Are probiotics and prebiotics safe for use during pregnancy and lactation? A systematic review and meta-analysis ================================================================================================================ * Hauna Sheyholislami * Kristin L. Connor ## Abstract Probiotic and prebiotic products are increasingly popular due to their potential health benefits, including in pregnancy where probiotic supplementation has been associated with prevention of gestational diabetes and mastitis. The incidence of adverse effects in pregnant people and their infants associated with probiotic, prebiotic and/or synbiotic product intake, however, remains unclear. The objectives of this study were to evaluate the evidence on adverse effects of maternal probiotic, prebiotic and/or synbiotic supplementation before and during pregnancy and lactation and interpret the findings to help inform clinical decision-making and care of pre-pregnant, pregnant and lactating people. A systematic review was conducted following PRISMA guidelines. Scientific databases were searched using pre-determined terms, and risk of bias assessments were conducted to determine study quality. Inclusion criteria were English language studies, human studies, access to full-text, and probiotic/prebiotic/synbiotic supplementation to the mother and not the infant. 11/70 eligible studies reported adverse effects and were eligible for inclusion in quantitative analysis, and data were visualised in a GOfER diagram. Probiotic and prebiotic products are safe for use during pregnancy and lactation. Only one study reported increased risk of vaginal discharge and changes in stool consistence (Relative Risk [95% CI]: 3.67 [1.04, 12.2]) when administering *Lactobacillus rhamnosus* and *L. reuteri*. Adverse effects associated with probiotic and prebiotic use do not pose any serious health concerns to mother or infant. Our findings and knowledge translation visualisations provide healthcare professionals and consumers with information to make evidence-informed decisions about the use of pre- and probiotics. Keywords * Pregnancy * infant * probiotics * prebiotics * synbiotics * safety * adverse effects ## Introduction The use and acceptance of probiotic products is increasing globally due to their documented health benefits1,2. Probiotics are live microorganisms that, when administered in adequate amounts, confer a health benefit on the host3. Prebiotics, compounds that induce growth or activity of beneficial microorganisms, such as bacteria and fungi4, and synbiotics, combinations of probiotics and prebiotics5, are also products that have the ability to influence an individual’s microbiomes4,5. In non-pregnant individuals, probiotics have been shown to support a healthy gut and digestive tract by treating or preventing infectious and persistent diarrhea6-8, irritable bowel syndrome6, abdominal pain and bloating9, and necrotizing enterocolitis10. Additionally, probiotics provide health benefits in pregnancy, such as the prevention of gestational diabetes11, mastitis12, constipation13, post-partum depression14, and growth of Group B *Streptococcus* bacteria15. Some probiotic products can alter vaginal and breastmilk microbial composition, and therefore have been used to prevent the recurrence of bacterial vaginosis16 and support the gut health of newborn infants by influencing the composition of their gut microbiomes17. Despite some clear benefits, there is a lack of certainty about the effectiveness of probiotics, prebiotics and synbiotics for human health, largely because many studies do not consider that the effects are likely strain-, dose- and condition-specific, and an individual’s response to an intervention may be unique, in part determined by their health status, age, and composition of their gut microbiome18. Critically, many studies fail to report on adverse effects or do not provide specific details regarding adverse effects, such as timing, duration, and severity of symptoms19. Lack of safety data collection and reporting may contribute to unwanted or unexpected health outcomes, which may be of particular concern for vulnerable populations, such as pregnant individuals. Indeed, up-to-date and easy to understand evidence on the safety of probiotic, prebiotic and synbiotic use before and during pregnancy and lactation is currently limited. This is a critical gap in knowledge, given that many physicians consider probiotics to be safe20 but want to learn more about them21,22, and women of childbearing age are highly receptive to taking probiotic-containing products, including to treat gastrointestinal symptoms23. Importantly, studies investigating probiotic, prebiotic and synbiotic use on health outcomes often do not present findings in an easy to understand manner for those who can use or be informed by the data, further contributing to ineffective communication of the efficacy and safety of these products. For these reasons, investigating adverse effect incidence following probiotic, prebiotic or synbiotic supplementation around the time of pregnancy, and synthesizing this evidence in an accessible way, is currently needed to determine whether there are risks that may be associated with their intake for populations that are vulnerable, and to ensure informed decision-making about their potential use to support an individual’s health. This study therefore aimed to 1. systematically synthesize the evidence on adverse effects of maternal probiotic, prebiotic or synbiotic supplementation before and during pregnancy and during lactation; 2. determine if these interventions are safe and 3. interpret findings to help inform clinical decision-making and care of pre-pregnant, pregnant and lactating people. ## Methods Our study adhered to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) guidelines (Supplementary Figure 1 and Supplementary Table 1). View this table: [Supp Table 1.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/T1) Supp Table 1. Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA). This checklist was used as a guideline to determine eligibility criteria, information sources and search strategy, data collection and analysis, and risk of bias assessments and data synthesis. ![Supp. Fig. 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F6.medium.gif) [Supp. Fig. 1.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F6) Supp. Fig. 1. PRISMA flow diagram for article selection and review. ### Information sources and search term A literature search was conducted between June 5 and June 10, 2020 using the search engines Web of Science, ProQuest, PubMed and CINAHL. The following search terms were used to retrieve peer-reviewed articles: (pregnancy OR lactation OR prenatal OR pregnant OR preconception) AND (probiotics OR prebiotics OR synbiotics) AND (safety OR effect OR risk OR adverse OR outcome). This search yielded 1793 articles, of which 1519 articles were retained after reference manager duplication (EndNote basic reference manager), and 1185 articles were retained after manual deduplication (Supplementary Figure 1). ### Inclusion criteria Eligible study designs were randomized control trials (RCTs), case series, controlled clinical trials, case-control studies, cross-sectional studies, and cohort studies. Further inclusion criteria were: 1) English language studies, 2) human studies, 3) access to full-text, and 4) probiotic/prebiotic/synbiotic supplementation to the mother and not the infant. ### Article screening and data collection A 3-level screening process was performed by an independent reviewer. At level 1, 1185 articles were evaluated based on title and abstract, and articles were screened out if they were not in the English language, human studies, or one of the identified eligible study designs. The remaining 244 articles were carried forward for screening at level 2, where full texts were obtained. Articles were excluded if there was no full-text access and if probiotic/prebiotic/synbiotic supplementation was to the infant and not the mother, or to both mother and infant. Articles were further excluded if the population of interest was not pregnant or lactating people, if the information provided was insufficient for the requirements of this study, if supplementation was not probiotic, prebiotic or synbiotic-related, and if the article was not a primary research article, leaving 70 studies. Sample size and country of study information were extracted at this level. At level 3, data were extracted from the 70 studies to capture PICO information (patient/population, intervention [timing/duration, strain/supplement type], comparison, and outcome [timing of outcome, statistics related to outcome]. Our primary outcome of interest was adverse effects reporting. Of the 70 eligible studies, 27 studies reported information on adverse events, and of these, 11 studies reported adverse effects. *Adverse events* were identified as unfavorable or harmful outcomes that occur during or after the use of the intervention, but are not necessarily caused by it, whereas *adverse effects* were identified as adverse events for which the causal relationship between the intervention and the event is at least a reasonable possibility24. Data were obtained for the type, number and timing of adverse effects. In order to determine the magnitude of the adverse effects reported, the number of each incident in both intervention and non-intervention groups was reported as a risk ratio (RR)25. We also noted the primary outcome of interest of the 70 studies under review, and categorized each study based on the primary outcome into 9 groups: pregnancy outcome (e.g. prevention and incidence of preterm delivery, incidence of preeclampsia), maternal metabolic health, maternal microbiome and gastrointestinal health, maternal breast health and breastmilk, maternal mental health, infant metabolic health, infant microbiome and gastrointestinal health, infant allergy and immune health, and infant growth and development (Supplementary Table 2). View this table: [Supp. Table 2.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/T2) Supp. Table 2. Classification for primary outcome groups. ### Data synthesis and visualisation A Graphical Overview for Evidence Reviews (GOfER) diagram26 was created to visualise adverse effects reporting and to synthesize key data collected from studies, including timing of study outcome measurements and adverse effect measurements (if applicable), the number of adverse effect-related incidents in intervention and non-intervention groups, risk ratio calculations, intervention type (single factor intervention or multi-factorial intervention), strain type (if applicable), study design (RCT or cohort), study timeline, primary outcome groups and risk of bias assessment scores. Heatmaps and a world map were created (R Software Version 3.6.1) to represent the quality assessment scores, and the country of study, respectively, for all studies included in level 3. Alluvial diagrams were created (RAWGraphs27) to illustrate the relationships between the intervention types used, and the primary outcome groups of the studies. A bubble plot (JMP Software Version 15.1.0) was created to represent the number of studies that reported a specific adverse effect stratified by intervention type. Finally, a funnel plot28 was created with the log risk ratio values for each of the reported adverse effects against standard error to represent potential publication bias of all studies included in the meta-analysis. ### Methodological quality assessment Articles were assessed for methodological quality according to study design using the following scales: the Cochrane Collaboration’s Tool for Assessing Risk of Bias for randomised controlled trials29 (n=67) and the Newcastle-Ottawa Quality Assessment scale for cohort studies30 (n=3). For RCTs, “other bias” was defined as uptake of the intervention by participants. For cohort studies, comparability of the exposed/intervention group and not exposed/no intervention group was determined based on whether or not the study’s analyses controlled for 1) other dietary intakes and 2) supplement intake, as both dietary intake and other intake of supplements may alter the function(s) of the probiotic/prebiotic/synbiotic supplementation31. Risk of bias summary scores were calculated for each individual study to determine the overall quality of the study by dividing the number of items with “low risk of bias” by the total number of items included in the tool32. Risk of bias summary scores were not calculated for the cohort studies, as the number of cohort studies was low (n=3). ### Data analysis A random effects33 meta-analysis was performed on data from studies that reported the incidence of adverse effects in intervention and control groups (n=11). Risk ratio was chosen as an estimate for effect size33. Statistical significance was confirmed at a=0.05 and results are presented as risk ratio (95% CI). Summary risk ratio statistics were calculated separately for single factor intervention studies and multi-factorial intervention studies. One study (Mirghafourvand et al.) reported adverse effects weekly for four weeks13. The final week of the study was chosen as the clinically important timepoint and was used to calculate the risk ratio of the study, as per Cochrane guidelines34. This is because most of the symptoms reported following probiotic use begin early and taper off upon continuous ingestion of these products35. Additionally, this method allowed for consistent analysis of adverse effects across studies, where in all remaining studies, adverse effects were measured at the end of the intervention period. In another study (Rautava et al.) there were two intervention groups and a single control group. We dealt with this by evenly dividing the number of participants and adverse effects in the control group in half to serve as two separate comparison groups for each intervention group, as per Cochrane guidelines37. Finally, absolute risk difference (ARD) was chosen as a secondary effect estimate38 for studies included in the meta-analysis (n=11) (Supplementary Table 3). Positive ARD suggests decreased risk of an adverse effect, whereas negative ARD suggests increased risk of an adverse effect. View this table: [Supp. Table 3.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/T3) Supp. Table 3. Absolute risk difference (ARD) for studies (n=11) with reported adverse effects. ## Results ### Study location, demographics and design: 70 studies under initial review The articles under review included data from 23 countries (Supplementary Figure 2). Forty-seven percent of studies were from Europe (n=33), 36% from Asia (n=25), 11% from Oceania (n=8), and the remaining 6% from North America, South America and Africa (n=4). Finland and Iran had the highest representation in studies under review (n=16 each). ![Supp. Fig. 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F7.medium.gif) [Supp. Fig. 2.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F7) Supp. Fig. 2. World heatmap illustrating geographical location of each study included in the review. Data are shown for 70 studies from 23 countries. Increasing study counts are represented by darker colours. Fourty-seven percent of studies were from Europe (n=33), 36% from Asia (n=25), 11% from Oceania (n=8), and the remaining 6% from North America, South America and Africa (n=4). Finland and Iran had the highest representation in studies under review, representing 23% each (n=16 each). For the 70 studies under initial review, 36% (n=25, including all three of the observational cohort studies) did not include any information related to safety or adverse events/effects, 26% (n=18) deemed their intervention to be safe (stated that no adverse events/effects were reported or stated that the intervention was safe, without showing safety data), 39% (n=27) reported on adverse events, and of these 27 studies, 16% (n=11) reported on adverse effects. Seventy one percent (n=50) of the 70 studies used a single factor intervention (probiotics [n=37], prebiotics [n=2], synbiotics [n=7], questionnaire [n=2], or fermented foods containing prebiotic [n=1]), and 29% (n=20) used a multi-factorial intervention (diet and probiotic [n=13], multiple probiotic [n=2], vitamin D and probiotic [n=1], complex nutritional supplement and probiotic [n=1], antenatal care and probiotic [n=1], fish oil and probiotic [n=1]) (Supplementary Figure 3). All 11 studies included in the GOfER diagram that reported on adverse effects were randomized controlled trials. Of the 11 studies, 6 included single factor interventions (probiotics [n=5] and prebiotics [n=1]), and 5 included multi-factorial interventions (diet+probiotic [n=1], multiple probiotics [n=2], nutritional supplement+probiotic [n=1], and fish oil+probiotic [n=1]) (Figure 3A). ![Supp. Fig. 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F8.medium.gif) [Supp. Fig. 3.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F8) Supp. Fig. 3. Alluvial Diagram of intervention type and study primary outcomes, showing the flow of weighted links between intervention types and primary outcome groups (all 70 studies captured). ### Methodological quality assessments Among the RCTs under review (n=67), risk of bias assessment summary scores for each individual study showed that 56.7% (n=38) of studies had overall low risk of bias, 29.9% (n=20) of studies had overall unclear/moderate risk of bias, and 13.4% (n=9) of studies had overall high risk of bias (Figure 1). The cohort studies (n=3) largely met all criteria of the quality assessment tool, with the exception of controlling for other dietary intakes category (n=1), controlling for supplement intake category (n=1), and adequacy of follow up category (n=1) (Figure 1). ![Fig. 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F1.medium.gif) [Fig. 1.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F1) Fig. 1. Heatmap of methodological quality assessment scores for A. randomized controlled trials (n=67) and B. cohort studies (n=3). For RCTs, green = low risk of bias, yellow = unclear risk of bias, red = high risk of bias. For cohort studies, green = criteria met, yellow = N/A, and red = criteria not met. ### Characteristics of studies that reported on adverse effects In total, 20 different adverse effects were reported in 11 studies (Figure 2). The most frequently reported adverse effects were gastrointestinal symptoms (type of symptom undefined; n=3), nausea (n=3), diarrhea (n=4) and constipation or bloating (n=3). The majority of the adverse effects were documented in mothers (n=18, vs. n=2 documented in the infant). Of the 20 adverse effect types reported, 16 were associated with probiotic intake, one was associated with prebiotic intake and nine were associated with multi-factorial interventions that included probiotic supplementation (Figure 2). The duration of the interventions varied from the first trimester of pregnancy to one year postpartum depending on the study, with the exception of one study which did not provide any information on the start or end of the intervention39. Of the 11 studies included in the GOfER analysis, only two studies reported the timing of the adverse effect, whereas the other nine studies did not report any timing information (Figure 4). Further, while seven studies were conducted in healthy pregnant individuals, the other four studies were conducted in pregnant individuals with underlying health conditions. These health conditions included lactational mastitis, constipation, overweight/obesity, and intermediate-degree infections/asymptomatic bacterial vaginosis13,40-42. ![Fig. 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F2.medium.gif) [Fig. 2.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F2) Fig. 2. Bubble plot of type and number of adverse effects stratified by intervention type. Size of bubble represents the number of times the given adverse effect was reported for the corresponding intervention across all studies. ![Fig. 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F3.medium.gif) [Fig. 3.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F3) Fig. 3. Alluvial diagrams of A. general intervention type and primary outcome group and B. specific intervention (including probiotic products, prebiotic product, and multi-factorial interventions) and adverse effect type (11 studies with reported adverse effects captured). ![Fig. 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F4.medium.gif) [Fig. 4.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F4) Fig. 4. Summary of studies with reported adverse effects. Study characteristics, intervention type, study timeline, number of adverse effect events in each group and major findings are summarized. Risk ratios (RR) are calculated using the number of events in each experimental group. Red point estimates indicate significant RR. Risk of bias was determined for: A. sequence generat ion, B. allocation concealment, C. blinding of participants/researchers, D. blinding of outcome assessors, E. selective outcome reporting, F. incomplete outcome data, and G. uptake of intervention.![Graphic][1] = high risk of bias, ![Graphic][2]= unclear risk of bias, ![Graphic][3]= low risk of bias. Primary outcome groups for each study are shown: ![Graphic][4]= maternal and infant metabolic health; ![Graphic][5] = maternal and infant microbiome & gastrointestinal health; ![Graphic][6] = maternal breastmilk and breast health; ![Graphic][7] = maternal mental health; ![Graphic][8]= infant allergy & immune health; ![Graphic][9]. = infant growth & behaviour; ![Graphic][10]= pregnancy outcome. PC = Preconception. T1, T2, T3 = First, second, or third trimester of pregnancy. <1Y = From birth to 1 year of infant’s age. 1Y-6Y = From 1 year to 6 years of infant’s age. AE = Adverse effect. GOS = Galactooligosaccharide. FOS = Fructooligosaccharide. 1Events shown under each adverse effect represent the number of events reported at each week of intervention, over the 4 week intervention period. Risk ratio calculated based on week 4 values. 2Does not specify the start time of the intervention, this is why there is no arrow to show the study timeline. Study duration was 21 days. 3Most studies did not indicate when the adverse effect was reported. This is why it is not shown on the study timeline. Various intervention products were used in the studies that reported adverse effects. The probiotic species used were Lactobacillus rhamnosus (n=7), L. reuteri (n=2), L. acidophilus (n=2), Bifidobacterium lactis (n=5), B. longum (n=1), L. paracasei (n=1), L. fermentum (n=1), and L. salivarius (n=1). The prebiotic carbohydrates used were galacto-oligosaccharides (GOS) and fructo-oligosaccharides (FOS) (Figure 3B). Synbiotic products were not used as an intervention in any of the studies that reported on adverse effects. ### Probiotics are safe for use during pregnancy with minimal reported adverse effects Of the six studies that used either probiotics or prebiotics alone as an intervention, one study (Gille et al.) reported a statistically significant increased risk of an adverse effect in the intervention group: maternal intake of a probiotic containing L. rhamnosus and L. reuteri for eight weeks during the first and second trimesters of pregnancy was associated with an increased risk of increased vaginal discharge and changes in stool consistence (RR: 3.67, [1.04, 12.96], Figure 4; ARD: −5.00 [−9.44, −0.55], Supplementary Table 3)43. The methodological quality assessment summary score suggests that this study demonstrated an overall low risk of bias (Figure 1). In the study that reported adverse effects over four weeks (Mirghafourvand et al.), there was no statistically significant increased risk of an adverse effect in the intervention group in the fourth week of the intervention (our chosen clinically important timepoint for analysis). However, the number of adverse effect-related events in the intervention group of this study is much higher during the first week of the intervention, which is expected, given the likely mechanism of action of probiotics within the body35. Analysis of methodological quality assessment suggests that this study also showed an overall low risk of bias (Figure 1). The summary risk ratio statistic of all single factor intervention studies did not suggest an increased relative risk of adverse effect incidence associated with probiotic administration (RR: 0.90 [0.56, 1.47], Figure 4). Of the five articles that used multi-factorial interventions, none of the studies showed a significantly increased risk of any adverse effect associated with multi-factorial probiotic-related supplementation. The summary risk ratio statistic of all multi-factorial intervention studies also did not suggest an increased relative risk of adverse effect incidence associated with multi-factorial probiotic-related supplementation (RR: 1.30 [0.90, 1.89], Figure 4). Finally, low to moderate heterogeneity of the observed effect sizes across the studies was found (I2=37.38%). To examine this heterogeneity, an Egger regression analysis was conducted. Visual inspection of the funnel plot showed little asymmetry for studies included in the meta-analysis, and no publication bias was found across the included studies (Begg’s test, p=0.217; Figure 5), suggesting that the studies included in our meta-analysis are a representative sample of the available evidence. ![Fig. 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/25/2021.01.19.21250133/F5.medium.gif) [Fig. 5.](http://medrxiv.org/content/early/2021/01/25/2021.01.19.21250133/F5) Fig. 5. Funnel plot of log risk ratio values against standard error for each study with reported adverse effects (n=11 studies). The vertical line represents the summary effect size estimates (log risk ratio), and the slanted lines are pseudo 95% CIs. The combined effect size (CSE) is the log risk ratio of all studies combined. The adjusted combined effect size and accompanying confidence and prediction intervals in this plot represent the results of a trim-and-fill procedure. These adjusted statistics are based upon the set of initially included studies expanded with the imputed data points. Funnel plot shows little asymmetry. ## Discussion Here we describe the first systematic review and meta-analysis to report on adverse effects in individuals taking probiotic, prebiotic or synbiotic supplements before, during or after pregnancy and during lactation. We present these findings in an easy-to-understand manner to equip knowledge users to make evidence-informed decisions about the potential risks and documented benefits associated with probiotic and prebiotic intake for pre-pregnant, pregnant and lactating people. We found no mortality or serious adverse effects associated with intake of probiotics, prebiotics or multi-factorial interventions including probiotics in pregnant people. Twenty different adverse effects were reported in 11 studies that used probiotics, prebiotics or probiotic-related multi-factorial interventions. Most adverse effects were related to gastrointestinal health in mothers during the third trimester of pregnancy. Maternal intake of a probiotic product alone was associated with increased risk of increased vaginal discharge and changes in stool consistence in one study, although the increased risk of these adverse effects was minimal. Our data suggest that supplementation with probiotic and prebiotic products is relatively safe for use during and after pregnancy and during lactation, and is not associated with any serious health outcomes in the mother or infant. Our evidence synthesis revealed that most reported adverse effects were related to gastrointestinal health, and the majority were reported in the mother. In non-pregnant populations, adverse effects related to probiotic use have been reported, and include gastrointestinal side effects19 such as abdominal cramping, nausea, soft stools, flatulence and taste disturbance. These gastrointestinal symptoms are consistent with those documented in our study and suggest that presentation of these adverse effects following intake of some probiotics is not specific to pregnant individuals. Furthermore, while experiencing gastrointestinal-related adverse effects such as nausea, diarrhea, or vomiting during pregnancy may cause discomfort, gastrointestinal symptoms are common and expected during pregnancy44, and our analysis suggests that these effects are not increased due to consumption of probiotics or prebiotics. The most frequent gastrointestinal conditions in pregnancy are nausea and vomiting, which affect 50-80% of women44, followed by gastroesophageal reflux disease and constipation44. Although our meta-analysis showed slight increased risk of increased vaginal discharge and changes in stool consistence, these conditions can usually be managed with lifestyle and dietary modifications, and without the use of medications, under guidance of a physician. Because there are probiotic products with documented clinical benefits in pregnancy, such as preventing or treating gestational diabetes11 and mastitis12, and these products may contribute to an overall improved health status for pre-pregnant, pregnant and postpartum patients, their benefits likely outweigh the documented minimal risks. Nevertheless, it must be acknowledged that some women may not wish to experience the adverse effects associated with these products for long periods of time, especially if the symptom onset is severe. Reproductive aged women are one of the most common groups to take probiotics23, and the most common consumer concerns regarding their use are related to potential side effects and efficacy1,2. Additionally, many individuals are unaware of prebiotic products and how they function to benefit health2. Individuals may therefore turn to their healthcare providers for information on the health benefits and safety of these products, especially in countries where product information on the packaging is prohibited45. Yet, studies show that many healthcare professionals only have a medium understanding of what probiotics are and how they work, although they believe probiotics are somewhat beneficial to health and are not harmful21,46. This suggests that there is a need for knowledge dissemination tools to summarise and translate efficacy and safety information of probiotics and prebiotics to consumers and healthcare providers. Indeed, most healthcare professionals want to learn more about these products21,22. Our data visualisations, including the GOfER diagram, provide this information in an accessible manner, empowering the individual and care provider to make evidence-informed decisions for maternal-child health. Our up-to-date analysis of the safety of probiotic and prebiotic use before and during pregnancy and lactation and assessment of study quality is especially valuable given the prior lack of safety data and the number of studies with inadequate design and/or reporting. Our study complements critical existing efforts to translate scientific evidence on probiotic products into clinically relevant information that can be used to make evidence-informed clinical decisions for both healthcare providers and their patients47. The mechanism underlying adverse effect occurrence following probiotic and prebiotic administration is not completely clear. Most of the studies we reviewed that reported adverse effects used combinations of probiotic strains or prebiotics, making it difficult to ascertain whether the adverse effects occurred as a result of one strain alone, or complex interventions. Additionally, the timing and duration of intervention administration varied between each study, ranging from the first trimester of pregnancy, to one year postpartum. To better understand the factors contributing to adverse effects associated with probiotic intake, future research should assess individual strain, dose- and timing-specific effects on safety outcomes in pregnancy and postpartum. Additionally, none of the studies reporting on safety outcomes administered synbiotics, suggesting that there is a need for research on, and reporting of, adverse effects related to synbiotic intake, and also related to multi-factorial interventions during pregnancy and lactation. Despite the fact that our evidence review fills a major knowledge gap on safety, there are limitations to some of the studies that were captured. Firstly, while more than half of the RCT studies included in our analysis had an overall low risk of bias, 43.3% had moderate or high risk of bias. Secondly, of the 11 studies that reported adverse effects, the majority did not note the timing or duration of these adverse effects, along with the severity of the specified symptoms, which is concerning as this information is critical to understanding the overall nature of these adverse effects and how they may affect individuals who consume them. Thirdly, less than half of these 11 studies were conducted in pregnant populations with underlying health conditions, including lactational mastitis, bacterial vaginosis, overweight or obesity and history of allergic disease. Current research in non-pregnant populations show that an individual’s gut microbial composition may be influenced by the presence of underlying health conditions, such as inflammatory bowel disease, irritable bowel syndrome, obesity, type 2 diabetes and atopy48. Further, an individual’s gut microbiome can determine how drugs are metabolized, which varies on a person-to-person basis49,50. Because these underlying health conditions have the ability to alter an individual’s gut microbiome, and that probiotic-gut microbiome interactions, like drug-gut microbiome interactions, are likely highly individualized, it is plausible that the efficacy and adverse effects of a pro- or prebiotic intervention will in part be determined by host health status and gut microbiome composition and function. For this reason, a better understanding of the safety of probiotics and prebiotics is not only needed for healthy pregnant and lactating individuals, but also those with underlying health conditions. The global probiotics market size was estimated USD 51.12 billion in 201951, and will continue to grow rapidly over time due to increased interest of these products. While this increase is mostly driven by women, who are more likely to take probiotics than men2,52, overall consumer interest is increasing, including about the effectiveness of, and side effects associated with, probiotic consumption1,2. This is especially important for women of childbearing age, who may choose to take probiotics during pregnancy without being aware of the safety of these products. Further, clinicians would like to know more about probiotic products22,23 to translate this knowledge to their patients and aid them in making evidence-informed clinical decisions. Our evidence review and meta-analysis of the available data suggest a preliminary set of recommendations for research and clinical consideration: * Researchers should provide detailed and specific safety and adverse effects reporting plans prior to conducting a study, including information and data on: * Intervention characteristics: strain type, dosage, duration (e.g. relative to gestational age) * Participant information: age, underlying health conditions, diet * Adverse effects: type, timing, symptoms, severity, duration, withdrawal due to intervention * Results of studies, including detailed efficacy and safety data, should be published using accessible language and easy to interpret visuals that can be used by diverse audiences * Clinicians and consumers should refer to available scientific evidence47 on efficacy of probiotic products and their associated adverse effects when making decisions about their use or impact on patient health, and clinicians should disseminate safety information to patients accordingly Care of prepregnant and pregnant people using or intending to use probiotics, prebiotics or synbiotics can be better managed if we know these products are safe, and the potential adverse effects can be prevented or managed with lifestyle modifications. Prior to this review, comprehensive knowledge on adverse effects associated with probiotic and prebiotic intake in pregnancy and in the postpartum/lactation periods was limited. Scarce and ineffective knowledge translation further limits evidence-informed decision-making about probiotic and prebiotic use, which may impact patient care and an individual’s health. Our study provides a knowledge translation tool for reporting safety outcomes associated with probiotic and prebiotic use in vulnerable populations that are highly receptive to these products. Our analyses suggest that probiotics and prebiotics are safe to use before, during and after pregnancy and during lactation. ## Data Availability N/A ## Author Contributions and Notes H.S. and K.L.C. conceptualised the ideas presented in this paper, planned the methodology, and investigated the evidence. H.S. analysed the data and drafted the manuscript with K.L.C. Both authors contributed to the review and revision of the final paper. The authors declare no conflict of interest. ## Acknowledgments This research received no specific grant from any funding agency, commercial or not-for-profit sectors. * Received January 19, 2021. * Revision received January 19, 2021. * Accepted January 25, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Chin-Lee, B., Curry, W.J., Fetterman, J., Graybill, M.A. & Karpa, K. Patient experience and use of probiotics in community-based health care settings. Patient Prefer Adherence 8, 1513–1520 (2014). 2. 2.Betz, M., et al. Knowledge, use and perceptions of probiotics and prebiotics in hospitalised patients. Nutrition & Dietetics 72, 261–266 (2015). 3. 3.Hill, C., et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nature Reviews Gastroenterology & Hepatology 11, 506–514 (2014). 4. 4.Gibson, G.R., et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nature Reviews Gastroenterology & Hepatology 14, 491–502 (2017). 5. 5.Swanson, K.S., et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nature Reviews Gastroenterology & Hepatology (2020). 6. 6.Allen, S.J., Martinez, E.G., Gregorio, G.V. & Dans, L.F. Probiotics for treating acute infectious diarrhoea. Cochrane Database of Systematic Reviews, N.PAG-N.PAG (2010). 7. 7.Bernaola Aponte, G., Bada Mancilla, C.A., Carreazo, N.Y. & Rojas Galarza, R.A. Probiotics for treating persistent diarrhoea in children. Cochrane Database of Systematic Reviews (2013). 8. 8.Goldenberg, J.Z., et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database of Systematic Reviews (2013). 9. 9.Ritchie, M.L. & Romanuk, T.N. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS One 7, e34938 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0034938&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22529959&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 10. 10.AlFaleh, K., Anabrees, J., Bassler, D. & Al-Kharfi, T. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database of Systematic Reviews (2011). 11. 11.Chen, X., Jiang, X., Huang, X., He, H. & Zheng, J. Association between Probiotic Yogurt Intake and Gestational Diabetes Mellitus: A Case-Control Study. Iran J Public Health 48, 1248–1256 (2019). 12. 12.Fernández, L., et al. Prevention of Infectious Mastitis by Oral Administration of Lactobacillus salivarius PS2 During Late Pregnancy. Clin Infect Dis 62, 568–573 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/civ974&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26611780&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 13. 13.Mirghafourvand, M.R., A. H. Alizadeh, S. M. Fardiazar, Z. Shokri, K. The Effect of Probiotic Yogurt on Constipation in Pregnant Women: A Randomized Controlled Clinical Trial. Iranian Red Crescent Medical Journal 18(2016). 14. 14.Slykerman, R.F.H., F. Wickens, K. Thompson, J. M. D. Barthow, C. Murphy, R. Kang, J. Rowden, J. Stone, P. Crane, J. Stanley, T. Abels, P. Purdie, G. Maude, R. Mitchell, E. A. Probiotic Pregnancy Study, Grp. Effect of Lactobacillus rhamnosus HN001 in Pregnancy on Postpartum Symptoms of Depression and Anxiety: A Randomised Double-blind Placebo-controlled Trial. Ebiomedicine 24, 159–165 (2017). 15. 15.Ho, M., Chang, Y. Y., Chang, W. C., Lin, H. C., Wang, M. H., Lin, W. C., Chiu, T. H. Oral Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 to reduce Group B Streptococcus colonization in pregnant women: A randomized controlled trial. Taiwanese Journal of Obstetrics & Gynecology 55, 515–518 (2016). 16. 16.Parma, M., Stella Vanni, V., Bertini, M. & Candiani, M. Probiotics in the prevention of recurrences of bacterial vaginosis. Altern Ther Health Med 20 Suppl 1, 52–57 (2014). 17. 17.Korpela, K.S., A. Vepsäläinen, O. Suomalainen, M. Kolmeder, C. Varjosalo, M. Miettinen, S. Kukkonen, K. Savilahti, E. Kuitunen, M. de Vos, W. M. Probiotic supplementation restores normal microbiota composition and function in antibiotic-treated and in caesarean-born infants. Microbiome 6, 182 (2018). 18. 18.van Baarlen, P., et al. Human mucosal in vivo transcriptome responses to three lactobacilli indicate how probiotics may modulate human cellular pathways. Proceedings of the National Academy of Sciences of the United States of America 108 Suppl 1, 4562–4569 (2011). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoyMToiMTA4L1N1cHBsZW1lbnRfMS80NTYyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDEvMjUvMjAyMS4wMS4xOS4yMTI1MDEzMy5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.Doron, S. & Snydman, D.R. Risk and safety of probiotics. Clin Infect Dis 60 Suppl 2, S129–134 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/civ085&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25922398&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 20. 20.Valdovinos-García, L.R., Abreu, A.T. & Valdovinos-Díaz, M.A. Probiotic use in clinical practice: Results of a national survey of gastroenterologists and nutritionists. Revista de Gastroenterología de México (English Edition) 84, 303–309 (2019). 21. 21.Fijan, S., et al. Health Professionals’ Knowledge of Probiotics: An International Survey. Int J Environ Res Public Health 16(2019). 22. 22.Wilson, Z. & Whitehead, K. A cross sectional survey to assess healthcare professionals’ attitudes to and understanding of probiotics. Clinical Nutrition ESPEN 34, 104–109 (2019). 23. 23.Ford, A.C., et al. American College of Gastroenterology monograph on the management of irritable bowel syndrome and chronic idiopathic constipation. Am J Gastroenterol 109 Suppl 1, S2-26; quiz S27 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ajg.2014.187&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25091148&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 24. 24.1. Higgins JPT, T.J., 2. Chandler J, 3. Cumpston M, 4. Li T, 5. Page MJ, 6. Welch VA G, P., S, G., D, J., S, V. & Yk, L. Chapter 19: Adverse effects. in Cochrane Handbook for Systematic Reviews of Interventions version 6.1 (ed. Higgins JPT, T.J., Chandler J, Cumpston M, Li T, Page MJ, Welch VA) (Cochrane, 2020). 25. 25.de Guingand, D.L., Palmer, K.R., Snow, R.J., Davies-Tuck, M.L. & Ellery, S.J. Risk of Adverse Outcomes in Females Taking Oral Creatine Monohydrate: A Systematic Review and Meta-Analysis. Nutrients 12(2020). 26. 26.Stahl-Timmins, W. GOfER Guide v2.6 [Online]. (Peninsula Technology Assessment Group, European Center for Environment and Human Health., Exeter: University of Exeter, [cited 2019 July 9]). 27. 27.Mauri, M., Elli, T., Caviglia, G., Uboldi, G. & Azzi, M. RAWGraphs: A Visualisation Platform to Create Open Outputs.. (p. 28:21-28:25) (In Proceedings of the 12th Biannual Conference on Italian SIGCHI Chapter, New York, NY, USA: ACM., 2017). 28. 28.R, S., van Rhee, H. & T, H. Introduction, comparison and validation of Meta-Essentials : A free and simple tool for meta-analysis. Research Synthesis Methods 8, 537–553 (2017) 29. 29.Higgins, J.P.T., et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 343, d5928 (2011). [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE3OiIzNDMvb2N0MThfMi9kNTkyOCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAxLzI1LzIwMjEuMDEuMTkuMjEyNTAxMzMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 30. 30.Wells, G., et al. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. 31. 31.Andrews, C.N., Sidani, S. & Marshall, J.K. Clinical Management of the Microbiome in Irritable Bowel Syndrome. Journal of the Canadian Association of Gastroenterology (2020). 32. 32.Moseley, A.M., et al. Agreement between the Cochrane risk of bias tool and Physiotherapy Evidence Database (PEDro) scale: A meta-epidemiological study of randomized controlled trials of physical therapy interventions. PLoS One 14, e0222770 (2019). 33. 33.de Guingand, D.L., Palmer, K.R., Snow, R.J., Davies-Tuck, M.L. & Ellery, S.J. Risk of Adverse Outcomes in Females Taking Oral Creatine Monohydrate: A Systematic Review and Meta-Analysis. Nutrients 12, 1780 (2020). 34. 34.1. Higgins JPT, 2. Thomas J, 3. Chandler J, 4. Cumpston M, 5. Li T, 6. Page MJ, 7. Welch VA (editors Higgins JPT, Li T, Deeks JJ (editors). Chapter 6: Choosing effect measures and computing estimates of effect. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (editors). Cochrane Handbook for Systematic Reviews of Interventions version 6.1 (updated September 2020). Cochrane, 2020. Available from [www.training.cochrane.org/handbook](http://www.training.cochrane.org/handbook). 35. 35.Ciorba, M.A. A Gastroenterologist’s Guide to Probiotics. Clinical Gastroenterology and Hepatology 10, 960–968 (2012). 36. 36.Rautava, S.K., E. Salminen, S. Isolauri, E. Maternal probiotic supplementation during pregnancy and breast-feeding reduces the risk of eczema in the infant. Journal of Allergy and Clinical Immunology 130, 1355–1360 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2012.09.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23083673&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000311641100016&link_type=ISI) 37. 37.1. Higgins JPT, 2. Thomas J, 3. Chandler J, 4. Cumpston M, 5. Li T, 6. Page MJ, 7. Welch VA (editors Higgins JPT, Eldridge S, Li T (editors). Chapter 23: Including variants on randomized trials. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (editors). Cochrane Handbook for Systematic Reviews of Interventions version 6.1 (updated September 2020). Cochrane, 2020. Available from [www.training.cochrane.org/handbook](http://www.training.cochrane.org/handbook). 38. 38.Sackett, D.L., Rosenberg, W.M., Gray, J.A., Haynes, R.B. & Richardson, W.S. Evidence based medicine: what it is and what it isn’t. Bmj 312, 71–72 (1996). [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjExOiIzMTIvNzAyMy83MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAxLzI1LzIwMjEuMDEuMTkuMjEyNTAxMzMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 39. 39.Arroyo, R., Martín, V., Maldonado, A., Jiménez, E., Fernández, L., Rodríguez, J. M. Treatment of infectious mastitis during lactation: antibiotics versus oral administration of Lactobacilli isolated from breast milk. Clin Infect Dis 50, 1551–1558 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/652763&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20455694&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000277806200001&link_type=ISI) 40. 40.Martín, V.C., N. Ocaña, S. Marín, M. Arroyo, R. Beltrán, D. Badiola, C. Fernández, L. Rodríguez, J. M. Rectal and Vaginal Eradication of Streptococcus agalactiae (GBS) in Pregnant Women by Using Lactobacillus salivarius CECT 9145, A Target-specific Probiotic Strain. Nutrients 11(2019). 41. 41.Pellonpera, O.M., K. Houttu, N. Vahlberg, T. Koivuniemi, E. Tertti, K. Ronnemaa, T. Laitinen, K. Efficacy of Fish Oil and/or Probiotic Intervention on the Incidence of Gestational Diabetes Mellitus in an At-Risk Group of Overweight and Obese Women: A Randomized, Placebo-Controlled, Double-Blind Clinical Trial. Diabetes Care 42, 1009–1017 (2019). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czo5OiI0Mi82LzEwMDkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wMS8yNS8yMDIxLjAxLjE5LjIxMjUwMTMzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 42. 42.Krauss-Silva, L.M., M. E. L. Alves, M. B. Braga, A. Camacho, K. G. Batista, M. R. R. Almada-Horta, A. Rebello, M. R. Guerra, F. A randomised controlled trial of probiotics for the prevention of spontaneous preterm delivery associated with bacterial vaginosis: preliminary results. Trials 12(2011). 43. 43.Gille, C., Boer, B., Marschal, M., Urschitz, M. S., Heinecke, V., Hund, V., Speidel, S., Tarnow, I., Mylonas, I., Franz, A., Engel, C., Poets, C. F. Effect of probiotics on vaginal health in pregnancy. EFFPRO, a randomized controlled trial. American Journal of Obstetrics and Gynecology 215(2016). 44. 44.Body, C. & Christie, J.A. Gastrointestinal Diseases in Pregnancy: Nausea, Vomiting, Hyperemesis Gravidarum, Gastroesophageal Reflux Disease, Constipation, and Diarrhea. Gastroenterol Clin North Am 45, 267–283 (2016). 45. 45.Pettoello-Mantovani, M., et al. Pilot study for the understanding and use of probiotics by different paediatric healthcare professionals working in different European countries. Italian Journal of Pediatrics 45, 57 (2019). 46. 46.Oliver, L., Rasmussen, H., Gregoire, M.B. & Chen, Y. Health Care Providers’ Knowledge, Perceptions, and Use of Probiotics and Prebiotics. Topics in Clinical Nutrition 29, 139–149 (2014). 47. 47.Skokovic-Sunjic, D. Clinical Guide to Probiotic Products Available in Canada. Indications, Dosage Forms, and Clinical Evidence to Date - 2020 Edition.. Vol. 2020 (2020). 48. 48.Bull, M.J. & Plummer, N.T. Part 1: The Human Gut Microbiome in Health and Disease. Integr Med (Encinitas) 13, 17–22 (2014). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26770121&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 49. 49.Zimmermann, M., Zimmermann-Kogadeeva, M., Wegmann, R. & Goodman, A.L. Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 570, 462–467 (2019). [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F25%2F2021.01.19.21250133.atom) 50. 50.Javdan, B., et al. Personalized Mapping of Drug Metabolism by the Human Gut Microbiome. Cell 181, 1661-1679.e1622 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2020.05.001&link_type=DOI) 51. 51.Probiotics Market Size, Share & Trends Analysis Report By Product (Food & Beverages, Dietary Supplements), By Ingredient (Bacteria, Yeast), By End Use, By Distribution Channel, And Segment Forecasts, 2019 - 2025. (2019). 52. 52.Yilmaz-Ersan, L., Ozcan, T. & Akpinar-Bayizit, A. Assessment of socio-demographic factors, health status and the knowledge on probiotic dairy products. Food Science and Human Wellness 9, 272–279 (2020). [1]: F4/embed/inline-graphic-1.gif [2]: F4/embed/inline-graphic-2.gif [3]: F4/embed/inline-graphic-3.gif [4]: F4/embed/inline-graphic-4.gif [5]: F4/embed/inline-graphic-5.gif [6]: F4/embed/inline-graphic-6.gif [7]: F4/embed/inline-graphic-7.gif [8]: F4/embed/inline-graphic-8.gif [9]: F4/embed/inline-graphic-9.gif [10]: F4/embed/inline-graphic-10.gif