Posts

Episode 33: From probiotic mechanisms to applications, with Prof. Graciela Lorca PhD

This episode, we discuss how to advance from probiotic mechanisms to human applications, with Prof. Graciela Lorca PhD at the University of Florida in Gainesville, USA. Prof. Lorca talks about her experiences seeking out the mechanisms of action of a probiotic – including which molecules from bacteria may have beneficial effects – and bringing a probiotic through drug trials for use in Type 1 diabetes. They also discuss probiotic responders versus nonresponders and how dietary intake may provide clues about who will respond to an intervention.

Key topics from this episode:

  • Prof. Lorca’s lab is primarily concerned with discovering the mechanisms of action of specific probiotics, including the molecules they produce that can have beneficial effects on a host.
  • Knowing how a probiotic works allows scientists to select strains that are likely to be effective for a certain application.
  • Prof. Lorca’s lab found that L. johnsonii produces extracellular vesicles (EVs) and that a few proteins carried in these EVs may be important markers of where and how they affect the host. She triggered antibodies against these proteins, allowing them to be tracked in the host.
  • EVs are small protrusions from the bacterial membrane, and only some bacteria produce them. Evs have complex cargo, which mostly represents the metabolic state of the cell.
  • Prof. Lorca studied bacteria that appeared to affect autoimmunity in animal models. In humans, administering these bacteria changed immune markers; this intervention is now in a Phase II trial with humans who have Type 1 diabetes. The bacteria may be acting in the small intestine, but they don’t colonize permanently.
  • Extensive data on safety were required to advance the probiotic through to a Phase II trial. Although administering EVs could be an even safer approach, they are difficult to purify from bacteria. Prof. Lorca continues to investigate the bioactive components of these EVs to perhaps administer only those components.
  • Prof. Lorca is also interested in responders versus nonresponders to a probiotic intervention. One of her clinical trials showed that people had either high lactic acid bacteria (LAB) or low LAB at baseline. For those with high levels of LAB, the levels didn’t change much over time. But for those with initially low levels of LAB, the levels increased over time. The latter responded better to treatment. Furthermore, people with high LAB were shown to consume a diet with more long-chain fatty acids, which LAB can utilize. Overall, dietary intake may be a key part of uncovering responders and nonresponders.
  • Over the next ten years in this field, Prof. Lorca believes we will be able to increasingly personalize probiotics according to someone’s genetics and dietary intake. Regulatory aspects are complicated but continue to evolve.

Episode links:

Additional resources:

About Prof. Graciela Lorca PhD:

Dr. Graciela Lorca is currently a Professor in the Department of Microbiology and Cell Science at the University of Florida. She completed her Licentiate in Genetics studies at the National University of Misiones and later received her doctoral degree in Food Technology at the National University of Tucuman in Argentina. She completed her postdoctoral studies at the University of California San Diego in Molecular Microbiology and at the University of Toronto in Structural Biology and Gene Regulation. Since joining the Department of Microbiology and Cell Science at the University of Florida in 2007, Dr. Lorca has focused on the identification of environmental signals that modulate host-microbe interactions. Using multiomic approaches, her laboratory is investigating the bacterial components such as extracellular vesicles that target host pathways involved on those beneficial interactions in vitro and in vivo. Furthermore, Dr. Lorca’s laboratory is currently conducting human trials to evaluate the use of Lactobacillus johnsonii Type 1 Diabetes patients. Dr. Lorca currently teaches a graduate and undergraduate level Probiotics course. She is also in charge of the new concentration on Microbiome in health and disease within the Online Master program at Department of Microbiology and Cell Science.

Probiotic Administration in Preterm Infants: Scientific Statement

Board of Directors, International Scientific Association for Probiotics and Prebiotics

in collaboration with

Dr. Geoffrey Preidis MD PhD, Pediatric Gastroenterology, Hepatology & Nutrition

Prof. Andi L Shane MD MPH MSc, Pediatric Infectious Diseases

A recent report of a fatality in an extremely premature infant recipient of a probiotic product has resulted in a warning letter from the United States Food & Drug Administration (FDA) to healthcare practitioners about probiotic supplementation in preterm infants and a warning letter to the probiotic product manufacturer.

Publicly available information suggests that this fatality was the direct consequence of bacteremia resulting from ingestion of the probiotic organism Bifidobacterium longum subsp. infantis delivered in medium chain triglyceride oil. This situation differs from case reports of adverse events that resulted from extrinsic probiotic product contamination (1, 2). This is an important distinction, as the potential risks and mitigation strategies differ between etiologies. As complete details of this most recent fatality have not been released, specific factors that may have contributed to the adverse outcome are unknown. However, it is worth considering the context of this case report within the broader literature available on probiotic use in this population, including the wealth of data available on sepsis incidence.

Evidence from systematic reviews

Premature infants, especially those of <32 weeks gestation and with a birth weight <1500 g, are a vulnerable population at significant risk of morbidity and mortality.  Necrotizing enterocolitis (NEC) is highly prevalent (5-10% incidence) among very preterm infants, with mortality rates of 20-30% and high morbidity among survivors, including short gut syndrome, parenteral nutrition-associated liver disease, and neurocognitive delay.

A large body of literature exists on the use of probiotics in hospitalized preterm infants, with particular focus on the prevention of NEC. At least 85 randomised clinical trials (RCTs) (3) have been conducted to evaluate the use of probiotics in preterm infants for the prevention of diseases associated with prematurity, and a number of systematic reviews with meta-analyses have analysed these data in recent years. Most RCTs conducted in the neonatal intensive care unit (NICU) designate sepsis as one of the main outcome measures.

The most recent meta-analysis was published online October 2 in JAMA Pediatrics (3). This study included 106 trials on probiotic, prebiotic, synbiotic and lactoferrin interventions for either preterm infants <37 weeks and/or those with low birth weight (<2500 g). Administration of probiotics containing multiple strains were found to be most effective in the reduction of all-cause mortality (31% reduction), with a 62% decrease in incidence of severe NEC compared to placebo (moderate and high certainty evidence). Single strain probiotics combined with lactoferrin provided greatest efficacy in the reduction of late-onset sepsis incidence (67% risk reduction with moderate certainty evidence). It was noted that none of the included studies reported cases of probiotic-induced sepsis.

Other authors including groups from the Cochrane Collaboration, American Gastroenterological Association (AGA) and the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) have found similar results, and studies can be reviewed here:

Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants – Sharif, S – 2023 | Cochrane Library

Probiotics Reduce Mortality and Morbidity in Preterm, Low-Birth-Weight Infants: A Systematic Review and Network Meta-analysis of Randomized Trials – Gastroenterology (gastrojournal.org)

Probiotics for Preterm Infants: A Strain-Specific Systematic… : Journal of Pediatric Gastroenterology and Nutrition (lww.com)

No meta-analysis has attributed increased risk of sepsis to probiotic use in preterm infants – rather, in many cases a protective effect (or a trend toward protection) was reported. However, it is important to acknowledge the real but rare risk of probiotic-induced bacteremia in this population. In a recent review of case reports of probiotic-associated invasive infections in children, probiotic-induced bacteremia in premature infants were found to have resolved in most cases with use of effective antimicrobial therapy (4).

With data collected on over 10,000 preterm infants, substantial benefits demonstrated and a low level of risk identified, promise to improve outcomes in preterm infants who receive a probiotic product currently exists. Based on the evidence currently available, hospitals and NICUs across the globe have already adopted practices relating to probiotic use in preterm infants, some with significant health impacts (5, 6).

Risk benefit analysis and considerations for healthcare implementation

Further work needs to be done to support probiotic administration in the NICU. Collaborative efforts include recommendations for practical steps to improve probiotic product quality assurance specifically for NICU use, published in July 2023 in JAMA Pediatrics (7).

It is important to note that few (or possibly no) effective interventions are without an adverse event profile, and probiotics are no exception. Even food has a safety standard of reasonable certainty and on a regular basis, individuals suffer fatal foodborne infections. When considering the clinical indications for any intervention for an individual patient or a population of individuals, a thorough comparison of all available data on both the potential risks and the potential benefits is warranted.

The American Gastroenterological Association (8) and other major societies (including ESPGHAN and the World Gastroenterology Organisation) (9, 10) endorse probiotic products for the prevention of NEC among preterm low birth weight infants. The societies’ guidelines agree that the recommendation to use probiotics is conditional. Conditional recommendations are sensitive to patients’ values and preferences, and to the guideline panel’s perception of risk-benefit balance.  However, the recent FDA letter does not acknowledge these recommendations and further, recommends against probiotic use in preterm infants despite the robust efficacy data. With interventions such as probiotic administration, ideally shared clinical decision-making with patient and clinician would ensue. Regulatory warnings inform the risk-benefit calculation but typically do not invalidate a clinical recommendation.

Summary

  • Probiotic administration to preterm infants has been demonstrated to significantly reduce the risk of NEC, sepsis and death in large systematic reviews with meta-analyses.
  • Meta-analyses have not identified significant adverse events or safety concerns, although rare case reports have documented sepsis attributed to probiotics.
  • Stringent manufacturing standards are recommended for probiotics in vulnerable populations such as preterm infants.
  • Standardized comprehensive safety reporting across probiotic intervention studies is needed, along with funding for the conduct of long term studies.
  • The risks and benefits of probiotic administration should be considered in both the specific population and individual patients, with regulatory frameworks to enable implementation.
  • More information about this fatality should be immediately released so healthcare professionals and researchers can learn from this experience and continue to provide optimal evidence-based patient care.

To inquire about expert academic physicians available for media comment, please contact ISAPP’s Executive Director, Marla Cunningham, at marla@nullisappscience.org

See also:

NEC Society: Statement on FDA Warning of Probiotics in Preterm Infants

References

(1) Vallabhaneni S, Walker TA, Lockhart SR, et al. Notes from the field: Fatal gastrointestinal mucormycosis in a premature infant associated with a contaminated dietary supplement–Connecticut, 2014. MMWR Morb Mortal Wkly Rep. 2015;64(6):155-156.

(2) Bizzarro MJ, Peaper DR, Morotti RA, Paci G, Rychalsky M, Boyce JM. Gastrointestinal Zygomycosis in a Preterm Neonate Associated With Contaminated Probiotics. Pediatr Infect Dis J. 2021;40(4):365-367.

(3) Wang Y, Florez ID, Morgan RL, et al. Probiotics, Prebiotics, Lactoferrin, and Combination Products for Prevention of Mortality and Morbidity in Preterm Infants: A Systematic Review and Network Meta-Analysis. JAMA Pediatr. 2023 Oct 2:e233849.

(4) D’Agostin M, Squillaci D, Lazzerini M, et al. Invasive Infections Associated with the Use of Probiotics in Children: A Systematic Review. Children (Basel). 2021 Oct 16;8(10):924.

(5)  Rath CP, Athalye-Jape G, Nathan E, et al. Benefits of routine probiotic supplementation in preterm infants. Acta Paediatr. 2023 Jul 28.

(6) Bui A, Johnson E, Epshteyn M, Schumann C, Schwendeman C. Utilization of a High Potency Probiotic Product for Prevention of Necrotizing Enterocolitis in Preterm Infants at a Level IV NICU. The Journal of Pediatric Pharmacology and Therapeutics 2023;28(5):473–475.

(7)  Shane AL, Preidis GA. Probiotics in the Neonatal Intensive Care Unit-A Framework for Optimizing Product Standards. JAMA Pediatr. 2023 Sep 1;177(9):879-880.

(8) Su GL, Ko CW, Bercik P, et al. AGA Clinical Practice Guidelines on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology. 2020 Aug;159(2):697-705.

(9) WGO Practice Guideline: Probiotics and Prebiotics. Available from: https://www.worldgastroenterology.org/guidelines/probiotics-and-prebiotics

(10) van den Akker CHP, van Goudoever JB, Shamir R, et al. Probiotics and Preterm Infants: A Position Paper by the European Society for Paediatric Gastroenterology Hepatology and Nutrition Committee on Nutrition and the European Society for Paediatric Gastroenterology Hepatology and Nutrition Working Group for Probiotics and Prebiotics. J Pediatr Gastroenterol Nutr. 2020 May;70(5):664-680.

How to navigate probiotic evidence and guidelines for pediatric populations

Episode 20: How to navigate probiotic evidence and guidelines for pediatric populations

How to navigate probiotic evidence and guidelines for pediatric populations

 

The Science, Microbes & Health Podcast 

This podcast covers emerging topics and challenges in the science of probiotics, prebiotics, synbiotics, postbiotics and fermented foods. This is the podcast of The International Scientific Association for Probiotics and Prebiotics (ISAPP), a nonprofit scientific organization dedicated to advancing the science of these fields.

How to navigate probiotic evidence and guidelines for pediatric populations, with Dr. Hania Szajewska

Episode summary:

In this episode, the ISAPP podcast hosts talk about evidence and guidelines for probiotics in pediatric populations, with Prof. Hania Szajewska MD PhD, of the Department of Paediatrics at the Medical University of Warsaw, Poland. They talk about some of the inconsistencies between different medical organizations’ guidelines for pediatric probiotic use, and how clinicians can move forward with recommendations based on the best available evidence.

 

Key topics from this episode:

  • Guidelines exist on probiotic use for gastroenterological issues in children, but there are differences (especially regarding acute gastroenteritis) between guidelines from different medical societies: European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) and The American Gastroenterological Association (AGA).
  • Realistic expectations are necessary when prescribing probiotics. Different probiotics have different benefits, but they are not a ‘magic bullet’. For example, the evidence shows certain probiotics for acute gastroenteritis reduce diarrhea by an average of one day. This could have a big impact on the quality of life of the end user, but for clinicians it may not sound like a lot so they must set expectations accordingly.
  • The market is overflowing with probiotic products, many of which do not have proven efficacy. This makes it difficult for end users and healthcare professionals to distinguish the best products.
  • Always look for evidence-based probiotics with documented efficacy for the indication for which they are intended.
    • Physicians have the ethical duty to prescribe evidence-based products (that is, clinically proven, effective products).
    • The exact strains and doses matter.
  • Formal training and education of healthcare professionals regarding the beneficial effects of microbes, the microbiome, and probiotics are currently lacking.
  • Is it more valuable to know probiotics’ mechanism of action, or to have evidence from clinical trials that they are effective?
    • Ideally we would have both, but since we don’t know the exact mechanism for all probiotics, positive evidence from clinical trials is crucial. 
    • We also need to make clear to healthcare professionals and end users what to expect from taking probiotics. For example, some probiotics reduce the chances of developing antibiotic-associated diarrhea by 50%. For colic, some probiotics can reduce the crying time by half an hour. These are modest benefits but for the affected individual they may be impactful.
  • For vulnerable populations such as preterm infants, we need high-quality products with proven safety and efficacy.

 

Episode abbreviations and links:

 

About Prof. Hania Szajewska

Hania Szajewska, MD, is Professor and Chair of the Department of Paediatrics at the Medical University of Warsaw and the Chair of the Medical Sciences Council. Among her various functions, she served as the Editor-in-Chief of the Journal of Pediatric Gastroenterology and Nutrition; a member of the Council and then as the General Secretary of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN); the Secretary of the ESPGHAN Committee on Nutrition. Most recently, she joined the Board of Directors of the International Scientific Association for Probiotics and Prebiotics (ISAPP). Prof. Szajewska has broad interests in pediatric nutrition but her research focuses on the effects of early nutritional interventions on later outcome; and the gut microbiota modifications such as with various biotics (probiotics, prebiotics, synbiotics, postbiotics). She is or has been actively involved in several European Union-funded research projects. She is an enthusiastic advocate for the practice of evidence-based medicine. Prof. Szajewska has co-authored more than 400 peer-reviewed publications and 30 book chapters. Citations &gt;18,141. Hirsch index 72 (WoS, March 2023).

Episode 18: The definition of postbiotics

 

The Science, Microbes & Health Podcast 

This podcast covers emerging topics and challenges in the science of probiotics, prebiotics, synbiotics, postbiotics and fermented foods. This is the podcast of The International Scientific Association for Probiotics and Prebiotics (ISAPP), a nonprofit scientific organization dedicated to advancing the science of these fields.

The definition of postbiotics, with Dr. Gabriel Vinderola and Prof. Seppo Salminen

Episode summary:

In this episode, the ISAPP podcast hosts join guests Gabriel Vinderola, PhD, Principal Researcher at the
National Scientific and Technical Research Council (CONICET) and Associate Professor at University of Litoral in Argentina, and Seppo Salminen, PhD, Professor at University of Turku in Finland, to discuss the relatively recent definition of postbiotics and what kinds of substances are included in this category. They talk about the criteria for something to qualify as a postbiotic, common mechanisms of action for postbiotics, and how postbiotic science has brought new perspectives on the study of probiotics.

 

Key topics from this episode:

  • What are postbiotics? Dr. Vinderola and Prof. Salminen dive deep into the definition of postbiotics created in 2021 and what it entails.
  • Postbiotics, similar to probiotics, prebiotics, and synbiotics, must provide health benefits to the host.
  • The nature of the postbiotic preparation is important for its health benefits. When the inactivation process is changed, this can lead to altered health benefits, and clinical studies must be repeated to ensure the desired health benefits are maintained.
  • They explain why “inanimate” was chosen to describe the microorganisms / components in a postbiotic preparation. 
  • What is the mode of action, or how do postbiotics work? 
    • Postbiotics show similar mechanisms of action to probiotics, except for ones requiring viability, since postbiotics will not grow and produce metabolic byproducts in the host.
    • Postbiotics can benefit the host via physical interaction with the host epithelial and immune cells.
    • A primary mechanism of action is likely to be through activation of the immune system, through which postbiotics can affect inflammation and some disease conditions. 
    • Postbiotics may also affect the microbiome composition and ability to inhibit pathogens.
  • From a regulatory point of view, inanimate microorganisms may represent an easier category to prove safe for users. For industry, postbiotics may be more convenient with a longer shelf life.
  • Some controversy still exists around the ISAPP-led postbiotic definition, and this has led to valuable discussions that are crucial to scientific progress. So far the authors of the definition have defended their stance.

 

Episode abbreviations and links:

 

Additional Resources:

Postbiotics. ISAPP infographic (also available in Japanese and Spanish).

Behind the publication: Understanding ISAPP’s new scientific consensus definition of postbiotics. ISAPP blog post.

Definition of postbiotics: A panel debate in Amsterdam. ISAPP blog post.

 

About Dr. Gabriel Vinderola: 

Gabriel Vinderola graduated at the Faculty of Chemical Engineering from the National University of Litoral (Santa Fe, Argentina) in 1997. He obtained his Ph.D. in Chemistry in 2002 at the same University. He collaborated with several research teams in Canada, Spain, France, Italy, Germany, Brazil and Finland. He is presently Principal Researcher of the National Scientific and Technical Research Council (CONICET) and Associate Professor at the Food Engineering Department of his home Faculty. He participated in 1999 in the development of the first commercial cheese carrying probiotic bacteria in Latin America. In 2011, he was awarded the prize in Food Technology for young scientists, by the National Academy of Natural, Physic and Exact Sciences from Argentina. He published more than 120 original scientific publications in international refereed journals and book chapters. From 2020 to present, he serves as a member of the board of directors of the International Scientific Association for Probiotics and Prebiotcis (ISAPP). He is engaged in science communication to the general public through Instagram (@gvinde).

 

About Prof. Seppo Salminen: 

Seppo Salminen, MSc, MS, PhD, is a Senior Advisor, Functional Foods Forum (FFF) at the University of Turku. His areas of expertise are gut microbiota, probiotics and prebiotics, nutrition and food safety, and EU regulations. Seppo teaches the topics of lactic acid biotechnology, functional foods and EU legislation and conducts research into food and health, intestinal microbiota, probiotics, prebiotics, functional foods, food legislation, health claims, and novel foods.

Shaping microbial exposures and the immune system in childhood: Can sandboxes be probiotic?

By Prof. Seppo Salminen, University of Turku, Finland

Gut microbiota researchers have established that microbial exposures in early life can be influential on health later in life. Children who develop asthma in early childhood, for example, have an altered gut microbiota linked with exposure to less diverse microorganisms in their first year. The ‘biodiversity hypothesis’ has been advanced recently, suggesting that western lifestyles and low biodiversity in urban environments reduce contact with microbes both via food and via the natural environment, presenting fewer opportunities for children to be exposed to a diversity of microbes in their earliest years and increasing the risk of non-communicable diseases. If this is the case, the environments of daycare and kindergarten facilities come under scrutiny as a source of microbial exposures at a crucial time of life. So is it beneficial to intervene in children’s environments to ensure more diverse microbial exposures? Can we enhance gut microbial diversity and richness in children through environmental interventions?

A new study provided proof that shaping children’s microbial exposures may be possible. The study was the first of its kind – a placebo-controlled, double-blinded study on the effect of environmental exposures on gut microbiota diversity and immune parameters in young children. The study used playground sandboxes at daycare facilities as sources of environmental microbial diversity and explored whether these could have effects on the children.

Six day-care centers in southern Finland were enrolled in the study, with two randomly assigned to intervention and four to placebo. Identical-looking playground sandboxes were used. Intervention sandboxes were filled with sand of glacial origin enriched with a known biodiversity powder (including commercial soil, deciduous leaf litter, peat, and Sphagnum moss; described in detail by Hui et al., 2019 ; Grönroos et al, 2018). In control centers the sand was regular sandbox sand and placebo peat material. Altogether, 26 children ages 3-5 participated in supervised play for 20 minutes in the morning and afternoon for two weeks. Researchers measured the composition of gut and skin microbiota, as well as blood immune markers.

The results demonstrated that exposure to diverse environmental microbiota enhanced both the bacterial richness and diversity of the skin bacterial community. The microbiome of the skin changed only in those children who had played in a sandbox enriched with natural materials. The authors also found that the daily exposure to higher microbial biodiversity resulted in positive differences in immune response. For instance, the authors reported shifts in skin microbiota associated with IL-10 and T cell frequencies. This provides the first evidence from a placebo-controlled, double-blinded study in young children showing the differential effects on microbiota and immunity of daily exposure to defined microbial biodiversity.

An interesting follow-up could be using sandboxes to deliver probiotics with a proven health impact to children. Since the sandbox microbes were shown to influence children’s immune systems, could researchers go one step further and modulate children’s microbiota in a targeted manner? A probiotic must be defined, shown to have a health benefit and administered in an efficacious dose. In the case of sandboxes, the health benefit would need to be demonstrated for a certain level or duration of environmental exposure.

Playgrounds and sandboxes require materials that tolerate heavy wear and tear and are safe at the same time. Such materials need to be kept free of unnecessary contamination as sandboxes, for example, can also be good reservoirs of some detrimental bacteria. Therefore, it could be important to have defined natural materials for a positive impact on health. In the future, we may see many creative approaches to ensuring children receive appropriate health-supporting microbial exposures early in life. However, creating probiotic approaches requires identification of specific microbes in the biodiversity powder.

What is a strain in microbiology and why does it matter?

By Prof. Colin Hill, Microbiology Department and APC Microbiome Ireland, University College Cork, Ireland

At the recent ISAPP meeting in Sitges we had an excellent debate on the topic of ‘All probiotic effects must be considered strain-specific’. Notwithstanding which side of the debate prevailed, it does raise the question: what exactly is a strain? As a card-carrying microbiologist I should probably be able to simply define the term and give you a convincing answer, but I find that it is a surprisingly difficult concept to capture. It is unfortunately a little technical as a topic for a light-hearted blog, but here goes. Let me start by saying that the term ‘strain’ is important largely because we like to name things and then use those names when we share information, but that the concept of ‘strain’ may have no logical basis in nature where mutations and changes to a bacterial genome are constantly occurring events.

Let’s suppose I have a culture of Lactobacillus acidophilus growing in a test-tube, grown from a single colony. This clonal population is obviously a single strain that I will name strain Lb. acidophilus ISAPP2022. That was easy! I am aware of course that within this population there will almost certainly be a small number of individual cells with mutations (single nucleotide polymorphisms, or SNPs), cells that may have lost a plasmid, or cells that have undergone small genomic rearrangements. Nonetheless, because this genetic heterogeneity is unavoidable, I still consider this to be a pure strain. If I isolate an antibiotic resistant version of this strain by plating the strain on agar containing streptomycin and selecting a resistant colony I will now have an alternative clonal population all sharing a SNP (almost certainly in a gene encoding a ribosomal subunit). Even though there is a potentially very important genotypic and phenotypic difference I would not consider this to be a new strain, but rather it is a variant of Lb. acidophilus ISAPP2022. To help people in the lab or collaborators I might call this variant ISAPP2022SmR, or ISAPP2022-1. In my view, I could continue to make changes to ISAPP2022 and all of those individual clonal populations will still be variants of the original strain. So, the variant concept is that any change in the genome, no matter how small, creates a new variant. When I grow ISAPP2022 in my lab for many years, or share it with others around the word, it is my view that we are all working with the same strain, despite the fact that different variants will inevitably emerge over time and in different labs.

Where the strain concept becomes more difficult is when I isolate a bacterium from a novel source and I want to determine if it is the same strain as ISAPP2022. If the whole genome sequence (WGS) is a perfect match (100% average nucleotide identity or ANI) then both isolates are the same strain and both can be called ISAPP2022. If they have only a few SNPs then they are variants of the same strain. If the two isolates only share 95% ANI then they are obviously not the same strain and cannot even be considered as members of the same species (I am using a species ANI cut-off of 96% that I adopted from a recent paper in IJSEM.

Where it gets really tricky is when the ANI lies between 96% (so that we know that the isolates are both members of the same species) and 100% (where they are unequivocally the same strain). Where should we place the cut-off to define a strain? At what point is a threshold crossed and an isolate goes from being a variant to becoming a new strain? Should this be a mathematical decision based solely on ANI, or do we have to consider the functionality of the changes? If it is mathematical then we could simply choose a specific value, say 99.95% or 99.99% ANI, and declare anything below that value is a new strain. Remember that the 2Mb genome size of Lb. acidophilus would mean that two isolates sharing 99.99% ANI could differ by up to 200 SNPs. This could lead to a situation where an isolate with 199 SNPs compared to ISAPP2022 is considered a variant, but an isolate with 201 SNPs is a new strain (even though it only differs from the variant with 199 SNPs by two additional SNPs). This feels very unsatisfactory. But what about an isolate with only 50 SNPs, but one that has a very different phenotype to ISAPP2022 because the SNPs are located in important genes? Or what about an isolate with an additional plasmid, or missing a plasmid, or with a chromosomal deletion or insertion? I would argue we should not have a hard and fast cut-off based on SNPs alone, but we should continue to call all of these variants, and not define them as new strains.

So, by how much do two isolates have to differ before we no longer consider them as variants of one another, but as new strains? I will leave that question to taxonomists and philosophers since for me it falls into the territory of ‘how many angels can dance on the head of pin?’

All this may seem somewhat esoteric, but there are practical implications. Can we translate the findings from a clinical trial done with a specific variant of a strain to all other variants of the same strain? If Lactobacillus acidophilus ISAPP2022 has been shown to deliver a health benefit (and is therefore a probiotic), can we assume that Lb acidophilus ISAP2022-1 or any other variant will have the same effect? What if a variant has only one mutation, but that mutation eliminates an important phenotype required for the functionality of the original strain? I am afraid that at the end of all this verbiage I have simply rephrased the original debate topic from ‘All probiotic effects must be considered strain-specific’ to ‘All probiotic effects must be considered variant-specific’. Looks like we might be heading back to the debate stage in 2023!

ISAPP’s Guiding Principles for the Definitions of ‘Biotics’

By Mary Ellen Sanders, PhD, ISAPP Executive Science Officer

Articulating a definition for a scientific concept is a significant challenge. Inevitably, scientists have different perspectives on what falls inside and outside the bounds of a term. Prof. Glenn Gibson, ISAPP co-founder and longtime board member, recently published a paper that describes his path to coining the word ‘prebiotic’, with this observation: “One thing I have learned about definitions is that if you propose one, then be ready for it to be changed, dismissed or ignored!”

Mary Ellen Sanders with Glenn Gibson

Members of the ISAPP board, however, have remained steadfast in their belief that such definitions are worth creating. They are the basis for shared understanding and coordinated progress across a scientific field.

Developing the consensus definition papers on probiotics, prebiotics, synbiotics, postbiotics and fermented foods was demanding on the part of all involved. The objective of the panels that met to discuss these definitions was clear – to provide common ground for consistent use of this growing body of terms for all stakeholders. Although some disagreement among the broader scientific community exists about some of the definitions, ISAPP’s approach relied on important, underlying principles:

  • Don’t unnecessarily limit future innovation
  • Don’t unnecessarily limit mechanisms of action
  • Don’t unnecessarily limit scope (host, regulatory category, mechanism, site of action, etc.)
  • Require a health benefit on a target host to be demonstrated – otherwise, what is the value of these biotic substances? (Of course, fermented foods were the exception in this criterion, because the value of consuming fermented foods even in the absence of an established health benefit is evident.)
  • Limit to preparations that are administered, not substances produced by in situ activities

In my opinion, many published definitions, including previous ones for postbiotics (see supplementary table here), are untenable because they don’t recognize these principles. There may also be a tendency to rely on historical use of terms, rather than to describe what is justified by current scientific knowledge. A good example of this is provided by the first definition of probiotics, published in 1965. It was “substances secreted by one microorganism that stimulate another microorganism” (Lily and Stillwell, 1965), which is far from the current definition of “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host (Hill et al. 2014).

If you’re looking for a concise summary of the five published ISAPP definitions, see here for our definitions infographic.

Additional reflections: I noted with a smile Glenn’s views on ISAPP, specifically on the appropriate pronunciation of the abbreviation ‘ISAPP’. “My only negative is that everyone involved in the organisation aside from 2 or 3 of us pronounce its acronym wrongly.” Most board members, including myself, have always pronounced this as ‘eye-sap’. Glenn opines, “The abbreviation is not eye-SAPP, it is ISAPP (with the ‘I’ – remarkably enough – being spoken as it is in the word ‘International’).” I wonder how he pronounces IBM?

 

 

 

 

Decoding a Probiotic Product Label

By Mary Ellen Sanders, PhD

Interested in knowing what’s in your probiotic product? Unfortunately, there are many ways that probiotic product labels can fall short.

First, not all items labeled as “probiotic” truly meet the scientific criteria for a probiotic product. See here for information on what qualifies as a probiotic. Some fermented foods are marketed today claiming to be ‘probiotic’. But these products often have undefined microbial content and lack any studies documenting health effects, criteria that are required for a probiotic. Instead, such products could legitimately be labeled as containing ‘live, active cultures’. Dietary supplement products formulated with untested microbes should similarly not be labeled as probiotics.

Also, a label might not provide adequate information on what types of microbes are contained in the product. Genus and species may be listed, but no strain designation. Maybe only “bifidobacteria” or “lactobacilli” are listed.

They might not disclose the potency of individual strains in the product. Some may provide a total count of colony forming units (cfu)/dose or serving, which in the case of a single strain product is informative. But in the case of a multi-strain product – products may contain a couple or up to 30 strains – you don’t know if equal amounts of all strains are included, or perhaps the bulk of the count is made up of the strain in the formulation that is least expensive to manufacture rather than the one that will make the probiotic more effective. Some products may provide one count for “Lactobacillus” and another count for “Bifidobacterium”, a slightly more informative approach than just total count, but still lacking in detail. Many challenges exist for multi-strain products, including the lack of robust methods to quantify different strains once combined, especially viable ones. This topic was one that was covered in an ISAPP webinar, Current issues in probiotic quality: An update for industry.

The label may state that the count is “at time of manufacture”, a number that is no doubt inadequate if you purchase the product close to the end of its shelf-life. Most probiotic strains suffer cell count decline over the course of shelf-life, with some strains more susceptible than others. This situation almost guarantees that by the pull-by date for a multi-strain product, the ratio of cfu of strains to each other is likely much different than at the time of formulation.

Surveys of probiotic product labels

Additionally, it is difficult for consumers to know what products are backed by studies documenting a health benefit. If a product is not labeled sufficiently, it is impossible to link it to evidence. Two studies surveyed commercial probiotic products in the Washington DC area, Retail Refrigerated Probiotic Foods and Their Association with Evidence of Health Benefits and More Information Needed on Probiotic Supplement Product Labels. Results showed that 45% of retail dietary supplement products did not provide strain designations and an equal number did not provide a measure of potency through the end of shelf-life. Only 35% of products could be linked (based on strain and dose) to evidence of a health benefit. Food products fared a bit better, with 49% of refrigerated probiotic food products being linked to evidence of a health benefit. One clear indication from this study was that if your food product discloses the strain designation, it is likely to have evidence of a health benefit. An overall conclusion was that product labeling – at least in this region – needs improvement.

Historical context on guidelines for probiotic product labels

According to the FAO/WHO 2002 Working Group on Guidelines for the Evaluation of Probiotics in Food (page 39 of this combined document), the following information should be on probiotic labels:

– Genus, species and strain designation for each probiotic strain in the product.

– Minimum viable numbers of each probiotic strain at the end of the shelf-life, typically expressed in colony forming units (or cfu).

– The suggested serving size (or dose) must deliver the effective dose of probiotics related to any health benefit communicated on the label.

– Health claim(s) (as allowed by law and substantiated by studies)

– Proper storage conditions

– Corporate contact details for consumer information

These principles are developed and reiterated in “Best Practices Guidelines for Probiotics” (2017) developed by the Council for Responsible Nutrition and IPA.

Additional information

ISAPP created an infographic to explain the information on a probiotic labels. Our example portrays an imaginary dietary supplement for sale in the United States. Labels on foods containing a probiotic or a probiotic produced in another country would be labeled differently from this example to comply with applicable regulations. For those interested in probiotic labels in the EU, see the infographic ISAPP created for a probiotic product in the European Union. Also of interest, USP.org created an infographic on “How to Read a Dietary Supplement Label” for U.S. products.

Can control of body malodor using probiotic topical cream be considered as a health benefit?

By Victoria Onwuliri, Masters degree student, with Dr. Kingsley C. Anukam, Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus, Nnewi, Anambra State, Nigeria.

I recall years back as a teenager, axillary sweating and pubertal odor were one of the overwhelming challenges I experienced. It really affected how I related with the people around me, and I became socially withdrawn. I was introduced to the use of deodorants after a very attractive advertisement on television. Truthfully, though, these personal hygiene products were not a good match for my active teenage lifestyle because their effects waned easily and I was back to my sweaty self. This continued with all different products I used; they would wear off, leaving my axilla smelling stronger than before. Sharing my experience with other adolescents like me made me realize that we all were facing similar issues and were seeking a long-lasting solution. I am very certain many teenagers and young adults all over the world are currently experiencing same problem and are in need of a solution.

Later on, as a scientist, I wondered if I could explore a solution for this problem. First, what was causing the odor? Several fundamental studies have shown that the apocrine gland that is located in the hypodermis of the skin is responsible for secreting the odorless precursor molecules. These precursors are transformed by some bacteria residing on the skin into smelly molecules including but not limited to sulphanylalkanols, short volatile branched-chain fatty acids and some steroid derivatives. It should be noted that both males and females produce some levels of body odor, but the intensity varies as females have 75% more apocrine glands in their armpits than males but males have larger apocrine glands. The differences in the size and number of apocrine glands may explain why males tend to smell more than females. (I hope my male colleagues will not take offense for my sharing this fact). However, differences in hygiene habits such as regular shaving, use of antiseptic soap, use of deodorants and antiperspirants could also play a role.

I was involved in a study with Dr. Kingsley C. Anukam as my supervisor in 2019 on the effect of antiperspirants and deodorants on the axillary skin microbiome of adult male and female subjects. This study supported my teenage observation that I was worse off after using these products: the study showed that the resultant effect of the regular use of these personal hygiene products was an imbalance in the seemingly normal bacterial population of the axillary skin, thereby promoting the proliferation of malodor-producing organisms such as Corynebacterium, Cutibacterium, some Staphylococcus and Streptococcus species. Interestingly lactobacilli were also detected in the axilla of over 82% of female and over 81% of male subjects, though in low relative abundance which suggests that lactobacilli might be considered as part of the normal axillary bacterial community. From this work, an idea emerged on exploring the possible beneficial effect of probiotics in decreasing the relative abundance of malodor-producing bacteria in the axilla of healthy adult individuals.

Dr. Anukam and I set up a study and employed the use of oil-based topical cream, made from natural ingredients, fortified with a probiotic of Nigerian origin, Lactiplantibacillus pentosus KCA1 strain.

Since some species of Corynebacterium (particularly Corynebacterium striatum, and Corynebacterium jeikeium), and Staphylococcus haemolyticus, Staphylococcus hominis, and Staphylococcus lugdunensis isolated from human axilla have been implicated in the generation of malodour volatile substances 1,2, and the fact that we identified lactobacilli in low abundance in the axilla of healthy subjects compared with Corynebacterium and Staphylococcus species, Dr. Anukam, agreed that applying lactobacilli, (which are generally regarded as safe bacteria) to the skin might change the ecology to a state whereby some lactobacilli with probiotic characteristics can nestle on the axillary skin.

The data obtained from the study3 which has already been published in the Journal of Cosmetic Dermatology (https://doi.10.1111/jocd.13949) showed the positive impact of this probiotic-fortified topical cream on the human axillary skin microbiota, as a means of reducing axillary malodor. We drew this conclusion based on the fact that malodor-producing Staphylococcus and Corynebacterium species were significantly reduced in abundance after applying the probiotic cream. In addition, all the participants gave positive feedback as they reported not perceiving any malodor during the study period.  Another interesting in silico finding from the study was the down-regulation of the bacterial metabolic functional genes such as the PLP-dependent protein (K06997) and pyridoxal 5′-phosphate synthase pdxS subunit (K06215) after the application of the probiotic cream.

This appears much more desirable when compared to the effect of regular usage of antiperspirants and deodorants on the axillary skin microbiome.

However, some arguments have arisen whether reduction of body odor could be taken as a health benefit since probiotic definition stipulates that a probiotic must ‘confer a health benefit on the host’. We know that body malodor has some social and psychological implications to some people which might impact negatively on their mental health. We therefore suggest that using tested microorganisms to reduce body malodor may contribute to the wellbeing of individuals, so this would count as a probiotic intervention.

We are not saying that probiotic cream alone would completely solve the problem of axillary skin/body malodor, but we believe its positive effect outweighs that of the antiperspirants and deodorants. In addition, the potential beneficial effects of skin-based probiotics could be increasingly explored by the cosmetic and pharmaceutical industries. Regarding our work, further study involving a larger population and more insight on the functional malodor control attributes of lactobacilli are warranted. I know teenagers everywhere are waiting for this breakthrough.

Preparation of topical cream fortified with Lactiplantibacillus pentosus KCA1

(GeneBank Accession # NZ_CM001538.1)

The study used natural ingredients that have already-known benefits on the skin, in the preparation of the topical cream. During the preparation, ingredients were heated and purified, in order to maintain sterility and keep them in their oil forms before the incorporation of the lyophilized Lactiplantibacillus pentosus KCA1.

Finished product:

 

 

Ingredients:

Cocoa butter, coconut oil, lavender oil, shea butter, lyophilized Lactiplantibacillus pentosus KCA1

 

 

References

  1. Natsch A, Schmid J, Flachsmann F. Identification of odoriferous sulfanylalkanols in human axilla secretions and their formation through cleavage of cysteine precursors by a C-S lyase isolated from axilla bacteria. Chem Biodivers. 2004;1(7):1058–72
  2. Bawdon D, Cox DS, Ashford D, James AG, Thomas GH. Identification of axillary Staphylococcus sp. involved in the production of the malodorous thioalcohol 3-methyl-3-sufanylhexan-1-ol. FEMS Microbiology Letters 2015; 362: fnv111. doi: 10.1093/femsle/fnv111
  3. Onwuliri V, Agbakoba NR, Anukam KC. Topical cream containing live lactobacilli decreases malodor-producing bacteria and downregulates genes encoding PLP-dependent enzymes on the axillary skin microbiome of healthy adult Nigerians. J Cosmet Dermatol. 2021;00:1–10. https://doi.org/10.1111/jocd.13949

 

 

 

Victoria Onwuliri is a Master degree student in the Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus, Nnewi, Anambra State, Nigeria.

What do we mean by ‘conferring a health benefit on the host’?

By Prof. Colin Hill, University College Cork, Ireland

Four of the Consensus definitions produced by ISAPP in recent years (see 1-4 below) finish with a similar wording, insisting that probiotics, prebiotics, synbiotics and postbiotics must confer a health benefit on the host”. This proviso was included to explicitly reinforce the fact that the raison d’etre for these interventions is that they must demonstrably improve host health. It would perhaps be wise to just stop there and leave the interpretation of what this really means to each individual reader. But that would not make for a very long blog and I am not very wise. Furthermore, it is useful to be more precise for scientific and regulatory purposes. At least two aspects seem to be open to elaboration; what is meant by ‘host’ and what is a ‘health benefit’? I will base my thoughts on the probiotic definition, but the logic should apply equally to all four health-based definitions.

Host. According to the Google dictionary a host is an animal or plant on or in which a parasite or commensal organism lives’. This means there are millions of potential host species on our planet, something that could potentially create confusion. For example, if a well characterised microbe (or microbes) is shown to provide a measurable health benefit when administered in adequate amounts in a murine model (the host) then it clearly meets the stated definition of probiotic. But only for mice! It should not be referred to as a probiotic for other species, including humans, solely based on murine evidence. This creates a situation where the same microbe can clearly meet the criteria to be a probiotic for one host but not for another. This is not simply semantics; it is of vital importance that it should not be assumed that health benefits confirmed in one host will also be realised in another without supporting evidence. Since the majority of discussions of probiotics address human applications, it may serve all stakeholders well – even if not directly mandated by the definition – if the word ‘probiotic’ was only used without qualification for microbes with measurable benefits in humans while all others should be qualified with the target host; ‘equine probiotic’, ‘canine probiotic’, or even ‘plant probiotic’.

Health benefit. Health is of course a continuum from a desirable but almost certainly unattainable state where every organ is performing optimally (something I will term ‘ideal health’) to a point where death is imminent (that I will term ‘poor health’). Of course, health is multidimensional and far more complex than a straight line between ‘ideal’ and ‘poor’ but for simplicity I will treat it as such. If we place ideal health on the left end of our straight line and poor health at the right end, then obviously any shift towards the left can be considered a health benefit. It could even be reasonably argued that if someone is gradually progressing from left to right down our imaginary line (for example, as we age) then halting or slowing down that progression could also be considered a health benefit. From this perspective every individual (not just the unwell) could potentially derive a health benefit from a probiotic, prebiotic, synbiotic or postbiotic.

The issue of cosmetic benefits is more nuanced. If an intervention improves someone’s appearance (or reduces body odour for example) it might not be considered a health benefit per se, but of course it could well have a beneficial effect on an individuals’ mental health. I will leave it to the psychologists and psychiatrists to determine how this could be convincingly demonstrated.

There is also the issue of production characteristics where the host is a food animal or a crop. If a microbial-based intervention leads to faster growth rates and increased yields should this qualify as a health benefit? My own opinion is if the intervention leads to higher productivity by preventing infections it could be considered a health benefit, but not if it simply leads to faster growth rates by improving feed conversion for example.

Can changing the microbiome be considered a health benefit? A trickier question is whether a direct effect on the microbiome could be considered as a health benefit? Every host has a microbiome of a particular configuration, richness, and diversity. I don’t think we are yet at a point where measurable changes in these general indices of microbiome composition can be termed a health benefit in the absence of a link to a more established health outcome. The consequence of any change will be microbiome-specific in any event; a reduction in diversity in the vaginal microbiome might be desirable, whereas an increase in diversity in the gut microbiome might well be considered beneficial. But what if we can measure a reproducible reduction in a specific pathobiont like Clostridioides difficile, or an increase in a microbe that is associated with good health such as Bifidobacterium? In my opinion we are arriving at a point where we can begin to refer to these impacts as a health benefit. This will become more and more relevant as we establish direct causal links between individual commensal microbes and health outcomes. Equally, an intervention that preserves microbiome structure during a disruption (e.g. infection or antibiotic treatment) could also be considered as beneficial. I don’t know if regulators are yet at the point of accepting outcomes such as these as direct health benefits, but I believe a strong case can be made.

To finish, I believe that it is a very exciting time for all of us in the field of probiotics, prebiotics, synbiotics and postbiotics, but it is really important that all of this important science is not compromised by loose language or by literal interpretations that adhere to the letter of the definitions but not to the intent. If you want to fully understand the intent of the definitions, I encourage you to read the full text of the consensus papers.

 

  1. https://doi.org/10.1038/nrgastro.2014.66
  2. https://doi.org/10.1038/nrgastro.2017.75
  3. https://doi.org/10.1038/s41575-020-0344-2
  4. https://doi.org/10.1038/s41575-021-00440-6

A postbiotic is not simply a dead probiotic

By Dr. Gabriel Vinderola, PhD,  Associate Professor of Microbiology at the Faculty of Chemical Engineering from the National University of Litoral and Principal Researcher from CONICET at Dairy Products Institute (CONICET-UNL), Santa Fe, Argentina

Postbiotics, recently addressed in an ISAPP consensus panel paper, are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit on the host. Criteria to meet the postbiotic definition are summarized here. One noteworthy aspect of this definition is that the word ‘probiotic’ does not appear. Although in practice a probiotic strain may be used as a progenitor strain in the manufacture of a postbiotic, the simple process of inactivating a probiotic is not sufficient to be called a postbiotic. It cannot be assumed that any non-viable probiotic cells in a probiotic product are automatically considered a postbiotic component. If a probiotic strain is used as a progenitor of a postbiotic, an efficacy study must be redone using the inanimate preparation and a benefit must be demonstrated. A probiotic product displaying fewer than the labeled count of viable cells is merely a low-quality product; it is not a postbiotic.

Further, the ISAPP consensus definition on postbiotics recognizes that the process of making a postbiotic implies a deliberate step to inactivate the viable cells of the progenitor strain. This process can be achieved by different technological steps such as heat-treatment (perhaps the most feasible approach), high pressure, radiation or simply aerobic exposure for strict anaerobes. A corresponding efficacy study must be conducted on the preparation. Or at the very least, any postbiotic component of a probiotic product must be specifically shown to contribute to the health benefit conferred by the product.

In contrast to postbiotics, probiotics are live microorganisms which when administered in adequate amounts confer a health benefit on the host. Four minimum criteria should be met for a strain to be considered as a probiotic: (i) sufficiently characterized; (ii) safe for the intended use; (iii) supported by at least one positive human clinical trial conducted according to generally accepted scientific standards or as per recommendations and provisions of local/national authorities when applicable; and (iv) alive in the product at an efficacious dose throughout shelf life (Binda et al. 2020). This last requirement reflects the key difference between probiotics and postbiotics. Probiotics must deliver an efficacious number of viable cells through the shelf life of the product. In practice, probiotic products may display significant numbers of non-viable cells (Raymond & Champagne, 2015), as some cells may lose viability during the technological process of biomass production, while undergoing manufacture or preservation steps and through product storage prior to purchase. In order to provide the target dose until end of shelf life, an overage of 0.5 to 1 log order CFU above the expected counts of viable cells is commonly included in the product to compensate for potential losses during product storage and handling (Fenster et al. 2019).

Thus, some quantity of non-viable cells may be usually expected in certain probiotic products, especially supplement products claiming a long, room temperature stable shelf-life. However, they will be considered as probiotic products of quality as long as they are able to deliver the expected amount of viable cells until the end of the product shelf-life. It is worth mentioning that the probiotics are expected to be viable at the moment of their administration. After that, if exposure to different regions of the gut causes cells to die, it is not of consequence as long as a health effect is achieved.

Probiotics and postbiotics have things in common (the need of efficacy studies that demonstrates their benefits) and things that distinguish them (the former are administered alive, whereas the latter are administrated in their inanimate form), but no probiotic becomes a postbiotic just by losing cell viability during storage.

Do new product formats need new clinical trials?

By Marla Cunningham​, Metagenics Global R&D Innovation Manager and 2021 ISAPP Industry Advisory Committee representative

Let’s assume a hypothetical clinical study has been published with positive impacts of a yoghurt containing Lacticaseibacillus rhamnosus strain XYZ in children with atopic dermatitis. If the strain is now to be incorporated into a fruit drink, at the clinically trialled dose throughout shelf life, can it be expected to have the same health benefits? Can the probiotic yoghurt study provide primary support of efficacy claims on the probiotic fruit drink? Such a question is highly relevant to the challenges that food and supplement manufacturers within the ISAPP community face daily in product development.

This important scientific and regulatory question is addressed in a new ISAPP-driven collaborative article, originating from opinions and data presented at the industry-organised Learning Forum at the 2019 ISAPP annual meeting in Antwerp. The paper, published online April 21 in Trends in Food Science and Technology, reviewed preclinical and clinical evidence for an impact of product matrix on functionality of probiotics and prebiotics.

The article notes it is well-recognised that heat, pH and moisture are key factors causing degradation in probiotics and prebiotics, and such factors currently weigh heavily in formulation design and quality assurance processes for these products. Beyond such impacts on degradation, some evidence suggests that ingredients in the product matrix can affect probiotic and prebiotic functionality in vitro, for example via the binding of proteins or carbohydrates to structural components of prebiotics or altering activity of effector molecules on probiotics.

However, clinical trials do not provide convincing evidence that observed preclinical interactions are significant in vivo. Head-to-head clinical trials comparing product formats are rare, meaning that direct evidence that product formats can influence a clinical endpoint is lacking. To address this gap, researchers are encouraged to consider comparing different matrixes in future clinical trials. Yet, while differences in study factors (such as populations, interventions and doses) limit conclusions that can be drawn from comparing across clinical studies, meta-analyses in general suggest a robustness of effect across a broad range of delivery matrices for given clinical endpoints.

Preclinical assessments are useful, but limited. Attempts to replicate findings from highly controlled preclinical experiments often fail because preclinical assessments cannot capture the complexity of the physiology or the individual factors inherent to the human subject.  It makes sense that any impact of physicochemical interaction between probiotics or prebiotics with a product matrix may not be revealed in vivo. If we consider the almost infinite number of variations that could make up a study subject’s (or consumer’s) diet, probiotics and prebiotics are in fact being delivered in a variety of matrices every day, with substantially greater potential for physiochemical interactions in the digestive tract outside of product formulation variables. Add to this interindividual differences in human physiology and microbiome, and the overall impact of product formulation differences on the expression of a clinical effect in an end consumer may be smaller still.

This broader perspective suggests that even if it were ethically and practically possible, unrestrained investment into the repetition of clinical trials for each new product format may not be the answer to provide a high degree of confidence for translation of clinical trial evidence to any given consumer. Instead, research dollars may be better spent in the short term on mechanistic and clinical studies investigating the relative impact of factors determining individual response to probiotic and prebiotic intervention, including factors intrinsic to the host as well the product formulation.

Nonetheless, it is critical that any extrapolation of evidence across product formats is supported by a solid scientific rationale. As such, the article provides recommendations for a practical path forward to demonstrate essential equivalence between product formats, utilising in vitro and in vivo tests, and clinical trials where justified. Such an approach is intended to provide reasonable assurance of scientific substantiation and may also go some way to meeting the expectations of regulatory authorities across the globe (reviewed within).

The open access article can be found here.

 

Current status of research on probiotic and prebiotic mechanisms of action

By Mary Ellen Sanders, PhD, ISAPP Executive Science Officer

Human intervention studies in the fields of probiotics and prebiotics assess the health effects of these ingredients, whether it’s improving specific symptoms or preventing the occurrence of a health condition. Yet scientists in the field recognize the importance of learning the ‘chain of events’ by which probiotics and prebiotics are able to confer health benefits. Such mechanistic insights allow better probiotic selection and development of therapeutic approaches, as well as more precise dosing.

Mechanisms of action for probiotics and prebiotics are complex and often difficult to pinpoint, especially since any given health benefit may derive from multiple co-functioning mechanisms. However, scientists have made incremental gains in understanding these mechanisms. This scientific progress was covered in a recent webinar co-presented by ISAPP and ILSI-Europe, titled Understanding Prebiotic and Probiotic Mechanisms that Drive Health Benefits. Speakers for the webinar were:

  • Sarah Lebeer, University of Antwerp, Belgium
  • Colin Hill, University College Cork, Ireland
  • Karen Scott, University of Aberdeen, UK
  • Koen Venema, Maastricht University – campus Venlo, The Netherlands

The webinar was held live on September 17, 2020. Of the 499 webinar registrants, 357 attended the webinar live from 57 countries, from Australia to the US. ISAPP and ILSI-Europe hope the webinar will serve as a resource for people who want a rapid overview about mechanisms of action.

Watch the full webinar here, and read further for a summary of key points from these experts.

Prebiotic benefits and mechanisms of action

Prebiotics are defined as substrates that are “selectively utilized by host microorganisms conferring a health benefit”. ‘Utilization’ in the gut may involve crossfeeding, which means products produced by the first microbes degrading the prebiotic can then be used by different members of the host microbiota – so it may take a series of complex steps to get to a final health outcome. However, selective utilization and health benefit are always required for a substance to meet the definition of a prebiotic.

The health benefit of a prebiotic can be local (in the gut) or systemic. Locally, prebiotics can act via fecal bulking, as they are typically types of fiber. In addition, they can produce short-chain fatty acids (SCFAs), which reduce gut pH and thereby can discourage pathogenic and toxigenic activity of gut microbes, increase calcium ion absorption and provide energy for gut epithelial cells.

Systemic functions of prebiotic metabolism include them being used as substrates for microbes that produce or interact with host cells to produce molecules with neurochemical, metabolic or immune activity. Further, SCFAs can end up in the blood and can reach the liver, muscles and the brain. The SCFAs interact with specific host receptors and can lead to the release of satiety hormones or interact with receptors in the liver, adipose tissue and muscle tissue, leading to reduced inflammation. Prebiotics can also interact directly with immune cells.

Probiotic health effects and mechanisms of action

Health and disease are the end results of complex interactions on a molecular scale within a human or animal host.  Host molecules also interact with microbial molecules, including those molecules introduced with or produced by probiotics. Designing studies to discover probiotic mechanisms in human research is extremely challenging because both host and probiotic are very complex systems that most probably engage with one another on multiple levels. Probiotic molecules can have direct effects and downstream effects, and we are aware of only a few cases where a health effect can be tied to one specific probiotic molecule.

Probiotics can interact directly with the host, but also can act indirectly by influencing the microbiome. There may be many different mechanisms by which a given probiotic interacts with the host.

It is interesting to note that probiotics use some of the same types of mechanisms (pili, small molecule production, etc.) that are used by pathogens, microbes that have a detrimental effect on the host.  But these shared mechanisms are usually connected to surviving or colonising strategies, not those that cause damage to the host.

L. rhamnosus GG is a well-researched model probiotic, for which many mechanisms have been identified, including pili, immune modulators and lactic acid production, some mechanisms shared with other probiotic strains and species. Other studies have identified mechanisms for novel types of probiotics. For example, in mice and humans taking a strain of Akkermansia, heat killed cells had the same or even better effect on markers of metabolic health, which implies that the molecules (perhaps proteins in the bacteria, unaffected by heat treatment) are mediating the effect in this case.

See here to watch the webinar in full.

 

 

EFSA’s QPS committee issues latest updates

By Bruno Pot, PhD, Vrije Universiteit Brussel and Mary Ellen Sanders, PhD, Executive Science Officer, ISAPP

On July 2nd, the European Food Safety Authority (EFSA) published the 12th update of the qualified presumption of safety (QPS) list, a list of safe biological agents, recommended for intentional addition to food or feed, covering notifications from October 2019-March 2020. It was good news to all stakeholders to see that EFSA discussed the recent taxonomic changes within the genus Lactobacillus (see ISAPP blog here) as well as addressed some microbes being considered as potential, novel probiotics.

What is QPS?

In 2005 EFSA established a generic approach to the safety assessment of microorganisms used in food and feed, prepared by a working group of the former Scientific Committee on Animal Nutrition, the Scientific Committee on Food and the Scientific Committee on Plants of the European Commission. This group introduced the concept of “Qualified Presumption of Safety” (QPS), which described the general safety profile of selected microorganisms. The QPS process was mainly developed to provide a generic pre‐evaluation procedure harmonized across the EU to support safety risk assessments of biological agents performed by EFSA’s scientific panels and units. A QPS assessment is performed by EFSA following a market authorisation request of a regulated product requiring a safety assessment. Importantly, in the QPS concept, a safety assessment of a defined taxonomic unit is performed independently of the legal framework under which the application is made in the course of an authorisation process.

QPS status is granted to a taxonomic unit (most commonly a species), based on reasonable evidence. A microorganism must meet the following four criteria:

1.       Its taxonomic identity must be well defined.

2.       The available body of knowledge must be sufficient to establish its safety.

3.       The lack of pathogenic properties must be established and substantiated (safety).

4.       Its intended use must be clearly described.

Any safety issues, noted as ‘qualifications’, that are identified for a species assessed under QPS must be addressed at the strain or product level. Microorganisms that are not well defined, for which some safety concerns are identified or for which it is not possible to conclude whether they pose a safety concern to humans, animals or the environment, are not considered suitable for QPS status and must undergo a full safety assessment. One generic qualification for all QPS bacterial taxonomic units is the need to establish the absence of acquired genes conferring resistance to clinically relevant antimicrobials (EFSA, 2008).

If an assessment concludes that a species does not raise safety concerns, it is granted “QPS status”. Once EFSA grants a microorganism QPS status, it is included on the “QPS list” and no microorganism belonging to that group needs to undergo a full safety assessment in the European Union.

The QPS list is re‐evaluated every 6 months by the EFSA Panel on Biological Hazards based on three “Terms of Reference” (ToR)*. This evaluation is based on an extensive literature survey covering the four criteria mentioned above.

What happened to the genus Lactobacillus?

In April 2020, based on a polyphasic approach involving whole genome sequencing of more than 260 species of the former genus Lactobacillus, the genus was reclassified into 25 genera including the emended genus Lactobacillus, which includes host-adapted organisms that have been referred to as the L. delbrueckii group, the earlier described genus Paralactobacillus as well as 23 novel genera, named Acetilactobacillus, Agrilactobacillus, Amylolactobacillus, Apilactobacillus, Bombilactobacillus, Companilactobacillus, Dellaglioa, Fructilactobacillus, Furfurilactobacillus, Holzapfelia, Lacticaseibacillus, Lactiplantibacillus, Lapidilactobacillus, Latilactobacillus, Lentilactobacillus, Levilactobacillus, Ligilactobacillus, Limosilactobacillus, Liquorilactobacillus, Loigolactobacilus, Paucilactobacillus, Schleiferilactobacillus, and Secundilactobacillus. Read more in the original paper here or on the ISAPP blog here).

These name changes could have considerable economic, scientific and regulatory consequences, as discussed during an expert workshop organised by the Lactic Acid Bacteria Industrial Platform (LABIP). One of the points discussed during this workshop was the possible implication of the name change on the QPS list in Europe and the FDA’s GRAS list in the USA.

What did EFSA do?

In a 42-page document, which can be found here, amongst others, the species of the former genus Lactobacillus that were already listed on the QPS list, have been formally renamed at the genus level. The species names remained the same, as the taxonomic revision from April 2020 only affected the genus name. As a result, the genus names of 37 former Lactobacillus species on the QPS were updated, and now span 13 different genera. Table 1 delineates these nomenclature updates.

Table 1: Taxonomic revision of the 37 species formerly of the Lactobacillus genus present on the QPS list (published here).

Earlier denomination                                                      Updated denomination
Lactobacillus acidophilus                     Lactobacillus acidophilus
Lactobacillus alimentarius Companilactobacillus alimentarius
Lactobacillus amylolyticus Lactobacillus amylolyticus
Lactobacillus amylovorus Lactobacillus amylovorous
Lactobacillus animalis Ligilactobacillus animalis
Lactobacillus aviarius Ligilactobacillus aviarius
Lactobacillus brevis Levilactobacillus brevis
Lactobacillus buchneri Lentilactobacillus buchneri
Lactobacillus casei Lacticaseibacillus casei
Lactobacillus collinoides Secundilactobacillus collinoides
Lactobacillus coryniformis Loigolactobacillus coryniformis
Lactobacillus crispatus Lactobacillus crispatus
Lactobacillus curvatus Latilactobacillus curvatus
Lactobacillus delbrueckii Lactobacillus delbrueckii
Lactobacillus dextrinicus Lapidilactobacillus dextrinicus
Lactobacillus diolivorans Lentilactobacillus dioliovorans
Lactobacillus farciminis Companilactobacillus farciminis
Lactobacillus fermentum Limosilactobacillus fermentum
Lactobacillus gallinarum Lactobacillus gallinarum
Lactobacillus gasseri Lactobacillus gasseri
Lactobacillus helveticus Lactobacillus helveticus
Lactobacillus hilgardii Lentilactobacillus hilgardii
Lactobacillus johnsonii Lactobacillus johnsonii
Lactobacillus kefiranofaciens Lactobacillus kefiranofaciens
Lactobacillus kefiri Lentilactobacillus kefiri
Lactobacillus mucosae Limosilactobacillus mucosae
Lactobacillus panis Limosilactobacillus panis
Lactobacillus paracasei Lacticaseibacillus paracasei
Lactobacillus paraplantarum Lactiplantibacillus paraplantarum
Lactobacillus pentosus Lactiplantibacillus pentosus
Lactobacillus plantarum Lactiplantibacillus plantarum
Lactobacillus pontis Limosilactobacillus pontis
Lactobacillus reuteri Limosilactobacillus reuteri
Lactobacillus rhamnosus Lacticaseibacillus rhamnosus
Lactobacillus sakei Latilactobacillus sakei
Lactobacillus salivarius Ligilactobacillus salivarius
Lactobacillus sanfranciscensis Fructilactobacillus sanfranciscensis

EFSA further specifies that “To maintain continuity within the QPS list, all the strains belonging to a previous designed Lactobacillus species will be transferred to the new species. Both the previous and new names will be retained”. (Emphasis added.)

Impact of the QPS update on the probiotic field

The probiotic field can also take note of this current update for its review of two ‘next generation’ probiotic species evaluated for possible QPS status, Akkermansia muciniphila and Clostridium butyricumAkkermansia muciniphila has been actively researched as a probiotic to help manage metabolic syndrome (Depommier et al. 2019). A probiotic preparation containing both Akkermansia muciniphila and Clostridium butyricum has been studied in a randomized controlled trial for postprandial glucose control in subjects with type 2 diabetes (Perraudeau et al 2020). The committee’s decisions:

  • Akkermansia muciniphila is not recommended for QPS status due to safety concerns;
  • Clostridium butyricum is not recommended for QPS status because some strains contain pathogenicity factors; this species is excluded for further QPS evaluation.

The publication of the next scientific opinion updating the QPS list is planned for December 2020, based on the 6-month assessments carried out by the BIOHAZ Panel.

Conclusion

Due to its scientific rigor and continuous updates, the EFSA QPS efforts provide useful perspective for the global scientific community on safety of candidate microbes for use in foods. Their embrace of the new taxonomic status of lactobacilli signals to other stakeholders that it is time to start the process of doing the same. Further, their assessment of species being proposed and studies as ‘next generation’ probiotics is an important reminder that a microbe’s status as a human commensal is not a guarantee of its safety for use in foods.

 

*QPS Terms of Reference (ToR) (quoted from here):

ToR 1: Keep updated the list of biological agents being notified in the context of a technical dossier to EFSA Units such as Feed, Pesticides, Food Ingredients and Packaging (FIP) and Nutrition, for intentional use directly or as sources of food and feed additives, food enzymes and plant protection products for safety assessment.

ToR 2: Review taxonomic units previously recommended for the QPS list and their qualifications when new information has become available. The latter is based on a review of the updated literature aiming at verifying if any new safety concern has arisen that could require the removal of the taxonomic unit from the list, and to verify if the qualifications still efficiently exclude safety concerns.

ToR 3: (Re)assess the suitability of new taxonomic units notified to EFSA for their inclusion in the QPS list. These microbiological agents are notified to EFSA and requested by the Feed Unit, the FIP Unit, the Nutrition Unit or by the Pesticides Unit.

 

New publication addresses the question: Which bacteria truly qualify as probiotics?

Although the international scientific consensus definition of probiotics, published in 2014, is well known—”live microorganisms that, when administered in adequate amounts, confer a health benefit on the host”—the word is often used incorrectly in practice.

A recent article published in Frontiers in Microbiology builds on this definition and describes four criteria for accurate use of the word ‘probiotic’. Eight scientists co-authored the paper, including two ISAPP board members. The project was initiated by industry scientists affiliated with IPA Europe.

The authors explain why it’s important for scientists and companies to be sure the four identified criteria apply before using the term ‘probiotic’. Given the many misuses of the term that are evident today, however, consumers need to scrutinize ‘probiotic’ products to be sure they are legitimate.

Read the ISAPP press release on this publication here.

See an infographic summary of this publication here.

 

 

ISAPP’s popular educational videos now feature subtitles in multiple languages

ISAPP’s series of six English-language videos are a useful resource for helping consumers answer important questions about probiotics, prebiotics, and fermented foods. In order to make these popular educational videos accessible to a wider global audience, ISAPP has now updated them with subtitles in multiple languages: Dutch, French, German, Indonesian, Italian, Japanese, Russian, and Spanish.

Dr. Roberta Grimaldi, a principal clinical research scientist who served as ISAPP’s Industry Advisory Committee representative from 2017-2019, led the video subtitling efforts.

“The videos are a good way to communicate information about these products, which are still not fully understood by consumers,” says Grimaldi. She says that while consumers see “a lot of miscommunication and misleading information” online, the easy-to-understand ISAPP videos help bring the scientific perspective to a broad audience.

Multi-lingual members of the ISAPP community stepped up to help with the translations, with Grimaldi managing the task and co-ordinating with the video production agency. She says, “It was definitely an amazing team effort, which I think gave us really great results.”

Science Translation Committee head Dr. Chris Cifelli underlines how worthwhile the video subtitles project has been for ISAPP. “Since ISAPP is an international organization, we have been working hard to make our educational materials accessible to as many people as possible. The subtitles allow the information in these videos to be shared much more widely, ultimately helping consumers make more informed decisions about probiotics, prebiotics, and fermented foods.”

Many of ISAPP’s infographics are also available in multiple languages.

 

How to change the language subtitles on an ISAPP video:

Step 1 – On the ISAPP videos page, click on the video.

Step 2 – Press pause and click the gear-like ‘Settings’ icon, to the right of the ‘CC’ icon.

Step 3 – Click on ‘Subtitles’ and select the language subtitle you prefer.

Step 4 – Resume the video by pressing play.

What makes a synbiotic? ISAPP provides a sneak peek at the forthcoming international scientific consensus definition

By Kristina Campbell, science and medical writer

The word ‘synbiotic’ is found on the labels of many different products, from supplements to chocolate bars, and it has generally been understood to be a combination of a probiotic and a prebiotic. But what happens when scientists want to test whether these combination products really deliver any health benefits? Can these products be tailored to have specific effects on the body or on the human gut microbiota? Agreeing on a clear definition of synbiotics is needed to provide focus for scientific research in this area, to facilitate the design of studies, and to allow for progress wherein their health effects are uncovered.

The scientific definition of synbiotic was the central topic of the international scientific panel brought together by ISAPP in May 2019 in Antwerp, Belgium. Members of the panel, eleven of the top academic experts in the field of probiotics and prebiotics, gathered to clarify a scientifically valid approach for use of the word ‘synbiotic’, and to communicate this by position paper. The outcome of this consensus panel is currently in press at Nature Reviews Gastroenterology & Hepatology.

Kelly Swanson, Professor in the Department of Animal Sciences and Division of Nutritional Sciences at University of Illinois at Urbana-Champaign, chaired the panel and led the paper’s publication. Swanson has been studying gastrointestinal health in both humans, companion animals (dogs and cats) and rodent models for the past 20 years—and having followed the rapid advances in the field of probiotics and prebiotics during those two decades, he knew the task of creating a synbiotic definition would not be easy.

He says, “The field is highly complicated, so an interdisciplinary panel was essential. The main areas of expertise included microbiology and microbial ecology; gastrointestinal physiology; immunology; food science; nutritional biochemistry and host metabolism.”

A timely discussion

According to Swanson, an increase in research interest, built on a foundation of recent scientific and technical gains, made this the right time to come to consensus on a synbiotic definition. He says, “Over the past decade, technological advances have allowed scientists to study the gut microbiome at a molecular level. In addition to characterizing the composition of the gut microbes, researchers are learning more about their biological activity and how they may impact host health.”

Furthermore, clarity about the definition was urgently needed because of the rapidly growing synbiotics market. Consumers seem to be more aware of synbiotics than ever, but they face a bewildering array of product offerings labeled as ‘synbiotic’ without a clear understanding of what that term entails and with no framework for establishing scientific efficacy. Swanson says, “As the field has moved forward and the sales of probiotics and prebiotics have increased, there has been more interest in combining substances to enhance efficacy. Some of these combinations may function as synbiotics, but it is not guaranteed. Rather than randomly combining substances together, there should be scientific rationale supporting their use.”

Clarifying the concept

One of the first questions the panel members had to tackle was whether to stick to the idea of a synbiotic as ‘probiotic plus prebiotic’, thus leaning heavily on the ISAPP-led international consensus definitions of probiotics and prebiotics published in 2014 and 2017, respectively. But the panel members decided this narrow scope would ultimately limit innovation in the synbiotic category.

Swanson explains, “While many synbiotics may be composed of an established prebiotic and established probiotic, the panel did not want to restrict scientific advances in the synbiotic category by requiring use of components already established on their own.”

As a result, he says, previously untested live microbes and potential prebiotic substances could be considered a synbiotic if the combination showed efficacy, and if the health benefit came from administering both the live microbe and the substrate it utilized—that is, the microbe together with its ‘food’.

Another conclusion from the panel is that probiotics (with known health benefits) and prebiotics (with known health benefits) cannot be called synbiotics unless they have been tested together. “There should be a rationale supporting the combination used, and then testing of the combination to confirm its efficacy,” says Swanson.

The panel suggests a synbiotic may be composed of either of the following, as long as efficacy is demonstrated for the combination:

  • Established probiotic + established prebiotic (each component meeting the efficacy and mechanistic criteria for each)
  • Previously untested live microbe + a substrate that is selectively utilized by the co-administered live microbe

Further details, including two different ‘categories’ of synbiotics, will be provided in the published paper.

In addition to the definition, the publication will cover the history of synbiotic-type products, how these products can be characterized, levels of evidence that currently exist versus levels of evidence desired, points about safety documentation and reporting, and relevant characteristics of the target hosts.

A remaining challenge—not just for the expert group, but also across the field—is the difficulty of establishing causal links between substances’ effects on the gut microbiota (e.g. ‘selective utilization’ of a substrate) and health outcomes.

While the publication of the synbiotic definition will be an important milestone, Swanson anticipates further discussion in the years ahead. “As more is learned, I expect the criteria for assessing synbiotic efficacy will continue to change,” he says.

An update on the scientific consensus definition of synbiotic was presented to ISAPP members at the 2020 virtual meeting in June.

 

The science on gut microbiota and intestinal gas: Everything you wanted to know but didn’t want to ask

By Kristina Campbell, science and medical writer

Even on the days when you don’t eat a large meal of Boston baked beans, the inside of your intestines is a gas-generating factory. This serves a valuable purpose for the body when everything is working as it should, with gases being produced and eliminated through a complex set of physiological processes. But sometimes gas becomes a problem—and this is when it’s valuable to know not only what contributes to intestinal gas symptoms, but also how dietary adjustments can alleviate some of the problems.

Dr. Fernando Azpiroz, Chief of Gastrointestinal Research at the Vall d’Hebron Research Institute and Professor of Medicine, Autonomous University of Barcelona (Spain), is an expert in both the pathophysiology of the gas produced in the digestive tract and the clinical problems related to intestinal gas. Dr. Azpiroz is the author of a chapter on intestinal gas in the well-known textbook, Sleisenger and Fordtran’s Gastrointestinal and Liver Disease, which is now in its 10th edition. And for more than 20 years he has been conducting research on digestive tract function, intestinal gas, and the contributions of the gut microbiota.

ISAPP caught up with Dr. Azpiroz to ask him about everything you wanted to know—where intestinal gas comes from, when it becomes a problem, and the latest research on the dietary changes that can reduce symptoms of intestinal gas while keeping the gut microbiota intact.

In different parts of the digestive tract, where does intestinal gas come from?

For the most part, the gas in the digestive system comes from metabolic activities of the colonic microbiota.

In addition, some air enters the stomach during swallowing. Most of this air is eliminated by eructation (i.e. burping), so there is a homeostasis. There is a small air chamber in the stomach of about 20 mLs, and this is pure atmospheric air, or CO2 after recent consumption of carbonated drinks.

In the small intestine, the neutralization of acids and alkali can theoretically produce large amounts of CO2. However, it’s more in theory than measured in experimental conditions.

Other gases originate from the metabolism of the gut microbiota. The residues of the diet that are not absorbed in the small bowel pass into the colon. These are the parts of the diets that we, as humans, cannot use. These pass into the colon, and in the colon serve as substrates for colonic microbiota. Gas is produced during this process of colonic fermentation.

What types of gases are produced in the digestive tract?

The composition of intestinal gas depends on where in the gut it is produced.

In the stomach, the gas is virtually all atmospheric air or CO2.

In the small bowel, theoretically it should be CO2, although the hard data are very scarce.

And in the colon, the largest component is likely hydrogen and CO2. But the data on that are very limited, and it is not known for sure that these theories are really true. The measurements of gas composition, in the colon or even in the gas eliminated per anus are still uncertain but so far the main concept is that most of the gas is hydrogen and CO2, and methane in subjects that have a methanogenic microbiota.

How does this gas normally get eliminated?

Throughout the GI tract, particularly the colon, about 80% of the gases produced are absorbed through the mucosa, pass into the blood, and are eliminated by breath. So only about 20-25% of the volume of gases produced, particularly in the colon, are eliminated per anus.

What influences the amount of gas produced?

The amount of gas produced in different subjects depends on two factors: one is the diet—the amount of residues (i.e. fiber) in the diet—and the second is the type of microbiota, which is very individual. It varies a lot from one subject to another.

How much intestinal gas is ‘too much’?

From the point of view of patients, of symptoms, what might be relevant is the volume of gases produced, and possibly the type of gases. One evolving idea is that some gases, which are produced in very small quantities, might serve as neurotransmitters, might play a role, but so far the information we have about the role of these gases is very limited so we cannot extrapolate that to clinical use.

Very recent data indicate that symptoms occur when the GI tract has a poor tolerance to its contents, and particularly to gas.

So what is the factor that makes gas produce symptoms? Well there are two factors. One is the amount of gas, and the other one is the tolerance of the subject.

In healthy subjects, it will typically take a large amount of gas to develop symptoms, if at all.

But even small amounts of gas in patients that have a hypersensitivity of the gut and poor tolerance to contents might contribute to their symptoms. This can be seen because, if we reduce the amount of flatulogenic substances in their diet, the symptoms reduce.

This is if we just take into consideration the volume of gas produced, but there is also another factor.

If we give a high flatulogenic diet or a high-residue diet, we know, because we have measured, that we increase the volume of gas produced into the colon. However, we also increase the amount of the fecal content in the colon. So it could be that these diets produce symptoms because they increase the amount of gas, but also because other non-gaseous components, the fecal content of the colon, also are poorly tolerated.

How can someone manipulate their diet to change the amount of intestinal gas that’s produced?

A ‘challenge’ diet, or a high flatulogenic diet in healthy subjects, makes them sick. They go from being symptom-free to having some symptoms, particularly flatulence and bloating.

In patients, the effect of the diet is more accentuated. If patients consume a diet with high residues they get very symptomatic, and if these patients will reduce residues in the diet, they see quite an improvement.

What are the options for dietary change when someone has IBS or wants to reduce gas symptoms?

One thing that has been shown recently is that the effect of a low-residue diet is similar regardless of the type of diet. In the past ten years or so there’s been a major trend with the use of low-FODMAP fermentable oligo-, di-, mono-saccharides and polyols diets.

However, the effect of these complex diets is not better than the effect of any sensible and simple low-residue diet. So if you reduce legumes, veggies from the diet, and fruits, you get a similar improvement.

The problem with low-residue diets, in particular the low-FODMAP diet, is that they introduce a restriction of the substrates for the feeding of the microbiota, and this is deleterious to the microbiota. The microbiota is impoverished.

The other limitation of low-residue diet is that the moment that the patient returns to a normal diet, the symptoms come back.

There is an alternative that has been shown in the past few years, which is to use some type of prebiotics that initially—because they are fermented—produce symptoms, but after a few days they induce an adaptation of the microbiota towards a microbiota that produces less gas with normal fermentation. And down the road, these prebiotics have a positive effect on symptoms in patients. As a matter of fact, the effect on symptoms is similar to a restrictive diet. The advantage of the prebiotics is that, after interruption, …the effect is sustained at least over a few weeks. And this is because it has been shown that prebiotics serve as substrates for microbiota and induce a proliferation of beneficial organisms.

There is also some preliminary evidence that some probiotics reduce the volume of gas production and reduce digestive symptoms in patients.

Is it a good idea to test your gut microbiota when you have IBS or gas symptoms?

There are different companies that claim that by analyzing microbiota they can diagnose some functional conditions, for instance IBS. The practical application of this technology has not been demonstrated. Usually they are expensive techniques and of no value.

It’s important to understand the real value of these methodologies and take the myth out of ‘wonder’ techniques that make a diagnosis from the microbiota and claim it explains everything.

Hopefully, this might be true in the future, but not right now. And actually many of the technologies that are used for this type of analysis are suboptimal.

 A previous blog post by Dr. Bob Hutkins on diet, gut microbiota, and intestinal gas is ISAPP’s most-read blog post of all time. Read it here.

The FDA’s view on the term probiotics, part 1

By James Heimbach, Ph.D., F.A.C.N., JHEIMBACH LLC, Port Royal, VA

James Heimbach, food and nutrition regulatory consultant

Over the past 20 years as a food and nutrition regulatory consultant, I have filed about 40 GRAS notices with the United States Food and Drug Administration (FDA), including 15 strains of probiotic bacteria and 5 prebiotics. This fall I submitted notices dealing with 4 strains of bacteria and on January 16 received a telephone call from FDA that surprised me and initially infuriated me, but which I have come to understand.

The essence of the call was that FDA was declining to file my probiotic notices because the notices had identified the subject bacteria as “probiotics” or “probiotic bacteria.” FDA suggested that I resubmit without calling the subject microorganisms “probiotics.”

 

 

As I said, I was surprised and frustrated, and I still would prefer that when FDA makes a policy swerve they would do it in a way that does not make extra work for me and delay my clients’ ability to get to market in a timely manner.

What I have had to do here is remove my advocate’s hat and put on my regulator’s hat. (I worked for FDA for a decade . . . long ago [1978 to 1988], but I still remember how to think like a regulator.) And here is the issue. Recall that GRAS is concerned with safety, not efficacy (generally recognized as safe, or GRAS), and the information provided in a GRAS notice is focused on safety (although benefits may be more-or-less incidentally covered). The reviewers at FDA are charged with assessing whether the notice provides an adequate basis to conclude that there is a reasonable certainty that no harm will result from the intended use. They are not charged, and they are not equipped, to evaluate what benefits ingestion of the substance or microorganism might provide. So they are not in a position to say whether the subject microorganism will “confer a health benefit on the host,” which is to say, they are not in a position to say whether or not it may be regarded as a probiotic. Remember, probiotics are defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host (Hill et al. 2014).

Given that the FDA reviewers cannot say whether the notified microorganism is rightly called a probiotic, they are reluctant to sign off that they have no questions about a notice that calls it one. Regulatory agencies have to be careful; things sometimes come back to haunt them. Those who have been following FDA’s GRAS-notice response letters for a couple of decades will be aware that the agency is putting more and more disclaimers into the letters—about standards of identity, about potential labeling issues, about benefits shown in clinical trials, and about Section 201(II) of the FD&C Act.

One concern that FDA likely has is that if some issue comes up in the future regarding a claim made for benefits from use of a product containing the subject bacterium, someone may make the argument that FDA had accepted that the strain is indeed a probiotic and so it presumably confers probiotic benefits. In the case of probiotics, there are also some internal FDA politics. As ISAPP meeting attendees may already be aware, FDA’s Center for Biologics Evaluation and Research (CBER) would like to claim jurisdiction over all administration of live microorganisms, and the Center for Food Safety and Applied Nutrition (CFSAN) does not seem willing to have a confrontation.

I suspect that a similar situation obtains with the term “prebiotic.” Although I have filed a number of GRAS notices for prebiotics, they haven’t been called that; they have been called fructooligosaccharides, or tamarind seed polysaccharide, or polydextrose, or 2’-O-fucosyllactose. I don’t know how FDA would respond if a GRAS determination were filed with the substance labeled as a prebiotic.

So, I’ve decided that my sympathies lie with FDA. Until and unless a microorganism has been confirmed by competent authority to have probiotic properties when used as intended in a GRAS notice, FDA is probably correct in rejecting its right to be labeled a probiotic. If it’s any consolation, this new position by the FDA has its origin in their acknowledgment of the official scientific definition of the word “probiotic”.

When Mary Ellen Sanders (ISAPP’s Executive Science Officer) reviewed my first draft of this note, she asked what I had in mind by “competent authority,” to which I don’t have a good answer at the present time except to insist that it is not FDA’s Division of GRAS Notice Review. Thirty years ago, when I was at FDA, I was in the Office of Food Science and Nutrition, and that office was charged with making determinations of that type (although I don’t recall anything about probiotics coming before us). But FDA no longer has such an office. Until it does, or until it agrees on another source of authority on designation of microorganisms as non-CBER-domain probiotics, I suspect that CFSAN will continue to be very cautious in this area.

Read part 2 of this blog series here.

Defining emerging ‘biotics’

By Mary Ellen Sanders PhD

From its inception, ISAPP has been committed to clarity in both the definitions and the contextual use of terms in the fields of probiotics and prebiotics fields. This is reflected in the FAO/WHO probiotic guidelines working group conducted immediately prior to the first ISAPP meeting in 2002, as well as our more recent consensus panels convened on probiotics (2013), and prebiotics (2016). We also have additional panels in progress on synbiotics (convened in May 2019 in Antwerp), fermented foods (scheduled for September) and postbiotics (scheduled for December).

A recently published paper, Emerging Health Concepts in the Probiotics Field: Streamlining the Definitions, addresses definitions of many newer terms in the ‘biotics’ arena, including probiotics, prebiotics, synbiotic, pharmabiotics, postbiotics, probioceuticals, paraprobiotics, oncobiotics, psychobiotics, and live biotherapeutic products. In my opinion, although this paper provides useful discussion of issues surrounding the proliferation of terms in the ‘biotics’ area, it falls short of providing clear direction for the field and indeed may well add to confusion by introducing unnecessary, new and poorly defined terms.

For example, the term ‘symbiotics’ is perpetuated, presumably as a synonym to synbiotic. It was a missed opportunity to clarify that the term ‘synbiotic’ is derived from the Greek root ‘syn’ meaning ‘with’ or ‘together.’ The term ‘symbiotic’ is simply incorrect, adds nothing and should be eliminated altogether.

This paper fails to advance the ISAPP consensus definition of prebiotic, published in 2017, by lead author Glenn Gibson, co-inventor of the terms ‘prebiotic’ and ‘synbiotic’. It is not clear whether the authors disagree with the ISAPP consensus definition, and if so, on what basis. They state that the ISAPP consensus definition is “the most actual definition”, the meaning of which is not clear to me, but then use an outdated definition in their summary box.

Further is the failure to acknowledge the broad scope of the definition of probiotics. Live biotherapeutic products (LBPs), which the paper states is a term that was “recently” introduced by the FDA, has been in use for over at least 15 years by the FDA’s Center for Biologics Evaluation and Research. The authors equate LBPs (which are defined as drugs) with next generation probiotics, yet these do not have to fall under the drug category any more than traditional probiotics are necessarily foods. Next generation probiotics, traditional probiotics or just probiotics can fall under numerous regulatory categories including foods, infant formulas, drugs, supplements, animal feeds, medical foods, foods for special dietary uses, and perhaps even cosmetics or medical devices. Thus, regulatory category is not stipulated by the definition, which is appropriate.

One of the difficulties with sorting through these terms is the lack of any consistent basis for defining them. Some terms, such as pharmabiotics and LBPs, are linked to specific regulatory categories. Others are defined by the nature of how they are comprised: live cells, cell components, or fermentation endproducts. Others are defined by their physiological benefit: psychobiotic, oncobiotic, immunobiotic. Even still, others are defined by their state of innovation: traditional vs. next generation probiotics. This state of affairs makes is impossible to develop a logical framework for categorizing them. Instead, we are left with a long list of substances that might be related, but have little real value. Where does it all stop? Next we will have to sift through thera/metabo/gen/retro/plas/func-biotics or any other pointless terms that can be arbitrarily slapped in front of ‘biotic.’

Certainly, there is nothing to prevent any person from coining a new term for a niche development. The many stakeholders in the broader ‘biotics’ field will, I suppose, determine any given term’s utility. I believe it would have been worthwhile for this paper to make an appeal to scientists to refrain from muddying the water by proposing new terms, and instead use existing terms with appropriate modifiers. For example, use ‘immune-active probiotic’ instead of ‘immunobiotic’, or ‘probiotic drug’ instead of ‘live biotherapeutic product.’ This approach is clearer to regulators and international organizations such as Codex Alimentarius, the US Food and Drug Administration and European Food Safety Authority. To the extent that the definitions of terms need to be clarified, I believe that the ISAPP approach, using groups of 10 or more well-known academic experts in the field reaching a consensus after extensive background search, is preferred over unilateral proclamations as delivered by this paper.

The Art of Interpretation

By Prof. Gregor Reid, BSc Hons PhD MBA ARM CCM Dr HS, Lawson Research Institute, University of Western Ontario, Canada

It takes a certain degree of intelligence to become a scientist, and certainly hard work to be able to fund a lab and students. Yet, is it not bemusing when scientists cannot interpret simple things like definitions and the results of human studies?

I’ve written repeatedly, as have others, about the definition of probiotics (in case you forgot – “Live microorganisms that, (or which) when administered in adequate amounts, confer a health benefit on the host”),1,2 and yet people look at it and must think that ‘dead’ fits, as does ‘consume’, as does ‘colonize’. It beggar’s belief how such a simple definition can be so badly interpreted by intelligent people.

Time after time papers I review mis-write and/or misinterpret the definition. Conference after conference, I hear dieticians, pharmacists, physicians, scientists not only get the definition wrong, but say things like ‘the probiotics in kombucha’ when there are none, ‘we have lots of probiotics in our gut’ when you don’t unless you consumed them, ‘the lactobacilli need to colonize’ when this was never a prerequisite nor does it happen except in rare instances.

The interpretation gets more difficult when people use terms that are completely undefined like ‘psycho-biotics’ and ‘post-biotics’. Even ‘dead probiotics’ have been used in clinical trials – God help us when the authors can’t even define it. Why stop at killing probiotic strains? Why not just kill any bacterial strain? Even the gut-brain axis which is now mentioned everywhere in the literature is undefined and unproven. The vagus nerve links to many body sites as does the nervous system, making it exceedingly difficult to prove that brain responses are only due to the gut microbes.

Everyone can site a manuscript that has been badly analyzed, interpreted or peer-reviewed, or whose findings are overblown. But let’s not excuse this as ‘it’s just science’ or ‘it’s just the way it is.’ No, it is not. When a paper uses a product that is stated to be ‘probiotic’, there is an onus on the authors to make sure the product meets the appropriate criteria. These have been stated over and over again and reiterated this March, 2019.3

If scientists and science writers are really that smart, then how do they keep getting this wrong? How do we let a poorly analyzed paper get published and allow authors to say that Bacteroides fragilis is a probiotic that can treat autism?4,5 And when this leads to companies claiming probiotics can treat autism, why do other scientists convey cynicism for the field instead of against their colleagues and specific companies making the false claims?

Where does opinion cross the line with ignorance or stupidity? Martin Luther King Jr. must have predicted life today when he said, “Nothing in all the world is more dangerous than sincere ignorance and conscientious stupidity.”

Is it envy or anger that drives the anti-probiotic sentiments? It seems to go far beyond a difference of opinion. When the BBC and JAMA fail to comment on two much better and larger studies on the effects of probiotics published6,7 at the same time as the ones in Cell8,9 that were promoted by press releases, what is driving opinion? The science or the press releases? Are the journalists and communications’ people interpreting study results vigorously? One cannot believe they are.

In an era where anyone can write anything at any time and pass it along to the world, what are we recipients to do? Just go with our instincts? Soon, we will not know the difference between fact and fake news. The avatars will be so real, we will act on falsehoods without knowing. When all news is fake, where does that leave us as people, never mind scientists?

Manuscripts are sent for peer-review but how many reviewers are experts in bioinformatics, molecular genetics, clinical medicine, biostatistics and what happens on the front line of products to consumers or patients? Like it or not, poor studies will get out there and it will be the media who will tell the story and interpret the findings or press releases.

One must hope that confirmatory science will continue and if it fails, the writers and readers will stop citing the original incorrect report. But how often does that happen? And what are we left with?

It takes effort to object or fight back, but if we don’t then the fake news will become the norm.

Try interpreting that if you will.

 

Literature Cited

  1.  FAO/WHO. 2001. Probiotics in food.  http://www.fao.org/food/food-safety-quality/a-z-index/probiotics/en/
  2. Hill C. et al. 2014. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotics. Nat. Reviews Gastroenterol. Hepatol. 11(8):506-14.
  3. Reid G. et al. 2019. Probiotics: reiterating what they are and what they are not. Front. Microbiol. 10: article 424.
  4. Hsiao et al. 2013. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 155(7):1451-63.
  5. Sharon G, et al. 2016. The central nervous system and the gut microbiome. Cell. 167(4):915-932.
  6. Korpela K. et al. 2018. Probiotic supplementation restores normal microbiota composition and function in antibiotic-treated and in caesarean-born infants. Microbiome. 6(1):182.
  7. De Wolfe, T.J. et al. 2018. Oral probiotic combination of Lactobacillus and Bifidobacterium alters the gastrointestinal microbiota during antibiotic treatment for Clostridium difficile infection. PLoS One. 13(9):e0204253.
  8. Suez J. et al. (2018). Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018 Sep 6;174(6):1406-1423.e16.
  9. Zmora N. et al. 2018. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell. Sep 6;174(6):1388-1405.e21.
probiotics calendar

Probiotics in the Year 2018

Prof. Daniel Merenstein MD, Georgetown University School of Medicine

Messages about probiotics seem to be everywhere. It is difficult for me to keep up with the emails, links, and stories I am sent by friends and colleagues. I am regularly asked my opinion about new studies. Null trials seem to really generate the most interest, with some people looking for limitations of the study and others generally over-extrapolating the null results, seemingly at times to generate the brashest headlines.

Today I want to take a step back and share how I see probiotics in 2018.

I just reviewed a 109-page NIH grant focused on a probiotic intervention for use in a resource poor area. Throughout the grant, the authors never once defined probiotics—presumably because the definition is so commonly known. They did define ‘prebiotics’ but they never felt the need to define probiotics. Imagine that: 2018, and probiotics no longer need to be defined lest the authors seem pedantic. This would not have been the case even five years ago.

Probiotics are backed by real science, they are here to stay, and they are impacting both how we practice medicine and how consumers care for their own health. These are real products with some robust outcomes supported by well-done, independent studies. That is worth emphasizing: there is level 1 evidence for certain products and indications. On the other hand, the use of many probiotics is not evidence-based and expectations about some are not realistic. In the real world, products do not work for every indication or study population. Effect sizes and effectiveness for most indications are often small. One of my true hesitations about fecal microbial transplantation* is how nearly every study has over 90% effectiveness. That gives me cause for concern.

Thus, when there is a null trial the skeptics shouldn’t over extrapolate and the probiotic devotees should not attack the authors. We can look to studies on other treatments as an example: In November of this year NEJM published an article that showed a new antibiotic did not work well for gonorrheal pharyngeal infections. What I didn’t see were any headlines stating, “Antibiotics don’t work for pharyngeal infections.” But headlines involving probiotics often make erroneously broad generalizations. There clearly are indications for which no probiotic has been or will be shown to work. Selling a probiotic for that indication is clearly unethical. But considering the robust evidence base we have for the indication of probiotics for gastroenteritis, it is inappropriate – after 2 null trials – for headlines to read, “Probiotics Do Not Ease Stomach Flu” or “Probiotics No Better Than Placebo for Gastroenteritis”.

This fall I spoke about probiotics at two conferences, the annual meetings of the American Academy of Family Physicians (AAFP) and the annual meeting of the Academy of Nutrition and Dietetics  (FNCE). I had never spoken at either conference. With the help of a colleague, I gave two talks at AAFP; both were over-registered with all 600+ spots taken. At the FNCE, the talk was also over-registered with 350 in attendance. The level of interest in probiotics was astonishing.

What I learned from my talks is that as long as there are well-designed studies demonstrating benefits, professionals are open to probiotics and will use them correctly. Further, both the FNCE and AAFP audiences shared similar concerns: can you trust that probiotic product labels are truthful regarding contents, and are there any safety concerns? Good science and quality oversight need to continue to address these important concerns.

2018 was a great year for the advancement of probiotics in mainstream medicine. However, I think for physicians to fully embrace probiotics, the probiotic industry will better need to police itself and make sure the products they sell are what they say they are. Then they need to communicate this on the product label, using a valid quality seal (such as offered by USP), so physicians and consumers will be confident about what they are using. If the science continues to advance and we communicate about it responsibly, the use of probiotics will be used appropriately and more frequently – as they should be.

 

*For all my colleagues in the gastroenterology world who have fallen in love with fecal transplant for recurrent C. diff,  the totality of evidence as of this writing is:  187 total patients, 5 studies (2 enema, 2 colonoscopy and 1 via-nasoduodenal tube), and punchline, TWO studies were blinded. The one with the lowest rate of success was the only one that was placebo-controlled and blinded. The other blinded study was donor versus patients’ own stools. Stew on that and feel free to correct me.  

Minimum criteria for probiotics: ISAPP perspectives

By Mary Ellen Sanders PhD, Executive Science Officer, ISAPP

During its 2018 annual meeting (June 5-7), ISAPP convened a group of 30 participants from 13 countries to address issues associated with global harmonization of regulations for probiotics and prebiotics. This topic was of interest due to the broad international presence at this meeting, ISAPP’s first in Asia. The goal of this group was to provide regulators guidance derived from this assemblage of experts regarding the minimum criteria a probiotic food or supplement should meet. Drs. Seppo Salminen, Yuan-Kun Lee, and Gabriel Vinderola, who chaired this group, recently completed a summary titled “ISAPP position statement on minimum criteria for harmonizing global regulatory approaches for probiotics in foods and supplements”.

In December of 2017 the International Probiotic Association (IPA) presented a proposal to Codex Alimentarius – a recognized body that develops global standards and guidelines related to foods – regarding establishment of guidelines for probiotic foods. Codex Alimentarius accepted this proposal and requested that Argentina prepare draft guidelines to be considered in the 2018 session of the Codex Alimentarius  Committee on Nutrition and Foods for Special Dietary Use. ISAPP representatives and group coordinators (Sanders, Salminen and Vinderola) took part along with IPA in a scientific meeting in Argentina to present the ISAPP views to local authorities and experts.  IPA hopes that these efforts will lead to harmonized regulations since “this lack of harmonization in industry practice and legislation remains and often leads to serious issues and concerns for the probiotics industry, regulators, and even consumers in regard of quality, safety and labelling.” (Page1 of the proposal)

As the efforts of harmonization of regulations for probiotic foods through Codex progresses, ISAPP offers – through this summary document – its perspectives on minimum criteria for probiotics. The ISAPP group’s conclusions echo the principles outlined in the IPA proposal. Our hope is that this ISAPP document will provide useful perspective to local regulators. As of this writing, Prof. Salminen has delivered this document to the Codex representative at the Finnish Ministry of Agriculture and Food. We hope that further dissemination of the perspectives in this document will contribute to a science-based approach to global harmonization of regulations for probiotics.

See the document for the list of minimum criteria.

ISAPP Releases New Infographic – Probiotic Checklist: Making a Smart Selection

Not all products labelled “probiotic” are true probiotics. ISAPP just released a new infographic focused on helping consumers make smart selections when examining probiotic products. The infographic addresses identifying products backed by science, effective dosing, and more.

See and download the full infographic here.

See all ISAPP infographics here.

 

clinician_guides

Guides for use of probiotics in the clinic – some recent ISAPP initiatives

By Mary Ellen Sanders, PhD

At the ISAPP meeting earlier this month, Prof. Dan Merenstein, MD, presented a summary of recent ISAPP initiatives focused on helping translate the evidence of probiotics and prebiotics into clinical action.

A 2013 paper reported that 87% of hospital formularies surveyed in the United States carried at least one probiotic. Yet when Merenstein looked at the names of the products tested, many were not supported by evidence for such uses. This highlights the need for clinicians to have access to clear, evidence-based probiotic use guidelines.

ISAPP has worked through a variety of avenues to get information into the hands of clinicians. It has supported continuing education credit activities, webinars, collaboration with clinical organizations to develop guidelines, publications in clinical journals, presentations at clinical meetings, and simplified summaries using infographics and videos. Some examples include the following.

 

World Gastroenterology Organisation Global Guidelines – Probiotics and Prebiotics

This document is the most visited and downloaded of all WGO guidelines. In 2017, under the leadership of Prof. Francisco Guarner, MD PhD, this document was updated. Three current ISAPP board members were part of the process and ISAPP provided funding. See here.

 

Petitions

ISAPP petitioned the United States Preventive Services Task Force to examine the role of probiotics in preventing antibiotic-associated diarrhea. They considered the petition, but didn’t feel it fit their mission.

ISAPP petitioned American Academy of Family Physicians to consider reviewing the evidence for probiotics for AAD to include in their evidence-based guidelines. This is under consideration.

After attending 2017 ISAPP, Dr. Claire Merrifield BSc MBBS PhD led an effort to have NICE Clinical Knowledge Summaries mention probiotics for AAD in an effort to get local groups to adopt guidelines. This has met with limited success. See here.

 

CME or CE activities

On April 17, 2018, Merenstein and Mary Ellen Sanders PhD served as faculty for a CME-eligible webinar sponsored by Medscape on “Navigating the World of Probiotics. Helping Patients Make Good Choices”. The activity is available on Medscape’s website here.

In February 2018, Merenstein published a CE activity with the Pharmacy Times titled “The Expanding Health Benefits of Prebiotics and Probiotics”. See here

Upcoming in October 2018, Merenstein will present “Probiotics and the GI Tract. What Should a Busy Clinician Know” at the American Academy of Family Physicians Annual Conference. This conference is attended by over 4,000 physicians and is focused on clinical practice. The event, eligible for CME, will be recorded and made available after the live presentation.

ISAPP co-founder, Prof. Glenn Gibson has or will present 6 lectures over 2017 and 2018 on the topic of “The Learning Curve for Probiotics and Prebiotics.” These lectures are available for CME credit and are targeted to family doctors, gastroenterologists, pediatricians, and dieticians in the UK.

Numerous CME presentations over 2017-2018 have been given by ISAPP board members:

M.D. Cabana:

  • “Probiotics: Friend or Folly?”  American Academy of Pediatrics National Conference and Exhibition. Chicago, IL. September 17, 2017.  The audience was about 450-500 clinicians.
  • “Probiotics in Primary Care Pediatrics: Diarrhea, Colic & Eczema.” American Academy of Pediatrics California Chapter 1 Meeting. 300 clinicians
  • “Probiotics for Colic?” Zuckerberg San Francisco General Hospital. Department of Pediatrics Grand Rounds. San Francisco, CA.
  • “Probiotic Interventions for Colic” UCSF Benioff Children’s Hospital, Oakland.
  1. Reid:
  • “Effects and importance of microbiota on urogenital health in women.” 16th Annual Congress of Gynecology and Obstetrics, Antalya, Turkey. 300 obstetricians and gynecologists.
  • “Probiotics to whom for what?” Health World Ltd International Congress Natural Medicine 2017, Hunter Valley, New South Wales, Australia,.601 healthcare practitioners and naturopaths.
  • “The microbiome and how it relates to maternal/newborn care.” The Graham Chance Lectureship, Perinatal Research Day, London, ON. 100 neonatologists and pediatric experts.
  • “Microbes and the brain.” Integrative Healthcare Symposium, New York City. 500 naturopaths and various specialists.
  • “Probiotics and detoxification.” Environmental Health Symposium, Scottsdale, Arizona, 8th April. 500 naturopaths and various specialists.

 

Webinars

On June 28, ISAPP co-founder, Prof. Glenn Gibson, will present a webinar along with Profs. Ted Dinan and Ian Rowland titled “Why is everybody talking about gut microbiota?” Sponsored by the British Nutrition Foundation, this webinar will target healthcare professionals in the UK and Europe. See here.

 

Publications in clinical journals

Several ISAPP board members

  • Evidence-Based Probiotic Use in Family Medicine. Submitted, Journal of Family Practice. Merenstein/Sanders/Tancredi
  • Probiotics for Human Use. In press, Nutrition Bulletin. Sanders/Merenstein/Hutkins/Merrifield
  • Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Invited review in preparation, Nature Reviews Gastroenterology and Hepatology. Gibson/Reid/Sanders/Merenstein
  • Clinical perspectives of prebiotics and synbiotics. In preparation, Gastroenterology. Gibson/Quigley

 

Featured on ISAPPscience.org

Infographics

 

Videos

  • What is a probiotic?
  • Health benefits of probiotics
  • Are all probiotics the same?
  • How to choose a probiotic

 

General guidelines for choosing probiotics and prebiotics

Some initiatives that Merenstein championed were a direct result of ideas generated during the discussion group he led during the 2017 ISAPP meeting in Chicago.

 

Image courtesy of nursingschoolsnearme.com/