33
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Using Probiotics to Flatten the Curve of Coronavirus Disease COVID-2019 Pandemic

      discussion

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction Despite strategies based on social distancing, hygiene, and screening, COVID-19 is progressing rapidly throughout the world, with healthcare systems at risk of being overwhelmed. While identification of effective drug therapies is ongoing, vaccines will not be available in the near future. Therefore, additional preventive strategies are urgently needed. COVID-19 presents with a spectrum of disease severity, ranging from mild and non-specific flu-like symptoms, to pneumonia, and life-threatening complications such as acute respiratory distress syndrome (ARDS) and multiple organ failure. While transmission of SARS-CoV-2 is thought to occur mainly via respiratory droplets, the gut may also contribute toward the pathogenesis of COVID-19 (1). SARS-CoV-2 RNA has been detected in the gastrointestinal tract and stool samples from patients (2–4), and in sewage systems (5). Coronaviruses, including SARS-Cov-2 can invade enterocytes, thereby acting as a reservoir for the virus (4). Indeed, large clinical studies from China indicate that gastrointestinal symptoms are common in COVID-19, and are associated with disease severity (3, 4). Probiotics are live microorganisms that when administered in adequate amounts confer a health benefit on the host (6). Clinical evidence shows that certain probiotic strains help to prevent bacterial and viral infections, including gastroenteritis, sepsis, and respiratory tract infections (RTIs). The reason for adding probiotic strains to the overall prevention and care strategy is founded in science and clinical studies, albeit hitherto none directly on the etiological agent of this pandemic. Clinical Data Supporting the Use of Probiotics to Prevent Covid-19 Probiotics can prevent antibiotic-associated diarrhea, and infections in the gastrointestinal tract, but also infections at other sites, including sepsis, and RTIs (7–13). Meta-analyses are the gold standard for evidence-based medicine. In one analysis of more than 8,000 preterm infants included in randomized control trials (RCTs), patients receiving enteral supplementation with probiotics showed a reduction in necrotizing enterocolitis, nosocomial sepsis, and all-cause mortality (14). A well-conducted RCT including >4,000 newborns in India found a reduction in sepsis and lower RTIs in infants treated with a strain of Lactobacillus plantarum combined with prebiotics (which are growth substrates specific for beneficial microorganisms) (15). Viruses are etiologic agents of over 90% of upper RTIs. The positive impact of probiotics on prevention of upper RTIs is documented in a number of studies. A meta-analysis of 12 RCTs including 3,720 adults and children reported a 2-fold lower risk of developing upper RTI in subjects taking probiotics, and a small but significant reduction in disease severity in those infected. A randomized, double-blind, placebo-controlled intervention study of 479 adults showed that Lactobacillus gasseri PA 16/8, Bifidobacterium longum SP 07/3, and Bifidobacterium bifidum MF 20/5 with vitamins and minerals lowered not only the duration of common cold episodes but also days with fever (16). The impact of probiotics on prevention of upper RTIs caused by specific viruses has also been documented. An RCT including 94 preterm infants showed that galacto-oligosaccharide and polydextrose prebiotic mixture (1:1), or probiotic Lactobacillus rhamnosus GG given between 3 and 60 days of life lowered the incidence of clinically defined virus-associated RTI by 2- to 3-fold compared to placebo (17). The incidence of rhinovirus-associated episodes, which comprised 80% of all RTIs in this study, was also strongly reduced with probiotics or prebiotics. The incidence of influenza RTI was reduced following consumption of Lactobacillus brevis in an open label study of 1,783 school children (18). Pertinent to the pandemic affecting adults more than children, these positive findings were confirmed in an RCT that included 27 elderly subjects receiving Bifidobacterium longum or placebo (19). Furthermore, lactic acid bacteria, from which many probiotics are selected, are part of the upper respiratory tract microbiota in healthy people, and some strains are being considered for prevention of recurrent otitis media (20, 21). This makes their use for contributing to slow down progression of the coronavirus pandemic worthy of consideration. Probiotics have also been used to prevent bacterial lower RTIs in critically ill adults. Meta-analyses of RCTs including close to 2,000 patients found that probiotic strains reduce the incidence of ventilator-associated pneumonia (22, 23). But low quality of evidence and conflicting results among different studies calls for additional well-conducted RCTs. It should be noted that not all probiotics, even those with gastrointestinal benefits, necessarily contribute in every way to reducing the risk of respiratory infection. For example, Lactobacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis may contribute to intestinal benefits, but do not reduce the number of viruses in the nasopharynx (24). Examples of products that could be considered, depending on availability in a given country, are provided in Table 1. Table 1 The following are examples (not exclusive) of probiotic products, or web sites listing products, with documentation in human studies that may have relevance to reducing the burden of the coronavirus pandemic. Products Basis for inclusion When to administer References Lactobacillus casei DN-114 001; DanActive/Actimel Fermented drink, Danone Reduced incidence and duration of RTIs Once daily for duration of the pandemic (12, 13) Lactobacillus gasseri PA 16/8, Bifidobacterium longum SP 07/3, and B. bifidum MF 20/5; Tribion harmonis, Merck Lowering duration and severity of flu-like illness Once daily for duration of the pandemic (16) Lactobacillus rhamnosus GG; Culturelle or other brand names For digestive health and gut barrier integrity, and prevention of viral RTIs One capsule daily for duration of the pandemic (17) Lactobacillus plantarum DR7; Malaysia Prevention of upper RTIs, immune modulation 2 g sachet per day for duration of pandemic (25) Bifidobacterium breve Yakult, and Lactobacillus casei Shirota; available as fermented drinks Lower incidence of ventilator-associated pneumonia One of each day for duration of the pandemic (26) Bifidobacterium longum BB536; Morinaga, and sold in many formulations Enhances innate immunity, prevents influenza infection One each day for duration of the pandemic (19) Pediococcus pentosaceus 5-33:3, Leuconostoc mesenteroides 32-77:1, L. paracasei ssp. paracasei 19, L. plantarum 2,362 plus inulin, oat bran, pectin, and resistant starch; Medipharm, Sweden To reduce rate of SIRS, infections, sepsis, days of stay in the intensive care unit, days under mechanical ventilation, and mortality For COVID-19 patients (27) A list of probiotics available in Canada for various health issues; www.probioticchart.ca A list of probiotics available in the USA for various health issues; www.usprobioticguide.com We must emphasize that none have been tested or proven to have an effect against SARS-CoV2, the virus causing COVID-19, nor are they proven treatments or cures for this condition. Mechanistic Basis for the Action of Probiotics to Prevent Infections and Relevance to Covid-19 Mechanisms that might explain clinical success of probiotics include enhancement of the intestinal epithelial barrier, competition with pathogens for nutrients and adhesion to the intestinal epithelium, production of anti-microbial substances and modulation of the host immune system (28). An RCT of 55 infants showed that enteral supplementation with a combination of Bifidobacterium bifidum and Streptococcus thermophilus reduced the incidence of diarrhea and shedding of rotaviruses (29), an effect that has been confirmed in subsequent studies (30). This would indicate interference with viral entry into cells and/or inhibition of viral replication in the intestine. While this mechanism may have a role in reducing dissemination of coronavirus via the gut, the probiotic strains were not administered to the respiratory tract. So, direct inhibition may appear impossible at this site. Having said that, lungs have their own microbiota and a gut-lung connection has been described whereby host-microbe, microbe-microbe and immune interactions can influence the course of respiratory diseases (31). RTIs such as influenza are associated with an imbalance in the microbial communities of the respiratory and gastrointestinal tracts (32, 33). This dysbiosis may alter subsequent immune function and predispose to secondary bacterial infection. As reports from China indicate that COVID-19 might be associated with intestinal dysbiosis causing inflammation and poorer response to pathogens (34, 35), the case exists for probiotic strains that restore gut homeostasis (36). It is feasible that orally administered probiotic strains could further influence this gut-lung axis, as some can migrate from the gut to distant sites, such as the breast to treat mastitis (37). The gut microbiome has a critical impact on systemic immune responses, and immune responses at distant mucosal sites, including the lungs (38, 39). Administration of certain bifidobacteria or lactobacilli has beneficial impact on influenza virus clearance from the respiratory tract (39, 40). Probiotic strains improve levels of type I interferons, increase the number and activity of antigen presenting cells, NK cells, T cells, as well as the levels of systemic and mucosal specific antibodies in the lungs (16, 19, 39). There is also evidence that probiotic strains modify the dynamic balance between proinflammatory and immunoregulatory cytokines that allow viral clearance while minimizing immune response-mediated damage to the lungs. This might be particularly relevant to prevent ARDS, a major complication of COVID-19. An RCT with Lactobacillus plantarum DR7 showed suppression of plasma pro-inflammatory cytokines (IFN-γ, TNF-α) in middle-aged adults, and enhancement of anti-inflammatory cytokines (IL-4, IL-10) in young adults, along with reduced plasma peroxidation and oxidative stress levels (25). Given the cytokine storm that appears to occur in many COVID-19 patients, this type of modulation may prove to be very important. The manner in which orally administered probiotic strains contributes to this appears to involve the immune response emanating from the intestine, a focal point of the body's defenses. Therefore, probiotic strains documented to enhance the integrity of tight junctions, for example through increasing butyrate, a fuel for colonocytes could theoretically reduce SARS-Cov-2 invasion. Evidence for antiviral activity of probiotic strains against common respiratory viruses, including influenza, rhinovirus, and respiratory syncytial virus comes from clinical and experimental studies (17–19, 41). While none of these effects or mechanisms have been tested on the new SARS-CoV-2 virus, this should not negate considering this approach, especially when effects of probiotics against other coronavirus strains have been reported (42–45). Furthermore, patients are dying from secondary bacterial infections. A recent study in mice has shown that oral administration of Lactobacillus acidophilus CMCC878, started 24 h after pulmonary inoculation of Pseudomonas aeruginosa and Staphylococcus aureus reduced bacterial load in the lungs, and decreased lung damage and systemic inflammation (46). Safety of Probiotics Probiotics are generally safe, even in the most vulnerable populations and in intensive care settings (14, 47). Cases of probiotic-associated bacteremia and fungaemia have occurred on extremely rare occasions, mainly in premature and immunocompromised patients treated with preparations lacking adequate quality control (48, 49). Rather than consider intensive care patients too ill to receive probiotic and prebiotic therapy, RCTs of probiotics for the prevention of ventilator-associated pneumonia provide a reason to consider them (22, 23, 26). Moreover, in an RCT of 65 critically ill, mechanically ventilated, multiple trauma patients, the synbiotic Pediococcus pentosaceus 5-33:3, Leuconostoc mesenteroides 32-77:1, L. paracasei ssp. paracasei 19, L. plantarum 2,362 plus inulin, oat bran, pectin, and resistant starch resulted in reduced rate of infections, systemic inflammatory response syndrome, sepsis, days of stay in the intensive care unit, days under mechanical ventilation, and mortality (27). Summary In summary, orally administered probiotic strains can reduce the incidence and severity of viral RTIs. At a time when doctors are using drugs with little anti- COVID-19 data, probiotic strains documented for anti-viral and respiratory activities (not low-quality undocumented imitations) should become part of the armamentarium to reduce the burden and severity of this pandemic. Government funding is being used to test numerous drugs but just as important, they should fund probiotic trials. In addition, use of recognized prebiotics (e.g., fructans, galactans) to enhance propagation of probiotic strains and indigenous beneficial microbes should be recommended as part of the overall strategy to flatten the curve (11, 50). Author Contributions EG, DB, and VD contributed conception of the manuscript. EG and VD wrote the first draft. DB, GG, and GR wrote sections of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version. Conflict of Interest GG and GR provide advice to probiotic and prebiotic companies. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

          Related collections

          Most cited references51

          • Record: found
          • Abstract: found
          • Article: not found
          Is Open Access

          Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic.

          An expert panel was convened in October 2013 by the International Scientific Association for Probiotics and Prebiotics (ISAPP) to discuss the field of probiotics. It is now 13 years since the definition of probiotics and 12 years after guidelines were published for regulators, scientists and industry by the Food and Agriculture Organization of the United Nations and the WHO (FAO/WHO). The FAO/WHO definition of a probiotic--"live microorganisms which when administered in adequate amounts confer a health benefit on the host"--was reinforced as relevant and sufficiently accommodating for current and anticipated applications. However, inconsistencies between the FAO/WHO Expert Consultation Report and the FAO/WHO Guidelines were clarified to take into account advances in science and applications. A more precise use of the term 'probiotic' will be useful to guide clinicians and consumers in differentiating the diverse products on the market. This document represents the conclusions of the ISAPP consensus meeting on the appropriate use and scope of the term probiotic.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Viral load of SARS-CoV-2 in clinical samples

            An outbreak caused by a novel human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in Wuhan in December 2019, 1 and has since spread within China and to other countries. Real-time RT-PCR assays are recommended for diagnosis of SARS-CoV-2 infection. 2 However, viral dynamics in infected patients are still yet to be fully determined. Here, we report our findings from different types of clinical specimens collected from 82 infected individuals. Serial samples (throat swabs, sputum, urine, and stool) from two patients in Beijing were collected daily after their hospitalisation (patient 1, days 3–12 post-onset; patient 2, days 4–15 post-onset). These samples were examined by an N-gene-specific quantitative RT-PCR assay, as described elsewhere. 3 The viral loads in throat swab and sputum samples peaked at around 5–6 days after symptom onset, ranging from around 104 to 107 copies per mL during this time (figure A, B ). This pattern of changes in viral load is distinct from the one observed in patients with SARS, which normally peaked at around 10 days after onset. 4 Sputum samples generally showed higher viral loads than throat swab samples. No viral RNA was detected in urine or stool samples from these two patients. Figure Viral dynamics of SARS-CoV-2 in infected patients Viral load (mean [SD]) from serial throat swab and sputum samples in patient 1 (A) and patient 2 (B). (C) Viral load (median [IQR]) in throat and sputum samples collected from 80 patients at different stages after disease onset. (D) Correlation between viral load in throat swab samples and viral load in sputum samples. We also studied respiratory samples (nasal [n=1] and throat swabs [n=67], and sputum [n=42]) collected from 80 individuals at different stages of infection. The viral loads ranged from 641 copies per mL to 1·34 × 1011 copies per mL, with a median of 7·99 × 104 in throat samples and 7·52 × 105 in sputum samples (figure C). The only nasal swab tested in this study (taken on day 3 post-onset) showed a viral load of 1·69 × 105 copies per mL. Overall, the viral load early after onset was high (>1 × 106 copies per mL). However, a sputum sample collected on day 8 post-onset from a patient who died had a very high viral load (1·34 × 1011 copies per mL). Notably, two individuals, who were under active surveillance because of a history of exposure to SARS-CoV-2-infected patients showed positive results on RT-PCR a day before onset, suggesting that infected individuals can be infectious before them become symptomatic. Among the 30 pairs of throat swab and sputum samples available, viral loads were significantly correlated between the two sample types for days 1–3 (R2=0·50, p=0·022), days 4–7 (R2=0·93, p<0·001), and days 7–14 (R2=0·95, p=0·028). From 17 confirmed cases of SARS-CoV-2 infection with available data (representing days 0–13 after onset), stool samples from nine (53%; days 0–11 after onset) were positive on RT-PCR analysis. Although the viral loads were less than those of respiratory samples (range 550 copies per mL to 1·21 × 105 copies per mL), precautionary measures should be considered when handling faecal samples.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Epidemiological, clinical and virological characteristics of 74 cases of coronavirus-infected disease 2019 (COVID-19) with gastrointestinal symptoms

              Objective The SARS-CoV-2-infected disease (COVID-19) outbreak is a major threat to human beings. Previous studies mainly focused on Wuhan and typical symptoms. We analysed 74 confirmed COVID-19 cases with GI symptoms in the Zhejiang province to determine epidemiological, clinical and virological characteristics. Design COVID-19 hospital patients were admitted in the Zhejiang province from 17 January 2020 to 8 February 2020. Epidemiological, demographic, clinical, laboratory, management and outcome data of patients with GI symptoms were analysed using multivariate analysis for risk of severe/critical type. Bioinformatics were used to analyse features of SARS-CoV-2 from Zhejiang province. Results Among enrolled 651 patients, 74 (11.4%) presented with at least one GI symptom (nausea, vomiting or diarrhoea), average age of 46.14 years, 4-day incubation period and 10.8% had pre-existing liver disease. Of patients with COVID-19 with GI symptoms, 17 (22.97%) and 23 (31.08%) had severe/critical types and family clustering, respectively, significantly higher than those without GI symptoms, 47 (8.14%) and 118 (20.45%). Of patients with COVID-19 with GI symptoms, 29 (39.19%), 23 (31.08%), 8 (10.81%) and 16 (21.62%) had significantly higher rates of fever >38.5°C, fatigue, shortness of breath and headache, respectively. Low-dose glucocorticoids and antibiotics were administered to 14.86% and 41.89% of patients, respectively. Sputum production and increased lactate dehydrogenase/glucose levels were risk factors for severe/critical type. Bioinformatics showed sequence mutation of SARS-CoV-2 with m6A methylation and changed binding capacity with ACE2. Conclusion We report COVID-19 cases with GI symptoms with novel features outside Wuhan. Attention to patients with COVID-19 with non-classic symptoms should increase to protect health providers.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Public Health
                Front Public Health
                Front. Public Health
                Frontiers in Public Health
                Frontiers Media S.A.
                2296-2565
                08 May 2020
                2020
                08 May 2020
                : 8
                : 186
                Affiliations
                [1] 1Materno-Fetal and Obstetrics Research Unit, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne , Lausanne, Switzerland
                [2] 2Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne , Lausanne, Switzerland
                [3] 3Food and Nutritional Sciences, St Joseph's Hospital, The University of Reading , Reading, United Kingdom
                [4] 4Department of Microbiology and Immunology, The University of Western Ontario , London, ON, Canada
                [5] 5Canadian R&D Centre for Human Microbiome and Probiotics, Lawson Health Research Institute , London, ON, Canada
                Author notes

                Edited by: Zisis Kozlakidis, International Agency for Research on Cancer (IARC), France

                Reviewed by: Imad Omar Al Kassaa, Lebanese University, Lebanon; Irene Lenoir-Wijnkoop, Utrecht University, Netherlands

                *Correspondence: Eric Giannoni eric.giannoni@ 123456chuv.ch

                This article was submitted to Infectious Diseases - Surveillance, Prevention and Treatment, a section of the journal Frontiers in Public Health

                Article
                10.3389/fpubh.2020.00186
                7227397
                32574289
                e3d20f4e-4a20-4866-9d31-94cb6220488a
                Copyright © 2020 Baud, Dimopoulou Agri, Gibson, Reid and Giannoni.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 09 April 2020
                : 24 April 2020
                Page count
                Figures: 0, Tables: 1, Equations: 0, References: 50, Pages: 5, Words: 3921
                Categories
                Public Health
                Opinion

                covid 19,probiotics,prebiotics,sars-cov-2,pandemics,coronavirus,respiratory infection

                Comments

                Comment on this article