1,275
Views
0
CrossRef citations to date
0
Altmetric
Research Article

A Review of Probiotic Ingredient Safety Supporting Monograph Development Conducted by the United States Pharmacopeia (USP)

, , &

Abstract

The United States Pharmacopeia (USP) is an independent, nonprofit science-based organization whose mission is to improve global health through public standards and related products for medicines, food and dietary supplements. Probiotic-based dietary supplements are increasingly popular in the marketplace and USP has developed fourteen monographs specific to probiotic ingredients, including representatives from the Genera Lactobacillus, Bacillus, Streptococcus, and Bifidobacterium. These monographs include the definition of the article, tests for identification, quantification assays (enumeration in the case of probiotics), limits for contaminants, and other quality parameters when appropriate. In addition to quality, the USP also considers the safety of probiotics for monograph development. This report includes an overview of the USP admission evaluation process for probiotics as well as a tabular summary of the probiotic monographs currently available. Pharmacopeia monographs can guide manufacturers and brand owners and protect consumers through establishment of quality standards.

Introduction

The United States Pharmacopeia (USP) is an independent, nonprofit science-based organization whose mission is to improve global health through public standards and related products for medicines, food and dietary supplements (Oketch-Rabah et al. Citation2018). In so doing, the USP serves to improve global health through this standard setting process. Within dietary supplements, the USP has developed numerous monographs for probiotic ingredients and products over the past several years. In fact, due to the increasing number of probiotic strains proposed for monograph development, the USP established a Probiotic Expert Panel (EP) to advise on quality requirements and safety considerations specific for probiotics. To date, the Probiotic EP has published several scientific peer-reviewed manuscripts to help guide manufacturers of probiotic dietary supplements. In addition, there are USP general chapters that should also be used by anyone developing probiotic dietary supplements. A summary of these references and general chapters are provided in .

Table 1. A summary of USP peer-reviewed scientific references and general chapters related to quality requirements and safety considerations specific to probiotics.

The USP process of developing standards for dietary ingredients including probiotics begins with an ‘admission evaluation’ process conducted by the Dietary Supplement Admission Evaluation and Labeling Expert Committee (DSAEL EC). Included in the evaluation process is information on the identity and quality of material under evaluation, toxicity data, human clinical safety studies, relevant in vitro or mechanistic studies, in silico data and adverse event (AE) data (Oketch-Rabah et al. Citation2018). The probiotic monographs, once developed, include the definition of the article, tests for identification at the strain level, enumeration assays (quantification), limits for contaminants, and other quality parameters when appropriate.

The scope of this report includes an overview of the USP admission evaluation process for probiotics as well as a tabular summary of the various probiotic monographs developed to date. As a note to the reader of this article, by probiotic we are referring to the formal definition as established by the Joint FAO/WHO Expert Consultation which is as follows: Probiotics are live microorganisms that, when administered in adequate amounts, confer a health benefit on the host (Hill et al. Citation2014, FAO/WHO Citation2001). Additionally, data from unpublished self-affirmed Generally Recognized as Safe (GRAS) documents for which FDA did not have any questions about the GRAS self-determination based on scientific procedures for use as an ingredient in the specified conventional foods and dietary supplements were used in some cases to support conclusions on safety endpoints.

Comprehensive quality and safety assessment of USP probiotic monographs

As of the publishing of this report there are a total of 14 USP Probiotic Monographs (containing 19 strains for which admission evaluation was done) within the Dietary Supplements Compendium (DSC) which are summarized in . A brief description of the quality and safety elements included in USP’s review of probiotics under consideration for monograph development is provided below.

Table 2. A summary table of current USP probiotic monographs.

The USP pathway for setting standard does not differ based on the safety evaluation pathway. Although, the magnitude of the quality and safety assessment of probiotic strains relies upon the extensiveness of the documentation provided by the company (known as the sponsor) providing data to support monograph development. The evaluation is either comprehensive or abbreviated.

A comprehensive evaluation is performed in absence of a GRAS dossier or New Dietary Ingredient (NDI) notification that has been submitted to FDA for the probiotic strain. In some cases, a GRAS dossier may become available after a monograph is already in development. This is the case for Lacticaseibacillus paracasei Lpc-37 ().

USP researches and evaluates diverse sources of safety information including historical use of probiotic species and/or strain, global pharmacopeial or regulatory authority monographs, approved lists such as the Qualified Presumption of Safety (QPS) list from the European Food Safety Authority (EFSA) (EFSA Citation2021), non-clinical (e.g. toxicology) studies, human clinical trials, case studies, and adverse event reporting databases.

The abbreviated admission evaluation path is often taken when a GRAS dossier or NDI notification has been submitted to the FDA for review and not objected to by the Agency. Both dossiers generally contain the required information to assess the safety of the probiotic strain. Although, a comprehensive evaluation may still be performed in the event the dossiers lack safety and quality information required for the evaluation.

Identification

The first step in evaluating a probiotic is in careful identification of the microorganism. Thus, its taxonomic status should be unambiguously established. The organism under assessment should be identified at species level based on up-to-date methodologies and current knowledge. For probiotic bacteria, whole genome sequence (WGS) analysis is required for the identification and characterization of the microorganism. This can be achieved by computational approaches for taxonomic assignments (e.g. phylogenomics or average nucleotide identity (ANI)), or by comparing the sequences commonly used for taxonomic identification (e.g. 16S rRNA gene), or other characteristic genes (e.g. housekeeping genes) to relevant databases (Roe et al. Citation2022; EFSA Citation2018; Health Canada Citation2023). The sponsor must provide documentation indicating the sequencing strategy and instrumentation used, the assembly method applied (bioinformatic approach, de novo or re-sequencing strategy, statistical measure of sequence quality, number of contigs and their length, and the annotation protocol used). Additionally, the documentation must demonstrate a clear comparison between the genome of the strain under investigation and the genomes of closely related type strains, unequivocally affirming the identification of the species to which the strain belongs.

In addition to the genotypic characterization, the probiotic strain must be phenotypically characterized. Phenotyping must be assessed based on characteristics routinely used to distinguish the species from others. This includes a series of testing for sufficient confirmation of observable traits of the species such as Gram classification, motility, presence of spores, bacterial morphology, cluster type (chain, pair, single), aerobiosis status, fermentation pathway and biochemical profile using Analytical Profile Index (API).

Safety evaluation

While global regulatory authorities generally concur on the fundamental safety principles for probiotics, there exists limited consensus regarding the precise types of studies and methodologies to validate their safety. When assessing the safety of a probiotic strain, USP investigates the following: in silico analysis to verify for the presence of antibiotic resistance genes, virulence and toxigenic factors, history of safe use of the species in food and dietary supplements, any available toxicology studies (in vitro and in vivo), clinical studies (if safety parameters are included), case studies, and adverse event (AE)reporting data. These parameters are discussed in more detail below.

The WGS serves not only as a tool for identification as discussed above, but also plays a crucial role in safety assessment. It is imperative to choose probiotic strains that do not contribute to the dissemination of functional antibiotic resistance in food and supplements (Ouwehand et al. Citation2016). The potential for antibiotic resistance in probiotic strains raises concern, particularly if this resistance can be transferred directly or through intermediary microbes to potentially harmful members of the microbiota. This could result in the emergence of antibiotic-resistant pathogens that are difficult to treat. The revised EFSA guidance from 2018 emphasizes the necessity for conducting both a phenotypic (Minimum Inhibitory Concentration, MIC) and genotypic analysis of antimicrobial resistance. It does not confine the analysis to functional antimicrobial resistance surpassing a predetermined threshold or cutoff (EFSA Citation2018). This approach aligns with the safety standards adopted by numerous countries around the world. In addition to genotypic analysis, the sponsor must also provide phenotypic testing for antimicrobial susceptibility.

A safety assessment must also consider the potential for a probiotic to express any virulence or toxin genes, which could lead to disease. Therefore, the sponsor must provide data showing the strain does not possess genetic elements responsible for producing virulence factors and/or toxigenic activity such as biofilm production, hemolysis, or toxins. Utilizing highly curated databases and genomic-scale analytical resources such as the Pathosystems Resource Integration Center (PATRIC), are good bioinformatic tools enabling in identifying acquired virulence and toxigenic genes (Roe et al. Citation2022).

Traditional toxicology studies are sometimes available for various probiotics under consideration for monograph development. When available, these studies are incorporated into the safety assessment for a probiotic. However, the utility of traditional animal toxicology studies designed for testing chemicals for relevance in assessing probiotic safety has been questioned (Roe et al. Citation2022).

A history of safe use for many microbial species is more commonly used to establish safety for many probiotics. Various genera and species of the family Lactobacillaceae and species of the genus Bifidobacterium for example have been historically consumed in the diet and as such are recognized as safe. The safe use of these species in foods and supplement products worldwide and the occurrence for many as normal commensals of mammalian microbiota adds further to documentation of safety. Furthermore, a well-documented history of use through consumption in large quantities, on a daily basis as fermented foods is recognized by authority bodies such as EFSA (Bernardeau et al. Citation2006; EFSA Citation2007). Thus, the intended use and known intake levels of the probiotic under review is included in the evaluation.

Clinical trials can serve as a source of safety information for a particular probiotic species or strain if safety endpoints are clearly evaluated as part of the study. However, it should be recognized that the majority of human clinical studies with probiotics reported in the literature were not designed as safety and tolerability studies. Thus, adequate reporting of adverse events from the clinical study may not be reported. In addition, there is a lack of consistency in terms of what safety endpoints should be assessed in clinical trials with probiotics. The reporting of adverse events is a critical component of any human clinical trial. A clear definition of what and how adverse events will be collected must be clearly reported in the study report. In order for a clinical trial to be helpful in a review of a probiotic, the adverse events should be fully reported in terms of grade (mild, moderate, severe, life threatening) and a determination on its causal relationship to the treatment is critical (Ioannidis et al. Citation2004).

In addition to adverse events reported from clinical trials a literature search for any case reports of adverse events is also conducted for a given probiotic species/strain. Various international databases for capturing AE reports are also searched including the US FDA Center for Food Safety and Applied Nutrition Adverse Event Reporting (CAERS), Canada Vigilance Program, Australia’s Therapeutic Goods Administration (TGA), World Health Organization (WHO) Uppsala Monitoring Center VigiAccess, Medicines and Healthcare products Regulatory Agency Yellow Card Database, Europe’s EudraVigilance system, and New Zealand’s Medicines and Medical Devices Safety Authority (NZ MedSafe).

Quality parameters of a probiotic monograph review

In addition to review of the safety aspects of the probiotic strain under consideration for monograph development, the quality of the various assays provided by the sponsor are also reviewed.

The nucleic acid-based assay for strain-level identification is the centerpiece of the probiotic monograph. The sponsor is required to provide a comprehensive PCR validation report including details such as the assay design, PCR conditions (including the reaction formula breakdown and PCR amplification cycles), as well as the primer sets along with their corresponding acceptance criteria. The validation report must demonstrate specificity of their primers while using non-targeted samples, strains from other species, and targeted samples, closely related strains from the same species including the strains already published in the species monograph.

In addition to the section for species identification, the species monograph also includes a chapter dedicated to enumeration. Typically, lactobacilli enumeration assays follow the method outlined in Probiotic Tests ˂64>. The same holds true for bifidobacteria, with the additional inclusion of cysteine chloride in the specified media as outlined in the species monograph. For other genera, or if a sponsor necessitates the use of its own enumeration method, the company must provide a scientifically validated enumeration method for its probiotic strain. This should entail details on the peptone diluent, sample preparation, and incubation conditions. The acceptance criteria for this assay are a minimum of 100% of the labeled viable cell count in CFU/g.

By default, the contaminants assays refer to the methods listed under Probiotic Tests ˂64>. The main contaminants are total combined yeasts and molds, non-lactic acid bacteria, Escherichia coli and Salmonella species. The methods of analysis indicated are currently accepted methods commonly used in industry. Although, users may substitute such methods by other validated test methods.

Furthermore, the absence of Listeria monocytogenes, Staphylococcus aureus, or Pseudomonas aeruginosa in addition to the absence of the specified microorganisms mentioned above should be tested and confirmed if a probiotic ingredient or a finished product containing a probiotic ingredient poses a risk associated with the contamination of the above microorganisms based on formal risk assessment programs such as Hazard Analysis and Critical Control Points (HACCP). The absence of Clostridium perfringens and Cronobacter sakazakii in addition to the absence of the listed specified microorganisms should be tested and confirmed if a probiotic ingredient or finished product is intended for infant use. The test methods for Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Clostridium perfringens, and Cronobacter sakazakii must be officially accepted methods or appropriately validated methods.

Discussion

Probiotic dietary supplement products are increasingly popular among consumers. Marketplace projections have estimated that globally, probiotic sales could reach over 90 billion USD by 2026 (MarketsandMarkets Citation2021). With the increase in popularity of this type of dietary supplement it is important that pharmacopeia monographs are available to guide manufacturers and brand owners and to protect consumers through establishment of quality standards. The USP has developed 20 probiotic monographs through an ‘admission evaluation’ conducted by scientific expert volunteers who are members of the DSAEL EC. A summary of those probiotic monographs is presented herein.

The quality and safety parameters reviewed by the DSAEL EC and/or incorporated in to monographs for probiotics are summarized in . The EFSA QPS approach follows this same basic premise for safety assessment of probiotics (EFSA 2020 a and b). More recently, the International Scientific Association for Probiotics and Prebiotics convened a meeting of experts to review similar quality and safety parameters of probiotics. The recommendations of these experts are similarly aligned with the approaches presented herein (Merenstein et al. Citation2023).

Table 3. Essential parameters of a comprehensive quality and safety review for a USP probiotic monograph.

A final consideration by the DSAEL EC is whether to recommend cautionary labeling specific to the probiotic strain under review to mitigate known risk. Any specific cautions by regulatory authorities cited for a particular species or strain would be considered. Other cautionary statements that have been proposed in the literature or by regulatory authorities are related to the use of probiotics by immunocompromised individuals. For example, Sanders et al. (Citation2010) recommends; …’in patients with weakened intestinal barrier function, immune compromised state or central venous catheter use, probiotic consumption should in any case be carefully considered’. Health Canada monograph for probiotics has the following caution statements for the intake of probiotics; ‘Consult a health care practitioner/health care provider/health care professional/doctor/physician prior to use if you have fever, vomiting, bloody diarrhea or severe abdominal painand toStop use and consult a health care practitioner/health care provider/health care professional/doctor/physician if symptoms of digestive upset (e.g. diarrhea) occur, worsen and/or persists beyond 3 days’. In addition, the use of probiotics is contraindicated in immune-compromised individuals with the statement: ‘Do not use this product if you have an immune-compromised condition (e.g. AIDS, lymphoma, patients undergoing long-term corticosteroid treatment)’. To date, the DSAEL EC has not required cautionary labeling statements such as these in any of the probiotic monographs discussed herein. The DSAEL EC would typically base a decision on whether or not to add cautionary labeling statements to monographs based on careful review of adverse event reports.

The final step in the review process conducted by the DSAEL EC is to determine whether a dietary ingredient (e.g. probiotic strain) as a component of a dietary supplement qualifies for admission into the United States Pharmacopeia and National Formulary (USP-NF). The Admission Criteria and Safety Classification for Dietary Supplements Guideline establishes a Class A and Class B classification system that categorizes dietary ingredients according to the level of safety concern (USP Citation2021). Specifically, the safety classification and admission criteria are presented in below.

Table 4. The admission criteria and safety classification for dietary supplements guideline class A and Class B classification system.

Even when a dietary ingredient is classified as Class A and admitted for monograph development, USP continues to monitor for safety information related to that dietary ingredient. This monitoring is ongoing and may serve as a basis for possible safety reevaluation. It should be noted that USP’s evaluation of a dietary supplement ingredient is performed solely for determining admission into the compendia and should not be interpreted as any statement of intrinsic safety or effectiveness of the dietary ingredient under review.

Safety assessments of probiotic bacterial species can be challenging. There is a lack of standardized methodologies for assessing the safety of probiotics. As a result, not all strains have the same level of characterization. USP is working toward addressing the lack of standardized methodologies as evidenced by publications listed in . Additional efforts are ongoing by the USP Probiotics EP and will be published as available.

In summary, this report can serve as an overview of the USP admission evaluation process for probiotics including the key quality and safety parameters that are important for admission to the compendia. The tabular summary of the various probiotic monographs can serve as quick reference of those developed to date.

Authors’ contributions

MEB, NA, BK, and ALR collected and analyzed the data presented in this manuscript. All authors participated in writing aspects of this manuscript and have responsibility for final content.

Acknowledgments

The authors wish to thank Nidhi Kanchan (The Ohio State University College of Pharmacy) and Sarah Smiley (Lake Erie College of Osteopathic Medicine) for their help in developing the tabular summaries of probiotic monographs.

Disclosure statement

The authors are either employed by USP (NA, BK) or currently work (ALR) or have worked (MEB) for companies who manufacture and/or distribute dietary supplements including probiotic products.

Data availability statement

Data described in the manuscript are available through access to the USP-NF at https://www.uspnf.com

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

Notes on contributors

Marie-Eve Boyte

Marie-Eve Boyte is a microbiologist graduated from Université Laval. She is currently the vice-chair of the USP Probiotic Expert Panel and the co-chair of the International Probiotics Association (IPA) Scientific Committee. She has more than 15 years of experience in the dietary supplement industry with strong expertise in the probiotic field.

Nadeem Akhtar

Nadeem Akhtar is a toxicologist holding an MS (Pharm.) in Pharmacology & Toxicology from the National Institute of Pharmaceutical Education and Research (NIPER) in S.A.S. Nagar, Punjab, India, currently working as Senior Scientist II (Toxicology) in Dietary Supplements and Herbal Medicines (DSHM) team at USP. He has more than 9 years of experience in the drug, cosmetics, and dietary supplement areas.

Binu Koshy

Binu Koshy is a microbiologist with a Ph.D. in Microbiology and a Post doctoral research experience from Indian Institute of Science (IISc) Bangalore, India. He is currently working as Senior Scientist II in Dietary Supplements and Herbal Medicines (DSHM) team at USP with more than 15 years’ experience in dietary supplement areas with expertise in probiotics and proteins.

Amy L. Roe

Amy L. Roe is a board-certified (ABT) toxicologist with a PhD in toxicology from the University of Kentucky. She is currently Chair of the USP Dietary Supplements Admission Evaluation and Labeling Expert Committee. She has over 24 years of experience as a practicing toxicologist in industry across both the drug and dietary supplement spaces.

References

  • Aguirre M, Collins MD. 1993. Lactic acid bacteria and human clinical infection. J Appl Bacteriol. 75(2):95–107. doi:10.1111/j.1365-2672.1993.tb02753.x.
  • Aljuobori A, Abdullah N, Zulkifli I, Soleimani A, Liang J, Oskoueian E. 2014. Lactobacillus salivarius fermentation reduced glucosinolate and fibre in canola meal. JFR. 3(5):95. doi:10.5539/jfr.v3n5p95.
  • Allen SJ, Jordan S, Storey M, Thornton CA, Gravenor M, Garaiova I, Plummer SF, Wang D, Morgan G. 2010. Dietary supplementation with lactobacilli and bifidobacteria is well tolerated and not associated with adverse events during late pregnancy and early infancy. J Nutr. 140(3):483–488. doi:10.3945/jn.109.117093.
  • Banerjee C, Bustamante CI, Wharton R, Talley E, Wade JC. 1988. Bacillus infections in patients with cancer. Arch Intern Med. 148(8):1769–1774. doi:10.1001/archinte.1988.00380080059017.
  • Barnard JP, Stinson MW. 1996. The alpha-hemolysin of Streptococcus gordonii is hydrogen peroxide. Infect Immun. 64(9):3853–3857. doi:10.1128/iai.64.9.3853-3857.1996.
  • Barreto FM, Simão ANC, Morimoto HK, Lozovoy MAB, Dichi I, Miglioranza LHS. 2014. Beneficial effects of Lactobacillus plantarum on glycemia and homocysteine levels in postmenopausal women with metabolic syndrome. Nutrition. 30(7-8):939–942. doi:10.1016/j.nut.2013.12.004.
  • Bernardeau M, Guguen M, Vernoux JP. 2006. Beneficial lactobacilli in food and feed: long-term use, biodiversity and proposals for specific and realistic safety assessments. FEMS Microbiol Rev. 30(4):487–513. doi:10.1111/j.1574-6976.2006.00020.x.
  • Borriello SP, Hammes WP, Holzapfel W, Marteau P, Schrezenmeir J, Vaara M, Valtonen V. 2003. Safety of probiotics that contain Lactobacilli or Bifidobacteria. Clin Infect Dis. 36(6):775–780. doi:10.1086/368080.
  • Brettin T, Davis JJ, Disz T, Edwards RA, Gerdes S, Olsen GJ, Olson R, Overbeek R, Parrello B, Pusch GD, et al. 2015. RASTtk: a modular and extensible implementation of the RAST algorithm for building custom annotation pipelines and annotating batches of genomes. Sci Rep. 5(1):8365. doi:10.1038/srep08365.
  • Burton JP, Wescombe PA, Moore CJ, Chilcott CN, Tagg JR. 2006. Safety assessment of the oral cavity probiotic Streptococcus salivarius K12. Appl Environ Microbiol. 72(4):3050–3053. doi:10.1128/AEM.72.4.3050-3053.2006.
  • Burton JP, Cowley S, Simon RR, McKinney J, Wescombe PA, Tagg JR. 2011. Evaluation of safety and human tolerance of the oral probiotic Streptococcus salivarius K12: a randomized, placebo-controlled, double-blind study. Food Chem Toxicol. 49(9):2356–2364. doi:10.1016/j.fct.2011.06.038.
  • Cabana MD, McKean M, Caughey AB, Fong L, Lynch S, Wong A, Leong R, Boushey HA, Hilton JF. 2017. Early probiotic supplementation for eczema and asthma prevention: a randomized controlled trial. Pediatrics. 140(3):e20163000. doi:10.1542/peds.2016-3000.
  • Chaves BD, Brashears MM, Nightingale KK. 2017. Applications and safety considerations of Lactobacillus salivarius as a probiotic in animal and human health. J Appl Microbiol. 123(1):18–28. doi:10.1111/jam.13438.
  • Ciprandi G, Vizzaccaro A, Cirillo I, Tosca MA. 2005. Bacillus clausii exerts immuno-modulatory activity in allergic subjects: a pilot study. Eur Ann Allergy Clin Immunol. 37(4):129–134.
  • Clausen PT, Aarestrup FM, Lund O. 2018. Rapid and precise alignment of raw reads against redundant databases with KMA. BMC Bioinformatics. 19(1):307. doi:10.1186/s12859-018-2336-6.
  • Costa GN, Marcelino-Guimarães FC, Vilas-Bôas GT, Matsuo T, Miglioranza LHS. 2014. Potential fate of ingested Lactobacillus plantarum and its occurrence in human feces. Appl Environ Microbiol. 80(3):1013–1019. doi:10.1128/AEM.02588-13.
  • Coulin P, Farah Z, Assanvo J, Spillmann H, Puhan Z. 2006. Characterisation of the microflora of attiéké, a fermented cassava product, during traditional small-scale preparation. Int J Food Microbiol. 106(2):131–136. doi:10.1016/j.ijfoodmicro.2005.06.012.
  • Cuentas AM, Deaton J, Khan S, Davidson J, Ardita C. 2017. The effect of Bacillus subtilis DE111 on the daily bowel movement profile for people with occasional gastrointestinal irregularity. J Probiotics Health. 5(4):04. doi:10.4172/2329-8901.1000189.
  • Davis JJ, Wattam AR, Aziz RK, Brettin T, Butler R, Butler RM, Chlenski P, Conrad N, Dickerman A, Dietrich E, et al. 2020. The PATRIC Bioinformatics Resource Center: expanding data and analysis capabilities. Nucleic Acids Res. 48(D1):D606–D612. doi:10.1093/nar/gkz943.
  • Delorme C. 2008. Safety assessment of dairy microorganisms: Streptococcus thermophilus. Int J Food Microbiol. 126(3):274–277. doi:10.1016/j.ijfoodmicro.2007.08.014.
  • Dickerson F, Adamos M, Katsafanas E, Khushalani S, Origoni A, Savage C, Schweinfurth L, Stallings C, Sweeney K, Goga J, et al. 2018. Adjunctive probiotic microorganisms to prevent rehospitalization in patients with acute mania: A randomized controlled trial. Bipolar Disord. 20(7):614–621. doi:10.1111/bdi.12652.
  • Di Pierro F, Colombo M, Zanvit A, Rottoli AS. 2016. Positive clinical outcomes derived from using Streptococcus salivarius K12 to prevent streptococcal pharyngotonsillitis in children: a pilot investigation. DHPS. ume 8:77–81. doi:10.1016/j.fct.2011.06.038.
  • Duc LH, Hong HA, Barbosa TM, Henriques AO, Cutting SM. 2004. Characterization of Bacillus Probiotics Available for Human Use. Appl Environ Microbiol. 70(4):2161–2171. doi:10.1128/aem.70.4.2161-2171.2004.
  • EFSA. 2007. Introduction of a Qualified Presumption of Safety (QPS) approach for assessment of selected microorganisms referred to EFSA. doi:10.2903/j.efsa.2007.587. (Accessed 10 November 2023).
  • EFSA. 2012. Guidance on the assessment of bacterial susceptibility to antimicrobials of human and veterinary importance. Efsa J. 10(6):2740.
  • EFSA. 2018. Guidance on the characterisation of microorganisms used as feed additives or as production organisms. Efsa J. 16(3):5206. doi:10.2903/j.efsa.2018.5206.
  • EFSA. 2020a. Scientific Opinion on the update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA (2017-2019.)Efsa J. 18(2):5966. doi:10.2903/j.efsa.2020.5966.
  • EFSA. 2020b. Update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA 13: suitability of taxonomic units notified to EFSA until September 2020. Efsa J. 19(1):6377. doi:10.2903/j.efsa.2021.6377.
  • EFSA. 2021. Qualified presumption of safety (QPS). European food safety authority (EFSA). Available at: https://www.efsa.europa.eu/en/topics/topic/qualified-presumption-safety-qps. (Accessed 10 November 2023).
  • FAO/WHO. 2001. Report of a Joint FAO/WHO expert consultation on evaluation of health and nutritional properties of probiotics in food including poder milk with live lactic acid bacteria. https://www.fao.org/3/ao512e/a0512e.pdf. (Accessed 10 November 2023).
  • FDA, GRN No. 231. 2008. Lactobacillus casei subsp. rhamnosus strain GG. Mead Johnson & Company. Date of closure May 29, 2008. https://www.accessdata.fda.gov/scripts/fdcc/?set=GRASNotices&id=231&sort=GRN_No&order=DESC&startrow=1&type=basic&search=231. Accessed 15 November 2023.
  • FDA, GRN No. 254. 2008. Lactobacillus reuteri strain DSM 17938. BioGaia AB. Date of closure November 18, 2008. https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwjFq4OdmP2CAxW-m2oFHUGJBHkQFnoECBkQAQ&url=https%3A//downloads.regulations.gov/FDA-2021-S-0023-0004/content.pdf&usg=AOvVaw2r7VmeT5CxbIkvPIYYk77u&opi=89978449.
  • FDA, GRN No 288. 2009. Lactobacillus rhamnosus strain HN001 GRAS Notice 288, Lactobacillus rhamnosus strain HN001 (fda.gov). Accessed 20 November 2023
  • FDA, GRN No 865. 2010. Lactobacillus acidophilus NCFM ®. GRAS Noice 865, Lactobacillus acidophilus NCFM (fda.gov). Accessed 15 November 2023.
  • FDA, GRN No 357. 2011. Lactobacillus acidophilus NCFM GRAS Notice 357, Inactivated Lactobacillus acidophilus NCFM (fda.gov). Accessed 20 November 2023
  • FDA, GRN No. 429. 2012. Lactobacillus casei strain Shirota. Yakult Honsha Co., Ltd. Date of closure April 10, 2012. www.accessdata.fda.gov/scripts/fdcc/?set=GRASNotices&id=429&sort=GRN_No&order=DESC&startrow=1&type=basic&search=Lactobacillus%20casei. Accessed 14 November 2023.
  • FDA, GRN No 445. 2013. Bifidobacterium animalis subsp. lactis strains HN019, Bi-07, Bl-04 and B420 GRAS Notice 445, Bifidobacterium animalis subsp. lactis strains HN019, Bi-07, Bl-04 and B420 (fda.gov). Accessed 20 November 2023
  • FDA, GRN No 502. 2014. Lactobacillus acidophilus La-14 GRAS Notice 502, Lactobacillus acidophilus La-14 (fda.gov). Accessed 20 November 2023
  • FDA, GRN No 526. 2015. Bacillus coagulans strain Unique IS2 spores preparation GRAS Notice 526, Bacillus coagulans strain Unique IS2 spores preparation (fda.gov). Accessed 20 November 2023
  • FDA, GRN No. 591. 2015. GRAS Exemption Claim for Streptococcus salivarius K12. http://www.fda.gov/Food/IngredientsPackagingLabeling/GRAS/NoticeInventory/default.htm. Accessed 15 November 2023.
  • FDA, GRN No. 601. 2015. Bacillus coagulans SBC37-01 spore preparation. Sabinsa Corporation. Date of closure April 28, 2016. https://www.cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set=GRASNotices&id=601&sort=GRN_No&order=DESC&startrow=1&type=basic&search=Bacillus. Accessed 13 November 2023.
  • FDA, GRN No. 722. 2018. Lactobacillus plantarum Lp-115. Dupont Nutrition & Health. Date of closure on February 16, 2018. https://www.cfsanappsexternal.fda.gov/scripts/fdcc/?set=GRASNotices&id=722&sort=GRN_No&order=DESC&startrow=1&type=basic&search=LACTOBACILLUS%20PLANTARUM. Accessed 14 November 2023.
  • FDA, GRN No. 807. 2019. Streptococcus salivarius M18. BLIS Technologies Ltd. Date of closure June 9, 2019 https://www.cfsanappsexternal.fda.gov/scripts/fdcc/index.cfm?set=GRASNotices&id=807&sort=GRN_No&order=DESC&startrow=1&type=basic&search=Streptococcus%20salivarius. Accessed 13 November 2023.
  • FDA, GRN No. 831. 2019. FDA has no questions GRAS Notice GRN 831 Agency Response Letter - Bacillus subtilis DE111 (fda.gov). Accessed 15 November 2023.
  • Forsberg KJ, Patel S, Gibson MK, Lauber CL, Knight R, Fierer N, Dantas G. 2014. Bacterial phylogeny structures soil resistomes across habitats. Nature. 509(7502):612–616. doi:10.1038/nature13377.
  • Freedman KE, Hill JL, Wei Y, Vazquez AR, Grubb DS, Trotter RE, Wrigley SD, Johnson SA, Foster MT, Weir TL. 2021. Examining the gastrointestinal and immunomodulatory effects of the novel probiotic Bacillus subtilis DE111. IJMS. 22(5):2453. doi:10.3390/ijms22052453.
  • Gebert S, Davis E, Rehberger T, Maxwell CV. 2011. Lactobacillus brevis strain 1E1 administered to piglets through milk supplementation prior to weaning maintains intestinal integrity after the weaning event. Benef Microbes. 2(1):35–45. doi:10.3920/bm2010.0043.
  • Giske CG, Turnidge J, Cantón R, Kahlmeter G, EUCAST Steering Committee. 2021. Update From The European Committee On Antimicrobial Suseptibility Testing (EUCAST). J Clin Microbiol. 60(3):e0027621. doi:10.1128/jcm.00276-21.
  • Gøbel RJ, Larsen N, Jakobsen M, Mølgaard C, Michaelsen KF. 2012. Probiotics to adolescents with obesity: effects on inflammation and metabolic syndrome. J Pediatr Gastroenterol Nutr. 55(6):673–678. doi:10.1097/mpg.0b013e318263066c.
  • Gomes AC, de Sousa RG, Botelho PB, Gomes TL, Prada PO, Mota JF. 2017. The additional effects of a probiotic mix on abdominal adiposity and antioxidant Status: A double-blind, randomized trial. Obesity (Silver Spring). 25(1):30–38. doi:10.1002/oby.21671.
  • Gueimonde M, Sánchez B, G de Los Reyes-Gavilán C, Margolles A. 2013. Antibiotic resistance in probiotic bacteria. Front Microbiol. 4:202. doi:10.3389/fmicb.2013.00202.
  • Gupta RS, Patel S, Saini N, Chen S. 2020. Robust demarcation of 17 distinct Bacillus species clades, proposed as novel Bacillaceae genera, by phylogenomics and comparative genomic analyses: description of Robertmurraya kyonggiensis sp. nov. and proposal for an emended genus Bacillus limiting it only to the members of the Subtilis and Cereus clades of species. Int J Syst Evol Microbiol. 70(11):5753–5798. doi:10.1099/ijsem.0.004475.
  • Harnett J, Davey G, Patrick A, Caddick C, Pearce L. 2011. Lactic Acid Bacteria | Streptococcus thermophilus. In: Fuquay JW, ed. Encyclopedia of Dairy Sciences (Second Edition). San Diego: Academic Press, p. 143–148.
  • Health Canada. 2022. Probiotics monograph available at http://webprod.hc-sc.gc.ca/nhpid-bdipsn/atReq.do?atid=probio&lang=eng. Accessed 15 November 2023.
  • Health Canada. 2023. Monograph on Probiotics. https://webprod.hc-sc.gc.ca/nhpid-bdipsn/atReq.do?atid=probio&lang=eng. Accessed 31 October 2023.
  • Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, et al. 2014. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 11(8):506–514. doi:10.1038/nrgastro.2014.66.
  • Ioannidis JP, Evans SJ, Gotzsche PC, O’Neill RT, Altman DG, Schulz K, Moher D, Group C, CONSORT Group. 2004. Better reporting of harms in randomized trials: an extension of the CONSORT statement. Ann Intern Med. 141(10):781–788. doi:10.7326/0003-4819-141-10-200411160-00009.
  • Jackson SA, Schoeni JL, Vegge C, Pane M, Stahl B, Bradley M, Goldman VS, Burguière P, Atwater JB, Sanders ME. 2019. Improving end-user trust in the quality of commercial probiotic products. Front Microbiol. 10:739. doi:10.3389/fmicb.2019.00739.
  • N J, Sudha M R. 2015. Bacillus clausii-The Probiotic of Choice in the Treatment of Diarrhoea. J Yoga Phys Ther. 05(04):1. doi:10.4172/2157-7595.1000211.
  • Kim KH, Jeong CH, Joo SJ, Park JH, Moon JY, Cho EJ, Lee HT, Kwon HJ, Kim BW, Eom SH, et al. 2015. Single Dose Oral Toxicity of Bacillus subtilis JNS in ICR Mice. Journal of the Korean Society of Food Science and Nutrition. 44(1):24–28. doi:10.3746/jkfn.2015.44.1.024.
  • Labellarte G, Maher M. 2019. Tolerance and effect of a probiotic supplement delivered in capsule form. Food and Nutrition Sciences. 10(06):626. doi:10.3390/sports6030070.
  • Lakshmi SG, Jayanthi N, Saravanan M, Ratna MS. 2017. Safety assesment of Bacillus clausii UBBC07, a spore forming probiotic. Toxicol Rep,. 4:62–71. doi:10.1016/j.toxrep.2019.06.004.
  • Larsen N, Vogensen FK, Gøbel RJ, Michaelsen KF, Forssten SD, Lahtinen SJ, Jakobsen M. 2013. Effect of Lactobacillus salivarius Ls-33 on fecal microbiota in obese adolescents. Clin Nutr. 32(6):935–940. doi:10.1016/j.clnu.2013.02.007.
  • Lefevre M, Racedo SM, Ripert G, Housez B, Cazaubiel M, Maudet C, Jüsten P, Marteau P, Urdaci MC. 2015. Probiotic strain Bacillus subtilis CU1 stimulates immune system of elderly during common infectious disease period: a randomized, double-blind placebo-controlled study. Immun Ageing. 12(1):24. doi:10.1186/s12979-015-0051-y.
  • Madempudi RS, Neelamraju J, Ahire JJ, Gupta SK, Shukla VK. 2020. Bacillus coagulans Unique IS2 in Constipation: A Double-Blind, Placebo-Controlled Study. Probiotics Antimicrob Proteins. 12(2):335–342. doi:10.1007/s12602-019-09542-9.
  • Mattarelli P, Bonaparte C, Pot B, Biavati B. 2008. Proposal to reclassify the three biotypes of Bifidobacterium longum as three subspecies: Bifidobacterium longum subsp. longum subsp. nov., Bifidobacterium longum subsp. infantis comb. nov. and Bifidobacterium longum subsp. suis comb. nov. Int J Syst Evol Microbiol. 58(Pt 4):767–772. doi:10.1099/ijs.0.65319-0.
  • MarketsandMarkets. 2021. Probiotics Market by Applications (Functional Food & Beverages (Dairy Products, Non-dairy Beverages, Infant Formula, Cereals), Dietary Supplements, Feed), Ingredient (Bacteria, Yeast), Form (Dry, Liquid), End User, and Region – Global Forecast to 2026. Probiotics Market Growth Analysis, Trends, and Forecasts to 2026 | MarketsandMarkets. (Accessed 21 January 2022).
  • Merenstein D, Pot B, Leyer G, Ouwehand AC, Preidis GA, Elkins CA, Hill C, Lewis ZT, Shane AL, Zmora N, et al. 2023. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes. 15(1):2185034. doi:10.1080/19490976.2023.2185034.
  • Oketch-Rabah HA, Roe AL, Muldoon-Jacobs K, Giancaspro GI. 2018. Challenges and opportunities for improving the safety assessment of botanical dietary supplements: A United States Pharmacopeia perspective. Clin Pharmacol Ther. 104(3):426–429. doi:10.1002/cpt.1113.
  • Ouwehand AC, Forssten S, Hibberd AA, Lyra A, Stahl B. 2016. Probiotic approach to prevent antibiotic resistance. Ann Med. 48(4):246–255. doi:10.3109/07853890.2016.1161232.
  • Paineau D, Carcano D, Leyer G, Darquy S, Alyanakian MA, Simoneau G, Bergmann JF, Brassart D, Bornet F, Ouwehand AC. 2008. Effects of seven potential probiotic strains on specific immune responses in healthy adults: a double-blind, randomized, controlled trial. FEMS Immunol Med Microbiol. 53(1):107–113. doi:10.1111/j.1574-695x.2008.00413.x.
  • Piatek J, Sommermeyer H, Bernatek M, Ciechelska-Rybarczyk A, Oleskow B, Sommer Mikkelsen L, Bundvig Barken K. 2019. Persistent infection by Salmonella enterica servovar Typhimurium: are synbiotics a therapeutic option? - a case report. Benef Microbes. 10(2):211–217. doi:10.3920/bm2018.0080.
  • Pradhan D, Mallappa RH, Grover S. 2020. Comprehensive Approaches for Assessing the Safety of Probiotic Bacteria. Food Control. 108:106872. doi:10.1016/j.foodcont.2019.106872.
  • Quigley EMM. 2017. Chapter 16 - Bifidobacterium longum. In: Floch MH, Ringel Y, Allan Walker W, eds. The Microbiota in Gastrointestinal Pathophysiology, Boston: Academic Press, p. 139–141.
  • Roe AL, Boyte ME, Elkins C, Goldman VS, Heimbach J, Madden E, Oketch-Rabah H, Sanders ME, Sirois J, Smith A. 2022. Considerations for determining safety of probiotics: A USP perspective. Regul Toxicol Pharmacol. 136:105266. doi:10.1016/j.yrtph.2022.105266.
  • Sakata S, Kitahara M, Sakamoto M, Hayashi H, Fukuyama M, Benno Y. 2002. Unification of Bifidobacterium infantis and Bifidobacterium suis as Bifidobacterium longum. Int J Syst Evol Microbiol. 52(Pt 6):1945–1951. doi:10.1099/00207713-52-6-1945.
  • Salminen S, von Wright A, Morelli L, Marteau P, Brassart D, de Vos WM, Fondén R, Saxelin M, Collins K, Mogensen G, et al. 1998. Demonstration of safety of probiotics–a review. Int J Food Microbiol. 44(1-2):93–106. doi:10.1016/s0168-1605(98)00128-7.
  • Salminen MK, Tynkkynen S, Rautelin H, Saxelin M, Vaara M, Ruutu P, Sarna S, Valtonen V, Järvinen A. 2002. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clin Infect Dis. 35(10):1155–1160. doi:10.1086/342912.
  • Sanders ME, Akkermans LMA, Haller D, Hammerman C, Heimbach J, Hörmannsperger G, Huys G, Levy DD, Lutgendorff F, Mack D, et al. 2010. Safety assessment of probiotics for human use. Gut Microbes. 1(3):164–185. doi:10.4161/gmic.1.3.12127.
  • Sarlin S, Tejesvi MV, Turunen J, Vänni P, Pokka T, Renko M, Tapiainen T. 2021. Impact of Streptococcus salivarius K12 on nasopharyngeal and saliva microbiome: a randomized controlled trial. Pediatr Infect Dis J. 40(5):394–402. doi:10.1097/inf.0000000000003016.
  • Schnadower D, Tarr PI, Casper TC, Gorelick MH, Dean JM, O’Connell KJ, Mahajan P, Levine AC, Bhatt SR, Roskind CG, et al. 2018. Lactobacillus rhamnosus GG versus placebo for acute gastroenteritis in children. N Engl J Med. 379(21):2002–2014. doi:10.1056/nejmoa1802598.
  • Sommer MO, Dantas G, Church GM. 2009. Functional characterization of the antibiotic resistance reservoir in the human microflora. Science. 325(5944):1128–1131. doi:10.1126/science.1176950.
  • Sudha RM. 2012a. Efficacy of Bacillus coagulans strain unique IS-2 in the treatment of patients with acute diarrhea. International Journal of Probiotics & Prebiotics: a Journal of Basic and Clinical Research on Gut Microbiota for Promotion of Human and Animal Health. 7(1):33–37. doi:10.3920/bm2017.0129.
  • Sudha RM, Yelikar KA, Deshpande S. 2012b. Clinical study of Bacillus coagulans Unique IS-2 (ATCC PTA-11748) in the treatment of patients with bacterial vaginosis. Indian J Microbiol. 52(3):396–399. doi:10.1007/s12088-011-0233-z.
  • Sudha MR, Bhonagiri S, Kumar MA. 2013. Efficacy of Bacillus clausii strain UBBC-07 in the treatment of patients suffering from acute diarrhoea. Benef Microbes. 4(2):211–216. doi:10.3920/bm2012.0034.
  • Tapiovaara L, Lehtoranta L, Poussa T, Mäkivuokko H, Korpela R, Pitkäranta A. 2016. Absence of adverse events in healthy individuals using probiotics–analysis of six randomised studies by one study group. Benef Microbes. 7(2):161–169. doi:10.3920/bm2015.0096.
  • Tompkins TA, Hagen KE, Wallace TD, Fillion-Forté V. 2008. Safety evaluation of two bacterial strains used in Asian probiotic products. Can J Microbiol. 54(5):391–400. doi:10.1139/w08-022.
  • Toohey JC, Townsend JR, Johnson SB, Toy AM, Vantrease WC, Bender D, Crimi CC, Stowers KL, Ruiz MD, VanDusseldorp TA, et al. 2020. Effects of probiotic (Bacillus subtilis) supplementation during offseason resistance training in female division I athletes. J Strength Cond Res. 34(11):3173–3181. doi:10.1519/jsc.0000000000002675.
  • Townsend JR, Bender D, Vantrease WC, Sapp PA, Toy AM, Woods CA, Johnson KD. 2018. Effects of probiotic (Bacillus subtilis DE111) supplementation on immune function, hormonal status, and physical performance in division I baseball players. Sports (Basel). 6(3):70. doi:10.3390/sports6030070.
  • Unique Biotech, Ltd. 2016. Self-affirmed GRAS Assessment for Bacillus clausii UBBC-07. Prepared by Soni and Associates, Inc. Date: September, 2016.
  • Upadrasta A, Pitta S, Madempudi RS. 2016. Draft genome sequence of the spore-forming probiotic strain Bacillus coagulans Unique IS-2. Genome Announc. 4(2):1–2. doi:10.1128/genomeA.00225-16.
  • USP. 2021. Guideline for the Admission of Dietary Supplement Ingredients to the USP–NF Monograph Development Process. Available at https://www.usp.org/sites/default/files/usp/document/get-involved/submission-guidelines/guideline-for-the-admission-of-dietary-supplement-ingredients.pdf. 2021. Accessed 14 November 2023.
  • Wang B, Hylwka T, Smieja M, Surrette M, Bowdish DME, Loeb M. 2018. Probiotics to prevent respiratory infections in nursing homes: a pilot randomized controlled trial. J Am Geriatr Soc. 66(7):1346–1352. doi:10.1111/jgs.15396.
  • Weitzel MLJ, Vegge CS, Pane M, Goldman VS, Koshy B, Porsby CH, Burguière P, Schoeni JL. 2021. Improving and comparing probiotic plate count methods by analytical procedure lifecycle management. Front Microbiol. 12:693066. doi:10.3389/fmicb.2021.693066.
  • Zhang Y, Chen J, Wu J, Chalson H, Merigan L, Mitchell A. 2013. Probiotic use in preventing postoperative infection in liver transplant patients. Hepatobiliary Surg Nutr. 2(3):142–147. doi:10.3978/j.issn.2304-3881.2013.06.05.
  • Zheng J, Wittouck S, Salvetti E, Franz CMAP, Harris HMB, Mattarelli P, O’Toole PW, Pot B, Vandamme P, Walter J, et al. 2020. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int J Syst Evol Microbiol. 70(4):2782–2858. doi:10.1099/ijsem.0.004107.