Next Article in Journal
Drosophila Sexual Attractiveness in Older Males Is Mediated by Their Microbiota
Next Article in Special Issue
Emerging Treatment Options for Infections by Multidrug-Resistant Gram-Positive Microorganisms
Previous Article in Journal
Exploring the Ambiguous Status of Coagulase-Negative Staphylococci in the Biosafety of Fermented Meats: The Case of Antibacterial Activity Versus Biogenic Amine Formation
Previous Article in Special Issue
Impact of Chronic Obstructive Pulmonary Disease on Incidence, Microbiology and Outcome of Ventilator-Associated Lower Respiratory Tract Infections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bacterial, Gut Microbiome-Modifying Therapies to Defend against Multidrug Resistant Organisms

1
Infectious Disease Department, Ochsner Clinic Foundation, New Orleans, LA 70121, USA
2
The University of Queensland Faculty of Medicine, Ochsner Clinical School, New Orleans, LA 70121, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(2), 166; https://doi.org/10.3390/microorganisms8020166
Submission received: 29 December 2019 / Revised: 17 January 2020 / Accepted: 17 January 2020 / Published: 24 January 2020
(This article belongs to the Special Issue Multidrug-Resistant Pathogens)

Abstract

:
Antibiotics have revolutionized human and animal healthcare, but their utility is reduced as bacteria evolve resistance mechanisms over time. Thankfully, there are novel antibiotics in the pipeline to overcome resistance, which are mentioned elsewhere in this special issue, but eventually bacteria are expected to evolve resistance to most new compounds as well. Multidrug resistant organisms (MDROs) that cause infections increase morbidity, mortality, and readmissions as compared with susceptible organisms. Consequently, many research and development pipelines are focused on non-antibiotic strategies, including fecal microbiota transplantation (FMT), probiotics and prebiotics, and a range of therapies in between. Studies reviewed here focus on efforts to directly treat or prevent MDRO infections or colonization. The studies were collected through clinicaltrials.gov, PubMed, and the International Conference on the Harmonisation Good Clinical Practice website (ichgcp.net). While the gold standard of clinical research is randomized controlled trials (RCTs), several pilot studies are included because the field is so young. Although a vast preclinical body of research has led to studies in humans, animal and in vitro studies are not within the scope of this review. This narrative review discusses microbiome-modifying therapies targeting MDROs in the gut and includes current results, ongoing clinical trials, companies with therapies in the pipeline specifically for MDROs, and commentary on clinical implementation and challenges.

1. Introduction

The gut serves as a reservoir of multidrug resistant (MDR) pathogens, particularly in patients who have had extensive antibiotics or a long hospital stay [1]. While most patients who become colonized do not progress to a symptomatic infection, certain patient populations are particularly vulnerable to multidrug resistant organisms (MDRO) infections, including immunosuppressed patients [2,3], travelers [4], patients in intensive care units [5], and those in long-term care facilities [6]. Additionally, those who are colonized can transmit MDROs to patients vulnerable to infection in the hospital setting. Antibiotics kill subsets of commensal gut bacteria and allow for the unchecked growth of pathogenic species, causing antibiotic use to be a major risk factor for MDRO colonization and infection [7].
Most of the microbiome-modifying therapies, or non-antibiotic therapies intended to change the bacterial makeup of the gut microbiome, are meant to replace good bacteria and out-compete pathogenic bacteria to regrow a healthy microbiome [8,9,10]. This discussion excludes developments in the bacteriophage and lysin field, although these are also capable of modifying the gut microbiome. Fecal microbiota transplant (FMT) and donor-derived therapies are beginning to become more heavily regulated, while probiotic and prebiotic or food supplement-based strategies remain unregulated. Guidelines have been developed to use FMT safely and effectively [11]. While some probiotics have disease-specific recommendations [12], dosing is still difficult. While a handful of clinical trials demonstrate some efficacy for preventing colonization or transmission of MDROs, much more evidence is needed to support the use of FMT, probiotics, or prebiotics in specific patient populations with or without adjunctive therapies. This review is intended to give clinicians and clinical researchers a context for a number of cutting-edge therapies to reduce MDRO carriage and infection.

2. Methods

Clinicaltrials.gov was searched for trials containing the words “fecal”, “probiotic”, and “prebiotic” in the “other terms” field with the “interventional” filter selected. Additionally, the International Conference on Harmonisation Good Clinical Practice website (ichgcp.net) was searched for the same terms. The resulting 2117 studies were manually reviewed for terminology in the title, outcomes, and intervention that included any of the following terms: fecal transplant, fecal microbiota transplant, probiotic, prebiotic, antibiotic resistant, antimicrobial resistant, carbapenem, carbapenem-resistant enterobacteriaceae (CRE), vancomycin, vancomycin-resistant enterococci (VRE), extended spectrum beta-lactamase (ESBL), multidrug resistant organism, MDRO, drug resistant, Staphylococcus aureus, and MRSA. Additionally, referenced articles within those studies that directly addressed MDRO prevention, decolonization, or treatment were added to this review. Finally, companies that are developing non-antibiotic therapeutics that target the gut were reviewed and those that had clinical trials for MDRO treatment or prevention are included in this review.
Duplicates were removed, and 82 articles or clinical trial postings were reviewed for eligibility. Twenty-nine of these were removed for irrelevance or a lack of data regarding MDRO outcomes. Fifty-three studies were identified as measuring MDROs or MDRO colonization as the primary outcome. On pubmed.gov, studies were searched for the corresponding clinical trial numbers or study authors to find the results of trials that fit criteria within clinicaltrials.gov. While this is not an exhaustive list of every study with data on MDRO colonization, the studies selected represent interventional trials that address the use of gut microbiome-modifying therapies to alleviate the surge of MDRO colonization and infections.

3. Fecal Microbiota Transplantation

The concept of transplanting human fecal material is more than 1700 years old, but modern medicine has only recently begun to seriously consider this therapy [13] with rigorous clinical trials beginning in the 2010s [14]. Despite the overwhelming number of trials without a severe adverse event, this therapy is not without risks. Typically, side effects include abdominal discomfort, fever, nausea, bloating, gas, and diarrhea. However, in June 2019, the US Food and Drug Administration (FDA) released a warning regarding FMT after two immunocompromised patients became seriously ill and one died [15] drawing media attention [16]. There is an ongoing controversy over whether the FDA should heavily regulate FMT, which has traditionally been performed using stool from a friend or family member under the guidance of a physician. As regulations become more rigorous, smaller stool banks will not be able to keep operating while for-profit FMT companies can financially keep up with increased testing and storage restrictions. Increased oversight will ultimately benefit this young scientific field by ensuring that patients get the safest, most effective FMT source material through an effective route of administration.
As of December 2019, 197 studies in clinicaltrials.gov were completed or actively enrolling subjects for FMT therapy, mostly for Clostridioides difficile infection (CDI), which was the genesis for treating MDROs with FMT [17]. Twenty-one studies in clinicaltrials.gov are for MDRO decolonization or treatment (Table 1). Companies that are focusing on MDRO-specific indications for their products, or who could use their products for this purpose are listed in Table 2. Several recent reviews have discussed the usefulness of FMT for MDRO decolonization or treatment [18,19,20,21]. The majority are case studies and results are mixed; some studies show complete eradication of MDROs and some show very limited changes in colonization. In a case study of a man who was colonized with 12 MDROs and received FMT for CDI, the result was partial decolonization (four MDROs 15 weeks post-FMT), fewer episodes of sepsis and infections, and less antibiotic use [22]. While this case seemed promising, the only randomized controlled trial (RCT) directly addressing MDRO decolonization found a small, but not significant reduction in ESBL and CRE colonization after antibiotics and FMT [23]. However, the low enrolment number and variability in both colonization and treatment methods (e.g., FMT pill at some sites and nasogastric application at others) could have contributed to the high variability in results. Additionally, there was no consistency in donor stools, with two separate institutions recruiting multiple donors, and the authors note that antimicrobial resistance (AMR) genes could have been acquired following FMT given that the screening process was not exhaustive for all possible contaminants.
Another small retrospective case-matched study of 20 patients in France showed 80% (8/10) of patients cleared CRE or CRE/Acinetobacter colonization 14 days after FMT versus 20% (2/10) in the control group [24]. In other small studies (n = 8 to 20) with various endpoints, the effectiveness of decolonization ranged from 20% to 93% [1,25,26,27,28,29]. One study by Davido et al. suggests that based on an 87% decolonization rate of VRE at three months, FMT should be further explored to combat outbreaks within hospitals [30]. However, because only eight people were in the study, larger studies are needed to determine whether FMT could work in the face of a large outbreak of VRE infection.
Oddly, studies of probiotics have more consistently used placebo controls than FMT studies, as discussed in the probiotics section and shown in Table 1 and Table 3. More thorough RCTs are needed to determine the utility of FMT for specific MDROs using single-donor source material.

4. Probiotics and Prebiotics

Probiotics are not federally regulated unless companies undergo regulatory compliance checks on their own or want to market their product as a drug. Anyone can sell and market probiotics regardless of whether the bacteria are alive or even present. Most probiotic companies do not genotype their bacteria or assess the presence of AMR genes [32], although Cabana et al. noted that reputable companies follow good manufacturing procedures and test for AMR genes, which can be verified by consumers [33]. While the debate over the safety of probiotic use in healthy individuals is ongoing, several clinicians and researchers have voiced concern over probiotic use in immunocompromised populations [34,35,36,37]. Side effects, even in healthy populations, can include infections, immune stimulation, production of harmful byproducts, and the spread of AMR genes [38]. Because of the lack of regulation for these products, bacteria that are not listed on the label can be present in the product and cause unexpected side effects. Regardless of safety, only a few health conditions have been studied for efficacy rigorously enough to garner a recommendation by expert clinicians [12]. Most prominent in these recommendations for gastrointestinal (GI) disorders, immune response, and liver disease are lactobacillus GG (LGG) and VSL#3, which are also the most extensively studied for MDRO indications thus far. It should be noted that the product, VSL#3, developed by Claudio De Simone at VSL Pharmaceuticals is now sold as VISBIOME® and is materially different from the product that VSL still sells as VSL#3 [39]. Because of this covert change in formulation, several research studies of VSL#3 were terminated, and some countries and retailers pulled VSL#3 from circulation.
Currently, 20 studies have tested or are testing probiotics, and one is testing a prebiotic for MDRO decolonization or prevention (Table 3). Ten of these studies have shown no difference in colonization following probiotic therapy in subjects who were healthy volunteers [40], methicillin-resistant S. aureus (MRSA)-colonized [41], VRE-colonized [42,43], ESBL-colonized [31], MDR Escherichia coli-colonized [44], a mix of MDRO colonization [45], on antibiotics or a ventilator [46], preterm infants [47], or travelers [48]. While these results can seem dismal, most studies have used LGG or L. reuteri singularly, so much more remains to be explored.
One striking outlier is a study done by Manley et al., in 2007, in which pasteurized yogurt with and without LGG was administered [49]. Of the 27 patients enrolled, 23 subjects completed the trial and 11/11 (100%) in the treatment arm were negative for VRE colonization eight weeks after initiation of the treatment as compared with 1/12 (9%) in the control arm. Eight of the control patients were then crossed over to receive LGG treatment, and all of them cleared VRE within four weeks. While this study seemed extremely promising, a larger trial by Warrack et al. that started the following year concluded that there was no difference in VRE colonization rates in recently infected patients who used 6 grams of LGG. Colonization status was only available for a portion of the participants (36/50), and an even smaller number were colonized at the beginning of the study (9/36) [42]. At four weeks, two additional patients in the control arm had become colonized and one in the treatment group had been cleared of VRE. Despite these findings, the same group found some efficacy of a particular LGG strain called HN001. Supplementation with L. rhamnosus HN001 reduced MRSA colonization, but these results were mainly restricted to gut decolonization, not nasal or other sites [41,50]. In a study of 61 children colonized with VRE, one group found that LGG temporarily decolonized patients, but ultimately some would become recolonized after treatment stopped [51].
Ongoing trials include a large study at Taipei Medical University that is examining the efficacy of a probiotic cocktail on VRE colonization (NCT03822819), and unlike the Warrick study [42], the researchers directly recruit patients who are colonized with VRE. Another trial examining the efficacy of Bifidobacterium infantis in women with recurrent MDRO urinary tract infections is ongoing at Ochsner Medical Center (NCT03644966), but neither of the authors are involved in that work.
In a departure from probiotics, Kaleido Biosciences is assessing KB109 for its ability to clear VRE, ESBL, or CRE colonization (NCT03944369). Their technology consists of a library of nondigestible glycans branded as Microbiome Metabolic Therapies (MMT™) that can be assessed in thousands of patient samples ex vivo for a compounds ability to encourage the growth of certain bacteria that outcompete unhealthy bacteria or inflammatory bacteria (reviewed in [52]). Some of the assays that support the use of these compounds for eliminating MDRO carriage were recently presented [53]. Inulin, an oligosaccharide that is undigestible by humans but fermented by bacteria in the gut, is currently being used in an ongoing clinical trial at Columbia University (NCT03865706). Typically, prebiotics cause moderate gastrointestinal issues like bloating and gas, but are unlikely to increased AMR genes because they are not made with live bacteria. Thus, prebiotics could be a valuable therapeutic tool in the fight against MDRO colonization and infection, but their efficacy remains to be determined.

5. Clinical Considerations and the Future of Microbiome-Modifying Bacterial Therapies.

Clinical use of FMT has been somewhat regulated; the FDA has exercised enforcement discretion regarding the investigation of new drug requirements for CDI in the face of overwhelming evidence that FMT is highly effective in otherwise treatment-resistant or recurrent CDI. Guidelines published regarding the implementation of FMT for CDI [11] could translate to use for other diseases. However, there is a lack of diagnostic capacity or even a clear understanding of what constitutes a healthy microbiome. It is difficult to determine whether FMT directly causes reliable changes in the microbiome or if specific, rationally designed therapies would work more effectively in such a complex system. Additionally, many studies use related donors, a multitude of donors, or inconsistent screening processes, making it difficult to determine if fecal transplant from any healthy donor is effective or if more specific elements of FMT are necessary for successful engraftment. Recently, super-donors have drawn attention in the field [54], and in the case of irritable bowel syndrome, a super-donor could make the difference as to whether FMT works or not [55]. In this same vein, perhaps therapies could replicate the bacterial consortiums found in super-donor individuals and be used in a standardized manner to compare the therapy to decolonize different organisms. In the clinical world, a diagnostic to help determine when patients are likely to succeed or fail with various FMT products or when they would need additional doses could significantly increase the use and consistency of FMT therapy. In the basic science world, a commercially available assay, such as the platform that Kaleido uses to test the output of various combinations of bacteria, could accelerate new strategies for FMT, prebiotic, and probiotic approaches, and even precision medicine.
Overall, the challenges with clinical implementation of probiotic therapies for MDROs or other diseases stem from a lack of regulation and insufficient scientific studies to determine appropriate dosing or extensively compare strains and strain combinations for MDRO decolonization. Some new studies in animals show that whether the probiotics are living or dead may not even matter [56], and that there may be more efficacy in simply using bacterial byproducts for therapy. Piewngam et al. found that people colonized with Bacillus from their diet had zero incidence of colonization with MRSA, which was markedly different from that of the 25% of people without Bacillus who were colonized in the GI tract and nasal cavity. Through in vitro testing, they discovered that fengycins, a class of cyclic lipopeptides, are produced by Bacillus and directly inhibit the growth of MRSA [57]. While this type of therapy has yet to be developed for a clinical trial, it signifies the value of more rigorous preclinical assessment of probiotics to pinpoint the most efficacious mechanisms of action.
Microbiome-modifying therapies would benefit from robust RCTs that are laser focused on a specific pathogen or patient population and a rational dosing regimen. An equivalent assessment to pharmacokinetic and pharmacodynamic studies, such as daily sequencing of stool samples to assess engraftment, would assist in determining dose and frequency effects, and commercial companies (e.g., CosmosID, CoreBiome, and Viome) have optimized shotgun sequencing of the whole microbiome to be extremely cost effective. Furthermore, if any microbiome therapy is going to be used for MDRO treatment or prevention, clear guidelines for use in gram-positive versus gram-negative infections and various mechanisms of resistance are necessary, as FMT has not been particularly efficacious with MDR gram-negative bacteria so far [44,45]. Some have speculated that FMT could be less effective for ESBL decolonization, although the mechanism by which this is possible has yet to be defined [25,58]. Finally, research needs to show more than a lack of inferiority; rather, studies should be designed to measure outcomes where FMT or probiotic therapies could show an advantage beyond acute treatment (e.g., reduced readmission, fewer subsequent infections, and fewer long-term complications). A 2019 study from Finland has suggested that extensive treatment with antibiotics can induce Parkinson disease 10 to 15 years later [59], and although long-term studies are costly, perhaps the long-term implications are worth considering for lengthy therapy options.

6. Conclusions

Given a handful of positive results of bacterial therapy for MDRO colonization, this strategy is certainly worth pursuing in a systematic rigorous way. Studies that include single-source FMT or standard, regulated probiotic therapies for specific MDROs enable physicians and hospital systems to make rational decisions about how to best handle colonized or infected patients and healthcare workers. However, these treatments alone will not solve the issue of antibiotic resistance. Other therapies including bacteriophage [60,61], lytic enzymes [62], novel cleaning techniques [63], repurposed drugs with antibiotic activity [64], and bacterial byproducts [57] should be further developed. With enough duplicity in treatment options, hopefully physicians will be able to prevent global outbreaks of treatment-resistant infections.

Author Contributions

A.F. and J.G.-D. contributed to the writing and research of this article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors thank Kathleen McFadden for her expedient and thorough editing.

Conflicts of Interest

The authors are conducting clinical trials for Rebiotix, Seres, Kaleido, and Vendanta, but are not involved in any of the studies reviewed here. J.G.-D. is on the advisory board for Rebiotix. The authors use the services of CosmosID.

References

  1. Bilinski, J.; Grzesiowski, P.; Sorensen, N.; Madry, K.; Muszynski, J.; Robak, K.; Wroblewska, M.; Dzieciatkowski, T.; Dulny, G.; Dwilewicz-Trojaczek, J.; et al. Fecal microbiota transplantation in patients with blood disorders inhibits gut colonization with antibiotic-resistant bacteria: Results of a prospective, single-center study. Clin. Infect. Dis. 2017, 65, 364–370. [Google Scholar] [CrossRef] [PubMed]
  2. Medboua-Benbalagh, C.; Touati, A.; Kermas, R.; Gharout-Sait, A.; Brasme, L.; Mezhoud, H.; Touati, D.; Guillard, T.; de Champs, C. Fecal carriage of extended-spectrum beta-lactamase-producing enterobacteriaceae strains is associated with worse outcome in patients hospitalized in the pediatric oncology unit of Beni-Messous Hospital in Algiers, Algeria. Microb. Drug Resist. 2017, 23, 757–763. [Google Scholar] [CrossRef] [PubMed]
  3. Thacker, N.; Pereira, N.; Banavali, S.D.; Narula, G.; Vora, T.; Chinnaswamy, G.; Prasad, M.; Kelkar, R.; Biswas, S.; Arora, B. Alarming prevalence of community-acquired multidrug-resistant organisms colonization in children with cancer and implications for therapy: A prospective study. Indian J. Cancer 2014, 51, 442–446. [Google Scholar] [CrossRef] [PubMed]
  4. Arcilla, M.S.; van Hattem, J.M.; Haverkate, M.R.; Bootsma, M.C.J.; van Genderen, P.J.J.; Goorhuis, A.; Grobusch, M.P.; Lashof, A.M.O.; Molhoek, N.; Schultsz, C.; et al. Import and spread of extended-spectrum beta-lactamase-producing Enterobacteriaceae by international travellers (COMBAT study): A prospective, multicentre cohort study. Lancet Infect. Dis. 2017, 17, 78–85. [Google Scholar] [CrossRef]
  5. McConville, T.H.; Sullivan, S.B.; Gomez-Simmonds, A.; Whittier, S.; Uhlemann, A.C. Carbapenem-resistant Enterobacteriaceae colonization (CRE) and subsequent risk of infection and 90-day mortality in critically ill patients, an observational study. PLoS ONE 2017, 12, e0186195. [Google Scholar] [CrossRef]
  6. Lim, C.J.; Cheng, A.C.; Kennon, J.; Spelman, D.; Hale, D.; Melican, G.; Sidjabat, H.E.; Paterson, D.L.; Kong, D.C.; Peleg, A.Y. Prevalence of multidrug-resistant organisms and risk factors for carriage in long-term care facilities: A nested case-control study. J. Antimicrob. Chemother. 2014, 69, 1972–1980. [Google Scholar] [CrossRef]
  7. Wang, J.; Foxman, B.; Mody, L.; Snitkin, E.S. Network of microbial and antibiotic interactions drive colonization and infection with multidrug-resistant organisms. Proc. Natl. Acad. Sci. USA 2017, 114, 10467–10472. [Google Scholar] [CrossRef] [Green Version]
  8. Bakken, J.S.; Borody, T.; Brandt, L.J.; Brill, J.V.; Demarco, D.C.; Franzos, M.A.; Kelly, C.; Khoruts, A.; Louie, T.; Martinelli, L.P.; et al. Treating Clostridium difficile infection with fecal microbiota transplantation. Clin. Gastroenterol. Hepatol. 2011, 9, 1044–1049. [Google Scholar] [CrossRef] [Green Version]
  9. Borody, T.J.; Campbell, J. Fecal microbiota transplantation: Techniques, applications, and issues. Gastroenterol. Clin. North. Am. 2012, 41, 781–803. [Google Scholar] [CrossRef]
  10. Khoruts, A.; Sadowsky, M.J. Understanding the mechanisms of faecal microbiota transplantation. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 508–516. [Google Scholar] [CrossRef] [Green Version]
  11. Mullish, B.H.; Quraishi, M.N.; Segal, J.P.; McCune, V.L.; Baxter, M.; Marsden, G.L.; Moore, D.J.; Colville, A.; Bhala, N.; Iqbal, T.H.; et al. The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: Joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut 2018, 67, 1920–1941. [Google Scholar] [CrossRef] [Green Version]
  12. Floch, M.H.; Walker, W.A.; Sanders, M.E.; Nieuwdorp, M.; Kim, A.S.; Brenner, D.A.; Qamar, A.A.; Miloh, T.A.; Guarino, A.; Guslandi, M.; et al. Recommendations for Probiotic Use--2015 Update: Proceedings and Consensus Opinion. J. Clin. Gastroenterol. 2015, 49 (Suppl. 1), S69–S73. [Google Scholar] [CrossRef]
  13. Borody, T.J.; Warren, E.F.; Leis, S.M.; Surace, R.; Ashman, O.; Siarakas, S. Bacteriotherapy using fecal flora: Toying with human motions. J. Clin. Gastroenterol. 2004, 38, 475–483. [Google Scholar] [CrossRef] [PubMed]
  14. van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.; Tijssen, J.G.; et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. McSeveney, M. FDA Warns about Potential Risks Related to Investigational Use of FMT; U.S. Food and Drug Administration: Washington, DC, USA, 2019.
  16. Jacobs, A. How Contaminated Stool Stored in a Freezer Left a Fecal Transplant Patient Dead. The New York Times, 2019. [Google Scholar]
  17. Millan, B.; Park, H.; Hotte, N.; Mathieu, O.; Burguiere, P.; Tompkins, T.A.; Kao, D.; Madsen, K.L. Fecal microbial transplants reduce antibiotic-resistant genes in patients with recurrent Clostridium difficile infection. Clin. Infect. Dis. 2016, 62, 1479–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Gargiullo, L.; Del Chierico, F.; D’Argenio, P.; Putignani, L. Gut microbiota modulation for multidrug-resistant organism decolonization: Present and future perspectives. Front. Microbiol. 2019, 10, 1704. [Google Scholar] [CrossRef]
  19. Gopalsamy, S.N.; Woodworth, M.H.; Wang, T.; Carpentieri, C.T.; Mehta, N.; Friedman-Moraco, R.J.; Mehta, A.K.; Larsen, C.P.; Kraft, C.S. The use of microbiome restoration therapeutics to eliminate intestinal colonization with multidrug-resistant organisms. Am. J. Med. Sci. 2018, 356, 433–440. [Google Scholar] [CrossRef]
  20. Saha, S.; Tariq, R.; Tosh, P.K.; Pardi, D.S.; Khanna, S. Faecal microbiota transplantation for eradicating carriage of multidrug-resistant organisms: A systematic review. Clin. Microbiol. Infect. 2019, 25, 958–963. [Google Scholar] [CrossRef]
  21. Woodworth, M.H.; Hayden, M.K.; Young, V.B.; Kwon, J.H. The role of fecal microbiota transplantation in reducing intestinal colonization with antibiotic-resistant organisms: The current landscape and future directions. Open Forum Infect. Dis. 2019, 6. [Google Scholar] [CrossRef] [Green Version]
  22. Crum-Cianflone, N.F.; Sullivan, E.; Ballon-Landa, G. Fecal microbiota transplantation and successful resolution of multidrug-resistant-organism colonization. J. Clin. Microbiol. 2015, 53, 1986–1989. [Google Scholar] [CrossRef] [Green Version]
  23. Huttner, B.D.; de Lastours, V.; Wassenberg, M.; Maharshak, N.; Mauris, A.; Galperine, T.; Zanichelli, V.; Kapel, N.; Bellanger, A.; Olearo, F.; et al. A 5-day course of oral antibiotics followed by faecal transplantation to eradicate carriage of multidrug-resistant Enterobacteriaceae: A randomized clinical trial. Clin. Microbiol. Infect. 2019, 25, 830–838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Saidani, N.; Lagier, J.C.; Cassir, N.; Million, M.; Baron, S.; Dubourg, G.; Eldin, C.; Kerbaj, J.; Valles, C.; Raoult, D.; et al. Faecal microbiota transplantation shortens the colonisation period and allows re-entry of patients carrying carbapenamase-producing bacteria into medical care facilities. Int. J. Antimicrob. Agents 2019, 53, 355–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Battipaglia, G.; Malard, F.; Rubio, M.T.; Ruggeri, A.; Mamez, A.C.; Brissot, E.; Giannotti, F.; Dulery, R.; Joly, A.C.; Baylatry, M.T.; et al. Fecal microbiota transplantation before or after allogeneic hematopoietic transplantation in patients with hematologic malignancies carrying multidrug-resistance bacteria. Haematologica 2019, 104, 1682–1688. [Google Scholar] [CrossRef] [Green Version]
  26. Davido, B.; Batista, R.; Fessi, H.; Salomon, J.; Dinh, A. Impact of faecal microbiota transplantation to eradicate vancomycin-resistant enterococci (VRE) colonization in humans. J. Infect. 2017, 75, 376–377. [Google Scholar] [CrossRef] [PubMed]
  27. Davido, B.; Batista, R.; Michelon, H.; Lepainteur, M.; Bouchand, F.; Lepeule, R.; Salomon, J.; Vittecoq, D.; Duran, C.; Escaut, L.; et al. Is faecal microbiota transplantation an option to eradicate highly drug-resistant enteric bacteria carriage? J. Hosp. Infect. 2017, 95, 433–437. [Google Scholar] [CrossRef]
  28. Dinh, A.; Fessi, H.; Duran, C.; Batista, R.; Michelon, H.; Bouchand, F.; Lepeule, R.; Vittecoq, D.; Escaut, L.; Sobhani, I.; et al. Clearance of carbapenem-resistant Enterobacteriaceae vs vancomycin-resistant enterococci carriage after faecal microbiota transplant: A prospective comparative study. J. Hosp. Infect. 2018, 99, 481–486. [Google Scholar] [CrossRef] [PubMed]
  29. Singh, R.; de Groot, P.F.; Geerlings, S.E.; Hodiamont, C.J.; Belzer, C.; Berge, I.; de Vos, W.M.; Bemelman, F.J.; Nieuwdorp, M. Fecal microbiota transplantation against intestinal colonization by extended spectrum beta-lactamase producing Enterobacteriaceae: A proof of principle study. BMC Res. Notes 2018, 11, 190. [Google Scholar] [CrossRef] [Green Version]
  30. Davido, B.; Batista, R.; Fessi, H.; Michelon, H.; Escaut, L.; Lawrence, C.; Denis, M.; Perronne, C.; Salomon, J.; Dinh, A. Fecal microbiota transplantation to eradicate vancomycin-resistant enterococci colonization in case of an outbreak. Med. Mal. Infect. 2019, 49, 214–218. [Google Scholar] [CrossRef]
  31. Ljungquist, O.; Kampmann, C.; Resman, F.; Riesbeck, K.; Tham, J. Probiotics for intestinal decolonization of ESBL-producing Enterobacteriaceae: A randomized, placebo-controlled clinical trial. Clin. Microbiol. Infect. 2019. [Google Scholar] [CrossRef]
  32. Cohen, P.A. Probiotic safety-no guarantees. JAMA Intern. Med. 2018, 178, 1577–1578. [Google Scholar] [CrossRef]
  33. Cabana, M.; Salminen, S.; Sanders, M. Probiotic safety-reasonable certainty of no harm. JAMA Intern. Med. 2019, 179, 276. [Google Scholar] [CrossRef] [PubMed]
  34. Happel, A.U.; Barnabas, S.L.; Froissart, R.; Passmore, J.S. Weighing in on the risks and benefits of probiotic use in HIV-infected and immunocompromised populations. Benef. Microbes 2018, 9, 239–246. [Google Scholar] [CrossRef] [PubMed]
  35. Luong, M.L.; Sareyyupoglu, B.; Nguyen, M.H.; Silveira, F.P.; Shields, R.K.; Potoski, B.A.; Pasculle, W.A.; Clancy, C.J.; Toyoda, Y. Lactobacillus probiotic use in cardiothoracic transplant recipients: A link to invasive Lactobacillus infection? Transpl. Infect. Dis. 2010, 12, 561–564. [Google Scholar] [CrossRef] [PubMed]
  36. Roy, U.; Jessani, L.G.; Rudramurthy, S.M.; Gopalakrishnan, R.; Dutta, S.; Chakravarty, C.; Jillwin, J.; Chakrabarti, A. Seven cases of Saccharomyces fungaemia related to use of probiotics. Mycoses 2017, 60, 375–380. [Google Scholar] [CrossRef]
  37. Vanichanan, J.; Chavez, V.; Wanger, A.; De Golovine, A.M.; Vigil, K.J. Carbapenem-resistant Lactobacillus intra-abdominal infection in a renal transplant recipient with a history of probiotic consumption. Infection 2016, 44, 793–796. [Google Scholar] [CrossRef]
  38. Marteau, P.; Seksik, P. Tolerance of probiotics and prebiotics. J. Clin. Gastroenterol. 2004, 38, S67–S69. [Google Scholar] [CrossRef]
  39. Maryland Federal Jury Unanimously Finds Probiotic Sellers Liable for False Advertising and Awards Product Inventor More than $18 Million in Damages. In Business Wire; 2018; Available online: https://www.businesswire.com/news/home/20181126005445/en/Maryland-Federal-Jury-Unanimously-Finds-Probiotic-Sellers (accessed on 26 November 2018).
  40. Lund, B.; Edlund, C.; Barkholt, L.; Nord, C.E.; Tvede, M.; Poulsen, R.L. Impact on human intestinal microflora of an Enterococcus faecium probiotic and vancomycin. Scand. J. Infect. Dis. 2000, 32, 627–632. [Google Scholar] [CrossRef]
  41. Warrack, S.; Panjikar, P.; Duster, M.; Safdar, N. Tolerability of a probiotic in subjects with a history of methicillin-resistant Staphylococcus aureus colonisation. Benef. Microbes 2014, 5, 389–395. [Google Scholar] [CrossRef]
  42. Warrack, S.Z.M.; Duster, M.; Panjikar, P.; Safdar, N. A pilot randomized trial to determine the tolerability of a probiotic in patients colonized with vancomycin-resistant enterococcus. J. Prob. Health 2016, 4. [Google Scholar] [CrossRef] [Green Version]
  43. Doron, S.; Hibberd, P.L.; Goldin, B.; Thorpe, C.; McDermott, L.; Snydman, D.R. Effect of Lactobacillus rhamnosus GG administration on vancomycin-resistant enterococcus colonization in adults with comorbidities. Antimicrob. Agents Chemother. 2015, 59, 4593–4599. [Google Scholar] [CrossRef] [Green Version]
  44. Tannock, G.W.; Tiong, I.S.; Priest, P.; Munro, K.; Taylor, C.; Richardson, A.; Schultz, M. Testing probiotic strain Escherichia coli Nissle 1917 (Mutaflor) for its ability to reduce carriage of multidrug-resistant E. coli by elderly residents in long-term care facilities. J. Med. Microbiol. 2011, 60, 366–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Salomao, M.C.; Heluany-Filho, M.A.; Menegueti, M.G.; Kraker, M.E.; Martinez, R.; Bellissimo-Rodrigues, F. A randomized clinical trial on the effectiveness of a symbiotic product to decolonize patients harboring multidrug-resistant Gram-negative bacilli. Rev. Soc. Bras. Med. Trop. 2016, 49, 559–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kwon, J.H.; Bommarito, K.M.; Reske, K.A.; Seiler, S.M.; Hink, T.; Babcock, H.M.; Kollef, M.H.; Fraser, V.J.; Burnham, C.A.; Dubberke, E.R.; et al. Randomized controlled trial to determine the impact of probiotic administration on colonization with multidrug-resistant organisms in critically ill patients. Infect. Control. Hosp. Epidemiol. 2015, 36, 1451–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kurt, A.; Ecevit, A.; Ince, D.A.; Abbasoglu, A.; Azap, O.K.; Ecevit, Z.; Ogus, E.; Tarcan, A. 833 The effect of probiotics on colonization of resistant organisms in preterm infants. ArchDis Child. 2012, 97, A240. [Google Scholar] [CrossRef] [Green Version]
  48. Dall, L.B.; Lausch, K.R.; Gedebjerg, A.; Fuursted, K.; Storgaard, M.; Larsen, C.S. Do probiotics prevent colonization with multi-resistant Enterobacteriaceae during travel? A randomized controlled trial. Travel Med. Infect. Dis. 2019, 27, 81–86. [Google Scholar] [CrossRef] [PubMed]
  49. Manley, K.J.; Fraenkel, M.B.; Mayall, B.C.; Power, D.A. Probiotic treatment of vancomycin-resistant enterococci: A randomised controlled trial. Med. J. Aust. 2007, 186, 454–457. [Google Scholar] [CrossRef]
  50. Eggers, S.; Barker, A.K.; Valentine, S.; Hess, T.; Duster, M.; Safdar, N. Effect of Lactobacillus rhamnosus HN001 on carriage of Staphylococcus aureus: Results of the impact of probiotics for reducing infections in veterans (IMPROVE) study. BMC Infect. Dis. 2018, 18, 129. [Google Scholar] [CrossRef] [Green Version]
  51. Szachta, P.; Ignys, I.; Cichy, W. An evaluation of the ability of the probiotic strain Lactobacillus rhamnosus GG to eliminate the gastrointestinal carrier state of vancomycin-resistant enterococci in colonized children. J. Clin. Gastroenterol. 2011, 45, 872–877. [Google Scholar] [CrossRef]
  52. Koropatkin, N.M.; Cameron, E.A.; Martens, E.C. How glycan metabolism shapes the human gut microbiota. Nat. Rev. Microbiol. 2012, 10, 323–335. [Google Scholar] [CrossRef] [Green Version]
  53. LeBlanc, G.B.B.; Hartman, M.; Hecht, M.; Luong, H.; Pan, Z.; Sofen, S.; Lawrence, J.; Miller, K.; Meehan, B.; Mahowald, M.; et al. Novel glycans reduce carbapenem-resistant enterobacteriaceae (CRE) and vancomycin-resistant enterococcus (VRE) colonization in an ex vivo assay by supporting growth of commensal microbiota at the expense of multidrug-resistant (MDR) organisms. In Proceedings of the IDWeek, Washington, DC, USA, October 2019. [Google Scholar]
  54. Wilson, B.C.; Vatanen, T.; Cutfield, W.S.; O’Sullivan, J.M. The super-donor phenomenon in fecal microbiota transplantation. Front. Cell Infect. Microbiol. 2019, 9, 2. [Google Scholar] [CrossRef] [Green Version]
  55. El-Salhy, M.H.J.; Gilja, O.H.; Kristoffersen, A.; Hausken, T. Effects of faecal microbiota transplantation in patients with irritable bowel syndrome (IBS): A randomised, double-blind placebo-controlled study. In Proceedings of the UEG Week, Barcelona, Spain, October 2019. [Google Scholar]
  56. Wang, S.; Ahmadi, S.; Nagpal, R.; Jain, S.; Mishra, S.P.; Kavanagh, K.; Zhu, X.; Wang, Z.; McClain, D.A.; Kritchevsky, S.B.; et al. Lipoteichoic acid from the cell wall of a heat killed Lactobacillus paracasei D3-5 ameliorates aging-related leaky gut, inflammation and improves physical and cognitive functions: From C. elegans to mice. Geroscience 2019. [Google Scholar] [CrossRef] [PubMed]
  57. Piewngam, P.; Zheng, Y.; Nguyen, T.H.; Dickey, S.W.; Joo, H.S.; Villaruz, A.E.; Glose, K.A.; Fisher, E.L.; Hunt, R.L.; Li, B.; et al. Pathogen elimination by probiotic Bacillus via signalling interference. Nature 2018, 562, 532–537. [Google Scholar] [CrossRef] [PubMed]
  58. Galloway-Pena, J.R.; Jenq, R.R. The only thing that stops a bad microbiome, is a good microbiome. Haematologica 2019, 104, 1511–1513. [Google Scholar] [CrossRef] [PubMed]
  59. Mertsalmi, T.H.; Pekkonen, E.; Scheperjans, F. Antibiotic exposure and risk of Parkinson’s disease in Finland: A nationwide case-control study. Mov Disord 2019. [Google Scholar] [CrossRef] [PubMed]
  60. El Haddad, L.; Harb, C.P.; Gebara, M.A.; Stibich, M.A.; Chemaly, R.F. A systematic and critical review of bacteriophage therapy against multidrug-resistant ESKAPE organisms in humans. Clin. Infect. Dis. 2019, 69, 167–178. [Google Scholar] [CrossRef]
  61. Kakasis, A.; Panitsa, G. Bacteriophage therapy as an alternative treatment for human infections. A comprehensive review. Int. J. Antimicrob Agents 2019, 53, 16–21. [Google Scholar] [CrossRef]
  62. Becker, S.C.; Roach, D.R.; Chauhan, V.S.; Shen, Y.; Foster-Frey, J.; Powell, A.M.; Bauchan, G.; Lease, R.A.; Mohammadi, H.; Harty, W.J.; et al. Triple-acting lytic enzyme treatment of drug-resistant and intracellular staphylococcus aureus. Sci. Rep. 2016, 6, 25063. [Google Scholar] [CrossRef]
  63. Villacis, J.E.; Lopez, M.; Passey, D.; Santillan, M.H.; Verdezoto, G.; Trujillo, F.; Paredes, G.; Alarcon, C.; Horvath, R.; Stibich, M. Efficacy of pulsed-xenon ultraviolet light for disinfection of high-touch surfaces in an Ecuadorian hospital. BMC Infect. Dis. 2019, 19, 575. [Google Scholar] [CrossRef] [Green Version]
  64. Miro-Canturri, A.; Ayerbe-Algaba, R.; Smani, Y. Drug repurposing for the treatment of bacterial and fungal infections. Front. Microbiol. 2019, 10, 41. [Google Scholar] [CrossRef]
Table 1. Clinical trials for fecal microbiota transplantation (FMT) that directly target multidrug resistant organisms (MDRO) colonization or infection.
Table 1. Clinical trials for fecal microbiota transplantation (FMT) that directly target multidrug resistant organisms (MDRO) colonization or infection.
YearSponsorProductPlacebo/ControlledClinical StageTargetNumber of PatientsNCT NumberEffectConclusionsReference
2015Washington University School of MedicineFMT (enema)noPhase 1MDRO infections20NCT02312986 Ongoing
2015Jinling Hospital, ChinaFMT (nasointestinal tube)noN/AMRSA10NCT02390622 Ongoing
2016University of MiamiFMT (enema)noPhase 1MDRO20NCT02816437 Ongoing
2016Cepheid/ Emory UniversityAllogeneic FMTyesPhase 1MDRO20NCT02922816 Ongoing
2017Raymond Poincaré Teaching HospitalFMT (nasoduodenal tube)noN/ACRE/VRE8EudraCT 2014-003048-11++3/8 patients decolonized after 3 months[27]
2017Medical University of Warsawintraduodenal FMTnoN/AMDRO20 NCT02461199+++15/20 (75%) complete decolonization[1]
2017Raymond Poincaré Teaching HospitalFMT (nasoduodenal tube)noN/AVRE8NCT03029078++++87.5% eradication at 3 months[26]
2017Microbiome Health Research InstituteAutologous FMTyesPhase 1MDRO4NCT03061097 Completed. No results available.
2017Microbiome Health Research InstituteFMT (pill)yesPhase 2VRE9NCT03063437 Completed. No results available.
2017Seattle Children’s HospitalFMTnoPhase 1ESC-R Enterobacteriaceae20NCT02543866 Ongoing
2018Academic Medical Centre, AmsterdamFMTnoN/AESBL15ISRCTN48328635++20-40% clearance with one or two FMTs[29]
2018 Versailles Saint-Quentin UniversityFMT (nasoduodenal tube)noN/ACRE/VRE17 +++4/8 CRE clearance 7/8 VRE clearance at 3 months[28]
2018RebiotixRBX2660noPhase 1/2 Recurrent MDRO UTIs60NCT03367910 Ongoing
2018Chinese University of Hong KongFMT yesPhase 2CRE/VRE 40NCT03479710 Ongoing
2018Rambam Health Care CampusFMTnoN/ACRE60NCT03167398 Ongoing
2019University Hospital, GhentAllogenic vs Autologous FMTyesPhase 2/3Any MDRO150NCT04188743 Unknown
2019University Health Network, TorontoFMTyesPhase 2/3CRE40NCT03802461 Ongoing
2019Rambam Health Care CampusFMTnoN/ACRE60NCT03391674 Ongoing
2019University of British ColumbiaFMTnoN/AAny MDRO90NCT04181112 Ongoing
2019Vancouver Island Health AuthorityFMTnoN/AAny MDRO50NCT03834051 Ongoing
2019Raymond Poincaré Teaching HospitalFMT (nasoduodenal tube)noN/AVRE8 ++++87% decolonization after 3 months[30]
2019Saint Antoine Hospital, ParisFMT (enema or nasogastric tube)noN/ACRE/VRE 10 +++Generally safe and effective in these patients[25]
2019University of British ColumbiaFMT (enema) +/− antibioticnoN/AMDROs90NCT04181112 Ongoing
2019Geneva University HospitalsFMT (pill and nasopharengeal)yesPhase 2ESBL/CPE 39NCT02472600+Slight reduction in colonization[23]
2020Rambam Health Care CampusFMT (pills)yesPhase 2/3CRE60NCT04146337 Not yet recruiting
2022 *University of Wisconsin, MadisonFMT (pills)yesPhase 2CRE/VRE 90NCT03643887 Not started
Fecal microbiota transplantation (FMT), Lactobacillus rhamnosus GG (LGG), vancomycin-resistant enterococci (VRE), methicillin-resistant Staphylococcus aureus (MRSA), multidrug resistant (MDR), Lactobacillus casei rhamnosus (Lcr35), carbapenem resistant Klebsiella Pneumonia (CRKP), extended spectrum beta-lactamases (ESBL), carbapenem-resistant enterobacteriaceae (CRE), extended-spectrum resistant (ESC-R), urinary tract infections (UTIs). <25% improvement (+), 26% to 50% improvement (++), 51% to 75% improvement (+++), 76% to 100% improvement (++++). * Projected start date.
Table 2. Companies developing microbiome-modifying therapies that could be used for MDROs.
Table 2. Companies developing microbiome-modifying therapies that could be used for MDROs.
CompanyProduct Name or PrefixTherapy TypeProposed MechanismTrials Specifically for MDRO
RebiotixRBX2660FMT (enema)Displacement of MDROPhase 1/2
KaleidoKB109prebioticFeed healthy bacteria to out-compete MDROClinical Food Study
ExeGiVisbiome (US), Vivomixx (EU)probioticDisplacement of MDROYes [31]
VedantaVE707rationally selected microbiotaDisplacement of MDROPreclinical
SciBacSCBengineered probioticTransfer of plasmids to enhance good bacteriaPreclinical
Rise TherapeuticsRdelivery technology for protein therapiesImmune modulationPreclinical
FinchCP & FINFMT, rationally selected microbiotaDisplacement of MDRONo
OpenBiomeunbranded pillsFMT (pills)Displacement of MDRONo
SeresSERFMT, rationally selected microbiotaDisplacement of MDRONo
EveloEDPmonoclonal microbialsImmune modulationNo
EnteromeEBsmall moleculeImmune modulationNo
PureTech HealthnumeroushydrogelPhysical clearing of gutNo
AtterxC-1205lyophilized E. coliPrevents growth of MDROOn website, no NCT
AtterxGN-4474bacterial conjugation + killer plasmidTransfer of toxic plasmid to target bacteriaOn website, no NCT
Table 3. Clinical trials for probiotics and prebiotics that directly target MDRO colonization or infection.
Table 3. Clinical trials for probiotics and prebiotics that directly target MDRO colonization or infection.
Treatment TypeYearSponsorProductPlacebo/ControlledClinical StageTargetNumber of PatientsNCT NumberEffectConclusionsReference
Probiotic (live bacteria product)2000Karolinska Institutetprobiotic or vancomycinnoN/AHealthy volunteers40 0No effect on VRE colonization[40]
2003Tufts Medical CenterLGG or CulturelleyesPhase 1VRE11NCT007562620No effect on VRE colonization[43]
2003University Hospital, Clermont-FerrandLcr35unclearN/AMDR Pseudomonas aeruginosa400NCT00621803 Terminated
2007Austin Healthyogurt with LGGyesN/AVRE27 ++++100% cleared[49]
2007Oregon Health and Science Universityyogurt drinkyesN/AVRE8NCT00591474 Terminated
2008University of Wisconsin, MadisonVSL#3yesPhase 2VRE50NCT009335560No effect on VRE but tolerated probiotics well[42]
2008Hadassah Medical OrganizationVSL#3yesPhase 1/2CRKP60NCT00722410 Unknown
2009University Hospital, Clermont-FerrandLcr35yesPhase 4VRE24NCT00437580 Unknown
2009Bio-K Plus International L. acidophilus CL1285®yesPhase 2MRSA146NCT00941356 Unknown
2010University of Wisconsin, MadisonL. rhamnosus HN001yesPhase 2MRSA49NCT01112995+30-50% reduction at 4 weeks[41]
2011University of OtagoE. coli Nissle 1917 (Mutaflor)yesN/AMDR E. coli69 0No effect[44]
2011Baskent UniversityL. reuteriyesN/AAny MDRO76NCT021782670No difference in acquisition of MDROs[47]
2011Poznan University of Medical SciencesLGGyesN/AVRE (children)61 +/-Temporary elimination of VRE[51]
2011University of Wisconsin, MadisonL. rhamnosus HN001yesPhase 2MRSA113NCT01321606+Reduced odds of MRSA colonization of the gut[50]
2012Washington University School of Medicine/ CDCCulturelle (LGG)yesPhase 4Any MDRO103NCT015511860No effect of acquisition or loss of any MDROs[46]
2014Washington University School of Medicine/ CDCCulturelle (LGG)yesPhase 4Any MDRO87NCT020465120No effect of acquisition or loss of any MDROs[46]
2014Hospital Universitario La PazLactitol and LactobacillusnoPhase 2CRKP20NCT02307383 Suspended (Unable to recruit patients)
2016Universidade de São Paulosymbiotic productyesN/AMDR Gram-negative101 0Not effective for decolonizing[45]
2017Lund University/ ExeGiVivomixx (EU) /Visbiome (US)yesN/AESBL80NCT038604150Vivomixx® was not superior to placebo[31]
2018Procter and Gamble/ Ochsner Health SystemB. infantisyesN/AMDR urinary tract infections100NCT03644966 Ongoing
2018Hvidovre University HospitalLactobacillus yesN/AVRE162NCT03560700 Not yet recruiting
2019Aarhus University HospitalLGGyesN/AMDR Enterobacteriaceae61 0No difference in colonization[48]
2019Hospital Italiano de Buenos AiresprobioticyesN/ACRE228NCT03967301 Ongoing
2019Taipei Medical University /Delta Electronicsprobiotic cocktailyesN/AVRE100NCT03822819 Ongoing
2020University of BergenLabinic (R) probiotic yesPhase 3ESBL2000NCT04172012 Ongoing
Prebiotic/food2019Columbia UniversityInulinyesPhase 2Any MDRO90NCT03865706 Ongoing
2019Kaleido BiosciencesKB109yesN/AVRE, ESBL, or CRE64NCT03944369 Ongoing
Fecal Microbiota Transplant (FMT), Lactobacillus rhamnosus GG (LGG), vancomycin-resistant enterococci (VRE), methicillin-resistant S. aureus (MRSA), multidrug resistant (MDR), carbapenem-resistant enterobacteriaceae (CRE), extended spectrum beta-lactamases (ESBL), Lactobacillus casei rhamnosus (Lcr35), and carbapenem resistant Klebsiella Pneumonia (CRKP). No effect (0), mixed results (+/−), <25% improvement (+), 76% to 100% improvement (++++)

Share and Cite

MDPI and ACS Style

Feehan, A.; Garcia-Diaz, J. Bacterial, Gut Microbiome-Modifying Therapies to Defend against Multidrug Resistant Organisms. Microorganisms 2020, 8, 166. https://doi.org/10.3390/microorganisms8020166

AMA Style

Feehan A, Garcia-Diaz J. Bacterial, Gut Microbiome-Modifying Therapies to Defend against Multidrug Resistant Organisms. Microorganisms. 2020; 8(2):166. https://doi.org/10.3390/microorganisms8020166

Chicago/Turabian Style

Feehan, Amy, and Julia Garcia-Diaz. 2020. "Bacterial, Gut Microbiome-Modifying Therapies to Defend against Multidrug Resistant Organisms" Microorganisms 8, no. 2: 166. https://doi.org/10.3390/microorganisms8020166

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop