Next Article in Journal
Involvement of the Antioxidant Effect and Anti-inflammatory Response in Butyrate-Inhibited Vascular Smooth Muscle Cell Proliferation
Next Article in Special Issue
Effects of a Formula Containing Two Types of Prebiotics, Bifidogenic Growth Stimulator and Galacto-oligosaccharide, and Fermented Milk Products on Intestinal Microbiota and Antibody Response to Influenza Vaccine in Elderly Patients: A Randomized Controlled Trial
Previous Article in Journal
Ferric Citrate Hydrate as a Phosphate Binder and Risk of Aluminum Toxicity
Previous Article in Special Issue
The Overarching Influence of the Gut Microbiome on End-Organ Function: The Role of Live Probiotic Cultures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recommendations for Probiotic Use in Humans—A 2014 Update

Yale University School of Medicine, 333 Cedar Street, 1080 LMP, New Haven, CT 06850, USA
Pharmaceuticals 2014, 7(10), 999-1007; https://0-doi-org.brum.beds.ac.uk/10.3390/ph7100999
Submission received: 30 May 2014 / Revised: 23 September 2014 / Accepted: 24 September 2014 / Published: 10 October 2014
(This article belongs to the Special Issue Probiotics and Prebiotics 2015)

Abstract

:
Probiotics have gained worldwide use during the last two decades. However, which probiotic to use in which clinical condition has remained confusing in some clinical conditions. We convened a workshop at Yale in conjunction with Harvard in 2005, inviting a spectrum of probiotic authorities to discuss and reach conclusions on recommendations for use in common clinical conditions; the workshop was reconvened again in 2008 and in 2011. Each time the group of authorities was enlarged and varied depending on research studies. This article lists the recommendations updated from 2011 and is amended to bring it up to date in childhood and adult diarrhea, antibiotic-associated diarrhea, necrotizing enterocolitis, inflammatory bowel disorders, irritable bowel syndrome, allergic disorders, and radiation enteritis pending our 4th Triennial Yale/Harvard workshop to be convened in 2015.

1. Introduction

The burgeoning use of probiotics has proliferated during the past two decades [1]. Although history has shown us that probiotics were part of foods in the past, they have begun to be used therapeutically by clinicians in regular diets [2]. When the popularity of probiotics became apparent, we convened a workshop meeting at Yale University, which included experts in the field of probiotic organisms, to made recommendations for their clinical use. This first workshop meeting occurred in 2006, and we reconvened two years later with some of the same experts and leading authorities in the field, and the last meeting occurred in 2011 [3,4,5]. The amount of literature has exploded, and many more opinions have been expressed [6]. It is our intention to have a 4th Yale workshop meeting to bring the clinical recommendations up to date in 2015.

2. Necrotizing Enterocolitis

Table 1 lists the recommendations made in 2011 by the expert Yale University panel [5]. When we reviewed these, we added the important literature from the past few years. It is clear that necrotizing enterocolitis is one of the most hazardous conditions in any stage of life [7]. Fortunately, when correctly diagnosed, it has been controlled with the use of bovine lactoferrin in combination with probiotic Lactobacillus rhamnosus GG [7,8].
Table 1. Recommendations for Probiotic Use—Update 2011. Data from [5]. Copyright (2011) Lippincott Williams & Wilkins.
Table 1. Recommendations for Probiotic Use—Update 2011. Data from [5]. Copyright (2011) Lippincott Williams & Wilkins.
Clinical Condition EffectivenessSpecific Strain of Organism & Strain ReferencesAnalysis References
Diarrhea
Infectious childhood—treatment AS. boulardii [9], LGG [10,11], L. reuteri SD2112 [12][9,10,11,12]
Prevention of AADAS. boulardii [14], LGG [15], combination. of L. casei DN114 G01, L. bulgaricus, snf S. thermophiles [16][14,16]
Prevention of recurrent CDAD B/CS. boulardii [17], LGG [13], Bacteriotherapy [18][17]
Prevention of CDAD B/CLGG [17], S. boulardii [13][13,17]
IBD
Pouchitis
Preventing and maintaining remissionAVSL#3 [19,20,21][19,20,21]
Induce remissionCVSL#3 [22][22]
Ulcerative colitis
Inducing remissionBE. coli Nissle [23], VSL#3 [24][23,24]
MaintenanceAE. coli Nissle, VSL#3 [25,26][25,26]
Crohn’s CE. coli Nissle [27], S. boulardii [28], LGG [29][26,28,29]
IBS
BB. infantis B5624[30,31], VSL#3 [32,33,34][30,31,32,33,34] **
CBifidobacterium animalis [35][35]
L. plantarum 299V [36][36]
Necrotizing Enterocolitis
BLactobacillus acidophilus NCDO1748 [7,37] and Bifidobacterium bifidium NCDO1453 [8][37]
Recommendations from 2008 *
Immune Response
ALGG, L. acidophilus LAFT1, L. plantarum, Bifidobacterium lactis, Lactobacillus johnsonii[27,38]
Allergy
Atopic eczema associated with cow’s milk allergy
Treatment ALGG, B. lactis [39][39]
Prevention ALGG, B. lactis [39][39]
Radiation Enteritis
CVSL#3 [38], L. acidophilus [27][27,38]
Vaginosis and Vaginitis
CL. acidophilus [40], Lactobacillus rhamnosus GR-1 [41], L. reuteri RC14 [42][40,41,42]
AAD indicates antibiotic-associated diarrhea; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome; CDAD, Clostridium difficile-associated diarrhea; LGG, Lactobacillus GG. * Check 2008 references for further elaboration on strains used and their availability. ** Reference [39] was made available after the workshop meeting on April 8, 2011 but believed to be significant enough to qualify this probiotic to be in a B category. “A” recommendation is based on strong, positive, well-conducted, controlled studies in the primary literature, not abstract form. “B” recommendation is based on positive, controlled studies but the presence of some negative studies. “C” recommendation is based on some positive studies but clearly an inadequate amount of work to establish the certainty of “A” or “B”.

3. Childhood Diarrhea

The microbiota is maintained in a stable ecology, and it appears that probiotics are very helpful in shortening the course of acute gastroenteritis diarrhea [9,10,11,12]. There are numerous studies to establish this. It is clear that starting Saccharomyces boulardii, LGG, or strains of Lactobacillus reuteri are extremely helpful in shortening the course of the diarrhea [9,10,11,12].

4. Adult Diarrhea

All of the more recent studies, particularly the one done by Hickson and associates in England, found that Lactobacillus casei, L. bulgaricus, and S. thermophilus were capable in reducing the incidence of diarrhea as well as having some effect on Clostridium difficile. This study also showed the potential to decrease morbidity, healthcare costs, and mortality in patients over the age of 50 [16].

5. Antibiotic-associated Diarrhea

Review of the modern literature in our Yale workshops [3,4,5] and by the most recent Goldenberg Cochrane Database System Review [43] clearly indicate that probiotics are helpful in the prevention of C. difficile-associated diarrhea in both adults and children. This literature indicates S. boulardii, LGG, in combinations [5] are helpful in accomplishing this outcome in antibiotic-associated diarrhea. A review of the literature by experts indicates that antibiotic-associated diarrhea can be prevented by the use of numerous organisms [43].
This entire issue has been clouded by the development of fecal microbial transplantation (FMT). It is clear now that FMT can cure and prevent resistant C. difficile infection [44]; the literature on this is extensive [45,46,47,48]. Furthermore, there are now centers to transplant volunteer specimens [23]. They are even suggestions this become a routine or government approved product that can be used for the cure and prevention of resistant C. difficile diarrhea. Surely, more will be forthcoming on this in the coming years.

6. Inflammatory Bowel Disease

A review of the literature reveals that pouchitis can be prevented, and remission can be maintained with the use of the probiotic VSL#3 [19,20,21]. Although ulcerative colitis is universally treated with corticosteroids and other agents, probiotics have some successful therapy reports. Review of the literature reveals that there is a dysbiosis that occurs in simple ulcerative colitis [49,50,51]. This is the same dysbiosis that is treated with FMT. Both previous analysis of the literature [5] has shown that ulcerative colitis could be placed into remission by several probiotics, including Escherichia coli Nissle, S. boulardii and LGG; however, VSL#3 has been the most successful [24,25]. These are limited studies, and most clinicians still prefer the use of more acute, aggressive agents [24,25]. However, there are scattered reports, such as that of Brace and colleagues, who have shown that the dysbiosis, which is seen with C. difficile, can be corrected in selective ulcerative colitis patients and reduce symptom control [50]. Shen and colleagues conducted a careful analysis of 23 randomized controlled trials with a total of 1763 cases and found from their analysis that VSL#3 was the most effective in ulcerative colitis, but there is much less of an effect in Crohn’s disease [51]. Review of the literature confirms that Crohn’s disease does not respond as well as ulcerative colitis [52].

7. Irritable Bowel Syndrome (IBS)

IBS is a major international clinical problem. Review of the modern literature reveals that Bifidobacterium infantis B5624 evaluated in Ireland has given the best reports for relief of symptoms, but most investigators do not give it an “A” rating, but more like a “B” rating [30,31]. The same investigators did find that the probiotic had immunomodulary effects on the microbiota in humans, and it did seem to extend from the mucosal system to the systemic immune system. [31]. Although this probiotic did have these immune effects, it still only had a limited effect on IBS. The IBS literature reveals human effects by Lactobacillus plantarum and Bifidobacterium animalis on IBS [35,36]. Cha and colleagues conducted another study on IBS in which 50 patients with diarrhea IBS were randomized into placebo where probiotics mixtures, including Lactobacillus acidophilus, L. plantarum, Lactobacillus rhamnosus, Bifidobacterium breve, Bifidobacterium lactis, Bifidobacterium longum, and Streptococcus thermophilus were used. The treatment was daily for 8 weeks, and the outcome was adequate relief of overall IBS symptoms [53]. Secondary quality of life symptoms were also studied. There was some stabilization of the intestinal microbiota in that there were some higher concordance rates between bacterial compositions before and after treatment, but we must remember that this was a small group of patients with a limited effect. Effect on probiotics on IBS still remains controversial. At this point in time, we can recommend B. infantis B5624, and a large group of other organisms is mentioned in the references [30,31,35,36].

8. Allergic Disorders

Table 1 lists the organisms that are effective on the immune response in allergic condition. We definitely have a large number of organisms, and they should be used in accordance with the strains recommended in articles [39,54].

9. Radiation Enteritis

Table 1 lists fair results in treating radiation enteritis. Since those reviews, there has been extensive literature discussion and reviews by Hakansson and Molin [54]. All possible probiotic organisms that have been tried are listed extensively, but there is no one set protocol which is recommended to prevent radiation diarrhea. The clinician should carefully review the literature and then decide which should be used in which particular post-radiation treatment [27,38].
Table 1 outlines guidelines for probiotic use. It is updated in this manuscript from the previous report [4]. The recommendations for these guidelines are based on the discussions at the workshop and in the literature.
The field of probiotics and symbiotics has now grown tremendously. The interested clinician should search the literature, and use the probiotic in the particular clinical situation that is needed.

Conflicts of Interests

The author declares no conflict of interest.

References

  1. Floch, M.H.; Kim, A.S. Probiotics A Clinical Guide; SLACK Inc.: Thorofare, NJ, USA, 2010. [Google Scholar]
  2. Floch, M.H.; Kim, A.S. Clinical Insights Probiotics, Prebiotics and Gut Health; Future Medicine Ltd: London, UK, 2014. [Google Scholar]
  3. Floch, M.H.; Madsen, K.K.; Jenkins, D.J.A.; Guandalini, S.; Katz, J.A.; Onderdonk, A.; Walker, W.A.; Fedorak, R.N.; Camilleri, M. Recommendations for probiotic use. J. Clin. Gastroenterol. 2006, 40, 275–278. [Google Scholar]
  4. Floch, M.H.; Walker, W.A.; Guandalini, S.; Hibberd, P.; Gorbach, S.; Surawicz, C.; Sanders, M.E.; Garcia-Tsao, G.; Quigley, E.M.M.; Isolauri, E.; et al. Recommendations for probiotic use—2008. J. Clin. Gastroenterol. 2008, S104–S108. [Google Scholar]
  5. Floch, M.H.; Walker, W.A.; Madsen, K.; Sanders, M.E.; Macfarlane, G.T.; Flint, H.J.; Dieleman, L.A.; Ringel, Y.; Guandalini, S.; Kelly, C.P.; Brandt, L.J. Recommendations for probiotic use—2011 update. J. Clin. Gastroenterol. 2011, S168–S171. [Google Scholar]
  6. Guarner, F.; Khan, A.G.; Garisch, J.; Eliakim, R.; Gangl, A.; Thomson, A.; Krabshuis, J.; Lemair, T.; Kaufmann, P.; de Paula, J.A.; et al. World gastroenterology organisation guideline; probiotics and prebiotics. J. Clin. Gastroenterol. 2012, 48, 468–481. [Google Scholar]
  7. Lin, H.C.; Chyong-Hsin, H.; Hsiu-Lin, C.; Chung, M.Y.; Hsu, J.F.; Lien, R.I.; Tsao, L.Y.; Chen, C.H.; Su, B.H. Oral probiotics prevent necrotizing enterocolitis in very low birth rate preterm infants; a multicenter, randomized, controlled trial. Pediatrics 2008, 122, 693–700. [Google Scholar]
  8. Manzoni, P.; Meyer, M.; Stolfi, I.; Pugni, L.; Romeo, M.G.; Messner, H.; Stolfi, I.; Decembrino, L.; Laforgia, N.; Vagnarelli, F.; et al. Bovine lactoferrin supplementation for prevention of necrotizing enterocolitis in very-low-birth-weight neonates: A randomized clinical trial. Early Hum. Dev. 2014, 90, S60–S65. [Google Scholar]
  9. Szujewsku, H.; Skorka, A.; Dylag, M. Meta-analysis. Saccharomyces boulardii for treating acute diarrhea in children. Aliment. Pharmacol. Ther. 2007, 25, 257–264. [Google Scholar]
  10. Guandalini, S.; Pensabene, I.; Zikri, M.H.; Dias, J.A.; Casali, L.G.; Hoekstra, H.; Kolacek, S.; Massar, K.; Micetic-Turk, D.; Papadopoulou, A.; et al. Lactobacillus GG administered in oral rehydration solution to children with acute diarrhea: A multi-center European Trial. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 54–60. [Google Scholar]
  11. Guandalini, S. Probiotics for children with diarrhea: An update. J. Clin. Gastroenterol. 2008, 42, S53–S57. [Google Scholar]
  12. Shornikova, A.V.; Casus, I.; Isolauri, E.; Mykkänen, H.; Vesikari, T. Lactobacillus reuteri as a therapeutic agent in acute diarrhea in young children. J. Pediatr. Gastroenterol. Nutr. 1997, 27, 399–404. [Google Scholar]
  13. Katz, J.A. Probiotics for the prevention of antibiotic-associated diarrhea and Clostridium difficile diarrhea. J. Clin. Gastroenterol. 2006, 40, 249–255. [Google Scholar]
  14. Surawicz, C.M. Role of probiotics in antibiotic associated diarrhea, Clostridium difficile associated diarrhea and recurrent Clostridium difficile diarrhea. J. Clin. Gastroenterol. 2008, 42, S64–S70. [Google Scholar]
  15. Doron, S.; Hibberd, P.; Gorbach, S.L. Probiotics for prevention of antibiotic-associated diarrhea. J. Clin. Gastroenterol. 2008, 42, S58–S63. [Google Scholar]
  16. Hickson, M.; D’Souza, A.L.; Muthu, N.; Rogers, T.R.; Want, S.; Rajkumar, C.; Bulpitt, C.J. Use of probiotic Lactobacillus preparation to prevent diarrhea associated with antibiotics: randomized double blind placebo controlled trial. BMJ 2007, 355, 80. [Google Scholar]
  17. Nu, X.; Kelly, C. Probiotics in Clostridium difficile infection. J. Clin. Gastroenterol. 2011, 45, S154–S158. [Google Scholar]
  18. Brandt, L.J.; eddy, S.S. Fecal microbiotic transplantation for recurrent Clostridium difficile infection. J. Clin. Gastroenterol. 2011, 45, S159–S167. [Google Scholar]
  19. Gionchetti, P.; Rizzello, F.; Venturi, A.; Brigidi, P.; Matteuzzi, D.; Bazzocchi, G.; Poggioli, G.; Miglioli, M.; Campieri, M.; et al. Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo0controlled trial. Gastroenterology 2000, 119, 305–309. [Google Scholar]
  20. Gionchetti, P.; Rizzello, F.; Helwig, U.; Venturi, A.; Lammers, K.M.; Brigidi, P.; Vitali, B.; Poggioli, G.; Miglioli, M.; Campieri, M.; et al. Prophylactis of pouchitis onset with probiotic therapy: A double-blind, placebo-controlled trial. Gastroenterology 2003, 124, 1202–1209. [Google Scholar]
  21. Mimura, T.; Rizzollo, F.; Helwig, U.; Poggioli, G.; Schreiber, S.; Talbot, I.C.; Nicholls, R.J.; Gionchetti, P.; Campieri, M.; Kamm, M.A. Once-daily high-dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut 2004, 53, 108–114. [Google Scholar]
  22. Gionchetti, P.; Rizzello, F.; Morselli, C.; Poggioli, G.; Tambasco, R.; Calabrese, C.; Brigidi, P.; Vitali, B.; Straforini, G.; Campieri, M. High-dose probiotics for the treatment of active pouchitis. Dis. Colon. Rectum. 2007, 50, 2075–2084. [Google Scholar]
  23. Rembacken, F.J.; Snelling, A.M.; Hawkey, P.M.; Chalmers, D.M.; Axon, A.T.R. Nonpathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: A randomized trial. Lancet 1999, 354, 635–639. [Google Scholar]
  24. Bibiloni, R.; Fedorak, R.N.; Tannock, G.W.; Madsen, K.L.; Gionchetti, P.; Campieri, M.; De Simone, C.; Balfour Sartor, R. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am. J. Gastroenterol. 2005, 100, 1539–1546. [Google Scholar]
  25. Miele, E.; Pascarella, F.; Giannetti, E.; Quaglietta, L.; Baldassano, R.N.; Staiano, A. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am. J. Gastroenterol. 2009, 104, 437–443. [Google Scholar]
  26. Kruis, W.; Fric, P.; Pokrotnieks, J.; Lukáš, M.; Fixa, B.; Kaščák, M.; Kamm, M.A.; Weismueller, J.; Beglinger, C.; Stolte, M.; et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 2004, 53, 1617–1623. [Google Scholar]
  27. Salminen, E.; Eloman, I.; Minkkinen, J.; Vapaatalo, H.; Salminen, S. Preservation of intestinal integrity during radiotherapy using live Lactobacillus acidophilus cultures. Clin. Radiol. 1988, 39, 435–437. [Google Scholar]
  28. Guslandi, M.; Mezzi, G.; Sorghi, M.; Testoni, P.A. Saccharomyces boulardii in maintenance treatment of Crohn’s disease. Dig. Dis. Sci. 2000, 45, 1462–1464. [Google Scholar]
  29. Gupta, P.; Andrew, H.; Kirschner, B.S.; Guandalini, S. Is Lactobacillus GG helpful in children with Crohn’s disease? Results of a preliminary open-label study. J. Pediatr. Gastroenterol. Nutr. 2000, 31, 453–457. [Google Scholar]
  30. Whorwell, P.J.; Altringer, L.; Morel, J.; Bond, Y.; Charbonneau, D.; O'Mahony, L.; Kiely, B.; Shanahan, F.; Quigley, E.M.M. Efficacy of an encapsulated probiotic Bifidobacterium Infantis 35624 in women with irritable bowel syndrome. Am. J. Gastroenterol. 2006, 101, 1581–1590. [Google Scholar]
  31. Groeger, D.; O’Mahony, L.; Murphy, E.F.; Bourke, J.F.; Dinan, T.G.; Kiely, B.; Shanahan, F.; Quigley, E.M.M.; et al. Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes 2013, 4, 325–339. [Google Scholar]
  32. Kim, J.H.; Camilleri, M.; McKenzie, S.; Lempke, M.B.; Burton, D.D.; Thomforde, G.M.; Zinsmeister, A.R. A randomized controlled trial of a probiotic, VSL#3 on gut transit and symptoms in diarrhea-predominant IBS. Aliment. Pharmacol. Ther. 2003, 17, 895–904. [Google Scholar]
  33. Kim, J.H.; Vazquez Roque, M.I.; Camilleri, M.; Stephens, D.; Burton, D.D.; Baxter, K.; Thomforde, G.; Zinsmeister, A.R. A randomized controlled trial of probiotic combination VSL#3 and placebo in IBS with bloating. Neurogastroenterol. Motil. 2005, 17, 687–696. [Google Scholar]
  34. Guandalini, S.; Magazzu, G.; Chiaro, A.; La Balestra, V.; Di Nardo, G.; Gopalan, S.; Sibal, A.; Romano, C.; Canani, R.B.; Lionetti, P.; et al. VSL#3 improves symptoms in children with irritable bowel syndrome: A multicenter, randomized, placebo-controlled, double-blind, crossover study. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 24–30. [Google Scholar]
  35. Guyonnet, D.; Chassany, O.; Ducrotte, P.; Picard, C.; Mouret, M.; Mercier, C.H.; Matuchansky, C. Effect of a fermented milk containing Bifidobacterium animalis DN-173 010 on the health-related quality of life and symptoms in irritable bowel syndrome in adults in primary care: A multicentre, randomized, double-blind, controlled trail. Aliment. Pharmacol. Ther. 2007, 26, 475–486. [Google Scholar]
  36. Niedzielin, K.; Kordecki, H.; Birkenfeld, B. A controlled, double-blind, randomized study on the efficacy of Lactobacillus plantarum 299 V in patients with irritable bowel syndrome. Eur. J. Gastroenterol. Hepatol. 2001, 13, 1143–1147. [Google Scholar]
  37. Ganguli, K.; Walker, W.A. Probiotics in the prevention of necrotizing enterocolitis. J. Clin. Gastroenterol. 2011, 45, S133–S138. [Google Scholar]
  38. Delia, P.; Sansotta, G.; Donato, V.; Messina, G.; Frosina, P.; Pergolizzi, S.; De Renzis, C; Famularo, G. Prevention of radiation induced diarrhea with the use of VLS#3, a new high potency probiotic preparation. Am. J. Gastroenterol. 2002, 97, 2150–2152. [Google Scholar]
  39. Isolauri, E.; Salminen, S. Probiotics: Use in allergic disorders. J. Clin. Gastroenterol. 2008, 42, S91–S96. [Google Scholar]
  40. Hilton, F.; Isenberg, H.D.; Alperstein, P.; France, K.; Borenstein, M.T. Ingestion of yogurt containing Lactobacillus acidophilus as prophylaxis for candida vaginitis. Am. Intern. Med. 1992, 116, 353–357. [Google Scholar]
  41. Anukam, K.; Osazuwa, E.; Ahonkhai, I.; Ngwu, M.; Osemene, G.; Bruce, A.W.; Reid, G. Augmentation of antimicrobial metronidazole therapy of bacterial vaginosis with oral probiotic Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14: Randomized, double-blind, placebo controlled trial. Microbes Infect. 2006, 8, 1450–1454. [Google Scholar]
  42. Anukam, K.C.; Osazuwa, E.; Osemene, G.; Ehigiagbe, F.; Bruce, A.W.; Reid, G. Clinical study comparing probiotic Lactobacillus GR-1 and RC-14: with metronidazole vaginal gel to treat symptomatic bacterial vaginosis. Microbes Infect. 2006, 8, 2772–2776. [Google Scholar]
  43. Goldenberg, J.Z.; Ma, S.S.; Saxton, J.D.; Martzen, M.R.; Vandvik, P.O.; Thorlund, K.; Guyatt, G.H.; Johnston, B.C. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst. Rev. 2013, 5, CD006095. [Google Scholar]
  44. Garborg, K.; Waagsbo, B.; Stallemo, A.; Matre, J.; Sundoy, A. Results of fecal donor instillation therapy for recurrent Clostridium difficile-associated diarrhea. Scand. J. Infect. Dis. 2010, 42, 857–861. [Google Scholar]
  45. Floch, M.H. The power of poop: Probiotics and fecal microbial transplant. J. Clin. Gastroenterol. 2012, 46, 625–626. [Google Scholar]
  46. Eiseman, B.; Silen, W.; Bascom, C.S.; Kauvar, A.J. Fecal enema as a adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958, 44, 854–859. [Google Scholar]
  47. Aas, J.; Gessert, C.E.; Bakken, J.S. Recurrent Clostridium difficile colitis: Case series involving 18 patients treated with donor stool administered via a nasogastric tube. Clin. Infect. Dis. 2003, 36, 580–585. [Google Scholar]
  48. Hamilton, M.J.; Weingarden, A.R.; Sadowsky, M.J.; Khoruts, A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 761–767. [Google Scholar]
  49. Naidoo, K.; Gordon, M.; Fagbemi, A.O.; Thomas, A.G.; Akobeng, A.K. Probiotics for maintenance of remission in ulcerative colitis. Cochrane Database Syst. Rev. 2011, 12, CD007443. [Google Scholar]
  50. Brace, C.; Gloor, G.B.; Ropeleski, M.; Allen-Vercoe, E.; Petrof, E.O. Microbial composition analysis of Clostridium difficile infections in an ulcerative colitis patient treated with multiple fecal microbiota transplantations. J. Crohn’s Colitis. 2014, 8, 1133–1137. [Google Scholar]
  51. Shen, J.; Zuo, Z.X.; Mao, A.P. Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn’s disease, and pouchitis: Meta-analysis of randomized controlled trials. Inflamm. Bowel Dis. 2014, 20, 21–35. [Google Scholar]
  52. Malchow, H.A. Crohn’s disease and Escherichia coli: A new approach in therapy to maintain remission of colonic Crohn’s disease. J. Clin. Gastroenterol. 1997, 25, 653–658. [Google Scholar]
  53. Cha, B.K; Jung, S.M.; Choi, C.H.; Song, I.D.; Lee, H.W.; Kim, H.J.; Hyuk, J.D.; Kyung Chang, S.; Kim, K.; Chung, W.S.; et al. The effect of a multispecies probiotic mixture on the symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: A randomized, double-blind, placebo-controlled trial. J. Clin. Gastroenterol. 2012, 46, 220–227. [Google Scholar]
  54. Hakansson, A.; Molin, G. Gut microbiota and inflammation. Nutrients 2011, 3, 637–682. [Google Scholar]

Share and Cite

MDPI and ACS Style

Floch, M.H. Recommendations for Probiotic Use in Humans—A 2014 Update. Pharmaceuticals 2014, 7, 999-1007. https://0-doi-org.brum.beds.ac.uk/10.3390/ph7100999

AMA Style

Floch MH. Recommendations for Probiotic Use in Humans—A 2014 Update. Pharmaceuticals. 2014; 7(10):999-1007. https://0-doi-org.brum.beds.ac.uk/10.3390/ph7100999

Chicago/Turabian Style

Floch, Martin H. 2014. "Recommendations for Probiotic Use in Humans—A 2014 Update" Pharmaceuticals 7, no. 10: 999-1007. https://0-doi-org.brum.beds.ac.uk/10.3390/ph7100999

Article Metrics

Back to TopTop