You are on page 1of 10

Curr Hematol Malig Rep (2016) 11:19–28

DOI 10.1007/s11899-016-0302-9

STEM CELL TRANSPLANTATION (R MAZIARZ, SECTION EDITOR)

Microbiota Manipulation With Prebiotics and Probiotics


in Patients Undergoing Stem Cell Transplantation
Tessa M. Andermann 1 & Andrew Rezvani 2 & Ami S. Bhatt 2,3,4

Published online: 16 January 2016


# Springer Science+Business Media New York 2016

Abstract Hematopoietic stem cell transplantation (HSCT) is Introduction


a potentially life-saving therapy that often comes at the cost of
complications such as graft-versus-host disease and post- Hematopoietic stem cell transplantation (HSCT) is a potentially
transplant infections. With improved technology to under- life-saving therapy for patients with both malignant and non-
stand the ecosystem of microorganisms (viruses, bacteria, fun- malignant hematologic disorders. Transplantation, however,
gi, and microeukaryotes) that make up the gut microbiota, comes at the cost of potential complications from chemotherapy
there is increasing evidence of the microbiota’s contribution and radiation, graft-versus-host disease (GVHD), and post-
to the development of post-transplant complications. Antibi- transplant infections. Gastrointestinal (GI) toxicity is common
otics have traditionally been the mainstay of microbiota- after HSCT, with up to 50 % of transplant recipients experienc-
altering therapies available to physicians. Recently, interest ing clinically significant diarrhea. While many clinical factors
is increasing in the use of prebiotics and probiotics to support related to host factors and cancer-targeted pharmacological in-
the development and sustainability of a healthier microbiota. terventions are implicated in the pathogenesis of gastrointestinal
In this review, we will describe the evidence for the use of toxicity, an increasing awareness of the contribution of the envi-
prebiotics and probiotics in combating microbiota dysbiosis ronment to these disease phenotypes is being explored. One such
and explore the ways in which they may be used in future potentially modifiable component of the environment is the gut
research to potentially improve clinical outcomes and de- microbiota, and this is increasingly being explored. The micro-
crease rates of graft-versus-host disease (GVHD) and biota is comprised of a diverse ecosystem of microbial organ-
post-transplant infection. isms, including bacteria, viruses, and fungi, existing in symbiosis
with the human host [1–3]. New laboratory techniques have
enabled the enumeration of the number, types, and gene content
Keywords Hematopoietic stem cell transplantation . of microorganisms within these communities [4, 5•], and have
Graft-versus-host disease . Post-transplant infection led to a greatly improved understanding of the relationship be-
tween the microbiota and the host. Several microbial niches exist
This article is part of the Topical Collection on Stem Cell Transplantation throughout the human body, including the skin, lungs, nares,
vagina, and gastrointestinal tract, with the majority of organisms
* Ami S. Bhatt
residing in the colon. Four main phyla found in the GI tract are
asbhatt@stanford.edu
known to contribute to human health and disease: Firmicutes,
Bacteroidetes, Actinobacteria, and Proteobacteria, with the ma-
1
Department of Medicine, Division of Infectious Diseases, Stanford jority of species being non-pathogenic [2]. These commensal
University, Stanford, CA, USA microbes play an important role in immune regulation, nutrition,
2
Department of Medicine, Division of Blood and Marrow and maintenance of host barriers against pathogens [6, 7]. The
Transplantation, Stanford University, Stanford, CA, USA importance of commensal organisms in pathogen defense is well
3
Department of Medicine, Division of Hematology, Stanford illustrated in germ-free mice, which lack the microbial stimulus
University, 269 Campus Drive, Stanford, CA 94305, USA necessary for the development of a mature immune system and
4
Department of Genetics, Stanford University, Stanford, CA, USA are subsequently more susceptible to infection [8, 9].
20 Curr Hematol Malig Rep (2016) 11:19–28

Dysbiosis, or the imbalance of the microbial ecosystem, that maintain a more diverse microbiome. Still, our ability to
has been associated with many diseases including irritable manipulate the microbiota to improve GVHD and infectious
bowel syndrome, inflammatory bowel disease (IBD), obesity, outcomes has significant implications for clinical outcomes in
and mental health [10–13]. In HSCT recipients, dysbiosis has HSCT patients and further study to clarify the role of various
been associated with complications including diarrhea, microbiota manipulating interventions is indicated.
GVHD, and bacterial infections, often with high morbidity Host-microbiota interactions in stem cell transplant patients
and mortality [14, 15••, 16••, 17••]. GVHD is a major com- have been reviewed in detail previously [28–31]; this review
plication of allogeneic HSCT and a significant barrier to wider aims instead to focus on the ways in which we might use
and more successful use of allotransplantation. While overall microbiota-based preventative and therapeutic strategies to
outcomes in allogeneic HSCT have continuously improved improve clinical outcomes in HSCT patients. While
and the incidence of most complications has decreased over microbiota-based strategies have traditionally involved the
time [18], the incidence of chronic GVHD is actually increas- use of antibiotics, the regular use of antibiotics comes at the
ing, likely in part due to the increasing at-risk population liv- cost of eradication of potentially salutary commensal organ-
ing longer, as overall transplant outcomes improve [19]. Con- isms and rising rates of antibiotic resistance. In normal sub-
ventional approaches to the prevention and treatment of jects and patients with other GI-related diseases, alternative
chronic GVHD have been disappointing and ineffective: de- approaches to Bprecision microbiota manipulation^ have been
spite decades of pre-clinical and clinical investigation, the found to impact the microbiota positively and dramatically at
standard first-line treatment for chronic GVHD remains glu- much less cost to individual and public health. These alterna-
cocorticoids, and there is no standard or consistently effective tive strategies primarily include the use of prebiotics—non-
second-line therapy [20]. Thus, novel approaches to GVHD digestible carbohydrate supplements that promote the growth
prevention and treatment are urgently needed. of commensal organisms in the gut—and the ingestion of live
The microbial milieu of the gastrointestinal tract has long organisms as probiotics (Fig. 1). These two microbiota-
been linked to the development of GVHD following stem cell manipulating strategies, prebiotics and probiotics, will be
transplant. For example, in the 1970s, germ-free mice were reviewed here as potential strategies to improve outcomes in
shown to be less likely to develop GVHD [21, 22]. In the HSCT patients.
1980s, human gut decontamination in concert with protective
laminar airflow rooms were found to be potentially helpful in
decreasing GVHD [23]. In more recent publications, prophy- Diet and Prebiotics
laxis with broad-spectrum antibiotics in both children and
adults was shown to reduce the development of GVHD [24, While direct effects of dietary metabolites on the human gut
25]. Broad antibiosis has the benefit of reducing not only epithelium and secondary target tissues are known, the diet is
GVHD but also the risk for infections associated with HSCT also a major modulator of the structure and function of the
[24]. However, other experiments attempting to replicate these human microbiota. The role of diet and nutrition in HSCT
findings did not find a similar effect and the use of total gut patients is often underappreciated, but growing evidence sug-
decontamination especially in concert with laminar airflow gests a link between the diet, the microbiota, and clinical out-
isolation to prevent GVHD has fallen out of popular use comes [32]. Specific elements of our diet called prebiotics are
[26]. Indeed, more recent research has indicated that a loss particularly influential in the structure and function of the mi-
of microbial diversity following HSCT results in higher mor- crobiota [33]. The term Bprebiotic^ has traditionally been ap-
tality rates, indicating a potentially deleterious effect of anti- plied to indigestible carbohydrates metabolized by gut bacteria
biosis on the microbiome [16••]. Loss of microbial diversity to produce nutrients utilized by intestinal epithelial cells (IECs)
and commensal flora can result in overgrowth of pathogenic in such a way as to improve overall health [33]. A commonly
organisms, thereby increasing the risk for sepsis by antibiotic- cited benefit of a prebiotic is that of fiber in altering intestinal
resistant organisms. In addition, the loss of commensal organ- immunity. Indigestible plant oligo- and polysaccharides made
isms has been associated with higher levels of gastrointestinal up of chains of sugars of varying lengths are fermented in the
inflammation, which was found to be especially pronounced colon by commensal bacteria, resulting in the production of
in patients with GVHD [17••]. A decrease in anaerobic bacte- short-chain fatty acids (SCFAs) that serve as a source of energy
ria and, in particular, the genus Blautia through antibiosis was for colonocytes [34]. SCFAs—namely butyrate, acetate, and
associated with an increased incidence of GVHD in those propionate—have been implicated in promoting anti-
undergoing allogeneic HSCT [27••]. Unnecessary use of an- inflammatory cytokine production of helper T cells, intestinal
aerobically active antibiotics may therefore require a particu- barrier integrity, and overall regulation of intestinal immune
larly heightened level of scrutiny. These studies call into ques- function [34, 35]. SCFAs also promote resolution of intestinal
tion our routine use of broad-spectrum antibiotics in prophy- inflammation by increasing development of T regulatory cells
laxis and gut decontamination and argue for other strategies (T-regs) through interactions with G-protein-coupled receptors
Curr Hematol Malig Rep (2016) 11:19–28 21

Fig. 1 Effects of prebiotics,


probiotics, and antibiotics on the
microbiota in stem cell transplant
patients. Diet and antibiotics
interact with host factors
including the patient’s underlying
malignancy, prior chemotherapy,
and conditioning regimens to
affect the composition of the
microbiota. Antibiotics in this
model can lead to a decrease in
microbiota diversity and result in
the development of dysbiosis,
diarrhea, GVHD, and infection.
The microbiota can be returned to
a more symbiotic state
representative of a prior healthier
state of homeostasis with the use
of prebiotics and probiotics that
improve microbiota diversity

[36, 37]. Originally, the term prebiotic was restricted to the patients to oligofructose-enriched inulin or a control and found
polysaccharide inulin (obtained from vegetables such as the a similar increase in fecal bifidobacteria and decrease in dis-
Jerusalem artichoke) and its derivative fructo-oligosaccharide ease activity [44], although an earlier randomized double-blind
(FOS), termed inulin-type fructans (ITF). More recently, other controlled trial did not find any clinical benefit [45].
oligosaccharides have been found to function as beneficial pre- The role of dietary changes in improving outcomes pa-
biotics. Xylo-oligosaccharides and galacto-oligosaccharides al- tients undergoing HSCT is even less well understood than
so increase bifidobacterium growth and SCFA concentration in in IBD. Dysbiosis has been implicated in the pathogenesis
randomized clinical trials; although to date, clinical studies of IBD as well as GVHD [48]. Studies of the microbiome
have only included healthy volunteers [38–41]. in HSCT patients with GVHD show similarities with IBD;
Dietary effects on the microbiota have been relatively well for example, HSCT patients with GVHD demonstrate a
researched in healthy volunteers and are increasingly being decrease in microbial diversity with an associated effect
studied in mouse models of colitis and in patients with IBD, on mortality [16••]. In comparison to patients with IBD,
where diet has long been suspected as a contributing factor to even less is known about how diet might impact the mi-
illness. Very little is known about dietary changes in patients crobiota in patients with GVHD. To date, only one study
undergoing HSCT, but IBD provides a context for understand- has looked at the efficacy and safety of a prebiotic in pa-
ing potential diet-microbiota effects applicable to future re- tients undergoing HSCT. In 2014, Iyama et al. reported a
search in transplant patients. Experimental evidence in the retrospective cohort study in which 22 patients were given
pathogenesis of IBD demonstrates support for both an abnor- a combination of glutamine, fiber, and a fructo-
mal immune response to normal microbiota and a normal oligosaccharide (GFO) and compared to matched controls
immune response to an abnormal microbiota. Studies of pa- [49]. Researchers found that the use of GFO supplementa-
tients with IBD have demonstrated decreased diversity, re- tion correlated with a significant decrease in the days of
duced proportions of Firmicutes, and increased proportions moderate diarrhea (grade 2, p = 0.0001), severe diarrhea
of Proteobacteria and Actinobacteria [42]. Dietary supple- (grade 3–4, p = 0.001), and severe mucositis (grade 3–4,
ments like prebiotics—in particular inulin and fructo- p = 0.033). There was a significant difference in survival
oligosaccharides (ITFs)—have been studied in patients with between those receiving the supplement and those who
IBD in a small number of RCTs. The results from these RCTs did not (100 vs. 77 %, respectively, p = 0.0091), although
vary in their support for clinical improvement [43–45]. In one this finding must be interpreted cautiously considering the
pilot trial of patients with ulcerative colitis, the use of ITF was retrospective nature of the trial. No differences in rates of
associated with a significant decrease in calprotectin, a marker infections were observed and the prebiotic supplement was
of active disease [46]. A single-arm pilot trial of FOS in pa- well tolerated and without adverse effects. Notably, all pa-
tients with Crohn’s disease found an increase in fecal tients included in this trial received a preparation of the
bifidobacterial content and a decrease in subjective disease probiotic Lactobacillus throughout their transplant without
activity [47]. Another trial randomized Crohn’s disease any resulting documented infections. While this trial is
22 Curr Hematol Malig Rep (2016) 11:19–28

small and retrospective, it does suggest that a prebiotic in By the time patients develop severe steroid-refractory acute
HSCT patients is likely safe and may improve clinical out- GVHD, it is often too late for effective treatment and recovery
comes related to post-transplant diarrhea and mucositis. of gastrointestinal mucosal integrity and function. Given the
Little is known about the role of prebiotics or probiotics in dismal clinical outcomes associated with steroid-refractory
HSCT patients, with much of the limited research available GVHD, preventive approaches are essential. Perhaps a
being focused on patients’ overall diet. After HSCT, nutrition- thoughtful modulation of the gut microbiota through alteration
al support by the enteral route is preferred in order to maintain in nutrition before and after transplant may decrease the risk of
digestive function and the mucosal barrier, preventing bacte- the development and/or severity of GVHD, with little risk to
rial translocation [50]. Debate exists, however, about the role the patient. To this end, prospective randomized trials of pre-
of total parenteral nutrition (TPN) vs. enteral nutrition (EN), biotics and early enteral nutrition will be vital in furthering our
and the effects on the microbiota of long-term TPN are not knowledge of host-microbiota dynamics important in the pre-
well understood. General guidelines in HSCT patients suggest vention of GVHD.
the use of a graded GVHD diet, which transitions patients
gradually from a liquid diet to an increasingly expansive menu
of solid foods once the volume of diarrhea has decreased Probiotics
below 500 mL/day [51]. The proposed diet for HSCT patients
with GVHD is recommended to have limited amounts of fats, Probiotics, defined as the administration of live microorgan-
fiber, lactose, acidic items, and GI irritants, although little isms to improve health, have been used for centuries and like-
research has been done to support these recommendations ly for millennia. The most commonly used probiotic organ-
[51]. In fact, given our understanding of microbiota-based isms—Lactobacillus spp. and Bifidobacterium spp.—are con-
therapeutics in healthy patients and in those with IBD, patients sumed in large quantities as part of traditional foods across the
undergoing HSCT may benefit from the earlier dietary addi- globe. Examples include natto and miso (from soybean), sau-
tion of fiber, vegetables, and fruits in order to prevent GVHD erkraut (from cabbage), and kombucha (fermented tea),
and to assist in gut healing after GI GVHD. Immunomodula- among others. Alterations in the human microbiota are asso-
tory diets have been shown to improve immune system cell ciated with disease and therapeutic manipulation of the micro-
function and to reduce inflammation through regulation of T biota with probiotics has the potential to improve health and
cell responses [52]. The definition of prebiotics may need to well-being. IBD and GVHD are both associated with de-
be expanded to include other nutrients and small molecules, creased microbiota diversity. For example, there is moderately
which could then serve as dietary supplements with the goal of strong evidence supporting the use of probiotics to prevent
positively impacting microbiota. Possible therapeutic targets Clostridium difficile infection [54]. Probiotics have proven
for intervention include not only fiber and SCFAs but also efficacious in treating a wide variety of diarrheal illnesses,
small molecules that interact with G-protein-coupled recep- including antibiotic-associated diarrhea [55–57], infectious di-
tors, or fermentative enzymes in the production of SCFAs arrhea or gastroenteritis [58, 59], irritable bowel syndrome
[53]. It is therefore reasonable to consider these or similar [60–64], ulcerative colitis [65, 66], necrotizing enterocolitis
interventions in patients undergoing HSCT with the hope of [67–69], constipation [70], and pouchitis [71–73], although
reducing the incidence of GVHD and/or infection. they have not yet shown significant efficacy in the treatment
In the process of undergoing HSCT, diet and nutrition are of Crohn’s disease [74]. Proposed mechanisms for probiotic
often viewed as a less important form of support compared to efficacy include the microorganism-based remodeling of mi-
immunosuppressive medication and prophylactic antibiotics. crobial communities and suppression of pathogens through
Given our growing understanding of the importance of the direct antimicrobial effects, stimulation of immune responses
microbiota in the development of diarrhea, infections, and that lead to upregulation of anti-inflammatory cytokines and
GVHD, the role of the diet and nutrition may soon rise to IgA, and promotion of intestinal barrier function [75, 76].
the level of prevention and treatment in patients undergoing Interventional studies of probiotics have primarily focused
HSCT. We may want to consider earlier enteral feeding of on the use of organisms that fall within the genera of
HSCT patients and limited use of TPN. As seen in IBD, Bifidobacterium and Lactobacillus, most likely because of
TPN in HSCT patients may decrease diarrhea but likely does their predominance in fermentable foods, their overall safety,
not impact clinical outcomes and can negatively impact the and their ease of culture and production. More recent studies
microbiota [27••]. Just as in IBD patients, those with GVHD have gone beyond these organisms to encompass a combina-
may benefit from an early enteral diet even if they are unable tion of bacteria and to employ a more diverse group of bacte-
to tolerate solid foods, even in the setting of mucositis. This ria. In a systematic review of probiotics in patients with IBD,
elemental diet may also include prebiotics and other supple- three RCTs found improved outcomes in patients with ulcer-
ments that may positively influence our microbiota. The ative colitis using the probiotic VSL#3, a highly concentrated
timing of these dietary interventions may also be important. mixture of multiple species from the genera Bifidobacterium
Curr Hematol Malig Rep (2016) 11:19–28 23

and Lactobacillus plus Streptococcus thermophiles [77]. In guidelines for stool donor screening, although the literature to
mouse and human studies, VSL#3 has been demonstrated to date universally favors blood and stool screening for HIV, hep-
increase anti-inflammatory IL-10 production and decrease atitis A, hepatitis B, hepatitis C, stool C.difficile, parasitic stool
proinflammatory IL-12 production by dendritic cells [78]. analysis, and bacterial culture, at a very minimum, prior to FMT
Going beyond the use of a few select organisms, fecal mi- in non-immunocompromised patients. Guidelines for donor
crobiota transplantation (FMT) as the ultimate probiotic has gar- screening prior to FMT in immunocompromised patients will
nered increasing attention for its successful use in refractory need to be established and should consider the use of a more
C.difficile-associated disease (CDAD). CDAD is one of the comprehensive screening protocol including the organisms rec-
most frequently studied examples of dysbiosis, and one that is ommended in Table 2. Improving our understanding of the mi-
increasingly more common and refractory as a result of our croorganisms comprising the fecal transplant will undoubtedly
overuse of broad-spectrum antibiotics. FMT has been shown make this a safer procedure, although the possibility of endog-
to be safe and effective for CDAD, including in highly refractory enous potentially pathogenic organisms like Eschericia coli or
disease [79]. As a result, interest has grown in FMT as a possible Enterococcus spp. leading to bacterial translocation, sepsis, and
treatment for IBD and for CDAD in patients with IBD. Howev- bacteremia will persist regardless of screening. One option to
er, the enthusiasm for fecal transplant is somewhat attenuated in consider in HSCT patients is banking of patient stool prior to
this population; in a recent systematic review of case studies and HSCT or even prior to chemotherapy, to be used in an autolo-
case series of FMT in patients with IBD with or without CDAD, gous fashion to treat CDAD, antibiotic-associated diarrhea, or
several patients in these reports were noted to have developed even GVHD. Autologous FMT in HSCT patients is currently
adverse effects including bacteremia, worsening of their IBD, undergoing investigation and represents an exciting prospect in
and fevers [80]. No deaths were reported after FMT, even in the future of microbiota-based therapeutics in immunocompro-
patients who were immunosuppressed. Despite these concerns, mised patients (https://clinicaltrials.gov/ct2/show/study/
71 % of patients were noted to have had resolution or reduction NCT02269150).
of IBD or IBD/CDAD symptoms, generally with single fecal Apart from single-organism probiotic supplements or
infusions. These results are encouraging, although notably transplant of an entire microbiota via FMT, we may be able
poorer than those reported for FMT in patients with CDAD to synthesize an idealized defined microbial ecosystem using
alone (success rate ∼90 %) [79]. organisms that are less likely to become pathogenic. This no-
Given these findings, it is not surprising that both probiotics tion of microbial ecosystem therapeutics (MET) is not new,
and FMT are used with great hesitation in patients who have but has yet to enter widespread use. A study in 1989 by Tvede
undergone HSCT. There is understandable concern for sepsis as and Rask-Madsen used a mixture of ten strains of bacteria,
a result of bacterial translocation, or viral infection, and indeed including three strains of Clostridium spp., three of
case reports have described this phenomenon [81, 82]. However, Bacteroides spp., two strains of E.coli, Peptostreptococcus
mouse models have shown that Lactobacillus administered be- spp., and Enterococcus faecalis [87]. In the five patients
fore and after HSCT reduced GVHD and improved survival who received this enteral mixture for CDAD, all became
[83]. In three case reports to date, patients with refractory CDAD asymptomatic and tested negative for C.difficile toxin within
after HSCT have undergone FMT with success and with mini- 24 h. In 2013, an ecosystem of 33 strains of non-pathogenic
mal adverse effects [84–86] (See Table 1). In these cases, FMT bacteria was isolated from a stool donor and used to treat two
may be the only non-surgical option for patients failing antibi- patients who were subsequently cured of their C.difficile in-
otic therapy for CDAD, and the safety of this approach should fection [88]. The resolution of symptoms was robust in these
improve with the implementation of broader and more compre- individuals; even after receiving antibiotics for other infec-
hensive screening of donor stool. Currently, there are no formal tions, these patients did not experience recurrent CDAD.

Table 1 Case reports of patients post-HSCT who have undergone fecal microbiota transplant

Patient Donor Method of Follow-up CDAD status at Authors


delivery last follow-up

64-year-old male 11 months Unknown donors given Enema 7 months Resolved Mittal et al. 2015 [84]
post autologous HSCT for at 2 time points 6
diffuse large B cell lymphoma months apart
60 year-old female 13 months Two unidentified donors Push enteroscopy 10 months Resolved De Castro et al. 2015 [85]
post allogeneic HSCT for acute
lymphoblastic leukemia
21 year-old female 1 year post Husband Nasogastric tube 2 months Resolved Neemann et al. 2012 [86]
allogeneic HSCT for acute
lymphoblastic leukemia
24 Curr Hematol Malig Rep (2016) 11:19–28

Table 2 Stool donor screening of blood and stool in FMT studies for CDAD with at least one immunocompromised patient
Friedman- Potential
Aas et al Hamilton et Zainah et Duplessis et Pathak et Trubiano et
Moraco et al. comprehensive
2003[94] al. 2012[95] al. 2012[96] al. 2012[97] al. 2013[98] al. 2014[100]
2014[99] screening*
Blood
Hepatitis A
Hepatitis B
Hepatitis C
Hepatitis E
HIV 1/2
HTLV-I/II
Syphilis
CMV
EBV
Stool
Clostridium
difficile
Bacterial culture**
Helicobacter
pylori
Ova and parasites
Giardia
Cryptosporidium
Microsporidia
Stronglyoides
stercoralis
Entamoeba
histolytica
Cyclospora
Isospora
Dientamoeba
fragilis
Blastocystis
hominis
Schistosoma
Norovirus
Rotavirus
Adenovirus
JC Virus
*Donor screening prior to FMT in HSCT patients should include consideration for this more comprehensive screening. In the case of Strongyloides
stercoralis and Schistosoma, testing would be recommended only after assessment of donor exposure risk
**Bacterial culture includes the evaluation of standard pathogenic organisms (e.g. Salmonella spp., Shigella spp., Campylobacter spp., Vibrio spp., and
Aeromonas spp., and Escherichia coli 0157:H7). Several studies also screened for drug resistant organisms (specifically VRE and multi-drug resistant
Gram-negative bacteria) which would be recommended in this vulnerable population
EBV Epstein-Barr virus; CMV cytomegalovirus; HIV human immunodeficiency virus, HTLV human T-cell lymphotropic virus

The use of multiple non-pathogenic organisms as a synthetic complicated by its extreme sensitivity to oxygen, researchers
Bdesigner microbiota^ may be safer than FMT. To this end, have recently found a way to stabilize the organism in an
recent metagenomic studies of the gut microbiome have re- aerobic environment through the use of an inulin and
sulted in the identification of novel organisms—most of antioxidant-based matrix [91]. Exemplifying the use of a
which are likely obligate anaerobes—that may be used specif- Bdesigner microbiota,^ researchers in Japan used a mixture
ically to repopulate the human gut microbiota and improve of rationally selected strains of Clostridia to induce the expan-
gastrointestinal health. For example, Faecalibacterium sion and differentiation of T-regs in a mouse model of colitis
prausnitzii, a member of the Firmicutes phylum, was first [92]. Very recently, Clostridium scindens has been found to
identified in 2008 as an important anti-inflammatory com- provide resistance to CDAD through bile acid production
mensal [89]. In patients with Crohn’s disease, it was discov- [93••]. The use of F. prausnitzii in probiotic form along with
ered that F. prausnitzii levels were lower in patients who de- other strains such as Clostridium butyrate (another butyrate-
veloped endoscopic recurrence of the disease compared to producing organism) and the bile acid-producing C. scindens
those who remained in remission. In a mouse model of inflam- may positively influence the microbiota in immunocompro-
matory colitis, oral administration of F. prausnitzii or its su- mised patients with the possibility of decreased risk for sepsis
pernatant reduced the severity of colitis and contributed to the or bacteremia in comparison to currently available probiotics.
normalization of the microbiota through mechanisms involv- The discovery of these important organisms as major players
ing high levels of butyrate production and increased secretion in resistance against disease highlights the increasing focus on
of IL-10 [90]. While the administration of F. prauznitzii is precision microbiota manipulation applicable to
Curr Hematol Malig Rep (2016) 11:19–28 25

immunocompromised patients. Further research is required to and we anticipate great potential for the microbiota as a po-
determine disease-specific or even individually based tentially modifiable biomarker of disease in this vulnerable
probiotics targeted against a variety of dysbiotic states. patient population.

Acknowledgments This work was made possible by the following


awards: American Society of Hematology Scholar Award (A.S.B.),
Conclusion
Amy Strelzer Manasevit award (National Marrow Donor Program and
Be The Match foundation) (A.S.B.), NCI K08 CA184420 (A.S.B), the
A better understanding of our microbiota and its symbiotic American Cancer Society Mentored Research Scholar Grant 122663-
relationship with the human host holds significant promise MRSG-12-162-01-LIB (A. R.), and NIH T32 AI007502 (T.M.A.). The
authors would also like to thank Dr. Lucy Tompkins from the Division of
for improving health in HSCT recipients. Broad pharmacolog-
Infectious Diseases at Stanford University for her review of the
ic approaches aimed at immunomodulation for treatment of manuscript and figures.
GVHD and antimicrobials strategies for treatment of infection
in BMT patients have predominated for decades, with limited Compliance with Ethical Standards
success (particularly in chronic GVHD) and with negative
Conflict of Interest The authors declare that they have no competing
consequences for increasing susceptibility to opportunistic in- interests.
fections and the promotion of drug-resistant organisms. Anti-
biotics manipulate the microbiota in such a way that dysbiosis Human and Animal Rights and Informed Consent This article does
often results, leading to subsequent inflammation, increased not contain any studies with human or animal subjects performed by any
of the authors.
risk for infection, and possibly an increased risk of GVHD. A
completely new approach is offered by the burgeoning evi-
dence in mice and human studies that a more precise alteration
of the microbiota may be an even more effective tool in
preventing and treating disease, particularly in the setting of References
HSCT. This approach represents a major shift in our current
understanding of infectious diseases and host-microbe inter-
Papers of particular interest, published recently, have been
actions. Microbiota-based therapeutics will include the use of
highlighted as:
narrower, more precise antibiotics as well as dietary changes,
• Of importance
nutritional prebiotic supplements that support the growth of
•• Of major importance
commensal organisms, probiotics, and FMT.
In the future, we may be able to use increasing knowledge 1. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R,
of the microbiota to develop Bprecision^ prebiotics that are Gordon JI. The human microbiome project. Nature. 2007;449:
even more powerful in expanding commensal flora or that 804–10.
influence organisms towards a particular shift of the gastroin- 2. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L,
Sargent M, et al. Diversity of the human intestinal microbial flora.
testinal metabolome. Probiotics could potentially be Science. 2005;308:1635–8.
engineered to support the gut microbiota while also having a 3. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel
biological Boff-switch^ that could be triggered at the first sus- BS, et al. Metagenomic analysis of the human distal gut
picion of bacteremia. The development of designer microbiota microbiome. Science. 2006;312:1355–9.
4. Morgan XC, Huttenhower C. Meta’omic analytic techniques for
with non-pathogenic organisms may be of use even in highly
studying the intestinal microbiome. Gastroenterology. 2014;146:
immunocompromised patients undergoing HSCT. An early 1437–48.e1.
detection of dysbiosis could be treated with an individualized 5.• Bhatt AS, Freeman SS, Herrera AF, Pedamallu CS, Gevers D,
microbiota transfer of commensal organisms necessary to en- Duke F, et al. Sequence-based discovery of Bradyrhizobium
enterica in cord colitis syndrome. N Engl J Med. 2013;369:517–
hance diversity for the prevention of subsequent bacteremia
28. Bhatt et al. used next-generation shotgun sequencing in the
and sepsis. In addition, an improved understanding of the characterization of the microbiome in umbilical cord HSCT
human ecosystem may allow us to use patients’ microbiota transplant patients with cord colitis and identified a novel
composition as a biomarker of disease. For example, we can pathogen, Bradyrhizobium enterica, responsible for disease in
foresee a future in which a patient’s microbiotic signature these patients.
6. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell
serves as a predictor of risk for steroid-refractory GVHD, development by a commensal bacterium of the intestinal microbi-
allowing the earlier addition of secondary treatments. Overall, ota. Proc Natl Acad Sci U S A. 2010;107:12204–9.
microbiota-based therapeutics hold great promise for the pre- 7. Round JL, Mazmanian SK. The gut microbiota shapes intestinal
vention and treatment of GVHD and infections in HSCT pa- immune responses during health and disease. Nat Rev Immunol.
2009;9:313–23.
tients. Significant opportunity exists for further research into 8. Kamada N, Kim Y-G, Sham HP, Vallance BA, Puente JL, Martens
the development of targeted and individualized dysbiosis pre- EC, et al. Regulated virulence controls the ability of a pathogen to
vention and treatment regimens applicable to HSCT patients, compete with the gut microbiota. Science. 2012;336:1325–9.
26 Curr Hematol Malig Rep (2016) 11:19–28

9. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor 23. Navari RM, Buckner CD, Clift RA, Storb R, Sanders JE, Stewart
prevents intestinal inflammatory disease. Nature. 2008;453:620–5. P, et al. Prophylaxis of infection in patients with aplastic anemia
10. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, receiving allogeneic marrow transplants. Am J Med. 1984;76:
Gordon JI. An obesity-associated gut microbiome with increased 564–72.
capacity for energy harvest. Nature. 2006;444:1027–31. 24. Vossen JM, Guiot HFL, Lankester AC, Vossen ACTM, Bredius
11. Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, RGM, Wolterbeek R, et al. Complete suppression of the gut
Maza O, et al. Artificial sweeteners induce glucose intolerance microbiome prevents acute graft-versus-host disease following al-
by altering the gut microbiota. Nature. 2014;514:181–6. logeneic bone marrow transplantation. PLoS One. 2014;9:
12. Collins SM. A role for the gut microbiota in IBS. Nat Rev e105706.
Gastroenterol Hepatol. 2014;11:497–505. 25. Beelen DW, Elmaagacli A, Müller K-D, Hirche H, Schaefer UW.
13. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Influence of intestinal bacterial decontamination using metronida-
et al. Microbiota modulate behavioral and physiological abnor- zole and ciprofloxacin or ciprofloxacin alone on the development
malities associated with neurodevelopmental disorders. Cell. of acute graft-versus-host disease after marrow transplantation in
2013;155:1451–63. patients with hematologic malignancies: final results and long-
14. Taur Y, Pamer EG. The intestinal microbiota and susceptibility to term follow-up of an open-label prospective randomized trial.
infection in immunocompromised patients. Curr Opin Infect Dis. Blood. 1999;93:3267–75.
2013;26:332–7. 26. Russell JA, Chaudhry A, Booth K, Brown C, Woodman RC,
15.•• Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, Valentine K, et al. Early outcomes after allogeneic stem cell trans-
et al. Intestinal domination and the risk of bacteremia in patients plantation for leukemia and myelodysplasia without protective
undergoing allogeneic hematopoietic stem cell transplantation. isolation: a 10-year experience. Biol Blood Marrow Transplant J
Clin Infect Dis Off Publ Infect Dis Soc Am. 2012;55:905–14. Am Soc Blood Marrow Transplant. 2000;6:109–14.
Taur et al. showed the reduction of intestinal microbiota diver- 27.•• Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF,
sity following allogeneic HSCT. Those patients with reduced et al. Intestinal blautia is associated with reduced death from graft-
microbiota diversity often developed microbiota domination versus-host disease. Biol Blood Marrow Transplant J Am Soc
by a single bacterial taxon leading to an increased risk of bac- Blood Marrow Transplant. 2015;21:1373–83. Jenq et al. demon-
teremia with the dominant organism. strated that increased microbiota diversity during transplant
16.•• Taur Y, Jenq RR, Perales M-A, Littmann ER, Morjaria S, Ling L, was associated with the decreased acute GVHD and lower
et al. The effects of intestinal tract bacterial diversity on mortality GVHD-related mortality. This resistance to GVHD and
following allogeneic hematopoietic stem cell transplantation. GVHD-related mortality was mediated by the abundance of
Blood. 2014;124:1174–82. Taur et al. demonstrated that intes- Blautia within the intestinal microbiota.
tinal microbiota diversity was an independent predictor of 28. Manzo VE, Bhatt AS. The human microbiome in hematopoiesis
mortality in patients undergoing allogeneic HSCT. Patients and hematologic disorders. Blood. 2015;126:311–8.
with low microbiota diversity were found to have higher all- 29. Mathewson N, Reddy P. The microbiome and graft versus host
cause mortality. disease. Curr Stem Cell Rep. 2015;1:39–47.
17.•• Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, 30. Shono Y, Docampo MD, Peled JU, Perobelli SM, Jenq RR.
Peter K, et al. Metagenomic analysis of the stool microbiome in Intestinal microbiota-related effects on graft-versus-host disease.
patients receiving allogeneic stem cell transplantation: loss of di- Int J Hematol. 2015;101:428–37.
versity is associated with use of systemic antibiotics and more 31. Docampo MD, Auletta JJ, Jenq RR. Emerging influence of the
pronounced in gastrointestinal graft-versus-host disease. Biol intestinal microbiota during allogeneic hematopoietic cell trans-
Blood Marrow Transplant J Am Soc Blood Marrow Transplant. plantation: control the Gut and the body will follow. Biol Blood
2014;20:640–5. Holler et al. found an association between the Marrow Transplant J Am Soc Blood Marrow Transplant. 2015;21:
loss of diversity in the intestinal microbiota following the use of 1360–6.
antibiotics during allogeneic HSCT and the development of 32. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button
acute gastrointestinal GVHD. JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human
18. Gooley TA, Chien JW, Pergam SA, Hingorani S, Sorror ML, gut microbiome. Nature. 2014;505:559–63.
Boeckh M, et al. Reduced mortality after allogeneic 33. Roberfroid MB. Introducing inulin-type fructans. Br J Nutr.
hematopoietic-cell transplantation. N Engl J Med. 2010;363: 2005;93 Suppl 1:S13–25.
2091–101. 34. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA,
19. Arai S, Arora M, Wang T, Spellman SR, He W, Couriel DR, et al. Bohlooly-Y M, et al. The microbial metabolites, short-chain fatty
Increasing incidence of chronic graft-versus-host disease in allo- acids, regulate colonic Treg cell homeostasis. Science. 2013;341:
geneic transplantation: a report from the Center for International 569–73.
Blood and Marrow Transplant Research. Biol Blood Marrow 35. Peng L, He Z, Chen W, Holzman IR, Lin J. Effects of butyrate on
Transplant J Am Soc Blood Marrow Transplant. 2015;21:266–74. intestinal barrier function in a Caco-2 cell monolayer model of
20. Wolff D, Gerbitz A, Ayuk F, Kiani A, Hildebrandt GC, Vogelsang intestinal barrier. Pediatr Res. 2007;61:37–41.
GB, et al. Consensus conference on clinical practice in chronic 36. Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and
graft-versus-host disease (GVHD): first-line and topical treatment Bwestern-lifestyle^ inflammatory diseases. Immunity. 2014;40:
of chronic GVHD. Biol Blood Marrow Transplant J Am Soc 833–42.
Blood Marrow Transplant. 2010;16:1611–28. 37. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al.
21. Jones JM, Wilson R, Bealmear PM. Mortality and gross pathology Regulation of inflammatory responses by gut microbiota and
of secondary disease in germfree mouse radiation chimeras. chemoattractant receptor GPR43. Nature. 2009;461:1282–6.
Radiat Res. 1971;45:577–88. 38. Lecerf J-M, Dépeint F, Clerc E, Dugenet Y, Niamba CN, Rhazi L,
22. van Bekkum DW, Roodenburg J, Heidt PJ, van der Waaij D. et al. Xylo-oligosaccharide (XOS) in combination with inulin
Mitigation of secondary disease of allogeneic mouse radiation modulates both the intestinal environment and immune status in
chimeras by modification of the intestinal microflora. J Natl healthy subjects, while XOS alone only shows prebiotic proper-
Cancer Inst. 1974;52:401–4. ties. Br J Nutr. 2012;108:1847–58.
Curr Hematol Malig Rep (2016) 11:19–28 27

39. Childs CE, Röytiö H, Alhoniemi E, Fekete AA, Forssten SD, difficile-associated diarrhea in adults and children. Cochrane
Hudjec N, et al. Xylo-oligosaccharides alone or in synbiotic com- Database Syst Rev. 2013;5:CD006095.
bination with Bifidobacterium animalis subsp. lactis induce 55. Cremonini F, Di Caro S, Covino M, Armuzzi A, Gabrielli M,
bifidogenesis and modulate markers of immune function in Santarelli L, et al. Effect of different probiotic preparations on
healthy adults: a double-blind, placebo-controlled, randomised, anti-helicobacter pylori therapy-related side effects: a parallel
factorial cross-over study. Br. J. Nutr. 2014: 1–12. group, triple blind, placebo-controlled study. Am J
40. Bouhnik Y, Raskine L, Simoneau G, Vicaut E, Neut C, Flourié B, Gastroenterol. 2002;97:2744–9.
et al. The capacity of nondigestible carbohydrates to stimulate 56. Beniwal RS, Arena VC, Thomas L, Narla S, Imperiale TF,
fecal bifidobacteria in healthy humans: a double-blind, random- Chaudhry RA, et al. A randomized trial of yogurt for prevention
ized, placebo-controlled, parallel-group, dose–response relation of antibiotic-associated diarrhea. Dig Dis Sci. 2003;48:2077–82.
study. Am J Clin Nutr. 2004;80:1658–64. 57. Kotowska M, Albrecht P, Szajewska H. Saccharomyces boulardii
41. Vulevic J, Juric A, Walton GE, Claus SP, Tzortzis G, Toward RE, in the prevention of antibiotic-associated diarrhoea in children: a
et al. Influence of galacto-oligosaccharide mixture (B-GOS) on randomized double-blind placebo-controlled trial. Aliment
gut microbiota, immune parameters and metabonomics in elderly Pharmacol Ther. 2005;21:583–90.
persons. Br. J. Nutr. 2015: 1–10. 58. Guandalini S, Pensabene L, Zikri MA, Dias JA, Casali LG,
42. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Hoekstra H, et al. Lactobacillus GG administered in oral rehydra-
Pace NR. Molecular-phylogenetic characterization of microbial tion solution to children with acute diarrhea: a multicenter
community imbalances in human inflammatory bowel diseases. European trial. J Pediatr Gastroenterol Nutr. 2000;30:54–60.
Proc. Natl. Acad. Sci. U. S. A. 2007;104:13780–5. 59. Margreiter M, Ludl K, Phleps W, Kaehler ST. Therapeutic value of a
43. Hafer A, Krämer S, Duncker S, Krüger M, Manns MP, Bischoff Lactobacillus gasseri and Bifidobacterium longum fixed bacterium
SC. Effect of oral lactulose on clinical and immunohistochemical combination in acute diarrhea: a randomized, double-blind, con-
parameters in patients with inflammatory bowel disease: a pilot trolled clinical trial. Int J Clin Pharmacol Ther. 2006;44:207–15.
study. BMC Gastroenterol. 2007;7:36. 60. Niedzielin K, Kordecki H, Birkenfeld B. A controlled, double-
44. Joossens M, De Preter V, Ballet V, Verbeke K, Rutgeerts P, blind, randomized study on the efficacy of Lactobacillus
Vermeire S. Effect of oligofructose-enriched inulin (OF-IN) on plantarum 299V in patients with irritable bowel syndrome. Eur J
bacterial composition and disease activity of patients with Gastroenterol Hepatol. 2001;13:1143–7.
Crohn’s disease: results from a double-blinded randomised con- 61. Kajander K, Hatakka K, Poussa T, Färkkilä M, Korpela R. A
trolled trial. Gut. 2012;61:958. probiotic mixture alleviates symptoms in irritable bowel syndrome
45. Benjamin JL, Hedin CRH, Koutsoumpas A, Ng SC, McCarthy patients: a controlled 6-month intervention. Aliment Pharmacol
NE, Hart AL, et al. Randomised, double-blind, placebo-controlled Ther. 2005;22:387–94.
trial of fructo-oligosaccharides in active Crohn’s disease. Gut. 62. Enck P, Zimmermann K, Menke G, Müller-Lissner S, Martens U,
2011;60:923–9. Klosterhalfen S. A mixture of Escherichia coli (DSM 17252) and
46. Casellas F, Borruel N, Torrejón A, Varela E, Antolin M, Guarner F, Enterococcus faecalis (DSM 16440) for treatment of the irritable
et al. Oral oligofructose-enriched inulin supplementation in acute bowel syndrome–a randomized controlled trial with primary care
ulcerative colitis is well tolerated and associated with lowered physicians. Neurogastroenterol Motil Off J Eur Gastrointest Motil
faecal calprotectin. Aliment Pharmacol Ther. 2007;25:1061–7. Soc. 2008;20:1103–9.
47. Lindsay JO, Whelan K, Stagg AJ, Gobin P, Al-Hassi HO, 63. Cui S, Hu Y. Multistrain probiotic preparation significantly re-
Rayment N, et al. Clinical, microbiological, and immunological duces symptoms of irritable bowel syndrome in a double-blind
effects of fructo-oligosaccharide in patients with Crohn’s disease. placebo-controlled study. Int J Clin Exp Med. 2012;5:238–44.
Gut. 2006;55:348–55. 64. Dapoigny M, Piche T, Ducrotte P, Lunaud B, Cardot J-M,
48. Chow J, Lee SM, Shen Y, Khosravi A, Mazmanian SK. Host- Bernalier-Donadille A. Efficacy and safety profile of LCR35 com-
bacterial symbiosis in health and disease. Adv Immunol. plete freeze-dried culture in irritable bowel syndrome: a random-
2010;107:243–74. ized, double-blind study. World J Gastroenterol WJG. 2012;18:
49. Iyama S, Sato T, Tatsumi H, Hashimoto A, Tatekoshi A, Kamihara 2067–75.
Y, et al. Efficacy of enteral supplementation enriched with gluta- 65. Ishikawa H, Akedo I, Umesaki Y, Tanaka R, Imaoka A, Otani T.
mine, fiber, and oligosaccharide on mucosal injury following he- Randomized controlled trial of the effect of bifidobacteria-
matopoietic stem cell transplantation. Case Rep Oncol. 2014;7: fermented milk on ulcerative colitis. J Am Coll Nutr. 2003;22:
692–9. 56–63.
50. Szeluga DJ, Stuart RK, Brookmeyer R, Utermohlen V, Santos 66. Kruis W, Fric P, Pokrotnieks J, Lukás M, Fixa B, Kascák M, et al.
GW. Nutritional support of bone marrow transplant recipients: a Maintaining remission of ulcerative colitis with the probiotic
prospective, randomized clinical trial comparing total parenteral Escherichia coli Nissle 1917 is as effective as with standard
nutrition to an enteral feeding program. Cancer Res. 1987;47: mesalazine. Gut. 2004;53:1617–23.
3309–16. 67. Lin H-C, Hsu C-H, Chen H-L, Chung M-Y, Hsu J-F, Lien R, et al.
51. van der Meij BS, de Graaf P, Wierdsma NJ, Langius JAE, Janssen Oral probiotics prevent necrotizing enterocolitis in very low birth
JJWM, van Leeuwen PAM, et al. Nutritional support in patients weight preterm infants: a multicenter, randomized, controlled trial.
with GVHD of the digestive tract: state of the art. Bone Marrow Pediatrics. 2008;122:693–700.
Transplant. 2013;48:474–82. 68. Manzoni P, Mostert M, Leonessa ML, Priolo C, Farina D, Monetti
52. Lye AD, Hayslip JW. Immunonutrition: does it have a role in C, et al. Oral supplementation with Lactobacillus casei subspecies
improving recovery in patients receiving a stem cell transplant? rhamnosus prevents enteric colonization by Candida species in
Nutr Cancer. 2012;64:503–7. preterm neonates: a randomized study. Clin Infect Dis Off Publ
53. Albenberg LG, Wu GD. Diet and the intestinal microbiome: asso- Infect Dis Soc Am. 2006;42:1735–42.
ciations, functions, and implications for health and disease. 69. Fernández-Carrocera LA, Solis-Herrera A, Cabanillas-Ayón M,
Gastroenterology. 2014;146:1564–72. Gallardo-Sarmiento RB, García-Pérez CS, Montaño-Rodríguez
54. Goldenberg JZ, Ma SSY, Saxton JD, Martzen MR, Vandvik PO, R, et al. Double-blind, randomised clinical assay to evaluate the
Thorlund K, et al. Probiotics for the prevention of Clostridium efficacy of probiotics in preterm newborns weighing less than
28 Curr Hematol Malig Rep (2016) 11:19–28

1500 g in the prevention of necrotising enterocolitis. Arch Dis Clostridium difficile infection in an allogeneic stem cell transplant
Child Fetal Neonatal Ed. 2013;98:F5–9. patient. Transpl Infect Dis Off J Transplant Soc. 2012;14:E161–5.
70. Yang Y-X, He M, Hu G, Wei J, Pages P, Yang X-H, et al. Effect of 87. Tvede M, Rask-Madsen J. Bacteriotherapy for chronic relapsing
a fermented milk containing Bifidobacterium lactis DN-173010 Clostridium difficile diarrhoea in six patients. Lancet Lond Engl.
on Chinese constipated women. World J Gastroenterol WJG. 1989;1:1156–60.
2008;14:6237–43. 88. Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, Daigneault
71. Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, MC, et al. Stool substitute transplant therapy for the eradication of
Bazzocchi G, et al. Oral bacteriotherapy as maintenance treatment Clostridium difficile infection: BRePOOPulating^ the gut.
in patients with chronic pouchitis: a double-blind, placebo- Microbiome. 2013;1:3.
controlled trial. Gastroenterology. 2000;119:305–9. 89. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán
72. Gionchetti P, Rizzello F, Helwig U, Venturi A, Lammers KM, LG, Gratadoux J-J, et al. Faecalibacterium prausnitzii is an anti-
Brigidi P, et al. Prophylaxis of pouchitis onset with probiotic ther- inflammatory commensal bacterium identified by gut microbiota
apy: a double-blind, placebo-controlled trial. Gastroenterology. analysis of Crohn disease patients. Proc Natl Acad Sci U S A.
2003;124:1202–9. 2008;105:16731–6.
73. Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber S, Talbot 90. Miquel S, Martín R, Rossi O, Bermúdez-Humarán LG, Chatel JM,
IC, et al. Once daily high dose probiotic therapy (VSL#3) for Sokol H, et al. Faecalibacterium prausnitzii and human intestinal
maintaining remission in recurrent or refractory pouchitis. Gut. health. Curr Opin Microbiol. 2013;16:255–61.
2004;53:108–14. 91. Khan MT, van Dijl JM, Harmsen HJM. Antioxidants keep the
74. Van Gossum A, Dewit O, Louis E, de Hertogh G, Baert F, potentially probiotic but highly oxygen-sensitive human Gut bac-
Fontaine F, et al. Multicenter randomized-controlled clinical trial terium faecalibacterium prausnitzii alive at ambient Air. PLoS
of probiotics (Lactobacillus johnsonii, LA1) on early endoscopic ONE. 2014;9:e96097.
recurrence of Crohn’s disease after lleo-caecal resection. Inflamm 92. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa
Bowel Dis. 2007;13:135–42. H, et al. Treg induction by a rationally selected mixture of
75. Vitetta L, Briskey D, Alford H, Hall S, Coulson S. Probiotics, Clostridia strains from the human microbiota. Nature. 2013;500:
prebiotics and the gastrointestinal tract in health and disease. 232–6.
Inflammopharmacology. 2014;22:135–54. 93.•• Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne
76. Preidis GA, Versalovic J. Targeting the human microbiome with A, et al. Precision microbiome reconstitution restores bile acid
antibiotics, probiotics, and prebiotics: gastroenterology enters the mediated resistance to Clostridium difficile. Nature. 2015;517:
metagenomics era. Gastroenterology. 2009;136:2015–31. 205–8. Buffie et al. found that the intestinal organism
77. Ghouri YA, Richards DM, Rahimi EF, Krill JT, Jelinek KA, Clostridium scindens conferred resistance to Clostridium diffi-
DuPont AW. Systematic review of randomized controlled trials cile in patients undergoing allogeneic HSCT as well as in a
of probiotics, prebiotics, and synbiotics in inflammatory bowel murine model. They further demonstrated that Clostridium
disease. Clin Exp Gastroenterol. 2014;7:473–87. scindens resistance is dependent on the synthesis of secondary
78. Hart AL, Lammers K, Brigidi P, Vitali B, Rizzello F, Gionchetti P, bile salts, demonstrating a potential avenue for the develop-
et al. Modulation of human dendritic cell phenotype and function ment of precision microbiota-based therapeutics.
by probiotic bacteria. Gut. 2004;53:1602–9. 94. Aas J, Gessert CE, Bakken JS. Recurrent Clostridium difficile
79. Drekonja D, Reich J, Gezahegn S, Greer N, Shaukat A, colitis: case series involving 18 patients treated with donor stool
MacDonald R, et al. Fecal microbiota transplantation for clostrid- administered via a nasogastric tube. Clin Infect Dis Off Publ Infect
ium difficile infection: a systematic review. Ann Intern Med. Dis Soc Am. 2003;36:580–5.
2015;162:630–8. 95. Hamilton MJ, Weingarden AR, Sadowsky MJ, Khoruts A.
80. Ianiro G, Bibbò S, Scaldaferri F, Gasbarrini A, Cammarota G. Standardized frozen preparation for transplantation of fecal micro-
Fecal microbiota transplantation in inflammatory bowel disease: biota for recurrent Clostridium difficile infection. Am J
beyond the excitement. Medicine (Baltimore). 2014;93:e97. Gastroenterol. 2012;107:761–7.
81. Quera R, Espinoza R, Estay C, Rivera D. Bacteremia as an adverse 96. Zainah H, Silverman A. Fecal bacteriotherapy: a case report in an
event of fecal microbiota transplantation in a patient with Crohn’s immunosuppressed patient with ulcerative colitis and recurrent
disease and recurrent Clostridium difficile infection. J Crohns clostridium difficile infection. Case Rep Infect Dis. 2012;2012:
Colitis. 2014;8:252–3. 810943.
82. Schwartz M, Gluck M, Koon S. Norovirus gastroenteritis after 97. Duplessis CA, You D, Johnson M, Speziale A. Efficacious out-
fecal microbiota transplantation for treatment of clostridium diffi- come employing fecal bacteriotherapy in severe Crohn’s colitis
cile infection despite asymptomatic donors and lack of sick con- complicated by refractory Clostridium difficile infection.
tacts. Am J Gastroenterol. 2013;108:1367. Infection. 2012;40:469–72.
83. Gerbitz A, Schultz M, Wilke A, Linde H-J, Schölmerich J, 98. Pathak R, Enuh HA, Patel A, Wickremesinghe P. Treatment of
Andreesen R, et al. Probiotic effects on experimental graft- relapsing Clostridium difficile infection using fecal microbiota
versus-host disease: let them eat yogurt. Blood. 2004;103:4365–7. transplantation. Clin Exp Gastroenterol. 2013;7:1–6.
84. Mittal C, Miller N, Meighani A, Hart BR, John A, Ramesh M. 99. Friedman-Moraco RJ, Mehta AK, Lyon GM, Kraft CS. Fecal
Fecal microbiota transplant for recurrent Clostridium difficile in- microbiota transplantation for refractory Clostridium difficile co-
fection after peripheral autologous stem cell transplant for diffuse litis in solid organ transplant recipients. Am J Transplant Off J Am
large B-cell lymphoma. Bone Marrow Transplant. 2015;50:1010. Soc Transplant Am Soc Transpl Surg. 2014;14:477–80.
85. de Castro CG, Ganc AJ, Ganc RL, Petrolli MS, Hamerschlack N. 100. Trubiano JA, George A, Barnett J, Siwan M, Heriot A, Prince HM,
Fecal microbiota transplant after hematopoietic SCT: report of a et al. A different kind of Ballogeneic transplant^: successful fecal
successful case. Bone Marrow Transplant. 2015;50:145. microbiota transplant for recurrent and refractory Clostridium dif-
86. Neemann K, Eichele DD, Smith PW, Bociek R, Akhtari M, ficile infection in a patient with relapsed aggressive B-cell lym-
Freifeld A. Fecal microbiota transplantation for fulminant phoma. Leuk Lymphoma. 2015;56:512–4.

You might also like