35. doi: 10.1159/000321044
    1. Garcia GJM, Schroeter JD, Segal RA, Stanek J, Foureman GL, Kimbell JS. Dosimetry of nasal uptake of water-soluble and reactive gases: A first study of interhuman variability. Inhal Toxicol. 2009;21: 607–618. doi: 10.1080/08958370802320186
    1. Gizurarson S. Anatomical and Histological Factors Affecting Intranasal Drug and Vaccine Delivery. Curr Drug Deliv. 2012;9: 566–582. doi: 10.2174/156720112803529828
    1. Pires A, Fortuna A, Alves G, Falcão A. Intranasal Drug Delivery: How, Why and What for? J Pharm Pharm Sci. 2009;12: 288–311. doi: 10.18433/j3nc79
    1. Helassa N, Garnett JP, Farrant M, Khan F, Pickup JC, Hahn KM, et al.. A novel fluorescent sensor protein for detecting changes in airway surface liquid glucose concentration. Biochem J. 2014;464: 213–220. doi: 10.1042/BJ20141041
    1. Wagenmann M, Naclerio RM. Anatomic and physiologic considerations in sinusitis. J Allergy Clin Immun. 1992;90: 419–423. doi: 10.1016/0091-6749(92)90161-t
    1. Ari A. Practical strategies for a safe and effective delivery of aerosolized medications to patients with COVID-19. Resp Med. 2020;167: 105987. doi: 10.1016/j.rmed.2020.105987
    1. Figueroa JM, Lombardo ME, Dogliotti A, Flynn LP, Giugliano R, Simonelli G, 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 Medicine. 2021;Volume 14: 6277–6286. doi: 10.2147/IJGM.S328486

Source: PubMed

3
Tilaa