Anda di halaman 1dari 11

Biomedicine & Pharmacotherapy 111 (2019) 537–547

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Probiotic supplements might not be universally-effective and safe: A review T


a b,⁎ a,⁎⁎
Damini Kothari , Seema Patel , Soo-Ki Kim
a
Department of Animal Science and Technology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
b
Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, 92182, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Last few decades have witnessed the unprecedented growth in the application of probiotics for promoting the
Probiotics general gut health as well as their inception as biotherapeutics to alleviate certain clinical disorders related to
Immunomodulation dysbiosis. While numerous studies have substantiated the health-restoring potentials for a restricted group of
Compromised gut microbial species, the marketed extrapolation of a similar probiotic label to a large number of partially char-
Sepsis
acterized microbial formulations seems biased. In particular, the individuals under neonatal stages and/or those
Allergy
Dietary supplements
with some clinical conditions including malignancies, leaky gut, diabetes mellitus, and post-organ transplant
Adverse effects convalescence likely fail to reap the benefits of probiotics. Further exacerbating the conditions, some probiotic
At-risk population strains might take advantage of the weak immunity in these vulnerable groups and turn into opportunistic
pathogens engendering life-threatening pneumonia, endocarditis, and sepsis. Moreover, the unregulated and
rampant use of probiotics potentially carry the risk of plasmid-mediated antibiotic resistance transfer to the gut
infectious pathogens. In this review, we discuss the safety perspectives of probiotics and their therapeutic in-
terventions in certain at-risk population groups. The embodied arguments and hypotheses certainly will shed
light on the fact why probiotic usage should be treated with caution.

1. Introduction metastasis [13], depression [14], anxiety [15], type 2 diabetes [16],
and obesity [17], among others.
Probiotics (Greek; Pro: promotion, Biotic: life) are generally defined Dysbiosis, an imbalance in the composition of the microbes fol-
as “live microorganisms which, when administered in adequate lowing antibiotic usage, has largely propelled the therapeutic applica-
amounts, confer health benefits on the host” [1]. Traditionally, a vast tion of probiotics [18]. The past two decades have pushed probiotics to
array of fermented foods like yogurt, kefir, kimchi, sauerkraut, tempeh, the highest echelon of dietary supplements following the implementa-
miso, and kombucha are part of regular diet across different cultures tion of 'Dietary Supplement Health and Education Act, 1994' by the US
and ethnicities, ranging from oriental to occidental [2,3], serving as the Food and Drug Administration (USFDA), allowing their marketing and
conventional source of probiotic strains. In recent years, probiotics are over-the-counter sales as health supplements under relatively flexible
touted for modulating the immune system, through the restoration of laws [19]. Since probiotics are considered more as food or health
gut homeostasis [4]. As per the current state of knowledge, probiotics supplements, they do not undergo rigorous clinical trials, unlike cura-
encompass both bacteria (Lactobacillus, Lactococcus, Leuconostoc, Ped- tive drugs. Especially, the probiotics-fortified foods including curd,
iococcus, Propionibacterium, Bifidobacterium, Bacillus, some Streptococcus, yogurt, ice cream, and cereals are being promoted without mentioning
Enterococcus, Escherichia coli) and yeast (Saccharomyces) genera [5]. the adequate parameters like formulation, efficacy, and consumer
Once delivered to the host, probiotics are expected to antagonize pa- safety. Altogether these loose criteria have resulted in skewed policies
thogens such as Enterococcus faecalis, Salmonella enterica subsp. enterica for governing probiotic formulations with unsubstantiated health
serotype Enteritidis, Listeria monocytogenes, Staphylococcus aureus and E. claims, across different continents and countries. Only recently, the
coli by immune, hormonal, and neuronal manipulations [6]. To the safety profiles of different probiotics as a function of different genera,
present date, only oral [7,8] and gut [9,10] ameliorative roles of pro- species, and strains, coupled with their relevance to diverse individuals
biotics have been validated for clinical uses. However, more studies are or at-risk population, have attracted enough attention [20–22]. Of note,
increasingly claiming the probiotics-mediated alleviation of disorders sepsis in cardio-surgical patient [23], anaphylactic allergic response in
related to the immune system [11], cardiovascular health [12], cancer sensitive individuals [24], systemic fungemia in elderly


Corresponding author at: Bioinformatics and Medical Informatics Research Center, San Diego State University, 5500 Campanile Dr, San Diego, CA, 92182, USA.
⁎⁎
Corresponding author.
E-mail addresses: seemabiotech83@gmail.com, Patel3@rohan.sdsu.edu (S. Patel), sookikim@konkuk.ac.kr (S.-K. Kim).

https://doi.org/10.1016/j.biopha.2018.12.104
Received 4 July 2018; Received in revised form 9 December 2018; Accepted 23 December 2018
0753-3322/ © 2018 Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

immunocompromised patients [25], and the risk of horizontal gene due to an impaired intestinal barrier, immunosuppression, or gut pre-
transfer (HGT) of antibiotic resistance to pathogenic microbes [26], maturity [31]. Bacterial translocation to sterile niches has been ob-
among many other issues due to probiotic usage have been reported. served following stroke, cirrhosis, and trauma, among a range of other
Diverse groups of individuals respond differently to foods, drugs, and conditions [32,33]. It has been speculated that the intestinal mucosa
dietary supplements, depending on their age, gender, host factors, and adhesion capacity [34] or mucolytic activity [35] of probiotic strains
co-morbidities [27,28]. Hence, the present review presents a compre- might carry out a role in their translocation. Probiotic translocation
hensive scrutiny of the proclaimed health effects of probiotic formula- might result in some clinical infections, including systemic and loca-
tions, underscoring their rationale of clinical administration in certain lized infections [34]. Predominantly, infections associated with specific
population groups. Hence, we maintain that it is much better to be probiotics include sepsis, endocarditis, localized, and opportunistic in-
pragmatic than harboring an unsubstantiated notion about the ther- fections via bacteremia (bacteria in the blood) or fungemia (fungi in the
apeutic applications of probiotics. Although many studies have cohe- blood).
sively endorsed the health-building roles of probiotics, they certainly do
not present the holistic and consistent picture. In this regard, besides 2.1.1. Sepsis
the strain-specific health effects, dosage, efficacy, and potency of mi- Sepsis is a clinical syndrome characterized by systematic in-
crobial species formulations [21], the immune response of the con- flammation and circulatory malfunctions following the pathogenic in-
sumers must be comprehensively studied. With new discoveries and fection like bacteremia or fungemia. It is one of the most common
better grasp on the mechanistic of immune system, it has now become causes of morbidity and mortality in critically ill patients [34] and pre-
clear that the host biochemical milieus and the host-microbe interac- term infants [36]. These populations are indeed highly prone to develop
tions are the decisive factors which determine the probiotics efficacy bacterial and fungal sepsis due to several factors including gut dys-
and safety for individual recipients. It is a matter of concern as most biosis, weak immunity, use of invasive procedures and devices, use of
people think that probiotics are perfectly safe and widely efficacious. broad-spectrum antibiotics and other drugs, possible bacterial translo-
This is certainly not the case, and it would be more useful to present a cation, the lack of nutrients, and nosocomial infections [34,37].
genuine picture, since it appears that adverse events are poorly reported Growing pieces of evidence support the beneficial role of probiotics in
in the randomized clinical trials (RCTs) of probiotics [29]. Keeping this critically ill and neonates (Table 1). Nevertheless, the ingestion of live
in mind, the present review documents the current knowledge on the microorganisms (probiotics) in rare instances might surplus the risk of
safety of probiotic administration in the at-risk population as well as sepsis as evidenced by several studies (Table 2), raising the safety
identifies the priorities or needs for future research. Especially, the 'risk- concern of routine probiotic use in such population. Recently, the lar-
benefit' ratios for the clinical administration of specific probiotic for- gest trial for 'Probiotics in Very Pre-term Infants (PiPS)' revealed no
mulations among the at-risk population, including pre-term infants and difference in the risk of NEC and sepsis between the probiotics and
those with critical illness, AIDS, organ transplantation, necrotizing control treated groups. The results of the PiPS trial also questioned the
enterocolitis (NEC), cancer, inflammatory bowel diseases, diarrhea, and published meta-analyses those supported the routine probiotic therapy
allergy have mainly been focused. in pre-term infants. Of note, the authors did not endorse any harms of
probiotic intervention for treating pre-term infants [38].
2. Safety, efficacy, and associated risks of probiotics in the at-risk Among lactobacilli, L. rhamnosus strains are mostly associated with
population human sepsis, suggesting their high potential to translocate [31]. Lac-
tobacillus sp. bacteremia is rarely fatal, albeit it may serve as a marker of
Probiotics are generally considered safe for a healthy population. more serious underlying disease [39]. A review including 89 patients
Several pieces of evidence of benefits with probiotic therapy were also with Lactobacillus bacteremia reported the mortality rate of 26% in 1-
reported in at-risk population groups (Table 1). Although the precise month and 48% in 1-year following infection onset [40]. Few cases
mechanisms of probiotics mediated clinical benefits are unclear, they were reported for Bifidobacterium-mediated sepsis and bacteremia
are conjectured to act through revamping gastrointestinal tract (GIT) (Table 2). One study reported sepsis associated with E. coli NISSLE 1917
microbiota, maintaining/protecting intestinal epithelium, reducing in- probiotic strain in low-birth weight infants [41]. Saccharomyces bou-
flammation, and modulating the immune profile. Previously, the joint lardii (a subtype of S. cerevisiae) based probiotic supplementary therapy
FAO/WHO [30] guidelines on probiotic evaluation reported that pro- is well-established to prevent antibiotic-associated diarrhea and treat
biotics may theoretically be linked to four specific types of side effects recurrent Clostridium difficile associated diarrhea [25]. However, several
in patients with underlying medical conditions: "(1) systemic infections; cases of fungemia and sepsis associated with this probiotic have also
(2) deleterious metabolic activities; (3) excessive immune stimulation been reported in patients with chronic diseases or critical illness
in susceptible individuals; and (4) gene transfer". The at-risk population (Table 2). These findings have led to the inference that probiotics might
groups are broadly characterized by the weakened immune system, gut not be harmless to immunocompromised patients/consumers, unlike
dysbiosis and/or impaired intestinal barrier, and therefore, it is im- immune-competent individuals. Nevertheless, there are scarce reports
portant to carefully assess the safety associated with deliberate ad- describing the probiotic-mediated sepsis in humans, the other side of
ministration of living microorganisms (i.e., probiotics). Moreover, the the coin simply emphasizes the safe clinical administration or dietary
availability of limited or mixed evidences of benefits from several consumption of probiotics in humans. In other words, the problem can
probiotic intervention studies owing to the variability in a target po- be with the host and not with the probiotics strain. If bacteria are
pulation, probiotics formulations administered, clinical and statistical capable of restoring homeostasis, yet host gut is porous and inner
heterogeneity, study limitations, and small sample sizes further ne- mucus layer-devoid, bacteria will migrate to sterile regions. Some in-
cessitates the comprehensive safety evaluation for at-risk population dividuals have leaky gut where mucosal integrity is invasion-prone, and
groups. Below, we discuss these theoretical threats for clinical probiotic breaching of which can cause infections, sepsis, and/or fungemia [5].
administration in different at-risk population groups, though they are The evidences point toward the relation between low leukocyte count,
not separate but inter-linked. Also, the risks associated with the un- neutropenia, high inflammatory markers, and traversal of gut-dwelling
restricted dietary intake of probiotics have been cataloged in Table 2. probiotic strains into bloodstream. It is the reason most cases with
sepsis are immunocompromised patients, cancer patients, and pre-term
2.1. Infections caused by probiotic administration infants.

Bacterial translocation typically refers to the passage of viable in- 2.1.2. Infective endocarditis
digenous bacteria from the GIT to extraintestinal sites (bloodstream), Another noteworthy point highlighting the potentially adverse

538
D. Kothari et al.

Table 1
Efficacy studies of probiotic therapy in risk population.
Disease Study No. of RCTs and patients Population Outcome References

NEC Systematic review and 24 RCTs; 5529 patients Infants < 37 weeks gestational age or < 2500 g Effective, safe and well tolerated [92]
meta-analysis birth weight, or both
NEC Systematic review and 20 RCTs involving 5982 patients Very low birth weight (< 1500 g) & pre-term (< Reduced risk of NEC and mortality [93]
meta-analysis 37 weeks gestational age) infants
NEC Systematic review and 44 studies (30 RCTs and 14 Pre-term infants Prevented severe NEC and late-onset sepsis, and reduced mortality [94]
meta-analysis observational studies)
VAP Systematic review and 9 RCTs; 1119 patients Critically ill adults Reduced incidence of ICU-acquired pneumonia and shortened ICU-stay; safe [95]
meta-analysis with no adverse effects
VAP Systematic review and 8 RCTs; 1083 patients Critically ill adults Reduced VAP cases and shortened ICU-stay but no change in mortality rate; [96]
meta-analysis conferred benefits but insufficient evidence of safety and efficacy
VAP RCT 150 patients Critically ill children (≤ 12 years) Effective and safe to lower the incidence of VAP, shortened hospital and ICU- [97]
stay;
VAP RCT 235 patients Critically ill adults Effective and safe for preventing VAP and the acquisition of potentially [98]
pathogenic microorganism’s colonization.

539
VAP Systematic review and 30 RCTs; 2972 patients Critically ill adults Reduced incidence of infections including VAP [99]
meta-analysis
IBS RCT 84 patients 20-70 years Improved quality of life; no adverse effects [100]
IBS Systematic review and 15 RCTs; 1793 patients Adults and children Conferred benefits but insufficient evidence of safety and efficacy [101]
meta-analysis
IBS symptoms RCT 60 patients Colorectal cancer (> 20 years) Improved bowel symptoms and quality of life [102]
Diarrhea Systematic review and 17 RCTs & Case reports; 1530 Cancer Reduced chemotherapy-related diarrhea but significant safety concerns [103]
meta-analysis patients
Diarrhea Systematic review 39 RCTs; 9402 patients HIV patients Effective and no adverse effects [104]
Inflammation Clinical trial 20 patients HIV patients (27–72 years) Improved gut immunity and no adverse effects [105]
Postoperative infection RCT 67 patients Liver transplantation Reduced infection and shortened duration of antibiotic use; no adverse effects [106]
Postoperative infection Systematic review and 4 RCTs; 246 patients Liver transplantation (mean age 53 years) Reduced postoperative infections, shortened hospital or ICU-stay, duration of [107]
meta-analysis antibiotic use and no probiotic associated adverse effects
Postoperative infection RCT 55 patients Liver transplantation (≥18 years) Effective in postoperative infections and improved early biochemical [108]
parameters of allograft function; no effect on mortality

RCT: Randomized control trial; NEC: Necrotizing enterocolitis; VAP: Ventilator-associated pneumonia; IBS: Irritable bowel syndrome; HIV: Human immunodeficiency virus.
Biomedicine & Pharmacotherapy 111 (2019) 537–547
D. Kothari et al.

Table 2
Risks associated with rampant dietary intake of probiotics.
Category Microorganisms Population Mechanisms References

Transmissible antibiotic resistance Lactobacillus, Pediococcus and Leuconostoc sp. – Vancomycin resistance → Staphylococcus aureus [109]
genes Lactobacillus sp. – Vancomycin resistance → Enterococcus faecalis [110]
Lactobacillus sp. – Broad spectrum antibiotic resistance (vancomycin, [26,111]
streptomycin,
gentamicin, aztreonam, and ciprofloxacin) → Staphylococcus sp.
Systemic infection Lactobacillus rhamnosus GG Pre-term infant with short gut syndrome Lactobacillus bacteremia [112]
L. rhamnosus GG Critically ill children with antibiotic related diarrhea Sepsis [113]
L. rhamnosus GG 11-month old infant with short gut syndrome Lactobacillus bacteremia [114]
L. rhamnosus GG 17-year-old boy with ulcerative colitis Lactobacillus bacteremia [115]
Lactobacillus sp. 58-year-old immunocompetent with mechanical ventilation Lactobacillus bacteremia and sepsis [116]
Three strains of L. rhamnosus 24-year-old female cardiosurgical patient Probiotic sepsis [23]
Bifidobacterium longum 74-year-old man with polymetastatic prostatic adenocarcinoma Bifidobacterium bacteremia [117]
B. longum subspecies infantis Pre-term infants Bifidobacterium bacteremia [118]
B. longum Low birth-weight infants Bifidobacterium bacteremia [119]
B. breve 2-year-old boy with Philadelphia chromosome-positive acute B- Bifidobacterium sepsis [120]
cell lymphoblastic leukemia
E. coli NISSLE strain 1917 Pre-term infants Severe sepsis [41]
Saccharomyces cerevisiae 48-year-old diabetic with multiple co-morbidities Multiple organ failure and septic shock in association with toxic [121]
megacolon and probiotic fungemia

540
S. cerevisiae var. boulardii Immunocompromised 73-year-old patient on chemotherapy Fungemia [25]
S. cerevisiae var. boulardii 8-year-old boy with respiratory distress (Intensive care unit Fungemia [122]
patient)
S. cerevisiae var. boulardii Premature neonate receiving nutrition enterally Fungal septicemia [123]
S. cerevisiae var. boulardii Critically ill patients Fungemia [82]
L. plantarum 30-year-old male with rheumatic valve disease Endocarditis [124]
L. casei 53-year-old immunocompetent patient Endocarditis [125]
L. jensenii 47-year-old immunocompetent patient Endocarditis [126]
L. paracasei 77-year-old male patient with prostate cancer Endocarditis [127]
L. acidophilus 48-year-old male with heart disease and dental manipulations Endocarditis [45]
L. rhamnosus > 65-year-old patient with hemorrhagic telangiectasia (HHT) Endocarditis [128]
Localized infection L. casei 60-year-old with renal transplant patient Intra-abdominal abscess [129]
L. paracasei 65-year-old diabetic patient Bacteremia and liver abscess [47]
L. rhamnosus 11-month-old female with trisomy 21 with respiratory viral Probiotic associated pneumonia [48]
infection
Metabolic disturbances Lactobacillus spp. – Unregulated 'bile salt hydrolase (BSH)' activity [64]
L. lactis – Biogenic amine production [63]
Allergic response L. rhamnosus GG Children (up to 4 years) Allergic rhinitis and more serious asthma [68]
L. acidophilus LAVRI-A1 Infants (6 and 12 months) Atopic sensitization [72]
L. rhamnosus GG Pregnant (4-6 weeks before expected delivery) and breastfeeding Wheezing bronchitis; atopic sensitization [130]
mothers (up to 2 years)
Bowel ischemia L. acidophilus, L. casei, L. salivarius, L. lactis, B. Critically ill patients Increased local oxygen demand [55]
bifidum, and B. lactis (acute pancreatitis)
Biomedicine & Pharmacotherapy 111 (2019) 537–547
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

clinical implications of probiotics is the risk of infective endocarditis trial reported increased mortality (16% vs 6%) due to bowel ischemia
(IE). IE is an inflammation of the endocardium, that has been reported (9 vs 0) in patients group subjected to enteral probiotic administration
as a major health concern leading to life-threatening cardiogenic ail- while severe acute pancreatitis (SAD) treatment [55]. The mechanisms
ments, dysrhythmias, and septic shocks [42]. Endocarditis due to Lac- of bowel ischemia by probiotics were speculated to be the increased
tobacillus infection is rare but considerably a serious clinical state in local oxygen demand following the administration of 10 billion pro-
patients with high mortality rate averaging 30% [43]. The high mor- biotic bacteria per day, which might potentially aggravate the critically
tality was attributed to the potentially pathogenic characteristics of reduced blood flow engendering the excessive local inflammation
Lactobacilli, which include the ability to affect platelet aggregation in owing to the heightened bacterial load in small bowel wall. The dismal
humans, form complexes with blood components including fibronectin, results of the PROPATRIA trial discouraged many researchers through
fibrinogen, collagen types I and V, as well as the overwhelming pro- raising the concerns about the clinical implications of probiotics for
duction of enzymes including glycosidases and proteases at the infec- treating the critically ill patients with pancreatitis [55]. In another
tion sites. Several Lactobacillus spp. have been reported to cause IE, such retrospective study, no positive or negative impact of probiotic treat-
as L. rhamnosus, L. casei, L. acidophilus, L. jensenii, L. plantarum and L. ment was demonstrated in patients with predicted SAD without initial
paracasei (Table 2). The major risk factors linked to Lactobacillus organ failure [56].
mediated endocarditis include impaired immunity, structural heart
disease, recent surgery, prolonged antibiotic therapy, dental infections 2.2. Metabolic disturbances
or procedures, and severe comorbidities [44]. Recently, probiotics
containing Lactobacillus spp. have been reported as a new risk factor for Although the US Food and Drug Administration (FDA) designates
endocarditis [45]. Therefore, any probiotic formulation and their clin- probiotics as 'Generally Recognized as Safe' (GRAS) (http://www.fda.
ical administration should be carefully examined in the aforementioned gov/food/Ingredientspackaging Labeling/GRAS/), surprisingly very
at-risk populations. few systematic reports are available which signify their consumer safety
in vulnerable individuals with certain clinical conditions. For example,
2.1.3. Localized infection metabolism upsetting effects on the GIT is another concern against the
Apart from bloodstream infections, few cases of local infections clinical probiotic interventions. Any perturbation or refashioning of the
including liver abscess (intra-abdominal abscess) and pulmonary in- metabolic pathways can cause obesity, type 2 diabetes, or other meta-
fections (Lactobacillus pneumonia) were also reported following the bolic syndromes. This suspicion has arisen in the wake of the discovery
clinical administration of certain probiotic formulations. The liver ab- that gut microbiota composition affects nutrient acquisition, energy
scess is a collection of pus or infected fluid surrounded by inflamed harvest, and a myriad of host metabolic pathways [57]. Food poisoning
tissue inside the liver. Most of the probiotic-related liver abscesses are is a multi-factorial health concern, of which microbial biogenic amine
not confirmed by strain identification [46] except for Lactobacillus (BA) is one factor. Though lactic acid bacteria used in food fermenta-
paracasei diagnosed from a 65-year-old diabetic patient [47]. The po- tion are considered as GRAS, some of them produce BAs (e.g., cada-
tential risk factors linked to lactobacilli liver abscess are advanced age, verine, histamine, tyramine, agmatine, and putrescine) liberating into
diabetes mellitus, liver transplantation, underlying hepatobiliary dis- the fermented dairy products, pickles [58], and beverages [59]. BAs are
ease, and the history of malignancy [46]. Although probiotic-associated neurotransmitters and immune mediators. BAs can increase or decrease
pneumonia is very rare with only one case of Lactobacillus mediated blood flow to the central nervous system and might trigger headaches
pneumonia was reported till date due to probiotic supplementation in people sensitive to these substances [60]. Some probiotic strains such
[48]. This report speculated that being immunocompromised (espe- as Lactobacillus buchneri, Lactobacillus helveticus, Lactobacillus hilgardii,
cially having AIDS) to be the foremost primary risk factor for Lactoba- and Streptococcus thermophilus can produce histamine in dairy products
cillus pneumonia. [61,62]. The excess histamine can be absorbed into the bloodstream via
the lining of the GIT resulting in allergic reactions. Previously, a study
2.1.4. Opportunistic infection had reported that L. lactis in fermented foods might cause a significant
The opportunistic pathogens are those which infect host with accumulation of cadaverine (a toxic diamine compound) over a period
weakened/impaired immunity [49]. The stark distinction between safe of consumption [63].
probiotics and potentially pathogenic genera might be getting blurry, The bile salt hydrolase (BSH) activity of certain probiotic strains in
with emerging facts. A comparative genomic study of Clostridium bu- the small bowel is known to lower blood cholesterol levels through
tyricum DKU-01 strain isolated from infant feces displayed considerable hydrolyzing the bile salts; however, unregulated expression of this en-
metabolic similarity with Bifidobacterium and Streptococcus genera on zyme is suspected to affect host metabolism by impairing lipid meta-
fructo-oligosaccharide (FOS) utilization [50]. However, some strains of bolism, promoting gall stone depositions, and/or obesity [64,65]. In
Clostridium species have earlier been linked to infant's fatal botulism recent years, various Lactobacillus species such as L. acidophilus, L. fer-
and NEC [51]. The risk of Lactobacilli and Bifidobacteria-associated mentum, L. ingluviei, and L. gasseri have been reported to affect weight
opportunistic infections is highly unlikely, however, Streptococcus and gain in host species, including humans and livestock animals [66]. Al-
Enterococcus that are also touted as probiotics represent opportunistic together, the probiotic effect in body weight was species- and strain-
pathogens [52]. specific as well as host-specific, and hence, the tendency of these pro-
Till date, no evidence of hemolytic activity has been reported for ducts toward influencing the obesity in humans needs to be examined
lactobacilli and bifidobacteria. However, Enterococcus faecium, a normal [66,67]. Hence, we persuasively argue that any probiotic formulation
inhabitant of human gut was reported to produce a hemolysin [53]. It cannot be generalized under a single umbrella to have positive health
has been shown that β-hemolytic strains of Enterococcus increased five effects as it influences host metabolism differentially commensurate to
times the chances of death by bacteremia when compared with the the individual's physiological state.
patients with bacteremia caused by non-β-hemolytic strains [54]. Since
gut microbes are prone to HGT, the acquisition of hemolysin producing 2.3. Allergic response
genes is not an unlikely concern. Hence, the opportunistic infection
risks of probiotics ought not to be dismissed. Atopic dermatitis (AD) is a chronic inflammatory skin disease with
relapsing pruritic eczema, involving allergic and immunological ab-
2.1.5. Ischemia normalities. Previously, probiotic supplementation has been success-
Ischemia refers to the inadequate blood supply to tissues or organs fully reported for the prevention and treatment of AD in children [68]
and is an exceptional etiology of acute pancreatitis. The PROPATRIA and adults [69]; however, no empirical evidence was found which

541
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

supports the beneficial role of probiotics in infants (< 1 year old) [70]. depression, and pre-term birth, among many other pathologies [6,83].
In several other studies, the treatment is linked to many adverse effects So, the idea that replenishing the friendly bacteria will bring back
including a higher risk for allergic rhinitis and more serious asthma homeostasis is quite convincing. However, several arguments can be
[68]; increased risk of atopic sensitization [71,72]; and increased made regarding the risks that probiotics pose, of which some of the
wheezing bronchitis [71]. Moreover, Sanders et al. [21] hypothesized robust have been discussed below.
that mere administration of one or a few single probiotic strains in high Considering the probiotics to be a consortium of exogenous mi-
numbers does not warrant a natural approach to gut colonization, but crobes, different mechanisms of interactions and health effects for dif-
raises the possibility of long-term side effects, especially in atopic po- ferent genera, species, and strain within individual hosts are expected.
pulation. Moreover, the variations in probiotics formulations, dosage, and ad-
ministration modes may greatly affect the outcomes among the het-
2.4. Immunological effects erogeneous recipients [84]. Therefore, an appropriate strain and for-
mulation should be considered toward a particular health condition.
The effects of probiotics on human immune function are still con- In general, the potential probiotic strains are being screened from
troversial. Augmentation of natural killer activity, T cell functions, and all sorts of niches including soil, feces, and fermented foods making
cytokine production are some of the plausible mechanisms underlying them remarkably distinct from those residing endogenously in the
the immune regulatory activities of probiotics [73]. The elicited cyto- human gut. Once ingested, the radically different gut micro-environ-
kine production via probiotic supplementation might cause inordinate ment and immune factors might exert selection pressure on probiotic
immunological effects, leading to auto-immune phenomena or certain strains to develop potential virulence factors including adhesins [85],
inflammations. Therefore, the effect of probiotic administration during biogenic amines (histamine, putrescine, cadaverine, tyramine, trypta-
auto-immune disease development needs attention. However, this risk mine, phenylethylamine, spermine and spermidine), certain toxins
is purely theoretical and has not been reported in any human popula- (hemolysins), and harmful enzymes (glycosidases, proteases, muci-
tion [74,75]. nases, and gelatinases) [35,86]. As reported previously, bacterial gen-
omes readily undergo genome reshuffling to adapt to the changing
2.5. Horizontally-transferred antibiotic resistance environment [87]. This genomic change might lead to the emergence of
new drug-resistant strains.
One argument against the long-term intake of probiotics entails the Notably, the probiotic strain suspensions are frequently mixed with
risk of antibiotic selection pressure. Most strains of Lactobacillus, sodium alginate and extruded on to calcium chloride solution. The
Lactococcus, and Bifidobacteria used as probiotics are generally benign, calcium alginate beads are then packaged to be used as probiotics [88].
without any pathogenic risk, yet untoward exceptions like horizontal What if these capsules provoke the immune system in the host? Or what
transfer of antibiotic resistance genes to the opportunistic pathogens if the carbohydrates trigger the activity of glycoside hydrolases?
sharing the gut microbiome might have serious clinical implications Moreover, the risk associated with the probiotic intake in individuals
[76]. The unrestrained consumption of non-specific probiotics as health with poor gut integrity must be thoroughly examined. Although the
supplements coupled with the usage of broad-spectrum antibiotics ex- reports highlighting the benefits of probiotics far outnumber those
erts selective pressure on gut microbiome, potentially generating multi- substantiating their health risk, the propensity of at-risk population for
resistant bacteria or yeasts, exacerbating the unlikely events of HGT the associated health risk should also be considered. While strain-spe-
among gut commensals and pathogens (Table 2). The GIT is widely cific efficacy of probiotics has gained adequate empirical support, the
considered as the reservoir and hotspot, engendering HGT among mi- plasticity of strains towards lethal gene intake, crossing gut barrier,
crobial communities, as it houses trillions of microbes in close proxi- causing shock, etc. needs greater attention. Above all, the host factors
mity [77]. Hence, under certain conditions the antibiotic-resistant leading to probable vulnerability toward probiotics supplementation
probiotics strains might transfer antibiotic resistant genes (ABR) either ought to be probed using RCTs with emphasis on specific safety con-
directly or indirectly to a commensal microflora inhabiting the common cerns. We briefly summarized the various criteria that should be used
gut environ. Moreover, human gut microbiota is also suggested to be a for the safety evaluation of a probiotic with aided health benefits in
trove of ABR genes, transferable between the transient bacteria (in- Table 3.
cluding probiotics and pathogens) and the normal residents. Notably, Probiotics are assemblage of live bacteria for consumption, hence,
the tetracycline resistance genes have been well-documented to be their manufacturing, labeling, and distribution should be strictly
shared by gut microbes [78]. For instance, Lactococcus lactis has been regulated [75]. FDA also encourages health care providers who pre-
reported to transfer these ABR genes to Enterococcus faecalis [79], while scribe dietary supplements containing live bacteria or yeast as drugs
a number of such studies (in vitro and animal model) have been re- (for treating or preventing underlying medical conditions) to submit an
viewed which highlighted the transfer of ABR genes between lactoba- 'Investigational New Drug Application' to FDA for review [89]. We
cilli and lactococci as well as the gut microbial commensals and tran- argue that FDA/WHO should exercise more prescriptive guidelines with
sient pathogens [80,81]. Although studies supporting the safety of more stringent criteria, especially toward their clinical applications. In
probiotics strains far outnumber their risks, it clearly does not reflect this way, at-risk populations such as immunocompromised [25], neo-
the holistic picture. nates [37], critically ill [34] and those undergoing post-surgical treat-
The inherent risks of probiotics administration in vulnerable groups ments [23] might be spared of the unknown risks of the unregulated
have been illustrated in Fig. 1. clinical interventions of probiotics. The sporadic adverse effects of
strain-specific probiotics highlight the lack of explicit legislation in the
3. Safety considerations and future directions manufacture and distribution of probiotics for healthcare applications.
In the hype to report its beneficial effects, the negative traits have been
Different nations have maintained different policies for the safety of altogether skipped. For example, the ingestion of probiotic bacterium E.
dietary supplements and pharmaceuticals. European Food Safety coli NISSLE 1917 might offer relief in chronic inflammatory bowel
Authority (EFSA) considers all common species of probiotics as safe for diseases and colitis; however, this strain can also induce genotoxicity by
the general population. Irrespective of policies, there has always been a peptide-polyketide elaboration and modulation of cytokine profiles in
risk associated with the probiotics use among the vulnerable popula- the host [90]. This report indicated that an effective probiotic might
tions [82]. also have concomitant side effects like any other medication. Such
It is easy to comprehend why probiotics grabbed such a significant prudent reports are warranted as it is increasingly becoming clear that
place in healthcare. Gut microbiota controls heart diseases, obesity, ‘the host micro-environment’ largely determines the salutogenic or

542
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

Fig. 1. Schematic representation of the potential risks and their consequences associated with the clinical administration of probiotics in the at-risk population
groups. (A) Probiotic translocation by the violation of gut integrity: in leaky or inflamed or pre-mature gut, probiotics (Saccharomyces cerevisiae, Lactobacillus,
Bifidobacterium) might traverse the intestinal mucosa and enter into bloodstream or vital organs causing systemic or localized infections; (B) Virulence factors and
harmful metabolites of probiotics might lead to opportunistic infections and metabolic disturbances, respectively; (C) Horizontal transfer of antibiotic resistance
genes from probiotics to pathogenic gut bacteria and vice-versa might lead to the development of antibiotic resistant pathogens; and (D) Exaggerated immune
response through elicited cytokine production might lead to autoimmune disease or inflammation.

pathogenic behavior of microbes. In the absence of immune surveil- [43,91]. Gut microflora members contend with human immune pres-
lance, even a pathogen can shut down its virulence machinery, and in sure yet are not pathogenic owing to a million-years-long mutualistic
the absence of immune patrolling, even a benign bacterium can become co-evolution. Herein, we conjecture that the exogenously introduced
a potential pathogen. It has been observed that under in vitro conditions microbial species (probiotics) in at-risk population groups might dis-
in the culture medium, the potential pathogens like Streptococcus, En- rupt the endogenous native gut microflora pertaining to a variety of risk
terococcus, E. coli, S.boulardii tend to lose their virulence appendages factors. In a broader context, it can simply fail to survive and poop-out,

Table 3
Criteria for the safety evaluation of probiotics.
Safety parameters Methodology/techniques References

Strain identification DNA-based technology including genome sequencing and genetic based tools [5]
Transmissible antibiotic resistant genes Antibiotic resistant profiling, identification of antibiotic resistant genes via comparative proteomics and functional [26]
genomics; in vitro studies that mimic natural gut environment in the presence or absence of antibiotics
Toxicological studies In vitro and in vivo studies in relation to biogenic amine production, hemolysis and harmful enzyme secretion. [35]
Efficacy studies Functional characterization via in vitro and in vivo (animal and human) studies including clinical trials [86]
Target population Immune compromised (AIDS or on immunosuppressant therapy), infants (pre-term and low-birth weight), allergic, [21]
critically ill patients (bowel syndrome, central venous catheters, cardiac valve disease, severe acute pancreatitis, cancer
among many others), and pregnant women
Product formulation and labelling Composition, product purity, presence or absence of contaminants (biological or non-biological), doses information, health [131]
claims (general and specific), and adverse effects
Others Strain by-strain basis evaluation, long-term and high-dosage effects [22]

543
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

Fig. 2. Archetypal probiotics genera with potential virulence factors.

restore health or might act as a pathogen under certain clinical states in References
the host. We sketched the various probiotics candidates and their likely
associated virulence factors in Fig. 2. [1] J. Gerritsen, H. Smidt, G.T. Rijkers, W.M. de Vos, Intestinal microbiota in human
Nevertheless, the therapeutic potentials of probiotics remain pro- health and disease: the impact of probiotics, Genes Nutr. 6 (2011) 209–240,
https://doi.org/10.1007/s12263-011-0229-7.
mising if appropriate regulatory procedures with informative labeling [2] M.R. Swain, M. Anandharaj, R.C. Ray, R. Parveen Rani, Fermented fruits and
including warnings/precautions linked to the specific medications are vegetables of Asia: a potential source of probiotics, Biotechnol. Res. Int. (2014)
included. (2014) 250424, https://doi.org/10.1155/2014/250424.
[3] S.N. Chilton, J.P. Burton, G. Reid, Inclusion of fermented foods in food guides
around the world, Nutrients. 7 (2015) 390–404, https://doi.org/10.3390/
nu7010390.
4. Conclusions [4] L. Galland, The gut microbiome and the brain, J. Med. Food 17 (2014) 1261–1272,
https://doi.org/10.1089/jmf.2014.7000.
The term ‘probiotics’ has been marketed in such a manner that ev- [5] S. Fijan, Microorganisms with claimed probiotic properties: an overview of recent
literature, Int. J. Environ. Res. Public Health 11 (2014) 4745–4767, https://doi.
eryone has a presumption that the consumption of these products is org/10.3390/ijerph110504745.
always beneficial, almost a panacea, with zero risks. However, the ac- [6] M. Carabotti, A. Scirocco, M.A. Maselli, C. Severi, The gut-brain axis: interactions
tual scenario seems different from this idea, and hence both the facets between enteric microbiota, central and enteric nervous systems, Ann.
Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 28 (2015) 203–209 (Accessed
of the picture must be considered via risk versus benefit evaluation, to June 9 2015), http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=
better harness the benefits of probiotics. In this review, we highlight the 4367209&tool=pmcentrez&rendertype=abstract.
necessities for demarcating the regulation of probiotics for the general [7] A. Haukioja, Probiotics and oral health, Eur. J. Dent. 4 (2010) 348–355 (Accessed
September 11 2015), http://www.pubmedcentral.nih.gov/articlerender.fcgi?
dietary and toward clinical interventions. In our discussion, we em- artid=2897872&tool=pmcentrez&rendertype=abstract.
phasized that the probiotics with health claims to treat/prevent specific [8] A. Kandwal, A. Chatterjee, H. Bhattacharya, Probiotics in periodontal health and
ailments should be regulated and marketed as “drug” rather than disease, J. Indian Soc. Periodontol. 15 (2011) 23, https://doi.org/10.4103/0972-
124X.82260.
“dietary supplements” and thereby must conform to more rigorous re-
[9] M.C. Collado, E. Isolauri, S. Salminen, Y. Sanz, The impact of probiotic on gut
quirements. The regulatory bodies should be stricter in the market health, Curr. Drug Metab. 10 (2009) 68–78 (Accessed October 12 2015), http://
launch of a probiotic product either as a food supplement or a bio- www.ncbi.nlm.nih.gov/pubmed/19149514.
therapeutic agent. Delving deeper might reveal that like all GRAS mi- [10] G. Aragon, D.B. Graham, M. Borum, D.B. Doman, Probiotic therapy for irritable
bowel syndrome, Gastroenterol. Hepatol. (N.Y.) 6 (2010) 39–44 (Accessed July 31
crobes are not probiotics, all probiotics might not be safe for all in- 2015), http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2886445&
dividuals. Especially, the host factors can be variable, and for a tool=pmcentrez&rendertype=abstract.
particular milieu under certain clinical conditions, probiotics might be [11] S. Patel, R. Shukla, A. Goyal, Probiotics in valorization of innate immunity across
various animal models, J. Funct. Foods 14 (2015) 549–561, https://doi.org/10.
immunogenic and/or pathogenic. Hence, this review highlights the fact 1016/j.jff.2015.02.022.
that probiotics are no 'magic bullet', and their administration under the [12] R.M. Thushara, S. Gangadaran, Z. Solati, M.H. Moghadasian, Cardiovascular
current pretense of microbiota restorative and immunomodulator, benefits of probiotics: a review of experimental and clinical studies, Food Funct. 7
(2016) 632–642, https://doi.org/10.1039/c5fo01190f.
might not be universally safe and effective. [13] Z. Nouri, F. Karami, N. Neyazi, M.H. Modarressi, R. Karimi, M.R. Khorramizadeh,
B. Taheri, E. Motevaseli, Dual anti-metastatic and anti-proliferative activity as-
sessment of two probiotics on HeLa and HT-29 cell lines, Cell J. 18 (2016)
Conflict of interest 127–134.
[14] R. Huang, K. Wang, J. Hu, Effect of Probiotics on Depression: a systematic review
and meta-analysis of randomized controlled trials, Nutrients 8 (2016), https://doi.
There is no conflict of interest in submission of the manuscript. org/10.3390/nu8080483.
[15] H. Yang, X. Zhao, S. Tang, H. Huang, X. Zhao, Z. Ning, X. Fu, C. Zhang, Probiotics
reduce psychological stress in patients before laryngeal cancer surgery, Asia. J.
Acknowledgement Clin. Oncol. 12 (2016) e92–e96, https://doi.org/10.1111/ajco.12120.
[16] M.A. Kasińska, J. Drzewoski, Effectiveness of probiotics in type 2 diabetes: a meta-
analysis, Pol. Arch. Med. Wewn. 125 (2015) 803–813, https://doi.org/10.20452/
This work is supported by Korea Institute of Planning and pamw.3156.
Evaluation for Technology in Food, Agriculture, Forestry (IPET) [17] A. Prados-Bo, S. Gómez-Martínez, E. Nova, A. Marcos, Role of probiotics in obesity
through Agri-Bio industry Technology Development Program, funded management, Nutr. Hosp. 31 (Suppl. 1) (2015) 10–18, https://doi.org/10.3305/
nh.2015.31.sup1.8702.
by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (118051- [18] P.E. Wischmeyer, D. McDonald, R. Knight, Role of the microbiome, probiotics, and
03).

544
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

“dysbiosis therapy” in critical illness, Curr. Opin. Crit. Care 22 (2016) 347–353, Lactobacillus: a retrospective review of over 200 cases, Eur. J. Clin. Microbiol.
https://doi.org/10.1097/MCC.0000000000000321. Infect. Dis. 24 (2005) 31–40, https://doi.org/10.1007/s10096-004-1253-y.
[19] U.S. Food and Drug Administration, Dietary Supplement Health and Education Act [44] S. Yagi, M. Akaike, M. Fujimura, T. Ise, S. Yoshida, Y. Sumitomo, Y. Ikeda,
of 1994, US Dep. Heal. Hum. Serv., 1994. T. Iwase, K.I. Aihara, H. Azuma, A. Kurushima, Y. Ichikawa, T. Kitagawa,
[20] C. Hammerman, A. Bin-Nun, M. Kaplan, Safety of probiotics: comparison of two T. Kimura, T. Nishiuchi, T. Matsumoto, Infective endocarditis caused by
popular strains, BMJ. 333 (2006) 1006–1008, https://doi.org/10.1136/bmj. Lactobacillus, Intern. Med. 47 (2008) 1113–1116, https://doi.org/10.2169/
39010.630799.BE. internalmedicine.47.0744.
[21] M.E. Sanders, L.M.A. Akkermans, D. Haller, C. Hammerman, J. Heimbach, [45] C.O. Encarnacion, A.M. Loranger, A.G. Bharatkumar, G.H. Almassi, Bacterial en-
G. Hörmannsperger, G. Huys, D.D. Levy, F. Lutgendorff, D. Mack, P. Phothirath, docarditis caused by Lactobacillus acidophilus leading to rupture of sinus of
G. Solano-Aguilar, E. Vaughan, Safety assessment of probiotics for human use, Gut Valsalva Aneurysm, Tex. Heart Inst. J. 43 (2016) 161–164, https://doi.org/10.
Microbes 1 (2010) 164–185, https://doi.org/10.4161/gmic.1.3.12127. 14503/THIJ-15-5121.
[22] M. van den Nieuwboer, E. Claassen, L. Morelli, F. Guarner, R.J. Brummer, [46] M. Sherid, S. Samo, S. Sulaiman, H. Husein, H. Sifuentes, S. Sridhar, Liver abscess
Probiotic and synbiotic safety in infants under two years of age, Benef. Microbes 5 and bacteremia caused by Lactobacillus: Role of probiotics? Case report and review
(2014) 45–60, https://doi.org/10.3920/BM2013.0046. of the literature, BMC Gastroenterol. 16 (2016), https://doi.org/10.1186/s12876-
[23] P. Kochan, A. Chmielarczyk, L. Szymaniak, M. Brykczynski, K. Galant, A. Zych, 016-0552-y.
K. Pakosz, S. Giedrys-Kalemba, E. Lenouvel, P.B. Heczko, Lactobacillus rhamnosus [47] A. Pararajasingam, J. Uwagwu, Lactobacillus: the not so friendly bacteria, BMJ
administration causes sepsis in a cardiosurgical patient-is the time right to revise Case Rep. (2017) (2017), https://doi.org/10.1136/bcr-2016-218423.
probiotic safety guidelines? Clin. Microbiol. Infect. 17 (2011) 1589–1592, https:// [48] C.D. Doern, S.T. Nguyen, F. Afolabi, C.A.D. Burnham, Probiotic-associated as-
doi.org/10.1111/j.1469-0691.2011.03614.x. piration pneumonia due to Lactobacillus rhamnosus, J. Clin. Microbiol. 52 (2014)
[24] M. Martín Muñoz, T. Caballero, M. Caminoa, M. Belver, E. Rojas, S. Quirce, Allergy 3124–3126, https://doi.org/10.1128/JCM.01065-14.
to a probiotic compound, Allergy Eur. J. Allergy Clin. Immunol. 64 (2009) 200 [49] J.A. Fishman, Opportunistic infections–coming to the limits of immunosuppres-
http://www.embase.com/search/results?subaction=viewrecord&from=export& sion? Cold Spring Harb. Perspect. Med. 3 (2013) a015669, https://doi.org/10.
id=L70020425%5Cnhttps://doi.org/10.1111/j.1398-9995.2009.02076.x. 1101/cshperspect.a015669.
[25] M.C. Appel-da-Silva, G.A. Narvaez, L.R.R. Perez, L. Drehmer, J. Lewgoy, [50] S. Mo, B.-S. Kim, S.-J. Yun, J.-J. Lee, S.-H. Yoon, C.-H. Oh, Genome sequencing of
Saccharomyces cerevisiae var. boulardii fungemia following probiotic treatment, Clostridium butyricum DKU-01, isolated from infant feces, Gut Pathog. 7 (2015) 8,
Med. Mycol. Case Rep. 18 (2017) 15–17, https://doi.org/10.1016/j.mmcr.2017. https://doi.org/10.1186/s13099-015-0055-3.
07.007. [51] N. Cassir, S. Benamar, B. La Scola, Clostridium butyricum: from beneficial to a new
[26] M. Zheng, R. Zhang, X. Tian, X. Zhou, X. Pan, A. Wong, Assessing the risk of emerging pathogen, Clin. Microbiol. Infect. 22 (2016) 37–45, https://doi.org/10.
probiotic dietary supplements in the context of antibiotic resistance, Front. 1016/j.cmi.2015.10.014.
Microbiol. 8 (2017), https://doi.org/10.3389/fmicb.2017.00908. [52] G. Huys, N. Botteldoorn, F. Delvigne, L. De Vuyst, M. Heyndrickx, B. Pot,
[27] D.E. Nielsen, A. El-Sohemy, Applying genomics to nutrition and lifestyle mod- J.J. Dubois, G. Daube, Microbial characterization of probiotics-Advisory report of
ification, Per. Med. 9 (2012) 739–749, https://doi.org/10.2217/PME.12.79. the Working Group “8651 Probiotics” of the Belgian Superior Health Council
[28] M. Kussmann, L.B. Fay, Nutrigenomics and personalized nutrition: science and (SHC), Mol. Nutr. Food Res. 57 (2013) 1479–1504, https://doi.org/10.1002/mnfr.
concept, Per. Med. 5 (2008) 447–455, https://doi.org/10.2217/17410541.5.5. 201300065.
447. [53] H.A. Elsner, I. Sobottka, D. Mack, M. Claussen, R. Laufs, R. Wirth, Virulence fac-
[29] A. Bafeta, M. Koh, C. Riveros, P. Ravaud, Harms reporting in randomized con- tors of Enterococcus faecalis and Enterococcus faecium blood culture isolates, Eur. J.
trolled trials of interventions aimed at modifying microbiota, Ann. Intern. Med. Clin. Microbiol. Infect. Dis. 19 (2000) 39–42, https://doi.org/10.1007/
169 (2018) 240, https://doi.org/10.7326/M18-0343. s100960050007.
[30] FAO/WHO, Guidelines for the Evaluation of Probiotics in Food, Food and [54] M.M. Huycke, C.A. Spiegel, M.S. Gilmore, Bacteremia caused by hemolytic, high-
Agriculture Organization of the United Nations and World Health Organization level gentamicin-resistant Enterococcus faecalis, Antimicrob. Agents Chemother. 35
Group Report, London Ontario, Canada, 2002, https://doi.org/10.1111/j.1469- (1991) 1626–1634, https://doi.org/10.1128/AAC.35.8.1626.
0691.2012.03873 FAO Food Nutr. Pap. 85. [55] D.A.P.S. Group, Probiotic prophylaxis in predicted severe acute pancreatitis: a
[31] M.T. Liong, Safety of probiotics: translocation and infection, Nutr. Rev. 66 (2008) randomised, double-blind, placebo-controlled trial, Lancet 371 (2008) 651–659.
192–202, https://doi.org/10.1111/j.1753-4887.2008.00024.x. [56] M.C. Van Baal, P. Kohout, M.G. Besselink, H.C. Van Santvoort, Z. Benes, R. Zazula,
[32] R. Wiest, G. Garcia-Tsao, Bacterial translocation (BT) in cirrhosis, Hepatology. 41 G.T. Rijkers, H.G. Gooszen, Probiotic treatment with Probioflora in patients with
(2005) 422–433, https://doi.org/10.1002/hep.20632. predicted severe acute pancreatitis without organ failure, Pancreatology (2012)
[33] D. Stanley, L.J. Mason, K.E. MacKin, Y.N. Srikhanta, D. Lyras, M.D. Prakash, 458–462, https://doi.org/10.1016/j.pan.2012.08.004.
K. Nurgali, A. Venegas, M.D. Hill, R.J. Moore, C.H.Y. Wong, Translocation and [57] J.K. Nicholson, E. Holmes, J. Kinross, R. Burcelin, G. Gibson, W. Jia, S. Pettersson,
dissemination of commensal bacteria in post-stroke infection, Nat. Med. 22 (2016) Host-gut microbiota metabolic interactions, Science (80-.) 336 (2012) 1262–1267,
1277–1284, https://doi.org/10.1038/nm.4194. https://doi.org/10.1126/science.1223813.
[34] C.A. Jacobi, C. Schulz, P. Malfertheiner, Treating critically ill patients with pro- [58] G. Spano, P. Russo, A. Lonvaud-Funel, P. Lucas, H. Alexandre, C. Grandvalet,
biotics: beneficial or dangerous? Gut Pathog. 3 (2011), https://doi.org/10.1186/ E. Coton, M. Coton, L. Barnavon, B. Bach, F. Rattray, A. Bunte, C. Magni,
1757-4749-3-2. V. Ladero, M. Alvarez, M. Fernández, P. Lopez, P.F. de Palencia, A. Corbi, H. Trip,
[35] M.J. Kim, S. Ku, S.Y. Kim, H.H. Lee, H. Jin, S. Kang, R. Li, T.V. Johnston, J.S. Lolkema, Biogenic amines in fermented foods, Eur. J. Clin. Nutr. 64 (Suppl. 3)
M.S. Park, G.E. Ji, Safety evaluations of Bifidobacterium bifidum BGN4 and (2010) S95–S100, https://doi.org/10.1038/ejcn.2010.218.
Bifidobacterium longum BORI, Int. J. Mol. Sci. 19 (2018), https://doi.org/10.3390/ [59] J. Yongsawatdigul, W. Wongngam, B. Khampirat, V. Valderrey, A. Bonasera,
ijms19051422. S. Fredrich, S. Hecht, K. Tašev, V. Ivanova-Petropulos, M. Stefova, Z.S. Stojanović,
[36] L. Liu, H.L. Johnson, S. Cousens, J. Perin, S. Scott, J.E. Lawn, I. Rudan, E. Mehmeti, K. Kalcher, V. Guzsvány, D.M. Stanković, S.-S. YANG, Y.-N. YANG, X.-
H. Campbell, R. Cibulskis, M. Li, C. Mathers, R.E. Black, Child health epidemiology L. LI, Y.B. ZHANG, A.R. Shalaby, M.M. Anwar, E.M. Sallam, W.H. Emam, J. Płotka-
reference group of WHO and UNICEF, global, regional, and national causes of Wasylka, V. Simeonov, J. Namieśnik, R. Preti, S. Vieri, G. Vinci, P. Perez-Hurtado,
child mortality: an updated systematic analysis for 2010 with time trends since E. Palmer, T. Owen, C. Aldcroft, M.H. Allen, J. Jones, C.S. Creaser, M.R. Lindley,
2000, Lancet 379 (2012) 2151–2161, https://doi.org/10.1016/S0140-6736(12) M.A. Turner, J.C. Reynolds, J.L. Ordóñez, R.M. Callejón, A.M. Troncoso,
60560-1. M.C.D.C. García-Parrilla, R.M. Callejón, D. Nei, N. Nakamura, K. Ishihara,
[37] G.-Q. Zhang, H.-J. Hu, C.-Y. Liu, S. Shakya, Z.-Y. Li, Probiotics for preventing late- M. Kimura, M. Satomi, A.C. Manetta, L. Di Giuseppe, R. Tofalo, M. Martuscelli,
onset sepsis in preterm neonates: a PRISMA-compliant systematic review and M. Schirone, M. Giammarco, G. Suzzi, S. Mallick, F. Chandra, A.L. Koner,
meta-analysis of randomized controlled trials, Medicine (Baltimore). 95 (2016) A. Jastrzębska, S. Kowalska, E. Szłyk, A. Jastrzebska, A. Piasta, S. Kowalska,
e2581, https://doi.org/10.1097/MD.0000000000002581. M. Krzemiński, E. Szłyk, T. Janči, D. Valinger, J. Gajdoš Kljusurić, L. Mikac,
[38] K. Costeloe, U. Bowler, P. Brocklehurst, P. Hardy, P. Heal, E. Juszczak, A. King, S. Vidaček, M. Ivanda, W. Henao-Escobar, L. Del Torno-De Román, O. Domínguez-
N. Panton, F. Stacey, A. Whiley, M. Wilks, M.R. Millar, A randomised controlled Renedo, M.A. Alonso-Lomillo, M.J. Arcos-Martínez, A. Czajkowska-Mysłek,
trial of the probiotic Bifidobacterium breve BBG-001 in preterm babies to prevent J. Leszczyńska, C. Berbegal, I. Pardo, S. Ferrer, N. Benkerroum, S. Ata, M. Akyuz,
sepsis, necrotising enterocolitis and death: The probiotics in preterm infantS H. Cabuk, J.A.V. Albelda, A. Uzunoglu, G.N.C. Santos, L.A. Stanciu, A. Murray,
(PiPS) trial, Health Technol. Assess. (Rockv). 20 (2016), https://doi.org/10.3310/ K. Skene, K. Haynes, M. Griffin, J. Sobal, T.A. Lyson, J. Korhonen, A. Honkasalo,
hta20660 vii-83. J. Seppälä, N.B.D. Thi, G. Kumar, C.-Y. Lin, M. Mohammadi, M. Kamankesh,
[39] R.N. Husni, S.M. Gordon, J.A. Washington, D.L. Longworth, Lactobacillus bacter- Z. Hadian, A.M. Mortazavian, A. Mohammadi, R. Parchami, M. Kamalabadi,
emia and endocarditis: review of 45 cases, Clin. Infect. Dis. 25 (1997) 1048–1055 N. Alizadeh, F. Shen, Q.F. Wu, Y.Q. Wei, X.M. Liu, P.A. Tang, G.M. Khairy,
http://www.ncbi.nlm.nih.gov/pubmed/9402355. H.A. Azab, S.A. El-Korashy, M.S. Steiner, A. Duerkop, S. Sentellas, O. Nunez,
[40] M.K. Salminen, S. Tynkkynen, H. Rautelin, M. Saxelin, M. Vaara, P. Ruutu, J. Saurina, S. Chopra, A. Singh, P. Venugopalan, N. Singh, N. Kaur,
S. Sarna, V. Valtonen, A. Järvinen, Lactobacillus bacteremia during a rapid increase G.I. Mohammed, A.S. Bashammakh, A.A. Alsibaai, H. Alwael, M.S. El-Shahawi,
in probiotic use of Lactobacillus rhamnosus GG in Finland, Clin. Infect. Dis. 35 Y.Y. Hu, X.J. Ma, Y.B. ZHANG, Y.K. Che, J.C. Zhao, R. Preti, G. Vinci, L. Pinto,
(2002) 1155–1160, https://doi.org/10.1086/342912. C.H.D. Nieto, M.A. Zon, H. Fernandez, M.C.U. de Araujo, Y.Q. Fu, Z.H. Zhou,
[41] K. Guenther, E. Straube, W. Pfister, A. Guenther, A. Huebler, Sever sepsis after Y.L. Li, X. Lu, C.X. Zhao, G.W. Xu, N. Dey, A. Ali, S. Podder, S. Majumdar,
probiotic treatment with Escherichia coli NISSLE 1917, Pediatr. Infect. Dis. J. 29 D. Nandi, S. Bhattacharya, A.K.A. Salazar, J.J.L. Castro, W.W. Cheng, D.W. Sun,
(2010) 188–189, https://doi.org/10.1097/INF.0b013e3181c36eb9. J.H. Cheng, P. Kumar, M.K. Chatli, A.K. Verma, N. Mehta, O.P. Malav, D. Kumar,
[42] J.R. McDonald, Acute infective endocarditis, Infect. Dis. Clin. North Am. 23 (2009) N. Sharma, D. Restuccia, U.G. Spizzirri, M. De Luca, O.I. Parisi, N. Picci,
643–664, https://doi.org/10.1016/j.idc.2009.04.013. T. Chatzimitakos, V. Exarchou, S.A. Ordoudi, Y. Fiamegos, C. Stalikas, F. Aflaki,
[43] J.P. Cannon, T.A. Lee, J.T. Bolanos, L.H. Danziger, Pathogenic relevance of V. Ghoulipour, N. Saemian, S. Shiebani, M. Salahinejad, R.G. Cruz-Monterrosa,

545
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

V. Resendiz-Cruz, A.A. Rayas-Amor, M. Lopez, G.C. Miranda-de La Lama, pathogenic) bacteria. Bad genes in good bugs, Front. Microbiol. 5 (2014) 754,
W.P. Evangelista, T.M. Silva, L.R. Guidi, P.A.S. Tette, R.M.D. Byrro, P. Santiago- https://doi.org/10.3389/fmicb.2014.00754.
Silva, C. Fernandes, M.B.A. Gloria, M. Bashiry, A. Mohammadi, H. Hosseini, [80] P. Sharma, S.K. Tomar, P. Goswami, V. Sangwan, R. Singh, Antibiotic resistance
M. Kamankesh, S. Aeenehvand, Z. Mohammadi, E. Omanovic-Miklicanin, among commercially available probiotics, Food Res. Int. 57 (2014) 176–195,
S. Valzacchi, J. Huang, N. Gan, F.Y. Lv, Y.T. Cao, C.R. Ou, H.Q. Tang, I. Altieri, https://doi.org/10.1016/j.foodres.2014.01.025.
A. Semeraro, F. Scalise, I. Calderari, P. Stacchini, K.B. Biji, C.N. Ravishankar, [81] H. Abriouel, M. del C. Casado Muñoz, L. Lavilla Lerma, B. Pérez Montoro,
R. Venkateswarlu, C.O. Mohan, T.K.S. Gopal, R. Preti, R. Bernacchia, G. Vinci, W. Bockelmann, R. Pichner, J. Kabisch, G.S. Cho, C.M.A.P. Franz, A. Gálvez,
H. Dong, K.J. Xiao, B. Redruello, V. Ladero, B. Del Rio, M. Fernandez, M.C. Martin, N. Benomar, New insights in antibiotic resistance of Lactobacillus species from
M.A. Alvarez, N. Alizadeh, M. Kamalabadi, A. Mohammadi, D. Yang, A.X. Lu, fermented foods, Food Res, Int. 78 (2015) 465–481, https://doi.org/10.1016/j.
D. Ren, J.H. Wang, S.Y. Tseng, S.Y. Li, S.Y. Yi, A.Y. Sun, D.Y. Gao, D.H. Wan, foodres.2015.09.016.
E. Shumilina, R. Slizyte, R. Mozuraityte, A. Dykyy, T.A. Stein, A. Dikiy, K.M.A. El- [82] V. Venugopalan, K.A. Shriner, A. Wong-Beringer, Regulatory oversight and safety
Nour, E.T.A. Salam, H.M. Soliman, A.S. Orabi, T.M. Ngapo, L. Vachon, C. Schaude, of probiotic use, Emerg. Infect. Dis. 16 (2010) 1661–1665, https://doi.org/10.
C. Meindl, E. Frohlich, J. Attard, G.J. Mohr, T. Esatbeyoglu, A. Ehmer, D. Chaize, 3201/eid1611.100574.
G. Rimbach, Y.P. Cai, Z.W. Sun, G. Chen, X.M. Liu, J.M. You, C.Q. Zhang, Z.H. Li, [83] D. Festi, R. Schiumerini, L.H. Eusebi, G. Marasco, M. Taddia, A. Colecchia, Gut
K.S. Suslick, F. Gardini, Y. Ozogul, G. Suzzi, G. Tabanelli, F. Ozogul, T.R. Lin, microbiota and metabolic syndrome, World J. Gastroenterol. 20 (2014)
Y.R. Wu, Z.H. Li, Z.P. Song, L.Q. Guo, F.F. Fu, B. Kuswandi, A. Nurfawaidi, 16079–16094, https://doi.org/10.3748/wjg.v20.i43.16079.
S. Leonardo, M. Campas, Recent trends in the determination of biogenic amines in [84] V. Giacchi, P. Sciacca, P. Betta, Multistrain probiotics, Probiotics, Prebiotics, and
fermented beverages – a review, Food Chem. 26 (2016) 27–40, https://doi.org/10. Synbiotics, (2016), pp. 279–302, https://doi.org/10.1016/B978-0-12-802189-7.
1080/10498850.2014.964433. 00019-8.
[60] V.T. Martin, B. Vij, Diet and headache: part 1, Headache 56 (2016) 1543–1552, [85] S. Patel, N. Mathivanan, A. Goyal, Bacterial adhesins, the pathogenic weapons to
https://doi.org/10.1111/head.12953. trick host defense arsenal, Biomed. Pharmacother. 93 (2017) 763–771, https://
[61] H.M.L.J. Joosten, M.D. Northolt, Detection, growth, and amine-producing capa- doi.org/10.1016/j.biopha.2017.06.102.
city of Lactobacilli in cheese, Appl. Environ. Microbiol. 55 (1989) 2356–2359. [86] K. Papadimitriou, G. Zoumpopoulou, B. Foligné, V. Alexandraki, M. Kazou, B. Pot,
[62] Y. Gezginc, I. Akyol, E. Kuley, F. Özogul, Biogenic amines formation in E. Tsakalidou, Discovering probiotic microorganisms: in vitro, in vivo, genetic and
Streptococcus thermophilus isolated from home-made natural yogurt, Food Chem. omics approaches, Front. Microbiol. 6 (2015), https://doi.org/10.3389/fmicb.
138 (2013) 655–662, https://doi.org/10.1016/j.foodchem.2012.10.138. 2015.00058.
[63] E. Kuley, E. Balıkcı, I. Özoğul, S. Gökdogan, F. Ozoğul, Stimulation of cadaverine [87] S. Patel, Drivers of bacterial genomes plasticity and roles they play in pathogen
production by foodborne pathogens in the presence of Lactobacillus, Lactococcus, virulence, persistence and drug resistance, Infect. Genet. Evol. 45 (2016), https://
and Streptococcus spp, J. Food Sci. 77 (2012) M650–M658, https://doi.org/10. doi.org/10.1016/j.meegid.2016.08.030.
1111/j.1750-3841.2012.02825.x. [88] M. Chavarri, I. Maranon, M. Carmen, Encapsulation technology to protect pro-
[64] M. Begley, C. Hill, C.G.M. Gahan, Bile salt hydrolase activity in probiotics, Appl. biotic bacteria, Int. J. Probiotics Prebiotics (2012), https://doi.org/10.5772/
Environ. Microbiol. 72 (2006) 1729–1738, https://doi.org/10.1128/AEM.72.3. 50046.
1729. [89] M.E. Sanders, A.L. Shane, D.J. Merenstein, Advancing probiotic research in hu-
[65] A.Y. Bustos, L. Saavedra, G.F. de Valdez, R.R. Raya, M.P. Taranto, Relationship mans in the United States: challenges and strategies, Gut Microbes 7 (2016)
between bile salt hydrolase activity, changes in the internal pH and tolerance to 97–100, https://doi.org/10.1080/19490976.2016.1138198.
bile acids in lactic acid bacteria, Biotechnol. Lett. 34 (2012) 1511–1518, https:// [90] M. Olier, I. Marcq, C. Salvador-Cartier, T. Secher, U. Dobrindt, M. Boury,
doi.org/10.1007/s10529-012-0932-5. V. Bacquié, M. Penary, E. Gaultier, J.P. Nougayrède, J. Fioramonti, E. Oswald,
[66] M. Million, E. Angelakis, M. Paul, F. Armougom, L. Leibovici, D. Raoult, Genotoxicity of Escherichia coli NISSLE 1917 strain cannot be dissociated from its
Comparative meta-analysis of the effect of Lactobacillus species on weight gain in probiotic activity, Gut Microbes 3 (2012) 501–509, https://doi.org/10.4161/
humans and animals, Microb. Pathog. 53 (2012) 100–108, https://doi.org/10. gmic.21737.
1016/j.micpath.2012.05.007. [91] S. Patel, Letter to the editor on “The necessity of overhaul in perception of mi-
[67] A.L. Rouxinol-Dias, A.R. Pinto, C. Janeiro, D. Rodrigues, M. Moreira, J. Dias, crobiological culture methods”, Microb. Pathog. 102 (2017) A1–A2, https://doi.
P. Pereira, Probiotics for the control of obesity - its effect on weight change, Porto org/10.1016/j.micpath.2016.12.001.
Biomed. J. 1 (2016) 12–24, https://doi.org/10.1016/j.pbj.2016.03.005. [92] K. Alfaleh, J. Anabrees, Probiotics for prevention of necrotizing enterocolitis in
[68] M. Kalliomäki, S. Salminen, H. Arvilommi, P. Kero, P. Koskinen, E. Isolauri, preterm infants, Evid. Based Child Heal. 9 (2014) 584–671, https://doi.org/10.
Probiotics in primary prevention of atopic disease: a randomised placebo-con- 1002/ebch.1976.
trolled trial, Lancet 357 (2001) 1076–1079, https://doi.org/10.1016/S0140- [93] C.S.M. Lau, R.S. Chamberlain, Probiotic administration can prevent necrotizing
6736(00)04259-8. enterocolitis in preterm infants: a meta-analysis, J. Pediatr. Surg. 50 (2015)
[69] E. Iemoli, D. Trabattoni, S. Parisotto, L. Borgonovo, M. Toscano, G. Rizzardini, 1405–1412, https://doi.org/10.1016/j.jpedsurg.2015.05.008.
M. Clerici, E. Ricci, A. Fusi, E. De Vecchi, S. Piconi, L. Drago, Probiotics reduce gut [94] E. Dermyshi, Y. Wang, C. Yan, W. Hong, G. Qiu, X. Gong, T. Zhang, The “golden
microbial translocation and improve adult atopic dermatitis, J. Clin. Age” of Probiotics: a systematic review and meta-analysis of randomized and
Gastroenterol. 46 (2012) S33–S40, https://doi.org/10.1097/MCG. observational studies in preterm infants, Neonatology 112 (2017) 9–23, https://
0b013e31826a8468. doi.org/10.1159/000454668.
[70] S.-O. Kim, Y.-M. Ah, Y.M. Yu, K.H. Choi, W.-G. Shin, J.-Y. Lee, Effects of probiotics [95] D. Barraud, P.-E. Bollaert, S. Gibot, Impact of the administration of probiotics on
for the treatment of atopic dermatitis: a meta-analysis of randomized controlled mortality in critically ill adult patients: a meta-analysis of randomized controlled
trials, Ann. Allergy Asthma Immunol. 113 (2014) 217–226, https://doi.org/10. trials, Chest. 143 (2013) 646–655, https://doi.org/10.1378/chest.12-1745.
1016/j.anai.2014.05.021. [96] L. Bo, J. Li, T. Tao, Y. Bai, X. Ye, R.S. Hotchkiss, M.H. Kollef, N.H. Crooks, X. Deng,
[71] M. Kopp, Clinical potential for the use of probiotics in the management of re- Probiotics for preventing ventilator-associated pneumonia, Cochrane Database
spiratory conditions and cold- and influenza-like symptoms, Nutr. Diet. Suppl. 51 Syst. Rev. (2014) (2014), https://doi.org/10.1002/14651858.CD009066.pub2.
(2011), https://doi.org/10.2147/NDS.S7937. [97] B. Banupriya, N. Biswal, R. Srinivasaraghavan, P. Narayanan, J. Mandal, Probiotic
[72] A.L. Taylor, J.A. Dunstan, S.L. Prescott, Probiotic supplementation for the first 6 prophylaxis to prevent ventilator associated pneumonia (VAP) in children on
months of life fails to reduce the risk of atopic dermatitis and increases the risk of mechanical ventilation: an open-label randomized controlled trial, Intensive Care
allergen sensitization in high-risk children: a randomized controlled trial, J. Med. 41 (2015) 677–685, https://doi.org/10.1007/s00134-015-3694-4.
Allergy Clin. Immunol. 119 (2007) 184–191, https://doi.org/10.1016/j.jaci.2006. [98] J. Zeng, C.-T. Wang, F.-S. Zhang, F. Qi, S.-F. Wang, S. Ma, T.-J. Wu, H. Tian, Z.-
08.036. T. Tian, S.-L. Zhang, Y. Qu, L.-Y. Liu, Y.-Z. Li, S. Cui, H.-L. Zhao, Q.-S. Du, Z. Ma,
[73] K. Shida, M. Nanno, S. Nagata, Flexible cytokine production by macrophages and C.-H. Li, Y. Li, M. Si, Y.-F. Chu, M. Meng, H.-S. Ren, J.-C. Zhang, J.-J. Jiang,
T cells in response to probiotic bacteria: a possible mechanism by which probiotics M. Ding, Y.-P. Wang, Effect of probiotics on the incidence of ventilator-associated
exert multifunctional immune regulatory activities, Gut Microbes 2 (2011) pneumonia in critically ill patients: a randomized controlled multicenter trial,
109–114, https://doi.org/10.4161/gmic.2.2.15661. Intensive Care Med. 42 (2016) 1018–1028, https://doi.org/10.1007/s00134-016-
[74] P.R. Marteau, M. De Vrese, C.J. Cellier, J. Schrezenmeir, Protection from gastro- 4303-x.
intestinal diseases with the use of probiotics, Am. J. Clin. Nutr. (2001), https:// [99] W. Manzanares, M. Lemieux, P.L. Langlois, P.E. Wischmeyer, Probiotic and syn-
doi.org/10.1093/ajcn/73.2.430s. biotic therapy in critical illness: a systematic review and meta-analysis, Crit. Care
[75] S. Doron, D.R. Snydman, Risk and safety of probiotics, Clin. Infect. Dis. 60 (2015) 20 (2016), https://doi.org/10.1186/s13054-016-1434-y.
S129–S134, https://doi.org/10.1093/cid/civ085. [100] V. Lorenzo-Zúñiga, E. Llop, C. Suárez, B. Álvarez, L. Abreu, J. Espadaler, J. Serra,
[76] I.C.V.J. Imperial, J.A. Ibana, Addressing the antibiotic resistance problem with I.31, a new combination of probiotics, improves irritable bowel syndrome-related
probiotics: reducing the risk of its double-edged sword effect, Front. Microbiol. 7 quality of life, World J. Gastroenterol. (2014) 8709–8716, https://doi.org/10.
(2016), https://doi.org/10.3389/fmicb.2016.01983. 3748/wjg.v20.i26.8709.
[77] C.J. Dorman, H-NS-like nucleoid-associated proteins, mobile genetic elements and [101] T. Didari, S. Mozaffari, S. Nikfar, M. Abdollahi, Effectiveness of probiotics in ir-
horizontal gene transfer in bacteria, Plasmid 75 (2014) 1–11, https://doi.org/10. ritable bowel syndrome: updated systematic review with meta-analysis, World J.
1016/j.plasmid.2014.06.004. Gastroenterol. 21 (2015) 3072–3084, https://doi.org/10.3748/wjg.v21.i10.3072.
[78] Y. Hu, X. Yang, J. Qin, N. Lu, G. Cheng, N. Wu, Y. Pan, J. Li, L. Zhu, X. Wang, [102] J.Y. Lee, S.H. Chu, J.Y. Jeon, M.K. Lee, J.H. Park, D.C. Lee, J.W. Lee, N.K. Kim,
Z. Meng, F. Zhao, D. Liu, J. Ma, N. Qin, C. Xiang, Y. Xiao, L. Li, H. Yang, J. Wang, Effects of 12 weeks of probiotic supplementation on quality of life in colorectal
R. Yang, G.F. Gao, B. Zhu, Metagenome-wide analysis of antibiotic resistance cancer survivors: a double-blind, randomized, placebo-controlled trial, Dig. Liver
genes in a large cohort of human gut microbiota, Nat. Commun. 4 (2013) 2151, Dis. 46 (2014) 1126–1132, https://doi.org/10.1016/j.dld.2014.09.004.
https://doi.org/10.1038/ncomms3151. [103] M.G. Redman, E.J. Ward, R.S. Phillips, The efficacy and safety of probiotics in
[79] H. Aarts, A. Margolles, Antibiotic resistance genes in food and gut (non- people with cancer: a systematic review, Ann. Oncol. 25 (2014) 1919–1929,

546
D. Kothari et al. Biomedicine & Pharmacotherapy 111 (2019) 537–547

https://doi.org/10.1093/annonc/mdu106. E. Botelho-Nevers, Bifidobacterium species bacteremia: risk factors in adults and


[104] G.M. Carter, A. Esmaeili, H. Shah, D. Indyk, M. Johnson, M. Andreae, H.S. Sacks, infants, Clin. Infect. Dis. 61 (2015) 482–484, https://doi.org/10.1093/cid/civ347.
Probiotics in human immunodeficiency virus infection: a systematic review and [118] C. Bertelli, T. Pillonel, A. Torregrossa, G. Prod’hom, C. Julie Fischer, G. Greub,
evidence synthesis of benefits and risks, Open Forum Infect. Dis. 3 (2016), https:// E. Giannoni, Bifidobacterium longum bacteremia in preterm infants receiving pro-
doi.org/10.1093/ofid/ofw164. biotics, Clin. Infect. Dis. 60 (2015) 924–927, https://doi.org/10.1093/cid/ciu946.
[105] G. D’Ettorre, G. Ceccarelli, N. Giustini, S. Serafino, N. Calantone, G. De Girolamo, [119] A. Zbinden, R. Zbinden, C. Berger, R. Arlettaz, Case series of Bifidobacterium
L. Bianchi, V. Bellelli, T. Ascoli-Bartoli, S. Marcellini, O. Turriziani, longum bacteremia in three preterm infants on probiotic therapy, Neonatology 107
J.M. Brenchley, V. Vullo, Probiotics reduce inflammation in antiretroviral treated, (2015) 56–59, https://doi.org/10.1159/000367985.
HIV-infected individuals: results of the “Probio-HIV” clinical trial, PLoS One 10 [120] S.L. Avcin, M. Pokorn, L. Kitanovski, M.M. Premru, J. Jazbec, Bifidobacterium breve
(2015), https://doi.org/10.1371/journal.pone.0137200. sepsis in child with high-risk acute lymphoblastic leukemia, Emerg. Infect. Dis. 21
[106] Y. Zhang, J. Chen, J. Wu, H. Chalson, L. Merigan, A. Mitchell, Probiotic use in (2015) 1674–1675, https://doi.org/10.3201/eid2109.150097.
preventing postoperative infection in liver transplant patients, Hepatobiliary Surg. [121] F. Lestin, A. Pertschy, D. Rimek, [Fungemia after oral treatment with
Nutr. 2 (2013) 142–147, https://doi.org/10.3978/j.issn.2304-3881.2013.06.05. Saccharomyces boulardii in a patient with multiple comorbidities]. [German],
[107] T. Sawas, S. Al Halabi, R. Hernaez, W.D. Carey, W.K. Cho, Patients receiving Fungamie nach oraler Gabe von, Dtsch. Medizinische Wochenschrift 128 (2003)
prebiotics and probiotics before liver transplantation develop fewer infections 2531–2533.
than controls: a systematic review and meta-analysis, Clin. Gastroenterol. Hepatol. [122] S. Atıcı, A. Soysal, K. Karadeniz Cerit, Ş. Yılmaz, B. Aksu, G. Kıyan, M. Bakır,
13 (2015) 1567–1574, https://doi.org/10.1016/j.cgh.2015.05.027. Catheter-related Saccharomyces cerevisiae fungemia following Saccharomyces bou-
[108] M. Grąt, K.M. Wronka, Z. Lewandowski, K. Grąt, M. Krasnodębski, lardii probiotic treatment: in a child in intensive care unit and review of the lit-
J. Stypułkowski, W. Hołówko, Ł. Masior, I. Kosińska, M. Wasilewicz, J. Raszeja- erature, Med. Mycol. Case Rep. 15 (2017) 33–35, https://doi.org/10.1016/j.
Wyszomirska, S. Rejowski, E. Bik, W. Patkowski, M. Krawczyk, Effects of con- mmcr.2017.02.002.
tinuous use of probiotics before liver transplantation: a randomized, double-blind, [123] F.Z. Chioukh, H. Ben Hmida, K. Ben Ameur, A. Toumi, K. Monastiri, Septicémie à
placebo-controlled trial, Clin. Nutr. 36 (2017) 1530–1539, https://doi.org/10. Saccharomyces cerevisiae chez un prématuré traité par Ultra-Levure®, Médecine
1016/j.clnu.2017.04.021. Mal. Infect. 43 (2013) 359–360, https://doi.org/10.1016/j.medmal.2013.06.008.
[109] M.S. Ammor, A. Belén Flórez, B. Mayo, Antibiotic resistance in non-enterococcal [124] E. Shinar, E. Leitersdorf, R. Yevin, Lactobacillus plantarum endocarditis, Klin.
lactic acid bacteria and bifidobacteria, Food Microbiol. 24 (2007) 559–570, Wochenschr. 62 (1984) 1173–1174, https://doi.org/10.1007/BF01712185.
https://doi.org/10.1016/j.fm.2006.11.001. [125] L. Zé-Zé, R. Tenreiro, A. Duarte, M.J. Salgado, J. Melo-Cristino, L. Lito,
[110] C. Devirgiliis, P. Zinno, G. Perozzi, Update on antibiotic resistance in foodborne M.M. Carmo, S. Felisberto, G. Carmo, Case of aortic endocarditis caused by
Lactobacillus and Lactococcus species, Front. Microbiol. 4 (2013), https://doi.org/ Lactobacillus casei, J. Med. Microbiol. 53 (2004) 451–453, https://doi.org/10.
10.3389/fmicb.2013.00301. 1099/jmm.0.05328-0.
[111] G.W. Tannock, J.B. Luchansky, L. Miller, H. Connell, S. Thode-Andersen, [126] P.A. Fradiani, A. Petrucca, F. Ascenzioni, G. Di Nucci, A. Teggi, S. Bilancini,
A.A. Mercer, T.R. Klaenhammer, Molecular characterization of a plasmid-borne P. Cipriani, Endocarditis caused by Lactobacillus jensenii in an immunocompetent
(pGT633) erythromycin resistance determinant (ermGT) from Lactobacillus reuteri patient, J. Med. Microbiol. 59 (2010) 607–609, https://doi.org/10.1099/jmm.0.
100-63, Plasmid 31 (1994) 60–71, https://doi.org/10.1006/plas.1994.1007. 017764-0.
[112] A.N. Kunz, J.M. Noel, M.P. Fairchok, Two Cases of Lactobacillus bacteremia during [127] B. Franko, M. Vaillant, C. Recule, E. Vautrin, J.P. Brion, P. Pavese, Lactobacillus
probiotic treatment of short gut syndrome, J. Pediatr. Gastroenterol. Nutr. 38 paracasei endocarditis in a consumer of probiotics, Med. Mal. Infect. 43 (2013)
(2004) 457–458, https://doi.org/10.1097/00005176-200404000-00017. 171–173, https://doi.org/10.1016/j.medmal.2013.01.007.
[113] M.H. Land, K. Rouster-Stevens, C.R. Woods, M.L. Cannon, J. Cnota, A.K. Shetty, [128] E. Boumis, A. Capone, V. Galati, C. Venditti, N. Petrosillo, Probiotics and infective
Lactobacillus sepsis associated with probiotic therapy: Fig 1, Pediatrics 115 (2005) endocarditis in patients with hereditary hemorrhagic telangiectasia: a clinical case
178–181, https://doi.org/10.1542/peds.2004-2137. and a review of the literature, BMC Infect. Dis. 18 (2018), https://doi.org/10.
[114] M.A. De Groote, D.N. Frank, E. Dowell, M.P. Glode, N.R. Pace, Lactobacillus 1186/s12879-018-2956-5.
rhamnosus GG bacteremia associated with probiotic use in a child with short gut [129] J. Vanichanan, V. Chávez, A. Wanger, A.M. De Golovine, K.J. Vigil, Carbapenem-
syndrome, Pediatr. Infect. Dis. J. 24 (2005) 278–280, https://doi.org/10.1097/01. resistant Lactobacillus intra-abdominal infection in a renal transplant recipient
inf.0000154588.79356.e6. with a history of probiotic consumption, Infection 44 (2016) 793–796, https://doi.
[115] E. Vahabnezhad, A.B. Mochon, L.J. Wozniak, D.A. Ziring, Lactobacillus bacteremia org/10.1007/s15010-016-0903-1.
associated with probiotic use in a pediatric patient with ulcerative colitis, J. Clin. [130] M.V. Kopp, Probiotics in the prevention and therapy of atopic diseases,
Gastroenterol. 47 (2013) 437–439, https://doi.org/10.1097/MCG. Monatsschr. Kinderheilkd 156 (2008) 1084–1092, https://doi.org/10.1007/
0b013e318279abf0. s00112-008-1832-6.
[116] H. Kulkarni, C. Khoury, Sepsis associated with Lactobacillus bacteremia in a patient [131] M.E. Sanders, F. Guarner, R. Guerrant, P.R. Holt, E.M. Quigley, R.B. Sartor,
with ischemic colitis, Indian J. Crit. Care Med. 18 (2014) 606, https://doi.org/10. P.M. Sherman, E.A. Mayer, An update on the use and investigation of probiotics in
4103/0972-5229.140152. health and disease, Gut 62 (2013) 787–796, https://doi.org/10.1080/00224499.
[117] E. Weber, Q. Reynaud, F. Suy, A. Gagneux-Brunon, A. Carricajo, A. Guillot, 2018.1447639.

547

Anda mungkin juga menyukai