Skip to main content
The Cochrane Database of Systematic Reviews logoLink to The Cochrane Database of Systematic Reviews
. 2023 Oct 23;2023(10):CD013631. doi: 10.1002/14651858.CD013631.pub2

Synbiotics, prebiotics and probiotics for people with chronic kidney disease

Tess E Cooper 1,, Rabia Khalid 1,2, Samuel Chan 3, Jonathan C Craig 4,5, Carmel M Hawley 3, Martin Howell 1,2, David W Johnson 3, Allison Jaure 1,2, Armando Teixeira-Pinto 1,2, Germaine Wong 1,2,6
Editor: Cochrane Kidney and Transplant Group
PMCID: PMC10591284  PMID: 37870148

Abstract

Background

Chronic kidney disease (CKD) is a major public health problem affecting 13% of the global population. Prior research has indicated that CKD is associated with gut dysbiosis. Gut dysbiosis may lead to the development and/or progression of CKD, which in turn may in turn lead to gut dysbiosis as a result of uraemic toxins, intestinal wall oedema, metabolic acidosis, prolonged intestinal transit times, polypharmacy (frequent antibiotic exposures) and dietary restrictions used to treat CKD. Interventions such as synbiotics, prebiotics, and probiotics may improve the balance of the gut flora by altering intestinal pH, improving gut microbiota balance and enhancing gut barrier function (i.e. reducing gut permeability).

Objectives

This review aimed to evaluate the benefits and harms of synbiotics, prebiotics, and probiotics for people with CKD.

Search methods

We searched the Cochrane Kidney and Transplant Register of Studies up to 9 October 2023 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal and ClinicalTrials.gov.

Selection criteria

We included randomised controlled trials (RCTs) measuring and reporting the effects of synbiotics, prebiotics, or probiotics in any combination and any formulation given to people with CKD (CKD stages 1 to 5, including dialysis and kidney transplant). Two authors independently assessed the retrieved titles and abstracts and, where necessary, the full text to determine which satisfied the inclusion criteria.

Data collection and analysis

Data extraction was independently carried out by two authors using a standard data extraction form. Summary estimates of effect were obtained using a random‐effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes, and mean difference (MD) or standardised mean difference (SMD) and 95% CI for continuous outcomes. The methodological quality of the included studies was assessed using the Cochrane risk of bias tool. Data entry was carried out by one author and cross‐checked by another. Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach.

Main results

Forty‐five studies (2266 randomised participants) were included in this review. Study participants were adults (two studies in children) with CKD ranging from stages 1 to 5, with patients receiving and not receiving dialysis, of whom half also had diabetes and hypertension.

No studies investigated the same synbiotic, prebiotic or probiotic of similar strains, doses, or frequencies. Most studies were judged to be low risk for selection bias, performance bias and reporting bias, unclear risk for detection bias and for control of confounding factors, and high risk for attrition and other biases.

Compared to prebiotics, it is uncertain whether synbiotics improve estimated glomerular filtration rate (eGFR) at four weeks (1 study, 34 participants: MD ‐3.80 mL/min/1.73 m², 95% CI ‐17.98 to 10.38), indoxyl sulfate at four weeks (1 study, 42 participants: MD 128.30 ng/mL, 95% CI ‐242.77 to 499.37), change in gastrointestinal (GI) upset (borborymgi) at four weeks (1 study, 34 participants: RR 15.26, 95% CI 0.99 to 236.23), or change in GI upset (Gastrointestinal Symptom Rating Scale) at 12 months (1 study, 56 participants: MD 0.00, 95% CI ‐0.27 to 0.27), because the certainty of the evidence was very low.

Compared to certain strains of prebiotics, it is uncertain whether a different strain of prebiotics improves eGFR at 12 weeks (1 study, 50 participants: MD 0.00 mL/min, 95% CI ‐1.73 to 1.73), indoxyl sulfate at six weeks (2 studies, 64 participants: MD ‐0.20 μg/mL, 95% CI ‐1.01 to 0.61; I² = 0%) or change in any GI upset, intolerance or microbiota composition, because the certainty of the evidence was very low.

Compared to certain strains of probiotics, it is uncertain whether a different strain of probiotic improves eGFR at eight weeks (1 study, 30 participants: MD ‐0.64 mL/min, 95% CI ‐9.51 to 8.23; very low certainty evidence).

Compared to placebo or no treatment, it is uncertain whether synbiotics improve eGFR at six or 12 weeks (2 studies, 98 participants: MD 1.42 mL/min, 95% CI 0.65 to 2.2) or change in any GI upset or intolerance at 12 weeks because the certainty of the evidence was very low.

Compared to placebo or no treatment, it is uncertain whether prebiotics improves indoxyl sulfate at eight weeks (2 studies, 75 participants: SMD ‐0.14 mg/L, 95% CI ‐0.60 to 0.31; very low certainty evidence) or microbiota composition because the certainty of the evidence is very low.

Compared to placebo or no treatment, it is uncertain whether probiotics improve eGFR at eight, 12 or 15 weeks (3 studies, 128 participants: MD 2.73 mL/min, 95% CI ‐2.28 to 7.75; I² = 78%), proteinuria at 12 or 24 weeks (1 study, 60 participants: MD ‐15.60 mg/dL, 95% CI ‐34.30 to 3.10), indoxyl sulfate at 12 or 24 weeks (2 studies, 83 participants: MD ‐4.42 mg/dL, 95% CI ‐9.83 to 1.35; I² = 0%), or any change in GI upset or intolerance because the certainty of the evidence was very low. Probiotics may have little or no effect on albuminuria at 12 or 24 weeks compared to placebo or no treatment (4 studies, 193 participants: MD 0.02 g/dL, 95% CI ‐0.08 to 0.13; I² = 0%; low certainty evidence).

For all comparisons, adverse events were poorly reported and were minimal (flatulence, nausea, diarrhoea, abdominal pain) and non‐serious, and withdrawals were not related to the study treatment.

Authors' conclusions

We found very few studies that adequately test biotic supplementation as alternative treatments for improving kidney function, GI symptoms, dialysis outcomes, allograft function, patient‐reported outcomes, CVD, cancer, reducing uraemic toxins, and adverse effects.

We are not certain whether synbiotics, prebiotics, or probiotics are more or less effective compared to one another, antibiotics, or standard care for improving patient outcomes in people with CKD. Adverse events were uncommon and mild.

Keywords: Adult; Child; Humans; Dysbiosis; Dysbiosis/complications; Dysbiosis/therapy; Indican; Prebiotics; Probiotics; Probiotics/therapeutic use; Renal Insufficiency, Chronic; Renal Insufficiency, Chronic/complications; Renal Insufficiency, Chronic/therapy; Synbiotics; Uremic Toxins

Plain language summary

Prebiotics (dietary fibre), probiotics (good bacteria) or synbiotics (prebiotics plus probiotics) for people with chronic kidney disease

Key messages

Chronic kidney disease (CKD) is a serious health problem that affects over 850 million people worldwide. People with kidney disease have an unhealthy balance of good and bad bacteria in their guts, called 'gut dysbiosis'. This imbalance arises because of the effects of reduced kidney function (retained toxic waste products, fluid retention causing the gut wall to swell), drugs frequently used in people with CKD (especially antibiotics), and dietary restrictions placed on people with CKD.

Gut dysbiosis can, in turn, cause or worsen CKD because bacteria can produce toxins that cross the bowel wall and damage the kidneys. Gut dysbiosis can also cause stomach problems (like bloating, cramping, constipation and diarrhoea) and reduce quality of life.

To improve the balance of the gut flora, good bacteria can be taken in tablets of high doses of prebiotics and probiotics. Prebiotics, or indigestible plant fibre, can encourage the growth of good bacteria. Synbiotics are a combination of prebiotics and probiotics. Some research suggests that taking high doses of the good bacteria can re‐balance the good bacteria in people's gut, thereby improving bowel symptoms and the conditions that lead to worsening of CKD.

What did we do?

We reviewed all of the evidence on synbiotics, prebiotics and probiotics to see whether they can improve outcomes in people who have CKD (all stages 1 to 5).

What did we find?

We found 45 studies randomising 2266 participants. Half of these looked at participants receiving dialysis (mostly haemodialysis), and the other half not receiving dialysis. Half also had diabetes and hypertension.

We are uncertain whether synbiotics, prebiotics, or probiotics improve bowel outcomes, quality of life, kidney toxin levels or kidney function.

The quality of the evidence that we found is low quality and very low certainty. All studies were conducted using moderate to poor‐quality methods with too few patients.

Summary

Currently, we do not have enough information from trials to know whether synbiotics, prebiotics or probiotics work to improve bowel symptoms, quality of life, kidney toxin levels, or kidney function in people with CKD. Ten studies are currently ongoing; therefore, it is possible that findings may change with the inclusion of these studies in future updates.

The evidence is up to date to 9 October 2023.

Summary of findings

Summary of findings 1. Synbiotic versus another synbiotic for people with chronic kidney disease.

Synbiotic versus another synbiotic for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: synbiotic 2
Comparison: synbiotic 1
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with synbiotic 1 Risk with synbiotic 2
eGFR
Albuminuria or proteinuria
Indoxyl sulfate
Change in any GI upset or intolerance
Microbiota composition
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; CKD: chronic kidney disease; eGFR: (estimated) glomerular filtration rate; GI: gastrointestinal; RCT: randomised controlled trial; RR: risk ratio.
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

No data were reported for these outcomes under this comparison.

Summary of findings 2. Synbiotic versus prebiotic for people with chronic kidney disease.

Synbiotic versus prebiotic for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: synbiotic
Comparison: prebiotic
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with prebiotic Risk with synbiotic
eGFR
(mL/min)
Follow‐up: 4 weeks
CKD stage 3Ga (transplant) 34 (1) ⊕⊝⊝⊝
very low1
The mean eGFR was 3.8 mL/min lower (17.98 lower to 10.38 higher) with synbiotic compared prebiotic
Albuminuria or proteinuria
Indoxyl sulfate
(ng/mL)
Follow‐up: 4 weeks
CKD stage G5D 42 (1) ⊕⊝⊝⊝
very low1
The mean indoxyl sulfate was 128.3 ng/mL higher (242.77 lower to 499.37 higher) with synbiotic compared prebiotic
Change in any GI upset (borborygmi)
Follow‐up: 4 weeks
CKD stage 3Ga (transplant) RR 15.26
(0.99 to 236.23) 34 (1) ⊕⊝⊝⊝
very low1
0 per 1,000 0 per 1,000
(0 to 0)
Change in any GI upset (GSRS total index)
Follow‐up: 12 months
CKD stage 3 to 4   56 (1) ⊕⊝⊝⊝
very low1
The mean change in any GI upset was 0.00 (0.27 lower to 0.27 higher) with synbiotic compared prebiotic
Microbiota composition
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; CKD: chronic kidney disease; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

Summary of findings 3. Prebiotic versus another prebiotic for people with chronic kidney disease.

Prebiotic versus another prebiotic for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: prebiotic 2
Comparison: prebiotic 1
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with prebiotic 1 Risk with prebiotic 2
eGFR
(mL/min)
Follow‐up: 12 weeks
CKD stage 3 to 5 (non‐dialysis) 50 (1) ⊕⊝⊝⊝
very low1
The mean eGFR was 0 mL/min lower (1.73 lower to 1.73 higher) with prebiotic 2 compared to prebiotic 1
Albuminuria or proteinuria
Indoxyl sulfate
(μg/mL)
Follow‐up: 6 weeks
CKD stage G5D with diabetes 64 (2) ⊕⊝⊝⊝
very low1
The mean indoxyl sulfate was 0.2 μg/mL lower (1.01 lower to 0.61 higher) with prebiotic 2 compared to prebiotic 1
Change in any GI upset or intolerance
Follow‐up: 4 weeks
CKD stage G5D with diabetes 24 (1) ⊕⊝⊝⊝
very low1
Burping The mean burping was 0.17 higher (0.5 lower to 0.84 higher) with prebiotic 2 compared to prebiotic 1
Cramping The mean cramping was 0.17 lower (0.5 lower to 0.16 higher) with prebiotic 2 compared to prebiotic 1
Distension The mean distension was 0.33 higher (0.04 lower to 0.7 higher) with prebiotic 2 compared to prebiotic 1
Flatulence The mean flatulence was 1 higher (0.25 higher to 1.75 higher) with prebiotic 2 compared to prebiotic 1
Nausea The mean nausea was 0 (0 to 0 ) with prebiotic 2 compared to prebiotic 1
Reflux The mean reflux was 0 (0.5 lower to 0.5 higher) with prebiotic 2 compared to prebiotic 1
Rumblings The mean rumblings was 0.5 higher (0.14 lower to 1.14 higher) with prebiotic 2 compared to prebiotic 1
Microbiota composition
Follow‐up: 4 weeks
CKD stage G5D with diabetes 24 (1)
Actinobacteria The mean Actinobacteria was 1.26 lower (4.46 lower to 1.94 higher) with prebiotic 2 compared to prebiotic 1
Bacteriodetes The mean Bacteriodetes was 3.23 higher (8.24 lower to 14.7 higher) with prebiotic 2 compared to prebiotic 1
Proteobacteria The mean Proteobacteria was 0.11 higher (1.61 lower to 1.83 higher) with prebiotic 2 compared to prebiotic 1
Firmicutes The mean Firmicutes was 2.44 lower (14.19 lower to 9.31 higher) with prebiotic 2 compared to prebiotic 1
Synergistetes The mean Synergistetes was MD 0.25 lower (0.89 lower to 0.39 higher) with prebiotic 2 compared to prebiotic 1
Verrucomicrobia The mean Verrucomicrobia was 0.96 higher (1.36 lower to 3.28 higher) with prebiotic 2 compared to prebiotic 1
Faecal acetate The mean faecal acetate was 69.91 lower (203.95 lower to 64.13 higher) with prebiotic 2 compared to prebiotic 1
Faecal propionate The mean faecal propionate was 19.35 lower (63.87 lower to 25.17 higher) with prebiotic 2 compared to prebiotic 1
Faecal butyrate The mean faecal butyrate was 11.04 lower (39.57 lower to 17.49 higher) with prebiotic 2 compared to prebiotic 1
Faecal total short‐chain fatty acids The mean faecal total short‐chain fatty acids were 104.71 lower (293.34 lower to 83.92 higher) with prebiotic 2 compared to prebiotic 1
Faecal total short‐chain fatty acids The mean faecal total short‐chain fatty acids were 104.71 lower (293.34 lower to 83.92 higher) with prebiotic 2 compared to prebiotic 1
Faecal indoles The mean faecal indoles were 2.71 higher (69.78 lower to 75.2 higher) with prebiotic 2 compared to prebiotic 1
Faecal P‐cresol The mean faecal p‐cresol was 28.84 higher (105.07 lower to 162.75 higher) with prebiotic 2 compared to prebiotic 1
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

Summary of findings 4. Prebiotic versus probiotic for people with chronic kidney disease.

Prebiotic versus probiotic for people with chronic kidney disease
Patient or population: people with CKD
Settings: hospital or primary care
Intervention: prebiotic
Comparison: probiotic
Outcomes Illustrative comparative risks* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Quality of the evidence
(GRADE)
Assumed risk Corresponding risk
Risk with probiotic Risk with prebiotic
eGFR
Albuminuria or proteinuria
Indoxyl sulfate
Change in any GI upset or intolerance
Microbiota composition
Graft function
Graft infection
*The basis for the assumed risk (e.g. the median control group risk across studies) is provided in footnotes. The corresponding risk (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RCT: randomised controlled trial
GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

No data were reported for these outcomes under this comparison.

Summary of findings 5. Probiotic versus another probiotic for people with chronic kidney disease.

Probiotic versus another probiotic for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: probiotic 2
Comparison: probiotic 1
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with probiotic 1 Risk with probiotic 2
eGFR
(mL/min)
Follow‐up: 8 weeks
CKD stage G3 to G4 30 (1) ⊕⊝⊝⊝
very low1
The mean eGFR was 0.64 mL/min lower (9.51 lower to 8.23 higher) with probiotic 2 than probiotic 1
Albuminuria or proteinuria
Indoxyl sulfate
Change in any GI upset or intolerance
Microbiota composition
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

Summary of findings 6. Synbiotic versus placebo or no treatment for people with chronic kidney disease.

Synbiotic versus placebo or no treatment for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: synbiotic
Comparison: placebo or no treatment
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with placebo or no treatment Risk with synbiotic
eGFR
(mL/min)
Follow‐up: 6 or 12 weeks
CKD stage G3b 98 (2) ⊕⊝⊝⊝
very low1
The mean eGFR was 1.42 mL/min higher (0.65 higher to 2.2 higher) with synbiotic than placebo or no treatment
Albuminuria or proteinuria
Indoxyl sulfate
Change in any GI upset or intolerance
(GSRS)
Follow‐up: 12 weeks
CKD stage 3b 23 (1) ⊕⊝⊝⊝
very low1
Rumbling The mean rumbling was 0.54 GSRS lower (0.77 lower to 0.31 lower) with synbiotic than placebo or no treatment
Hard stools The mean hard stools was 0.09 GSRS lower (0.35 lower to 0.17 higher) with synbiotic than placebo or no treatment
Abdominal pain The mean abdominal pain was 0.22 GSRS higher (0.02 lower to 0.46 higher) with synbiotic than placebo or no treatment
Constipation syndrome The mean constipation syndrome was 0.17 GSRS lower (0.72 lower to 0.38 higher) with synbiotic than placebo or no treatment
Microbiota composition
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

Summary of findings 7. Prebiotic versus placebo or no treatment for people with chronic kidney disease.

Prebiotic versus placebo or no treatment for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: prebiotic
Comparison: placebo or no treatment
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with placebo or no treatment Risk with prebiotic
eGFR
Albuminuria or proteinuria
Indoxyl sulfate
(mg/L)
Follow‐up: 8 weeks
CKD stage G5D 75 (2) ⊕⊝⊝⊝
very low1
The mean indoxyl sulfate was 0.14 mg/L lower (0.6 lower to 0.31 higher) with prebiotic than placebo or no treatment
Any change in GI upset or intolerance
Microbiota composition
Follow‐up: 8 weeks
CKD stage 5GD 44 (1) ⊕⊝⊝⊝
very low1
Faecalibacterium The mean Faecalibacterium was 2.37 higher (0.23 higher to 4.51 higher) with prebiotic than placebo or no treatment
Parabacteroides The mean Parabacteroides was 0.22 higher (0.06 higher to 0.38 higher) with prebiotic than placebo or no treatment
Bifidobacteria The mean Bifidobacteria was 3.92 lower (9.83 lower to 1.99 higher) with prebiotic than placebo or no treatment
Ruminococcus The mean Ruminococcus was 3.86 higher (0.32 lower to 8.04 higher) with prebiotic than placebo or no treatment
Prevotella The mean Prevotella was 0.43 lower (1.45 lower to 0.59 higher) with prebiotic than placebo or no treatment
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

Summary of findings 8. Probiotic versus placebo or no treatment for people with chronic kidney disease.

Probiotic versus placebo or no treatment for people with chronic kidney disease
Patient or population: people with CKD
Setting: hospital or primary care
Intervention: probiotic
Comparison: placebo or no treatment
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) No. of participants
(RCTs) Certainty of the evidence
(GRADE)
Risk with placebo or no treatment Risk with probiotic
eGFR
(mL/min)
Follow‐up: 8, 12 or 15 weeks
CKD stage 1 with type 2 diabetes mellitus 40 (1) ⊕⊝⊝⊝
very low1
The mean eGFR was 12.1 mL/min higher (4.19 higher to 20.01 higher) with probiotic than placebo or no treatment
CKD stage 3A with diabetes and hypertension 28 (1)
The mean eGFR mL/min was 0.4 higher (4.15 lower to 4.95 higher) with probiotic than placebo or no treatment
CKD stage G5D with diabetes and hypertension 60 (1)
The mean eGFR was 0.02 mL/min higher (0.63 lower to 0.67 higher) with probiotic than placebo or no treatment
Albuminuria
(g/dL)
Follow‐up: 12 or 24 weeks
CKD stage G5D 33 (1) ⊕⊕⊝⊝
low2
The mean albuminuria was 0 g/dL (0.19 lower to 0.19 higher) with probiotic than placebo or no treatment
CKD stage G5D with diabetes and hypertension 160 (3)
The mean albuminuria was 0.03 g/dL higher (0.1 lower to 0.16 higher) with probiotic than placebo or no treatment
Proteinuria
(mg/dL)
Follow‐up: 12 or 24 weeks
CKD stages 1 to 5 with diabetic nephropathy 60 (1) ⊕⊝⊝⊝
very low1
The mean albuminuria was 15.6 g/dL (34.3 lower to 3.1 higher) with probiotic than placebo or no treatment
Indoxyl sulfate
(mg/dL)
Follow‐up: 12 or 24 weeks
CKD stage G5D 33 (1) ⊕⊝⊝⊝
very low1
The mean indoxyl sulfate was 6 mg/dL lower (15.02 lower to 3.02 higher) with probiotic than placebo or no treatment
CKD stage G5D with diabetes and hypertension 50 (1) ⊕⊝⊝⊝
very low1
The mean indoxyl sulfate was 3.14 mg/dL lower (10.26 lower to 3.98 higher) with probiotic than placebo or no treatment
Change in any GI upset or intolerance (abdominal pain/diarrhoea)
Follow‐up: 12 weeks
CKD stage G5D RR 2.13
(0.21 to 21.22) 33 (1) ⊕⊝⊝⊝
very low1
59 per 1,000 125 per 1,000
(12 to 1,000)
Change in any GI upset or intolerance (constipation)
Follow‐up: 12 weeks
CKD stage G5D RR 3.18
(0.14 to 72.75) 33 (1) ⊕⊝⊝⊝
very low1
0 per 1,000 0 per 1,000
(0 to 0)
Microbiota composition
Graft function
Graft infection
*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CKD: chronic kidney disease; CI: confidence interval; eGFR: estimated glomerular filtration rate; GI: gastrointestinal; RR: risk ratio; RCT: randomised controlled trial
GRADE Working Group grades of evidenceHigh certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

1 Downgraded twice for serious risk of bias, and once for sparse or small study data.

2 Downgraded twice for serious risk of bias.

Background

Description of the condition

Chronic kidney disease

Chronic kidney disease (CKD) is defined as abnormalities of kidney structure or function, present for three months with implications for health (KDIGO 2013). The current classification for CKD has five stages (Appendix 1) classified based on two markers: (1) evidence of kidney damage (presence of proteinuria, microalbuminuria, or structural abnormality) and (2) the sustained impairment of estimated glomerular filtration rate (eGFR) for at least three months. Stages 1 to 3 are considered to be early CKD, at which point patients may have no or limited symptoms (with only urine or blood tests detecting the presence of kidney abnormality). Patients with CKD stages 4 and 5 have advanced‐stage kidney disease and may require or undergo dialysis or transplantation (KDIGO 2013).

The global prevalence of CKD is high, affecting 11% to 13% of the population (Hill 2016). In 2017, an estimated 1.23 million people died from kidney failure, a 33.7% increase from 2007 (GBD 2017). CKD predisposes the patient to a wide range of complications, including cardiovascular disease (CVD), infection, and cancer. Often CKD does not display symptoms until the disease is advanced (Jha 2013) and is therefore often considered to be underestimated as a comorbidity, making the exact prevalence and burden difficult to calculate.

The gut microbiome

The human microbiome is the collective genomes of the micro‐organisms in a particular environment (Valdes 2018) and is of emerging high interest in chronic disease research. The human gut microbiota includes fungi, bacteria, archaea, protozoa, and viruses that all interact with each other and the host to affect the health and physiology of the host (Azad 2018). The human intestine contains more than 10 billion micro‐organisms, and the microbial composition changes from person to person, along both the digestive tract and within the urinary and kidney environments (Aron‐Wisnewsky 2016). Recent culture‐independent studies that use high‐throughput sequencing have indicated that any microbial imbalances (otherwise known as gut dysbiosis or leaky gut) may be associated with cardiometabolic diseases in the long term (such as allergy, asthma, inflammatory bowel disease, celiac disease, systemic lupus erythematosus, arthritis, CKD, diabetes, obesity, and CVD) (Aron‐Wisnewsky 2016; Bromberg 2015).

In people with advanced stages of CKD, uraemia and metabolic acidosis alter the biochemical milieu, promoting disturbances in gut microbiota, the community or micro‐organisms themselves (Valdes 2018) and the intestinal barrier (Mafra 2019). These disturbances, referred to as gut dysbiosis, are further exacerbated by fluid retention with intestinal wall oedema, dietary restrictions and exposure to pharmacologic agents (particularly antibiotics) (Sampai‐Maia 2016). Gut dysbiosis, in turn, frequently leads to gastrointestinal (GI) symptoms (Chan 2020) and has, in turn, been linked with the progression of CKD, in particular, the production of putative uraemic toxins (e.g. indoxyl sulfate, p‐cresol sulfate, phenylacetylglutamine, trimethylamine‐N‐oxide, kynurenine), increased gut permeability, and transmural movement of bacteria and endotoxins leading to inflammation and oxidative stress (Beerepoot 2016; Cao 2022; Cremon 2018; Lehto 2018; Luyckx 2018).

Description of the intervention

Early observational and intervention studies have been investigating food‐intake patterns and various synbiotic interventions (antibiotics, prebiotics, or probiotics) to measure the effects on microbiota in treating cardiometabolic diseases, in particular CKD (Aron‐Wisnewsky 2016).

Prebiotics

The International Scientific Association for Probiotics and Prebiotics (Gibson 2017) defines prebiotics as substrates, or non‐digestible dietary substances, that are selectively utilised and fermented within the small intestine by host micro‐organisms. Modifying or diversifying the host microbiota may induce a health benefit to the host.

Most types of prebiotics are subsets of carbohydrate groups and mostly oligosaccharide carbohydrates (Davani‐Davari 2019).

  • Fructans: inulin and fructo‐oligosaccharides (stimulate the enrichment of native probiotics Lactobacilli and Bifidobacteria)

  • Galacto‐oligosaccharides (also known as trans‐galacto‐oligosaccharides): (stimulate the enrichment of native probiotics Lactobacilli, Bifidobacteria, Enterobacteria, Bacteroidetes, and Firmicutes)

  • Starch and glucose‐derived oligosaccharides: resistant starch, polydextrose

  • Other oligosaccharides: pectic‐oligosaccharide (from the polysaccharide pectin)

  • Non‐carbohydrate oligosaccharides: cocoa‐derived flavanols.

Natural sources of prebiotics can be obtained in peas, beans, cow's milk, human breast milk, soybean, rye, tomato, barley, wheat, honey, banana, onion, chicory, garlic, sugar beet, asparagus, and artichoke.

Probiotics

The term probiotics is used to describe live micro‐organisms that are intended to confer health benefits on the host when administered in adequate quantities (FAO/WHO 2002). The living bacteria may modulate the existing composition of gut microbiota to improve the health of the GI tract, the immune system, the inflammatory state and the "bioavailability of micronutrients" (Cremon 2018).

The key microbial organisms often found in probiotic treatments are:

  • Lactobacillus

  • Bifidobacterium

  • Saccharomyces

  • Streptococcus

  • Enterococcus

  • Escherichia

  • Bacillus

Natural sources of probiotics can be obtained in fermented foods such as yoghurt, kimchi, kombucha, sauerkraut, miso, pickles, raw apple cider vinegar, kefir, tempeh, some cheeses, and some sourdough breads.

Synbiotics

Synbiotics are the combination of prebiotics and probiotics in one treatment with the intention of producing a superior effect compared to either agent alone (Pan 2018). The effect is currently unknown.

Synthetic versions of synbiotics, prebiotics, and probiotics are available as oral capsules, tablets, liquids, or powder forms over‐the‐counter in most developed countries (Cremon 2018).

How the intervention might work

Growing research suggests that high doses of synbiotics, prebiotics, and probiotics are able to modify and improve dysbiosis of gut micro‐organisms by altering the population of the micro‐organisms. With the right balance of gut flora, a primary benefit is (believed to be) the suppression of pathogens through immunostimulation and gut barrier enhancement (reduced permeability of the gut) (Cremon 2018).

Gut microbiota ferments prebiotics and produces short‐chain fatty acids (lactic acid, butyric acid, propionic acid), which have positive effects on the airways and dendritic cells in the bone marrow, and decrease the pH of the colon (Davani‐Davari 2019). Prebiotics also decrease the gut pH, resulting in the butyrogenic effect ‐ where a slight change in the unit of change in pH alters the entire composition or population of acid‐sensitive species (Bacterioides) and promotes butyrate formation of Firmicutes (Davani‐Davari 2019).

Probiotics alter the intestinal pH, inhibit pathogens (via the generation of antibacterial compounds, competitively eliminating pathogens in receptor binding sites and competing for available nutrients), inhibit the production of mutagenic and carcinogenic substances, and maintain the intestinal barrier (Kato 2008).

Why it is important to do this review

Prebiotics and probiotics are freely available as over‐the‐counter purchases in most high‐income countries and are being used as therapeutic supplements for improving the function and balance of gut microbiota in the general population. Whilst many positive effects have been identified, the exact mechanism of action by which these compounds exert their beneficial actions in humans is only partially understood (Cremon 2018). In the general population, there is no definitive data to support the use of synbiotics, prebiotics, or probiotics. In CKD, there are uncertain effects in people with reduced kidney function because of the risk of catastrophic infections from the live micro‐organisms in patients who are immunocompromised. The efficacy of these interventions and the certainty of the evidence in CKD patients remains unknown, and it is imperative to synthesise the benefits and harms associated with these treatments.

Objectives

This review aims to look at the benefits and harms of synbiotics, prebiotics, and probiotics for people with CKD.

Methods

Criteria for considering studies for this review

Types of studies

All randomised controlled trials (RCTs) and quasi‐RCTs (RCTs in which allocation to treatment was obtained by alternation, use of alternate medical records, date of birth or other predictable methods) and cluster RCTs were included. Unblinded, single and double‐blind studies were included.

Cross‐over studies were included, and only data from the first phase was used for analysis.

Full journal publication and peer review were required. Unpublished clinical studies with online results available were, however, included.

Studies in any healthcare setting were included.

Excluded study designs: abstracts, single‐arm studies, commentaries, editorials, and clinical observations.

Types of participants

Inclusion criteria
  • Adults and children with CKD (stages 1 to 5), receiving dialysis, and kidney transplant recipients.

  • Studies of populations with altered GI function and diabetic kidney disease were included and analysed as subgroups.

Exclusion criteria
  • Studies of populations receiving enteral nutrition.

  • Adults and children who have signs of systemic illness (such as fever, loin pain, toxicity).

Types of interventions

  • Any synbiotic, prebiotic, or probiotic treatment compared to another, other pharmacological, non‐pharmacological, placebo, or no treatment

  • Any route of administration, dose, duration, or frequency

  • Formulations such as oral tablets and capsules, liquids, and powders

  • Combination therapies of synbiotics, prebiotics, or probiotics with other pharmacological treatments or non‐pharmacological treatments were analysed as separate comparisons.

Participants receiving concurrent pharmacological medications for co‐morbidities, such as blood glucose medications, blood pressure medications, and immunosuppressants, were included, and we planned to analyse these as subgroups.

Studies of high‐dose prebiotics for the purpose of purgation and studies of dietary changes were excluded.

Comparison pairs for analysis
  • A synbiotic, prebiotic, or probiotic treatment versus another synbiotic, prebiotic, or probiotic

  • A synbiotic, prebiotic, or probiotic treatment versus any other pharmacological comparator (antibiotics, immunosuppressants, other medicines)

  • A synbiotic, prebiotic, or probiotic treatment versus another non‐pharmacological comparator (dietary, educational, behavioural, vitamin or herbal supplements, Traditional Chinese Medicine)

  • Any synbiotic, prebiotic, or probiotic treatment versus placebo

  • Any synbiotic, prebiotic, or probiotic treatment versus no treatment

  • Any synbiotic, prebiotic, or probiotic treatment versus a combination treatment (any of the above)

  • Any synbiotic, prebiotic, or probiotic treatment in combination with any of the above versus any of the above comparators.

For each of these comparisons, synbiotics, prebiotics, and probiotics were analysed as separate comparisons.

Types of outcome measures

This review did not exclude studies based on non‐reporting of outcomes of interest.

The outcomes selected include the relevant SONG core outcome sets as specified by the Standardised Outcomes in Nephrology initiative (SONG 2017).

Primary outcomes
  1. Kidney function: eGFR; serum creatinine (SCr); albuminuria; proteinuria; infection (including pyelonephritis or urosepsis)

  2. Uraemic toxins: urea; indoxyl sulfate; p‐cresyl sulfate; trimethylamine N‐oxide; phenylacetylglutamine; kynurenine

  3. GI function: change in any GI upset or intolerance; microbiota composition; faecal characteristics (such as the Bristol Stool Chart) (Lewis 1997); colonic transit time.

Secondary outcomes
  1. Dialysis outcomes: peritoneal dialysis (PD) or haemodialysis (HD) infection; vascular access; technique survival; dialysis failure

  2. Transplant function: need for transplant; graft survival/health

  3. Patient‐reported outcomes: pain rating using any validated pain scale; quality of life (QoL) (using any validated scale); fatigue; life participation

  4. Adverse events: any adverse events (including infection); serious adverse events (including death); withdrawals due to adverse events. Potential (but not limited to) events include GI responses, nausea, vomiting, diarrhoea, and constipation

  5. CVD

  6. Cancer

Search methods for identification of studies

Electronic searches

We searched the Cochrane Kidney and Transplant Register of Studies up to 9 October 2023 through contact with the Information Specialist using search terms relevant to this review. The Register contains studies identified from the following sources.

  1. Monthly searches of the Cochrane Central Register of Controlled Trials (CENTRAL)

  2. Weekly searches of MEDLINE OVID SP

  3. Searches of kidney and transplant journals and the proceedings and abstracts from major kidney and transplant conferences

  4. Searching of the current year of EMBASE OVID SP

  5. Weekly current awareness alerts for selected kidney and transplant journals

  6. Searches of the International Clinical Trials Registry Platform (ICTRP) Search Portal and ClinicalTrials.gov.

Studies contained in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE based on the scope of Cochrane Kidney and Transplant. Details of search strategies, as well as a list of handsearched journals, conference proceedings and current awareness alerts, are available on the Cochrane Kidney and Transplant website.

See Appendix 2 for search terms used in strategies for this review.

Searching other resources

  1. Reference lists of review articles, relevant studies, and clinical practice guidelines.

  2. Contacting relevant individuals/organisations seeking information about unpublished or incomplete studies.

  3. Grey literature sources (e.g. abstracts, dissertations, and theses), in addition to those already included in the Cochrane Kidney and Transplant Register of Studies, were not searched.

Data collection and analysis

Selection of studies

The search strategy described was used to obtain titles and abstracts of studies that may be relevant to the review. The titles and abstracts were screened independently by two authors, who discarded studies that were not applicable; however, studies and reviews that might include relevant data or information on studies were retained initially. Two authors independently assessed the retrieved abstracts and, where necessary, the full text of these studies to determine which studies satisfied the inclusion criteria. Disagreements were resolved in consultation with a third author.

Data extraction and management

Data extraction was carried out independently by two authors using standard data extraction forms. Disagreements were resolved in consultation with a third author. Studies reported in non‐English language journals were translated before assessment. Where more than one publication of one study existed, reports were grouped together, and the publication with the most complete data was used in the analyses. Where relevant outcomes were only published in earlier versions, these data were used. Any discrepancy between published versions was to be highlighted.

Assessment of risk of bias in included studies

The following items were independently assessed by two authors using the risk of bias assessment tool (Higgins 2022) (see Appendix 3).

  • Was there adequate sequence generation (selection bias)?

  • Was allocation adequately concealed (selection bias)?

  • Was knowledge of the allocated interventions adequately prevented during the study?

    • Participants and personnel (performance bias)

    • Outcome assessors (detection bias)

  • Were incomplete outcome data adequately addressed (attrition bias)?

  • Are reports of the study free of suggestion of selective outcome reporting (reporting bias)?

  • Was the study apparently free of other problems that could put it at risk of bias?

Measures of treatment effect

For dichotomous outcomes (e.g. progression to CKD stage), comparisons between groups were based on risk ratios (RR), the number needed to treat for an additional beneficial outcome (NNT) and pooled differences as absolute measures of effect with 95% confidence intervals (CI).

Where continuous scales of measurement were used to assess the effects of treatment (e.g. pain or decline in kidney function), the mean difference (MD) was used, or the standardised mean difference (SMD) if different scales had been used.

Where possible, we used the mean change score from baseline. We anticipated that some studies may only report the mean endpoint score of which we planned to use the final time point available.

Unit of analysis issues

We only accepted randomisation of the individual participants. For multiple‐dose studies, we used data for the first dose only. For cross‐over studies, we used data from the first phase only.

Dealing with missing data

Any further information required from the original author was requested by written correspondence (e.g. emailing the corresponding author), and any relevant information obtained in this manner was included in the review. Evaluation of important numerical data such as screened, randomised patients as well as intention‐to‐treat (ITT), as‐treated and per‐protocol population was carefully performed. Attrition rates, for example, drop‐outs, losses to follow‐up and withdrawals, were investigated. Issues of missing data and imputation methods (for example, last‐observation‐carried‐forward) were critically appraised (Higgins 2022).

Assessment of heterogeneity

We first assessed the heterogeneity by visual inspection of the forest plot. We quantified statistical heterogeneity using the I² statistic, which describes the percentage of total variation across studies that was due to heterogeneity rather than sampling error (Higgins 2003). A guide to the interpretation of I² values is as follows.

  • 0% to 40%: might not be important

  • 30% to 60%: may represent moderate heterogeneity

  • 50% to 90%: may represent substantial heterogeneity

  • 75% to 100%: considerable heterogeneity.

The importance of the observed value of I² depends on the magnitude and direction of treatment effects and the strength of evidence for heterogeneity (e.g. P‐value from the Chi² test, or a CI for I²) (Higgins 2022).

Assessment of reporting biases

Where possible, funnel plots were planned to be used to assess for the potential existence of small study bias (Higgins 2022).

Data synthesis

Data were pooled using random‐effects models. Fixed‐effect models were also fitted to investigate potential discrepancies with the random‐effects models (e.g., the influence of large studies on the pooled estimates).

Subgroup analysis and investigation of heterogeneity

Subgroup analysis and meta‐regression were planned to be used to explore possible sources of heterogeneity where there was sufficient data. Heterogeneity among participants could be related to the distribution of age, co‐morbidities, and cause of kidney disease. Heterogeneity in treatments could be related to prior agent(s) used and the agent, dose, and duration of therapy. Adverse effects were tabulated and assessed with descriptive techniques, as they were likely to be different for the various agents used (this was conducted as part of the meta‐analysis as adverse events were a secondary outcome). Where possible, the risk difference (RD) with 95% CI was calculated for each adverse event, either compared to no treatment or to another agent.

Planned subgroups where sufficient data were available.

  • CKD stage: 1 to 5 pre‐dialysis, dialysis, transplant, diabetes, hypertension

  • Dose (to be determined upon presentation of available data)

  • Timepoint: short‐term, long‐term (to be determined upon presentation of available data)

  • Level of GI function or GI issues (to be determined upon presentation of available data)

  • Age: children (< 18 years), adults (> 18 years).

Sensitivity analysis

We planned to undertake sensitivity analyses (however, this was not possible) to explore the influence of the following factors on effect size.

  • Repeating the analysis, excluding unpublished studies

  • Repeating the analysis, taking account of the risk of bias, as specified

  • Repeating the analysis, excluding any very long or large studies to establish how much they dominate the results

  • Repeating the analysis excluding studies using the following filters: diagnostic criteria, language of publication, source of funding (industry versus other), and country.

Summary of findings and assessment of the certainty of the evidence

We presented the main results of the review in 'Summary of findings' tables. These tables present key information concerning the certainty of the evidence, the magnitude of the effects of the interventions examined, and the sum of the available data for the main outcomes (Schünemann 2022a).

The 'Summary of findings' tables also include an overall grading of the evidence related to each of the main outcomes using the GRADE (Grades of Recommendation, Assessment, Development and Evaluation) approach (GRADE 2008; GRADE 2011). The GRADE approach defines the certainty of a body of evidence as the extent to which one can be confident that an estimate of effect or association is close to the true quantity of specific interest. This was assessed by two authors. A summary of the assessment process is in Appendix 4. The certainty of a body of evidence involves consideration of within‐trial risk of bias (methodological quality), directness of evidence, heterogeneity, precision of effect estimates and risk of publication bias (Schünemann 2022b). We planned to present the following outcomes in the 'Summary of findings' tables.

  • Changes in kidney function: eGFR

  • Changes in kidney function: kidney damage (albuminuria, proteinuria)

  • Uraemic toxins: free and protein‐bound concentrations of serum indoxyl sulfate

  • GI function: change in any GI upset or intolerance

  • GI function: microbiota composition

  • Transplant function: graft function

  • Transplant function: graft infection

Results

Description of studies

The following section contains broad descriptions of the studies considered in this review. For further details on each individual study, please see the characteristics of studies tables (Characteristics of included studies; Characteristics of excluded studies; Characteristics of studies awaiting classification; Characteristics of ongoing studies).

Results of the search

Our search of the Specialised Register up to 9 October 2023 identified a total of 161 records. After screening titles and abstracts and full‐text review, 45 studies (88 reports) were included, 21 studies (29 reports) were excluded, 12 studies were identified as ongoing, and 12 abstracts (17 reports) are awaiting classification. We will include ongoing and abstracts awaiting classification in a future update of this review (Figure 1).

1.

1

Study flow diagram.

Included studies

Forty‐five studies, randomising 2274 participants, met our inclusion criteria (Characteristics of included studies).

Thirty‐seven studies were single‐centre, and eight studies were multicentre. All studies took place in a research centre or hospital outpatient setting in Australia, Belgium, Brazil, Canada, China, France, Indonesia, Iran, Italy, Mexico, Saudi Arabia, South Africa, and the USA.

Sample sizes ranged from 13 (Biruete 2017) to 124 (Xie 2015a) participants.

Twenty‐five studies investigated participants with CKD stage 5D (dialysis), 18 studies investigated participants with stages 1 to 5 non‐dialysis, and two studies investigated kidney transplant or simultaneous pancreas‐kidney (SPK) transplant recipients (Guida 2017; PREBIOTIC 2022). Twenty‐nine studies included participants who had diabetes, and 24 studies included participants who had hypertension.

Forty‐three studies investigated adults 18 years and older, and two studies investigated adults and children of all ages (Mirzaeian 2020; Xie 2015a).

Thirty‐two studies compared two parallel arms, three studies compared three parallel arms (Elamin 2017; Haghighat 2019; Xie 2015a), 10 studies compared a cross‐over treatment with either a washout period (Biruete 2017; de Andrade 2021; Esgalhado 2018; He 2022; Li 2020; Natarajan 2014; Poesen 2016; SYNERGY 2014) or no washout period (Bliss 1992; Ranganathan 2009).

Comparisons

Twenty‐eight studies contributed data to our meta‐analyses, 11 studies did not report data for our planned outcomes or data was not reported in a useable way to be meta‐analysed (de Araujo 2022; Ebrahim 2022; Elamin 2017; Kooshki 2019; Lydia 2022; Miraghajani 2017; Pan 2018; PREBIOTIC 2022; Shariaty 2017; Soleimani 2017; Xie 2015a), and six were cross‐over studies and did not report separate data for the first phase of the study (de Andrade 2021; He 2022; Natarajan 2014; Poesen 2016; Ranganathan 2009; SYNERGY 2014).

Excluded studies

Following full‐text review, we excluded 21 studies (Characteristics of excluded studies). Eleven studies were undertaken in the wrong population, nine studies investigated the wrong intervention, and one study was of ineligible study design.

Ongoing studies and studies awaiting classification

Twenty‐six ongoing studies were identified on trial registries or had published an a priori protocol (Characteristics of ongoing studies), and 12 abstracts were identified (Characteristics of studies awaiting classification). These studies will be assessed in a future update of this review.

Risk of bias in included studies

See Figure 2 for a graphical summary of the risk of bias assessment within each study.

2.

2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Most studies were characterised by an unclear risk of bias across most domains due to the lack of information and detail provided to permit judgement.

Using funnel plots to detect publication bias was not feasible due to the lack of available data to analyse quantitatively.

Allocation

Random sequence generation

Thirty‐six studies were judged to have a low risk of bias for providing an adequate description of how their randomisation methods were undertaken.

Nine studies were judged to have an unclear risk of bias due to a lack of information provided on their randomisation methods, although stated to be randomised.

Allocation concealment

Twenty‐four studies were judged to have a low risk of bias for providing an adequate description of how allocations were concealed.

Two studies were judged to be at high risk of bias due to being open‐label and appeared to have no approach to concealing allocation (Ebrahim 2022; Meng 2019).

Nineteen studies were judged to have an unclear risk of bias due to a lack of information provided on how allocations were concealed.

Blinding

Performance bias

Twenty‐six studies were judged to have a low risk of bias for clearly reporting how both the study participants were kept blind to treatments as well as the study personnel.

Ten studies were judged to be at high risk of bias due to being open‐label (Bliss 1992; Cosola 2021; Ebrahim 2022; Elamin 2017; Lopes 2018; Meng 2019; Miranda Alatriste 2014; Pan 2021; ProbiotiCKD 2019; Xie 2015a).

Nine studies were judged to have an unclear risk of bias due to a lack of information provided on blinding methods, although stated to be double‐blind.

Detection bias

Fifteen studies were judged to have a low risk of bias for clearly reporting how the outcomes were kept blind to treatments.

Nine studies were judged to be at high risk of bias due to being open‐label (Bliss 1992; Cosola 2021; Elamin 2017; Lopes 2018; Meng 2019; Miranda Alatriste 2014; Pan 2021; ProbiotiCKD 2019; Xie 2015a). Ebrahim 2022 was blinded to investigators and therefore judged unclear rather than high risk.

Twenty‐one studies were judged to have an unclear risk of bias due to a lack of information provided on blinding methods for outcome assessors, although stated to be double‐blind.

Incomplete outcome data

Eleven studies were judged to be at low risk of bias for adequately accounting for all participants throughout the study, providing reasons for withdrawals, having low attrition rates, and undertaking an ITT analysis.

Thirty‐one studies were judged to be at high risk of bias due to either not accounting for all participants throughout the study and providing reasons for withdrawals, high attrition rates (ranging from 40% to 50% and higher), or for not undertaking an ITT analysis.

Three studies were judged to have unclear risk of bias as no information about withdrawals, attrition, or ITT analysis could be identified in the available text (Elamin 2017; Esgalhado 2018; Kooshki 2019).

Selective reporting

Twenty‐six studies were judged to be at low risk of bias for providing a trial registration number or had an a priori published protocol.

Five studies were judged to be at high risk of bias due to serious changes to study outcomes and analyses that were not explained (Abbasi 2017; Cosola 2021; Dehghani 2016; Miranda Alatriste 2014; Viramontes‐Horner 2015).

Fourteen studies were judged to have an unclear risk of bias as no information was available about trial registration or an a priori published protocol.

Other potential sources of bias

Control for confounding factors

Seventeen studies were judged to be at low risk of bias for adequately describing control for standardising dietary intake and/or ingesting synbiotics, prebiotics, or probiotics where possible for the pre‐study run‐in and during study periods.

Twenty‐eight studies were judged to have an unclear risk of bias for not reporting any information about whether diet or medications were controlled for pre‐ or during study periods.

Other biases

Seventeen studies were judged to be at low risk of bias due to providing information on funding (non‐conflicting) and reporting disclosures (non‐conflicting).

Twenty‐two studies were judged to be at high risk of bias due to conflicting funding (industry or otherwise).

Six studies were judged to have unclear risk of bias as no information was available about funding or disclosures, or only information about one of these was provided.

Effects of interventions

See: Table 1; Table 2; Table 3; Table 4; Table 5; Table 6; Table 7; Table 8

Adverse events are reported in Appendix 5.

1: Synbiotic versus synbiotic

Haghighat 2019 compared the same probiotics (Lactobacillusacidophilus T16, Bifidobacteriumbifidum BIA‐6, Bifidobacteriumlactis BIA‐6, and Bifidobacteriumlongum LAF‐5) with two different prebiotics (fructo‐oligosaccharides plus galacto‐oligosaccharides plus inulin compared to maltodextrin).

Patient‐reported outcomes
Health‐related quality of life

Haghighat 2019 reported no difference in health‐related QoL (HRQoL‐14) at 12 weeks in patients receiving synbiotics compared to another synbiotic of a different dose (Analysis 1.1 (1 study, 46 participants): MD 1.98, 95% CI ‐11.2 to 15.08; very low certainty evidence).

1.1. Analysis.

1.1

Comparison 1: Synbiotic versus synbiotic, Outcome 1: Patient‐reported outcomes: HRQoL at 12 weeks

Adverse events

Adverse events were minimal and non‐serious, and withdrawals were not related to the study interventions (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for this comparison.

2: Synbiotic versus prebiotic

Six studies contributed to this comparison.

  • Guida 2014

    • Synbiotic: Lactobacillusplantarum, Lactobacilluscasei subsp. rhamnosus, Lactobacillus gasseri, Bifidobacteriuminfantis,Bifidobactetriumlongum, Lactobacillusacidophilus, Lactobacillussalivarius,Lactobacillussporogenes and Streptococcusthermophilus plus prebiotics inulin and tapioca‐resistant starch

    • Prebiotic: tapioca‐resistant starch

  • Guida 2017

    • Synbiotic: Lactobacillusplantarum,Lactobacilluscasei subsp. rhamnosus,Lactobacillusgasseri, Bifidobacteriuminfantis,Bifidobacteriumlongum, Lactobacillusacidophilus, Lactobacillussalivarius, Lactobacillussporogenes, Streptococcusthermophilus plus inulin and tapioca‐resistant starch

    • Prebiotic: cellulose

  • Haghighat 2019

    • Synbiotic 1: Lactobacillusacidophilus T16, Bifidobacteriumbifidum BIA‐6, Bifidobacteriumlactis BIA‐6, and Bifidobacteriumlongum LAF‐5 plus fructo‐oligosaccharides, galacto‐oligosaccharides and inulin

    • Synbiotic 2: Lactobacillusacidophilus T16, Bifidobacteriumbifidum BIA‐6, Bifidobacteriumlactis BIA‐6, and Bifidobacteriumlongum LAF‐5 plus maltodextrin

    • Prebiotic: maltodextrin

  • Lopes 2018

    • Synbiotic: Bifidobacterium longum BL‐G301 plus extruded sorghum flakes

    • Prebiotic: extruded corn flakes

  • Mirzaeian 2020

    • Synbiotic: Lactobacillus casei, Lactobacillus acidophilus, Lactobacillus rhamnosus, Lactobacillus bulgaricus, Bifidobacterium breve, Bifidobacterium longum, and Streptococcus thermophiles plus fructo‐oligosaccharide lactose

    • Prebiotic: maltodextrin

  • SYNERGY II 2021

    • Synbiotic: Lactobacillus, Bifidobacteria, and Streptococcus plus high‐resistant starch fibre supplement

    • Prebiotic: maltodextrin.

Kidney function
Estimated glomerular filtration rate

Guida 2017 reported no difference in eGFR at four weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.1(1 study, 34 participants): MD ‐3.80 mL/min/1.73 m², 95% CI ‐17.98 to 10.38; very low certainty evidence).

2.1. Analysis.

2.1

Comparison 2: Synbiotic versus prebiotic, Outcome 1: Kidney function: eGFR at 4 weeks

SYNERGY II 2021 reported that patients receiving synbiotic supplementation resulted in a mean reduction in eGFR of ‐3.14 mL/min/1.73 m2 (95% CI ‐6.23 to ‐0.06) at 12 months from baseline (prebiotics had a mean increase of 2.61 mL/min/1.73 m2; 95% CI ‐0.41 to 5.63) (n = 56; P < 0.01).

Serum creatinine

Mirzaeian 2020 reported no difference in SCr at eight weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.2 (1 study, 42 participants): MD ‐0.20 mg/dL, 95% CI ‐1.50 to 1.10; very low certainty evidence).

2.2. Analysis.

2.2

Comparison 2: Synbiotic versus prebiotic, Outcome 2: Kidney function: serum creatinine at 8 weeks

SYNERGY II 2021 reported synbiotic supplementation resulted in a mean increase in SCr concentration of 20.8 μmol (95% CI 2.97 to 38.5) at 12 months from baseline (prebiotics had a mean decrease of ‐9.79 μmol; 95% CI 21.7 to 2.09) (n = 56; P < 0.01).

Uraemic toxins
Urea

Mirzaeian 2020 reported no difference in urea at four weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.3 (1 study, 42 participants): MD ‐2.10 mg/dL, 95% CI ‐13.93 to 9.73; very low certainty evidence).

2.3. Analysis.

2.3

Comparison 2: Synbiotic versus prebiotic, Outcome 3: Uraemic toxins: urea at 4 weeks

Indoxyl sulfate

Mirzaeian 2020 reported no difference in indoxyl sulfate at four weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.4 (1 study, 42 participants): MD 128.30 ng/mL, 95% CI ‐242.77 to 499.37; very low certainty evidence).

2.4. Analysis.

2.4

Comparison 2: Synbiotic versus prebiotic, Outcome 4: Uraemic toxins: indoxyl sulfate at 4 weeks

SYNERGY II 2021 reported no difference in total indoxyl sulfate at 12 months in patients receiving synbiotic supplementation (mean change 1.5 μmol/L; 95% CI ‐3.25 to 6.26) compared to prebiotics (mean change ‐3.07 μmol/L; 95% CI ‐9.14 to 2.99) mean change from baseline (n = 56; P = 0.96).

SYNERGY II 2021 reported no difference in free indoxyl sulfate at 12 months in patients receiving synbiotic supplementation (mean change 0.1 μmol/L; 95% CI ‐0.14 to 0.34) compared to prebiotics (mean change ‐0.09 μmol/L; 95% CI ‐0.34 to 0.17) mean change from baseline (n = 56; p = 0.25).

P‐cresyl sulfate

Guida 2014 reported p‐cresyl sulfate was lower at four weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.5 (1 study, 34 participants): MD ‐2.10 μg/mL, 95% CI ‐3.92 to ‐0.28; very low certainty evidence).

2.5. Analysis.

2.5

Comparison 2: Synbiotic versus prebiotic, Outcome 5: Uraemic toxins: p‐cresol sulfate at 4 weeks

SYNERGY II 2021 reported no difference in total p‐cresyl sulfate at 12 months in patients receiving synbiotic supplementation (mean change 28.8 μmol/L; 95% CI ‐6.32 to 64.0) compared to prebiotics (mean change ‐17.2 μmol/L; 95% CI ‐49.8 to 15.3) mean change from baseline (n = 56; P = 0.15).

SYNERGY II 2021 reported no difference in free p‐cresyl sulfate at 12 months in patients receiving synbiotic supplementation (mean change 0.98 μmol/L; 95% CI 0.17 to 1.79) compared to prebiotics (mean change ‐0.17 μmol/L; 95% CI ‐1.18 to 0.84) mean change from baseline (n = 56; P = 0.08).

Gastrointestinal function
Change in any gastrointestinal upset or intolerance

Guida 2017 reported prebiotics decreased borborygmi at four weeks compared to synbiotics (Analysis 2.6 (1 study, 34 participants): RR 15.26, 95% CI 0.99 to 236.23; very low certainty evidence).

2.6. Analysis.

2.6

Comparison 2: Synbiotic versus prebiotic, Outcome 6: GI function: change in any GI upset or intolerance (prevalence of borborygmi) at 4 weeks

SYNERGY II 2021 reported no difference in change in GI symptoms according to the Gastrointestinal Symptom Rating Scale (GSRS) Total Index (scale of 1 to 7, a higher score indicates worse discomfort) at 12 months in patients receiving synbiotics compared to prebiotics (Analysis 2.7 (1 study, 56 participants): MD 0.00, 95% CI ‐0.27 to 0.27; very low certainty evidence). SYNERGY II 2021 also reported no difference in the subdomains of the GSRS in patients receiving synbiotics compared to prebiotics, respectively (mean score ± SD): reflux (1.4 ± 0.8, 1.3 ± 0.4); abdominal pain (1.2 ± 0.4, 1.3 ± 0.4); indigestion (1.7 ± 0.9, 1.6 ± 0.5); constipation (1.4 ± 1.0, 1.5 ± 0.8); diarrhoea (1.3 ± 0.5, 1.2 ± 0.4).

2.7. Analysis.

2.7

Comparison 2: Synbiotic versus prebiotic, Outcome 7: GI function: change in any GI upset or intolerance (GSRS Total Index at 12 months)

Microbiota composition

SYNERGY II 2021 reported no difference in change in microbiota composition (Richness and Shannon's index) at 12 months in patients receiving synbiotics compared to prebiotics (n = 56). SYNERGY II 2021 reported, "The coverage of the microbiota diversity for all samples was high with a rarefied sequencing depth of 4 million reads with 298 species identified".

Faecal characteristics

Lopes 2018 reported faecal pH at seven weeks was lower in patients receiving synbiotics compared to prebiotics (Analysis 2.8 (1 study, 58 participants): MD ‐0.63, 95% CI ‐1.13 to ‐0.13; very low certainty evidence).

2.8. Analysis.

2.8

Comparison 2: Synbiotic versus prebiotic, Outcome 8: GI function: faecal pH at 7 weeks

Guida 2017 reported no difference in stool shape and characteristics on the Bristol Stool Chart (scale of 1 to 7) at four weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.9 (1 study, 34 participants): MD ‐0.50, 95% CI ‐1.15 to 0.15; very low certainty evidence).

2.9. Analysis.

2.9

Comparison 2: Synbiotic versus prebiotic, Outcome 9: GI function: faecal characteristics (Bristol Stool Chart) at 4 weeks

SYNERGY II 2021 reported no difference in stool consistency on the Bristol Stool Chart at 12 months in patients receiving synbiotics compared to prebiotics (Analysis 2.10 (1 study, 56 participants): MD 0.50, 95% CI ‐0.18 to 1.18; very low certainty evidence).

2.10. Analysis.

2.10

Comparison 2: Synbiotic versus prebiotic, Outcome 10: GI function: faecal characteristics (Bristol Stool Chart) at 12 months

SYNERGY II 2021 reported no difference in stool frequency (patient‐reported number of times their bowels opened in the previous 24‐hour period) at 12 months in patients receiving synbiotics compared to prebiotics, respectively (1.8 ± 1.0, 1.6 ± 0.9; n = 56).

Patient‐reported outcomes
Health‐related quality of life

Haghighat 2019 reported no difference in HRQoL (HRQoL‐14) scores at 12 weeks in patients receiving synbiotics compared to prebiotics (Analysis 2.11 (2 studies, 65 participants): MD 6.38, 95% CI ‐4.88 to 17.64; very low certainty evidence).

2.11. Analysis.

2.11

Comparison 2: Synbiotic versus prebiotic, Outcome 11: Patient‐reported outcomes: HRQoL at 24 weeks

SYNERGY II 2021 reported no difference in HRQoL (Assessment of Quality of Life‐4 Dimension, 5‐point scale (AQoL‐4D)) at 12 months in patients receiving synbiotics compared to prebiotics, respectively (median (IQR): 16.0 (13.0 to 18.0), 15.0 (13.0 to 17.0); n = 56).

Adverse events

It is uncertain whether synbiotics decreased adverse events compared to prebiotics (Analysis 2.12 (5 studies, 279 participants): very low certainty evidence). It is uncertain whether synbiotics decreased withdrawals due to adverse events compared to prebiotics (Analysis 2.13 (4 studies, 211 participants): very low certainty evidence).

2.12. Analysis.

2.12

Comparison 2: Synbiotic versus prebiotic, Outcome 12: Adverse events: any adverse event

2.13. Analysis.

2.13

Comparison 2: Synbiotic versus prebiotic, Outcome 13: Withdrawals due to adverse events

Mirzaeian 2020 reported one death in each arm; however, they were not related to the study interventions.

The remaining adverse events were minimal and non‐serious, and withdrawals were not related to study treatment (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

Subgroups were not estimable for the remaining analyses in this comparison. In Analysis 2.12 and Analysis 2.13, totals were turned off due to zero event rate data and are also not estimable.

3: Prebiotic versus prebiotic

Five studies contributed to this comparison.

  • Biruete 2017 compared inulin to maltodextrin

  • Bliss 1992 compared gum Arabic to pectin

  • Li 2020 compared 50:50 inulin plus oligofructose to maltodextrin

  • Ramos 2019 compared fructo‐oligosaccharide to maltodextrin

  • Sirich 2014 compared high‐amylose corn starch to high‐amylopectin starch.

Kidney function
Estimated glomerular filtration rate

Ramos 2019 reported no difference in eGFR at 12 weeks in patients receiving fructo‐oligosaccharide compared to maltodextrin of a different dose (Analysis 3.1 (1 study, 50 participants): MD 0.00 mL/min, 95% CI ‐1.73 to 1.73; very low certainty evidence).

3.1. Analysis.

3.1

Comparison 3: Prebiotic versus prebiotic, Outcome 1: Kidney function: eGFR at 12 weeks

Uraemic toxins
Indoxyl sulfate

It is uncertain whether prebiotic 1 (inulin or high‐amylose corn starch) improves indoxyl sulfate at six weeks compared to prebiotic 2 (maltodextrin or high‐amylopectin starch) (Analysis 3.2 (2 studies, 64 participants): MD ‐0.20 μg/mL, 95% CI ‐1.01 to 0.61; I² = 0%; very low certainty evidence).

3.2. Analysis.

3.2

Comparison 3: Prebiotic versus prebiotic, Outcome 2: Uraemic toxins: indoxyl sulfate at 6 weeks

Uraemic toxins
P‐cresyl sulfate

It is uncertain whether prebiotic 1 (inulin or high‐amylose corn starch) improves indoxyl sulfate at six weeks compared to prebiotic 2 (maltodextrin or high‐amylopectin starch) (Analysis 3.3 (2 studies, 64 participants): SMD ‐0.04 μg/mL, 95% CI ‐0.53 to 0.45; I² = 0%; very low certainty evidence).

3.3. Analysis.

3.3

Comparison 3: Prebiotic versus prebiotic, Outcome 3: Uraemic toxins: p‐cresol sulfate at 6 weeks

Gastrointestinal function
Change in any gastrointestinal upset or intolerance

In very low certainty evidence, Biruete 2017 reported no difference in patients receiving inulin compared to maltodextrin at four weeks for burping (Analysis 3.4 (1 study, 24 participants): MD 0.17, 95% CI ‐0.50 to 0.84), cramping (Analysis 3.5 (1 study, 24 participants): MD ‐0.17, 95% CI ‐0.50 to 0.16), distension (Analysis 3.6 (1 study, 24 participants): MD 0.33, 95% CI ‐0.04 to 0.70), flatulence (Analysis 3.7 (1 study, 24 participants): MD 1.00, 95% CI 0.25 to 1.75), nausea (Analysis 3.8 (1 study, 24 participants): MD 0.00, 95% CI ‐0.00 to 0.00), reflux (Analysis 3.9 (1 study, 24 participants): MD 0.00, 95% CI ‐0.50 to 0.50), and rumbling (Analysis 3.10 (1 study, 24 participants): MD 0.50, 95% CI ‐0.14 to 1.14).

3.4. Analysis.

3.4

Comparison 3: Prebiotic versus prebiotic, Outcome 4: GI function: change in any GI upset or intolerance (burping) at 4 weeks

3.5. Analysis.

3.5

Comparison 3: Prebiotic versus prebiotic, Outcome 5: GI function: change in any GI upset or intolerance (cramping) at 4 weeks

3.6. Analysis.

3.6

Comparison 3: Prebiotic versus prebiotic, Outcome 6: GI function: change in any GI upset or intolerance (distension) at 4 weeks

3.7. Analysis.

3.7

Comparison 3: Prebiotic versus prebiotic, Outcome 7: GI function: change in any GI upset or intolerance (flatulence) at 4 weeks

3.8. Analysis.

3.8

Comparison 3: Prebiotic versus prebiotic, Outcome 8: GI function: change in any GI upset or intolerance (nausea) at 4 weeks

3.9. Analysis.

3.9

Comparison 3: Prebiotic versus prebiotic, Outcome 9: GI function: change in any GI upset or intolerance (reflux) at 4 weeks

3.10. Analysis.

3.10

Comparison 3: Prebiotic versus prebiotic, Outcome 10: GI function: change in any GI upset or intolerance (rumblings) at 4 weeks

Microbiota composition

In very low certainty evidence, Biruete 2017 reported no difference in patients receiving inulin compared to maltodextrin at four weeks for actinobacteria Analysis 3.11 (1 study, 24 participants): MD ‐1.26, 95% CI ‐4.46 to 1.94), bacteriodetes (Analysis 3.12 (1 study, 24 participants): MD 3.23, 95% CI ‐8.24 to 14.70), proteobacteria (Analysis 3.13 (1 study, 24 participants): MD 0.11, 95% CI ‐1.61 to 1.83), firmicutes (Analysis 3.14 (1 study, 24 participants): MD ‐2.44, 95% CI ‐14.19 to 9.31), synergistetes (Analysis 3.15 (1 study, 24 participants): MD ‐0.25, 95% CI ‐0.89 to 0.39), verrucomicrobia (Analysis 3.16 (1 study, 24 participants): MD 0.96, 95% CI ‐1.36 to 3.28), faceal acetate (Analysis 3.17 (1 study, 24 participants): MD ‐69.91, 95% CI ‐203.95 to 64.13), faecal propionate (Analysis 3.18 (1 study, 24 participants): MD ‐19.35, 95% CI ‐63.87 to 25.17), faecal butyrate (Analysis 3.19 (1 study, 24 participants): MD ‐11.04, 95% CI ‐39.57 to 17.49), faecal total short‐chain fatty acids (Analysis 3.20 (1 study, 24 participants): MD ‐104.71, 95% CI ‐293.34 to 83.92), faecal indoles (Analysis 3.21 (1 study, 24 participants): MD 2.71, 95% CI ‐69.78 to 75.20), and faecal p‐cresol (Analysis 3.22 (1 study, 24 participants): MD 28.84, 95% CI ‐105.07 to 162.75).

3.11. Analysis.

3.11

Comparison 3: Prebiotic versus prebiotic, Outcome 11: GI function: microbiota composition (Actinobacteria) at 4 weeks

3.12. Analysis.

3.12

Comparison 3: Prebiotic versus prebiotic, Outcome 12: GI function: microbiota composition (Bacteriodetes) at 4 weeks

3.13. Analysis.

3.13

Comparison 3: Prebiotic versus prebiotic, Outcome 13: GI function: microbiota composition (Proteobacteria) at 4 weeks

3.14. Analysis.

3.14

Comparison 3: Prebiotic versus prebiotic, Outcome 14: GI function: microbiota composition (Firmicutes) at 4 weeks

3.15. Analysis.

3.15

Comparison 3: Prebiotic versus prebiotic, Outcome 15: GI function: microbiota composition (Synergistetes) at 4 weeks

3.16. Analysis.

3.16

Comparison 3: Prebiotic versus prebiotic, Outcome 16: GI function: microbiota composition (Verrucomicrobia) at 4 weeks

3.17. Analysis.

3.17

Comparison 3: Prebiotic versus prebiotic, Outcome 17: GI function: microbiota composition (faecal acetate) at 4 weeks

3.18. Analysis.

3.18

Comparison 3: Prebiotic versus prebiotic, Outcome 18: GI function: microbiota composition (faecal propionate) at 4 weeks

3.19. Analysis.

3.19

Comparison 3: Prebiotic versus prebiotic, Outcome 19: GI function: microbiota composition (faecal butyrate) at 4 weeks

3.20. Analysis.

3.20

Comparison 3: Prebiotic versus prebiotic, Outcome 20: GI function: microbiota composition (faecal total short‐chain fatty acids) at 4 weeks

3.21. Analysis.

3.21

Comparison 3: Prebiotic versus prebiotic, Outcome 21: GI function: microbiota composition (faecal indoles) at 4 weeks

3.22. Analysis.

3.22

Comparison 3: Prebiotic versus prebiotic, Outcome 22: GI function: microbiota composition (faecal p‐cresol) at 4 weeks

Faecal characteristics

In very low certainty evidence, Biruete 2017 reported no difference in patients receiving inulin compared to maltodextrin at four weeks for bowel movements (Analysis 3.23 (1 study, 24 participants): MD ‐0.27, 95% CI ‐1.23 to 0.69), ease of passage (Analysis 3.24 (1 study, 24 participants): MD 0.00, 95% CI ‐0.90 to 0.90; very low certainty evidence), and consistency (Analysis 3.25 (1 study, 24 participants): MD ‐0.19, 95% CI‐1.34 to 0.96).

3.23. Analysis.

3.23

Comparison 3: Prebiotic versus prebiotic, Outcome 23: GI function: faecal characteristics (bowel movements) at 4 weeks

3.24. Analysis.

3.24

Comparison 3: Prebiotic versus prebiotic, Outcome 24: GI function: faecal characteristics (ease of passage) at 4 weeks

3.25. Analysis.

3.25

Comparison 3: Prebiotic versus prebiotic, Outcome 25: GI function: faecal characteristics (consistency) at 4 weeks

Adverse events and withdrawals

It is uncertain whether prebiotic 1 (gum Arabic or inulin plus oligofructose) decreased adverse events compared to prebiotic 2 (pectin or maltodextrin) (Analysis 3.26 (2 studies, 79 participants): RR 2.92, 95% CI 0.10 to 88.03; I² = 77%; very low certainty evidence).

3.26. Analysis.

3.26

Comparison 3: Prebiotic versus prebiotic, Outcome 26: Adverse events: number participants reporting an event

It is uncertain whether inulin plus oligofructose decreased withdrawals due to adverse events compared to maltodextrin (Analysis 3.27 (1 study, 39 participants): RR 0.70, 95% CI 0.13 to 3.75; very low certainty evidence).

3.27. Analysis.

3.27

Comparison 3: Prebiotic versus prebiotic, Outcome 27: Withdrawals due to adverse events

Adverse events were minimal and non‐serious, and withdrawals were not related to the study interventions (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for this comparison.

4: Prebiotic versus probiotic

Two studies contributed to this comparison.

  • Natarajan 2014 compared a 1:1 blend of cream of wheat and psyllium husk to Streptococcus thermophilus KB19, Lactobacillus acidophilus KB 27, and Bifidobacteriumlongum KB 31.

  • Wang 2015a compared maltodextrin to Bifidobacteriumbifidum A218, Bifidobacteriumcatenulatum A302, Bifidobacteriumlongum A101, Lactobacillusplantarum A87.

Dialysis outcomes
Peritonitis

Wang 2015a reported no difference in the occurrence of peritonitis at eight weeks in patients receiving prebiotics compared to probiotics (Analysis 4.1 (1 study, 47 participants): very low certainty evidence).

4.1. Analysis.

4.1

Comparison 4: Prebiotic versus probiotic, Outcome 1: Dialysis outcomes: peritonitis at 24 weeks

Adverse events

Natarajan 2014 reported one death not related to the study intervention: myocardial infarction at home from underlying cardiovascular conditions. Cross‐over numbers and denominators are unclear. Wang 2015a reported one patient in the placebo group died due to a head injury.

The remaining adverse events were minimal and non‐serious, and withdrawals were not related to the study interventions (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for this comparison.

5: Low versus high dose probiotic

Miranda Alatriste 2014 compared a low dose of Lactobacillus casei Shirota (8 x 109 CFU) to a higher dose of Lactobacillus casei Shirota (16 x 109 CFU).

Kidney function
Estimated glomerular filtration rate

Miranda Alatriste 2014 reported no difference in eGFR at eight weeks in patients receiving a low dose of Lactobacillus casei Shirota compared to a higher dose (Analysis 5.1 (1 study, 30 participants): MD ‐0.64 mL/min, 95% CI ‐9.51 to 8.23; very low certainty evidence).

5.1. Analysis.

5.1

Comparison 5: Low versus high dose probiotic, Outcome 1: Kidney function: GFR at 8 weeks

Serum creatinine

Miranda Alatriste 2014 reported no difference in SCr at eight weeks in patients receiving a low dose of Lactobacillus casei Shirota compared to a higher dose (Analysis 5.2 (1 study, 30 participants): MD ‐0.13 mg/dL, 95% CI ‐0.89 to 0.63; very low certainty evidence).

5.2. Analysis.

5.2

Comparison 5: Low versus high dose probiotic, Outcome 2: Kidney function: serum creatinine at 8 weeks

Urea

Miranda Alatriste 2014 reported no difference in urea rate at eight weeks in patients receiving a low dose of Lactobacillus casei Shirota compared to a higher dose (Analysis 5.3 (1 study, 30 participants): MD 4.57 mg/dL, 95% CI ‐9.52 to 18.66; very low certainty evidence).

5.3. Analysis.

5.3

Comparison 5: Low versus high dose probiotic, Outcome 3: Uraemic toxins: urea at 8 weeks

Adverse events

Adverse events were minimal and non‐serious, and withdrawals were not related to the study treatment (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for this comparison.

6: Synbiotic versus placebo or no treatment

Three studies contributed to this comparison.

  • Cosola 2021 compared a synbiotic (Lactobacillus casei LC4P1, Bifidobacteriumanimalis BLC1 plus fructo‐oligosaccharides, inulin and natural antioxidants) to a placebo.

  • Dehghani 2016 compared a synbiotic (Lactobacilluscasei, Lactobacillusacidophilus, Lactobacillusbulgaricus, Lactobacillusrhamnosus, Bifidobacteriumbreve, Bifidobacterium longum, Streptococcusthermophilus plus fructo‐oligosaccharides) to a placebo.

  • Viramontes‐Horner 2015 compared a synbiotic (Lactobacillus acidophilus, Bifidobacteriumlactis Bi‐07 plus inulin, omega‐3 fatty acids and vitamins) to a placebo.

Kidney function
Estimated glomerular filtration rate

It is uncertain whether synbiotics improve eGFR at six or 12 weeks compared to placebo (Analysis 6.1 (2 studies, 98 participants): MD 1.42 mL/min, 95% CI 0.65 to 2.2; very low certainty evidence).

6.1. Analysis.

6.1

Comparison 6: Synbiotic versus placebo, Outcome 1: Kidney function: eGFR at 6 or 12 weeks

Serum creatinine

It is uncertain whether synbiotics improve SCr at six or 12 weeks compared to placebo (Analysis 6.2 (2 studies, 98 participants): MD ‐0.57 mg/dL, 95% CI ‐1.08 to 0.07; very low certainty evidence).

6.2. Analysis.

6.2

Comparison 6: Synbiotic versus placebo, Outcome 2: Kidney function: SrCr at 6 or 12 weeks

Uraemic toxins
Urea

It is uncertain whether synbiotics improve urea at six or 12 weeks compared to placebo, with no differences found between subgroups (Analysis 6.3 (2 studies, 117 participants): MD 3.34 mg/dL, 95% CI ‐15.65 to 22.32; very low certainty evidence; test for subgroup differences P = 0.15).

6.3. Analysis.

6.3

Comparison 6: Synbiotic versus placebo, Outcome 3: Uraemic toxins: urea at 6 or 12 weeks

Gastrointestinal function
Change in any gastrointestinal upset or intolerance

In very low certainty evidence, Cosola 2021 reported no difference at 12 weeks in patients receiving synbiotics compared to placebo on the GSRS scale for rumbling (Analysis 6.4 (1 study, 23 participants): MD ‐0.54, 95% CI ‐0.77 to ‐0.31), hard stools (Analysis 6.5 (1 study, 23 participants): MD ‐0.09, 95% CI ‐0.35 to 0.17), abdominal pain (Analysis 6.6 (1 study, 23 participants): MD 0.22, 95% CI ‐0.02 to 0.46) or constipation syndrome (Analysis 6.7 (1 study, 23 participants): MD ‐0.17, 95% CI ‐0.72 to 0.38).

6.4. Analysis.

6.4

Comparison 6: Synbiotic versus placebo, Outcome 4: GI function: GSRS Item 6 (rumbling) at 12 weeks

6.5. Analysis.

6.5

Comparison 6: Synbiotic versus placebo, Outcome 5: GI function: GSRS Item 13 (hard stools) at 12 weeks

6.6. Analysis.

6.6

Comparison 6: Synbiotic versus placebo, Outcome 6: GI function: abdominal pain at 12 weeks

6.7. Analysis.

6.7

Comparison 6: Synbiotic versus placebo, Outcome 7: GI function: constipation syndrome at 12 weeks

Adverse events

Adverse events were minimal and non‐serious, and withdrawals were not related to the study interventions (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for the remaining analyses other than Analysis 6.3.

7: Prebiotic versus placebo or no treatment

Three studies contributed to this comparison.

  • Meng 2019 compared a high‐resistant starch, low‐protein flour to no treatment.

  • Esgalhado 2018 compared a resistant starch to a placebo.

  • Khosroshahi 2018 compared high‐amylose maize‐resistant starch to a placebo.

Kidney function
Serum creatinine

It is uncertain whether prebiotics improves SCr at eight or 12 weeks compared to placebo or no treatment (Analysis 7.1 (3 studies, 145 participants): MD 0.52 mg/dL, 95% CI ‐2.39 to 3.44; very low certainty evidence).

7.1. Analysis.

7.1

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 1: Kidney function: serum creatinine at 8 or 12 weeks

A difference was identified across subgroups by CKD stage. Participants receiving placebo or no treatment showed increased SCr in patients with CKD stage A2 with diabetic nephropathy (70 participants) compared to CKD stage G5D (75 participants); however, heterogeneity was very high I² = 79% (P = 0.03).

Uraemic toxins
Urea

Meng 2019 reported no difference no difference to urea at 12 weeks in patients receiving prebiotics compared to no treatment (Analysis 7.2 (1 study, 70 participants): MD ‐0.40 mmol/L, 95% CI ‐0.86 to 0.06; very low certainty evidence).

7.2. Analysis.

7.2

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 2: Uraemic toxins: urea at 12 weeks

Indoxyl sulfate

It is uncertain whether prebiotics improve indoxyl sulfate at eight weeks compared to placebo or no treatment (Analysis 7.3 (2 studies, 75 participants): SMD ‐0.14 mg/L, 95% CI ‐0.60 to 0.31; very low certainty evidence).

7.3. Analysis.

7.3

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 3: Uraemic toxins: indoxyl sulfate at 8 weeks

P‐cresyl sulfate

Khosroshahi 2018 reported no difference in p‐cresyl sulfate at eight weeks in patients receiving prebiotics compared to placebo (Analysis 7.4 (1 study, 44 participants): MD ‐1.68 mg/L, 95% CI ‐14.30 to 10.94; very low certainty evidence).

7.4. Analysis.

7.4

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 4: Uraemic toxins: p‐cresyl sulfate at 8 weeks

Gastrointestinal function
Microbiota composition

In very low certainty evidence, Khosroshahi 2018 reported no difference at eight weeks in prebiotics compared to placebo to the microbiota compositions of Facealibacterium (Analysis 7.5 (1 study, 44 participants): MD 2.37, 95% CI 0.23 to 4.51), Parabacteroides (Analysis 7.6 (1 study 44 participants): MD 0.22, 95% CI 0.06 to 0.38), Bifidobacteria (Analysis 7.7 (1 study 44 participants): MD ‐3.92, 95% CI ‐9.83 to 1.99), Ruminococcus (Analysis 7.8 (1 study 44 participants): MD 3.86, 95% CI ‐0.32 to 8.04), and Prevotella (Analysis 7.9 (1 study 44 participants): MD ‐0.43, 95% CI ‐1.45 to 0.59).

7.5. Analysis.

7.5

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 5: GI function: microbiota composition (Facealibacterium) at 8 weeks

7.6. Analysis.

7.6

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 6: GI function: microbiota composition (Parabacteroides) at 8 weeks

7.7. Analysis.

7.7

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 7: GI function: microbiota composition (Bifidobacteria) at 8 weeks

7.8. Analysis.

7.8

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 8: GI function: microbiota composition (Ruminococcus) at 8 weeks

7.9. Analysis.

7.9

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 9: GI function: microbiota composition (Prevotella) at 8 weeks

Adverse events

Khosroshahi 2018 reported no difference in any adverse events (number of participants reporting an event) in patients receiving prebiotics compared to placebo (Analysis 7.10 (1 study, 50 participants): RR 2.50, 95% CI 0.53 to 11.70; very low certainty evidence).

7.10. Analysis.

7.10

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 10: Adverse events: any adverse event (number of participants)

Khosroshahi 2018 reported no difference in withdrawals due to adverse events in patients receiving prebiotics compared to placebo (Analysis 7.11 (1 study, 50 participants): RR 3.0, 95% CI 0.13 to 70.30; very low certainty evidence).

7.11. Analysis.

7.11

Comparison 7: Prebiotic versus placebo or no treatment, Outcome 11: Withdrawals due to adverse events

Adverse events were minimal and non‐serious, and withdrawals were not related to the study interventions (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

There were no subgroups to analyse for the remaining analyses.

8: Probiotic versus placebo or no treatment

Nine studies contributed to this comparison.

  • Abbasi 2017 compared soy milk plus Lactobacillusplantarum A7 to soy milk.

  • Borges 2018 compared Streptococcusthermophilus, Lactobacillusacidophilus, and Bifidobacterialongum strains to a placebo.

  • Eidi 2018 compared LactobacillusRhamnosus to infant formula.

  • Lim 2021 compared Lactococcuslactis subspecies (Lactis LL358, Lactobacillussalivarius LS159, and Lactobaccilluspentosus LPE588) to a placebo.

  • Liu 2020 compared Bifidobacteriumlongum NQ1501, Lactobacillusacidophilus YIT2004 and Enterococcifaecalis YIT0072 to a placebo.

  • Mafi 2018 compared Lactobacillusacidophilus, Bifidobacteriumbifidum ZT‐B1, Lactobacillusreuteri ZT‐Lre and Lactobacillusfermentum ZT‐L3 to a placebo

  • Mazruei Arani 2019 compared honey containing a viable and heat‐resistant probiotic Bacillus coagulans T4 (IBRC‐N10791) to control honey.

  • ProbiotiCKD 2019 compared different probiotics over different weeks (week 1, weeks 2 to 3, weeks 4 to 15) to placebo.

  • Soleimani 2017 compared Lactobacillus acidophilus, Lactobacillus casei, and Bifidobacteria bifidum to a placebo.

Kidney function
Estimated glomerular filtration rate

It is uncertain whether probiotics improve eGFR at eight, 12, or 15 weeks compared to placebo or no treatment (Analysis 8.1 (3 studies, 128 participants): MD 2.73 mL/min, 95% CI ‐2.28 to 7.75; I² = 78%; very low certainty evidence). A difference was identified across subgroups by CKD stage. Participants receiving placebo or no treatment showed decreased eGFR in patients with CKD stage 1 with type 2 diabetes mellitus (40 participants) compared to CKD stage 3A with diabetes and hypertension (28 participants) and CKD stage G5D with diabetes and hypertension (60 participants); however, heterogeneity was very high I² = 77.6% (P = 0.01).

8.1. Analysis.

8.1

Comparison 8: Probiotic versus placebo or no treatment, Outcome 1: Kidney function: GFR at 8, 12 or 15 weeks

Serum creatinine

Probiotics may have little or no effect on SCr at eight, 12, or 24 weeks compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.2 (6 studies, 303 participants): MD ‐0.51 mg/dL, 95% CI ‐1.06 to 0.04; I² = 96%; low certainty evidence; test for subgroup differences P = 0.55).

8.2. Analysis.

8.2

Comparison 8: Probiotic versus placebo or no treatment, Outcome 2: Kidney function: serum creatinine at 8, 12 or 24 weeks

Albuminuria

Probiotics may have little or no effect on albuminuria at eight, 12, or 24 weeks compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.3 (4 studies, 193 participants): MD 0.02 g/dL, 95% CI ‐0.08 to 0.13; I² = 0%; low certainty evidence; test for subgroup differences P = 0.80).

8.3. Analysis.

8.3

Comparison 8: Probiotic versus placebo or no treatment, Outcome 3: Kidney function: albuminuria at 12 or 24 weeks

Proteinuria

Mafi 2018 reported no difference in proteinuria at 12 weeks in patients receiving probiotics compared to placebo or no treatment (Analysis 8.4 (1 study, 60 participants): MD ‐15.60 mg/dL, 95% CI ‐34.30 to 3.10; very low certainty evidence).

8.4. Analysis.

8.4

Comparison 8: Probiotic versus placebo or no treatment, Outcome 4: Kidney function: proteinuria at 12 weeks

Uraemic toxins
Urea

Probiotics may have little or no effect on urea at 12 or 24 weeks compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.5 (5 studies, 263 participants): MD ‐1.11 mg/dL, 95% CI ‐2.58 to 0.35; I² = 0%; low certainty evidence; test for subgroup differences P = 0.73).

8.5. Analysis.

8.5

Comparison 8: Probiotic versus placebo or no treatment, Outcome 5: Uraemic toxins: urea at 12 or 24 weeks

Indoxyl sulfate

It is uncertain whether probiotics improved indoxyl sulfate at 12 or 24 weeks compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.6 (2 studies, 83 participants): MD ‐4.42 mg/dL, 95% CI ‐9.83 to 1.35; I² = 0%; very low certainty evidence; test for subgroup differences P = 0.63).

8.6. Analysis.

8.6

Comparison 8: Probiotic versus placebo or no treatment, Outcome 6: Uraemic toxins: indoxyl sulfate at 12 or 24 weeks

P‐cresyl sulfate

It is uncertain whether probiotics improved p‐cresyl sulfate at four, 12 or 24 weeks compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.7 (3 studies, 125 participants): MD ‐2.12 mg/dL, 95% CI ‐5.19 to 0.95; I² = 16%; very low certainty evidence; test for subgroup differences P = 0.21).

8.7. Analysis.

8.7

Comparison 8: Probiotic versus placebo or no treatment, Outcome 7: Uraemic toxins: p‐cresyl sulfate at 4, 12 or 24 weeks

Indole‐3‐acetic acid

Borges 2018 reported no difference in indole‐3‐acetic acid at 12 weeks in patients receiving probiotics compared to placebo or no treatment (Analysis 8.8 (1 study, 33 participants): MD ‐288.10 μg/L, 95% CI ‐464.41 to ‐111.79; very low certainty evidence).

8.8. Analysis.

8.8

Comparison 8: Probiotic versus placebo or no treatment, Outcome 8: Uraemic toxins: indole‐3‐acetic acid acid at 12 weeks

Gastrointestinal function
Change in any gastrointestinal upset or intolerance

Borges 2018 reported no difference at 12 weeks in patients receiving probiotics compared to placebo or no treatment for abdominal pain or diarrhoea (Analysis 8.9 (1 study, 33 participants): RR 2.13, 95% CI 0.21 to 21.22; very low certainty evidence), or constipation (Analysis 8.10 (1 study, 33 participants): RR 3.18, 95% CI 0.14 to 72.75; very low certainty evidence).

8.9. Analysis.

8.9

Comparison 8: Probiotic versus placebo or no treatment, Outcome 9: GI function: any GI upset or intolerance (abdominal pain/diarrhoea) at 12 weeks

8.10. Analysis.

8.10

Comparison 8: Probiotic versus placebo or no treatment, Outcome 10: GI function: any GI upset or intolerance (constipation) at 12 weeks

Faecal characteristics

Borges 2018 reported no difference in faecal pH at 12 weeks in patients receiving probiotics compared to placebo or no treatment (Analysis 8.11 (1 study, 20 participants): MD ‐2.10, 95% CI ‐7.28 to 3.08; very low certainty evidence).

8.11. Analysis.

8.11

Comparison 8: Probiotic versus placebo or no treatment, Outcome 11: GI function: faecal pH at 12 weeks

Adverse events

It is uncertain whether probiotics decrease adverse events compared to placebo or no treatment, with no difference found between subgroups (Analysis 8.12 (4 studies, 205 participants): RR 1.96, 95% CI 0.63 to 6.12; I² = 13%; very low certainty evidence; test for subgroup differences P = 0.17), and withdrawals due to adverse events, with no difference found between subgroups (Analysis 8.13 (4 studies, 205 participants): RR 4.96, 95% CI 1.13 to 21.77; I² = 0%; very low certainty evidence; test for subgroup differences P = 1.00).

8.12. Analysis.

8.12

Comparison 8: Probiotic versus placebo or no treatment, Outcome 12: Adverse events: any adverse event (number of participants)

8.13. Analysis.

8.13

Comparison 8: Probiotic versus placebo or no treatment, Outcome 13: Withdrawals due to adverse events

Lim 2021 reported one death in each arm but not related to the study treatment, "No significant gastrointestinal side effects were reported or recorded throughout the study in either group."

The remaining adverse events were minimal and non‐serious, and withdrawals were not related to study treatment (see Appendix 5).

The studies in this comparison did not report data on our remaining primary and secondary outcomes.

Discussion

Summary of main results

Forty‐five studies (2266 randomised participants) were included in this review. Study participants were predominantly adults (two studies included children) with CKD ranging from stages 1 to 5, with patients receiving and not receiving dialysis, of whom half also had diabetes and hypertension.

No studies investigated the same synbiotic, prebiotic or probiotic of similar strains, doses, or frequencies. The risk of bias in the included studies varied between low, unclear or high risk across each of the domains.

  • Synbiotics versus synbiotics: compared to synbiotics of a different dose, it is uncertain whether synbiotics improve HRQoL because the certainty of the evidence was very low.

  • Synbiotics versus prebiotics: compared to prebiotics, it is uncertain whether synbiotics improve eGFR, SCr, urea, indoxyl sulfate, p‐cresyl sulfate, change in any GI upset or intolerance, microbiota composition, faecal characteristics, or HRQoL because the certainty of the evidence was very low.

  • Prebiotics versus prebiotics: compared to prebiotics of a different dose, it is uncertain whether prebiotics improves eGFR, indoxyl sulfate, p‐cresyl sulfate, change in any GI upset or intolerance, microbiota composition, or faecal characteristics because the certainty of the evidence was very low.

  • Prebiotics versus probiotics: compared to probiotics, it is uncertain whether prebiotics prevent peritonitis because the certainty of the evidence was very low.

  • Probiotics versus probiotics: compared to probiotics of a different dose, it is uncertain whether probiotics improve eGFR, SCr, or urea because the certainty of the evidence was very low.

  • Synbiotics versus placebo or no treatment: compared to placebo or no treatment, it is uncertain whether synbiotics improve eGFR, SCr, urea, or change in any GI upset or intolerance because the certainty of the evidence was very low.

  • Prebiotics versus placebo or no treatment: compared to placebo or no treatment, it is uncertain whether prebiotics improve SCr, urea, indoxyl sulfate, p‐cresyl sulfate, or microbiota composition because the certainty of the evidence is very low. For SCr, a difference was identified across subgroups by CKD stage. Participants receiving placebo or no treatment showed an increase in SCr in patients with CKD stage A2 with diabetic nephropathy (70 participants) compared to CKD stage G5D (75 participants); however, heterogeneity was very high I² = 79% (P = 0.03).

  • Probiotics versus placebo or no treatment: compared to placebo or no treatment, it is uncertain whether probiotics improve eGFR, proteinuria, indoxyl sulfate, p‐cresyl sulfate, indole‐3‐acetic acid, change in any GI upset or intolerance, or faecal characteristics because the certainty of the evidence is very low. For eGFR, a difference was identified across subgroups by CKD stage. Participants receiving placebo or no treatment showed decreased eGFR in patients with CKD stage 1 with type 2 diabetes mellitus (40 participants) compared to CKD stage 3A with diabetes and hypertension (28 participants) and CKD stage G5D with diabetes and hypertension (60 participants); however, heterogeneity was very high I² = 77.6% (P = 0.01). Probiotics may have little or no effect on SCr, albuminuria, or urea compared to placebo or no treatment, as the certainty of the evidence is low.

For all comparisons, adverse events were poorly reported and were minimal (flatulence, nausea, diarrhoea, abdominal pain) and non‐serious, and withdrawals were not related to the study treatment.

Overall completeness and applicability of evidence

This review identified a substantial number of RCTs testing the efficacy of synbiotics, prebiotics or probiotics in people with CKD. However, the severe lack of congruence across the studies highlights the lack of high‐quality RCTs testing similar biotics, doses, strains, frequencies, and standardised outcomes.

This field contains emerging trials of diverse and assorted interventions. No studies investigated the same synbiotic, prebiotic or probiotic of similar strains or doses.

We have identified significant gaps in the evidence with the meta‐analyses undertaken containing limited data from small studies. It is critically important for this emerging field to conduct trials of alternative interventions in a standardised way to help better inform whether these agents favourably modify gut dysbiosis and its associated morbidity and QoL in people with CKD.

The four major issues around the completeness of the evidence were:

  1. Limited sample size and insufficient power

  2. Standardised dosing of synbiotics, prebiotics or probiotics

  3. Standardised measuring

  4. Reporting of outcomes.

The three major issues around the applicability of the evidence were:

  1. Participant criteria: the spread of patients across CKD stages 1 to 5, dialysis and non‐dialysis, diabetes, and hypertension

  2. Outcome measures varied greatly by definition of units, scale, and time points.

  3. Lack of patient‐important and patient‐centred outcomes.

Quality of the evidence

Overall, the quality of the evidence was low to very low. Studies were judged to have a spread of low to unclear to high risks of bias across most domains (Figure 2). Of the available evidence, meta‐analyses undertaken were of limited data from small studies. Data were sparse and addressed few primary and secondary outcomes.

Across all comparisons, GRADE evaluations were judged to be low or very low certainty evidence. This evidence was downgraded primarily due to the risk of bias and sparse data from small study sizes. The evidence was also downgraded to very low for single‐study data or incomplete or no data reported for an outcome.

Very low certainty evidence implies that we are very uncertain about results (not estimable due to lack of data or poor quality). We have no evidence to support or refute the use of synbiotics, prebiotics, or probiotics in people with CKD, and findings should be viewed with caution.

Potential biases in the review process

This review was conducted as per the protocol following prespecified inclusion criteria and used comprehensive literature searches to find all relevant studies. We do not believe there are any other potential biases in this review process.

Agreements and disagreements with other studies or reviews

This review is the most up‐to‐date synthesis of the evidence that examines synbiotics, prebiotics, and probiotics in people with CKD.

A recent systematic review has identified similar RCTs to those of an earlier search date (McFarlane 2019). The included studies are similar, and the results of the meta‐analyses also found, from very low certainty evidence, that synbiotic, prebiotic or probiotic interventions made little or no difference to some uraemic toxins (urea, indoxyl sulfate, p‐cresyl sulfate). Similarly, the review concludes that further high‐quality, standardised trials are needed to develop answers within this emerging field.

In contrast, Zheng 2020 and colleagues identified improvement in lipids, oxidative stress and inflammatory biomarkers from treatment with biotic supplementation. The meta‐analysis appears to have combined synbiotics, prebiotics and probiotics involving entirely different treatment options, doses and strains. Further to this, the included studies have been rated as mostly low risk of bias across the domains. Our review disagrees with this quality assessment. GRADE rating was also not used for assessing the certainty of the evidence. Zheng 2020 and colleagues may prematurely promote the use of biotic interventions in this population.

A further two systematic reviews focus on patients receiving dialysis and conclude similar results. Chen 2022 and colleagues found from low‐quality evidence that supplementation with biotics may reduce some inflammatory biomarkers (C‐reactive protein, interleukin‐6), and uraemic toxins (indoxyl sulfate) and increase HDL in dialysis patients. March 2020 and colleagues have similarly reported that supplementation with biotics may reduce endotoxins, uraemic toxins (indoxyl‐sulphate, p‐cresyl sulfate), and reduced GI symptoms. However, the review correctly identifies that these were low‐quality, high‐risk of bias studies.

Authors' conclusions

Implications for practice.

We found very few studies that adequately test biotic supplementation as an alternative treatment to improve GI symptoms, kidney function, uraemic toxins, allograft function, dialysis outcomes, QoL, CVD, cancer, and adverse events for people with CKD.

There is currently no evidence to support or refute the use of synbiotics, prebiotics, or probiotics in people with CKD, and findings should be viewed with caution.

We are not certain whether synbiotics, prebiotics, or probiotics are more or less effective compared to one another, antibiotics, or standard care for improving patient outcomes in people with CKD. Adverse events were uncommon and mild. Twenty‐six studies are currently ongoing; therefore, it is possible findings may change with the inclusion of these studies in future updates.

Implications for research.

To effectively examine the efficacy of synbiotics, prebiotics, or probiotics in people with CKD, future research in this field requires adequately powered, longer‐term RCTs comparing synbiotics, prebiotics, and probiotics separately (and with a placebo) measuring standardised strains, dosing and frequency as well as a standard set of core kidney transplant and core CKD outcomes (SONG 2017).

History

Protocol first published: Issue 5, 2020

Acknowledgements

We wish to acknowledge the assistance of the Cochrane Kidney and Transplant Information Specialists, Gail Higgins, Ruth Mitchell and Anh Kieu. The authors are grateful to the following peer reviewers for their time and comments: Gabrielle Williams (Melanoma Institute Australia), Deirdre Hahn (Department of Nephrology, The Children’s Hospital Westmead, Australia).

The Methods section of this review is based on a standard template used by Cochrane Kidney and Transplant.

Appendices

Appendix 1. Chronic kidney disease (CKD) definitions

The US Kidney Disease: Improving Global Outcomes (KDIGO) organization developed clinical practice guidelines and classifications for CKD in 2002. In 2012, these guidelines and classifications were updated and adopted by the international Kidney Disease Improving Global Outcomes (KDIGO) CKD guideline development work group (KDIGO 2013).

  Persistent albuminuria categories
Description and range
A1 A2 A3
Normal to mildly increased Moderately increased Severely increased
ACRa < 30 mg/g ACR30 to 300 mg/g ACR > 300 mg/g
eGFRb categories (mL/min/1.73 m2)
Description and range
G1 Normal or highc ≥ 90 1 if CKD 1 2
G2 Mildly decreasedc 60 to 89 1 if CKD 1 2
G3a Mildly to moderately decreased 45 to 59 1 2 3
G3b Moderately to severely decreased 30 to 44 2 3 3
G4 Severely decreased 15 to 29 3 3 4+
G5 Kidney failure < 15 4+ 4+ 4+

aACR ‐ albumin‐creatinine ratio

beGFR ‐ estimated glomerular filtration rate

cIn the absence of evidence of kidney damage, neither eGFR category G1 nor G2 fulfil the criteria for CKD (KDIGO 2013).

Classification is based on two markers: evidence of kidney damage (such as the presence of microalbuminuria, proteinuria or structural abnormality); and the sustained impairment of eGFR for at least 3 months. Normal eGFR in young adults is around 100 to 120 mL/min/1.73 m2.

Early CKD is described as stages 1 to 3 of the KDIGO 2012 classification. At these stages, a patient may have no outward symptoms or signs of illness and only testing such as dipstick urine measurement for proteinuria/haematuria, or blood test may detect the presence of a kidney abnormality.

Appendix 2. Electronic search strategies

Database Search terms
CENTRAL
  1. synbiotic*:ti,ab,kw

  2. probiotic*:ti,ab,kw

  3. prebiotic*:ti,ab,kw

  4. (bifido* or lactobacill*):ti,ab,kw

  5. bacillus:ti,ab,kw

  6. enterococcus:ti,ab,kw

  7. escherichia:ti,ab,kw

  8. oligosaccharide*:ti,ab,kw

  9. saccharomyces:ti,ab,kw

  10. streptococcus next thermophilus:ti,ab,kw

  11. (resistant next starch):ti,ab,kw

  12. polydextrose:ti,ab,kw

  13. (dietary next (fiber or fibre)):ti,ab,kw

  14. "gum arabic":ti,ab,kw

  15. "hi‐maize":ti,ab,kw

  16. oligosaccharides:ti,ab,kw

  17. familact:ti,ab,kw

  18. "probinul neutro":ti,ab,kw

  19. inulin:ti,ab,kw

  20. {OR #1‐#19}

  21. (kidney next disease* or renal next disease* or kidney next failure or renal next failure):ti,ab,kw

  22. renal next insufficiency:ti,ab,kw

  23. dialysis:ti,ab,kw

  24. (h*emodialysis or h*emofiltration or h*emodiafiltration):ti,ab,kw

  25. "renal replacement therapy":ti,ab,kw

  26. (ESRF or ESKF or ESRD or ESKD):ti,ab,kw

  27. (CKF or CKD or CRF or CRD):ti,ab,kw

  28. (CAPD or CCPD or APD):ti,ab,kw

  29. (predialysis or "pre‐dialysis"):ti,ab,kw

  30. ((kidney or renal) next (transplant* or graft*)):ti,ab,kw

  31. {OR #21‐#30

  32. #20 and #31 in Trials

MEDLINE
  1. Synbiotics/

  2. Probiotics/

  3. Prebiotics/

  4. synbiotic*.tw.

  5. probiotic*.tw.

  6. prebiotic*.tw.

  7. or/1‐6

  8. Bifidobacterium bifidum/

  9. exp Lactobacillus/

  10. Streptococcus Thermophilus/

  11. exp Saccharomyces/

  12. exp Bacillus/

  13. exp Enterococcus/

  14. Oligosaccharides/

  15. Escherichia/

  16. (bifido* or lactobacill*).tw.

  17. streptococcus thermophilus.tw.

  18. saccharomyces.tw.

  19. bacillus.tw.

  20. enterococcus.tw.

  21. eschericia.tw.

  22. Dietary Fiber/

  23. Gum Arabic/

  24. (dietary fiber or dietary fibre).tw.

  25. resistant starch.tw.

  26. polydextrose.tw.

  27. gum arabic.tw.

  28. hi‐maize.tw.

  29. oligosaccharides.tw.

  30. familact.tw.

  31. probinul neutro.tw.

  32. Inulin/

  33. 33 inulin.tw.

  34. or/8‐33

  35. or/7,34

  36. exp Kidney Diseases/

  37. exp Renal Replacement Therapy/

  38. dialysis.tw.

  39. (hemodialysis or haemodialysis).tw.

  40. (hemofiltration or haemofiltration).tw.

  41. (hemodiafiltration or haemodiafiltration).tw.

  42. (kidney disease* or renal disease* or kidney failure or renal failure).tw.

  43. (ESRF or ESKF or ESRD or ESKD).tw.

  44. (CKF or CKD or CRF or CRD).tw.

  45. (CAPD or CCPD or APD).tw.

  46. (predialysis or pre‐dialysis).tw.

  47. (diabet* adj5 (kidney or renal)).tw.

  48. DKD.tw.

  49. (glomerulo* or glomerular).tw.

  50. (nephritis or nephrotic).tw.

  51. (PKD or ADPKD).tw.

  52. ((kidney or renal) adj1 (transplant* or graft*)).tw.

  53. or/36‐52

  54. and/35,53

EMBASE
  1. synbiotic agent/

  2. prebiotic agent/

  3. exp probiotic agent/

  4. synbiotic*.tw.

  5. probiotic*.tw.

  6. prebiotic*.tw.

  7. or/1‐6

  8. bifidobacterium bifidum/

  9. exp Lactobacillus/

  10. exp bacillus/

  11. exp enterococcus/

  12. escherichia/

  13. oligosaccharide/

  14. exp saccharomyces/

  15. streptococcus thermophilus/

  16. (bifido* or lactobacill*).tw.

  17. bacillus.tw.

  18. enterococcus.tw.

  19. escherichia.tw.

  20. oligosaccharide*.tw.

  21. saccharomyces.tw.

  22. streptococcus thermophilus.tw.

  23. dietary fiber/

  24. gum arabic/

  25. gum arabic.tw.

  26. (dietary adj (fiber or fibre)).tw.

  27. resistant starch.tw.

  28. polydextrose.tw.

  29. hi‐maize.tw.

  30. familact.tw.

  31. probinul neutro.tw.

  32. Inulin/

  33. inulin.tw.

  34. or/8‐33

  35. or/7,34

  36. exp renal replacement therapy/

  37. kidney disease/

  38. chronic kidney disease/

  39. kidney failure/

  40. chronic kidney failure/

  41. mild renal impairment/

  42. stage 1 kidney disease/

  43. moderate renal impairment/

  44. severe renal impairment/

  45. end stage renal disease/

  46. renal replacement therapy‐dependent renal disease/

  47. diabetic nephropathy/

  48. (hemodialysis or haemodialysis).tw.

  49. (hemofiltration or haemofiltration).tw.

  50. (hemodiafiltration or haemodiafiltration).tw.

  51. dialysis.tw.

  52. (CAPD or CCPD or APD).tw.

  53. (kidney disease* or renal disease* or kidney failure or renal failure).tw.

  54. (CKF or CKD or CRF or CRD).tw.

  55. (ESRF or ESKF or ESRD or ESKD).tw.

  56. (predialysis or pre‐dialysis).tw.

  57. exp kidney transplantation/

  58. ((kidney or renal) adj1 (transplant* or graft*)).tw.

  59. or/36‐58

  60. and/35,59

Appendix 3. Risk of bias assessment tool

Potential source of bias Assessment criteria
Random sequence generation
Selection bias (biased allocation to interventions) due to inadequate generation of a randomised sequence
Low risk of bias: Random number table; computer random number generator; coin tossing; shuffling cards or envelopes; throwing dice; drawing of lots; minimisation (minimisation may be implemented without a random element, and this is considered to be equivalent to being random).
High risk of bias: Sequence generated by odd or even date of birth; date (or day) of admission; sequence generated by hospital or clinic record number; allocation by judgement of the clinician; by preference of the participant; based on the results of a laboratory test or a series of tests; by availability of the intervention.
Unclear: Insufficient information about the sequence generation process to permit judgement.
Allocation concealment
Selection bias (biased allocation to interventions) due to inadequate concealment of allocations prior to assignment
Low risk of bias: Randomisation method described that would not allow investigator/participant to know or influence intervention group before eligible participant entered in the study (e.g. central allocation, including telephone, web‐based, and pharmacy‐controlled, randomisation; sequentially numbered drug containers of identical appearance; sequentially numbered, opaque, sealed envelopes).
High risk of bias: Using an open random allocation schedule (e.g. a list of random numbers); assignment envelopes were used without appropriate safeguards (e.g. if envelopes were unsealed or non‐opaque or not sequentially numbered); alternation or rotation; date of birth; case record number; any other explicitly unconcealed procedure.
Unclear: Randomisation stated but no information on method used is available.
Blinding of participants and personnel
Performance bias due to knowledge of the allocated interventions by participants and personnel during the study
Low risk of bias: No blinding or incomplete blinding, but the review authors judge that the outcome is not likely to be influenced by lack of blinding; blinding of participants and key study personnel ensured, and unlikely that the blinding could have been broken.
High risk of bias: No blinding or incomplete blinding, and the outcome is likely to be influenced by lack of blinding; blinding of key study participants and personnel attempted, but likely that the blinding could have been broken, and the outcome is likely to be influenced by lack of blinding.
Unclear: Insufficient information to permit judgement
Blinding of outcome assessment
Detection bias due to knowledge of the allocated interventions by outcome assessors.
Low risk of bias: No blinding of outcome assessment, but the review authors judge that the outcome measurement is not likely to be influenced by lack of blinding; blinding of outcome assessment ensured, and unlikely that the blinding could have been broken.
High risk of bias: No blinding of outcome assessment, and the outcome measurement is likely to be influenced by lack of blinding; blinding of outcome assessment, but likely that the blinding could have been broken, and the outcome measurement is likely to be influenced by lack of blinding.
Unclear: Insufficient information to permit judgement
Incomplete outcome data
Attrition bias due to amount, nature or handling of incomplete outcome data.
Low risk of bias: No missing outcome data; reasons for missing outcome data unlikely to be related to true outcome (for survival data, censoring unlikely to be introducing bias); missing outcome data balanced in numbers across intervention groups, with similar reasons for missing data across groups; for dichotomous outcome data, the proportion of missing outcomes compared with observed event risk not enough to have a clinically relevant impact on the intervention effect estimate; for continuous outcome data, plausible effect size (difference in means or standardised difference in means) among missing outcomes not enough to have a clinically relevant impact on observed effect size; missing data have been imputed using appropriate methods.
High risk of bias: Reason for missing outcome data likely to be related to true outcome, with either imbalance in numbers or reasons for missing data across intervention groups; for dichotomous outcome data, the proportion of missing outcomes compared with observed event risk enough to induce clinically relevant bias in intervention effect estimate; for continuous outcome data, plausible effect size (difference in means or standardized difference in means) among missing outcomes enough to induce clinically relevant bias in observed effect size; ‘as‐treated’ analysis done with substantial departure of the intervention received from that assigned at randomisation; potentially inappropriate application of simple imputation.
Unclear: Insufficient information to permit judgement
Selective reporting
Reporting bias due to selective outcome reporting
Low risk of bias: The study protocol is available and all of the study’s pre‐specified (primary and secondary) outcomes that are of interest in the review have been reported in the pre‐specified way; the study protocol is not available but it is clear that the published reports include all expected outcomes, including those that were pre‐specified (convincing text of this nature may be uncommon).
High risk of bias: Not all of the study’s pre‐specified primary outcomes have been reported; one or more primary outcomes is reported using measurements, analysis methods or subsets of the data (e.g. sub‐scales) that were not pre‐specified; one or more reported primary outcomes were not pre‐specified (unless clear justification for their reporting is provided, such as an unexpected adverse effect); one or more outcomes of interest in the review are reported incompletely so that they cannot be entered in a meta‐analysis; the study report fails to include results for a key outcome that would be expected to have been reported for such a study.
Unclear: Insufficient information to permit judgement
Other bias
Bias due to problems not covered elsewhere in the table
Low risk of bias: The study appears to be free of other sources of bias.
High risk of bias: Had a potential source of bias related to the specific study design used; stopped early due to some data‐dependent process (including a formal‐stopping rule); had extreme baseline imbalance; has been claimed to have been fraudulent; had some other problem.
Unclear: Insufficient information to assess whether an important risk of bias exists; insufficient rationale or evidence that an identified problem will introduce bias.

Appendix 4. The GRADE approach (Grades of Recommendation, Assessment, Development, and Evaluation)

The GRADE approach assesses the certainty of a body of evidence, rating it in one of four grades (GRADE 2008).

  • High: we are very confident that the true effect lies close to that of the estimate of the effect

  • Moderate: we are moderately confident in the effect estimate; the true effect is likely to be close the estimate of effect, but there is a possibility that it is substantially different

  • Low: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect

  • Very low: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

We decreased the certainty of evidence if there was (Balshem 2011):

  • serious (‐1) or very serious (‐2) limitation in the study design or execution (risk of bias);

  • important inconsistency of results (‐1);

  • some (‐1) or major (‐2) uncertainty about the directness of evidence;

  • imprecise or sparse data (‐1) or serious imprecision (‐2); or

  • high probability of publication bias (‐1).

We increased the certainty of evidence if there was (GRADE 2011):

  • a large magnitude of effect (direct evidence, relative risk (RR) = 2 to 5 or RR = 0.5 to 0.2 with no plausible confounders) (+1); very large with RR > 5 or RR < 0.2 and no serious problems with risk of bias or precision; more likely to rate up if effect is rapid and out of keeping with prior trajectory; usually supported by indirect evidence (+2);

  • evidence of a dose response gradient (+1); or

  • all plausible residual confounders or biases would reduce a demonstrated effect, or suggest a spurious effect when results show no effect (+1).

Appendix 5. Adverse events

Adverse events

Note: all numerals are 'number of participants reporting an event' (not number of events reported) unless stated otherwise.

Study ID Synbiotic Prebiotic Probiotic Placebo No treatment/the control starch etc. Notes from study
Abbasi 2017 Any AE: 4/22 (flatulence: 4)
Withdrawal due to AE: 0
Any AE: 5/22 (flatulence: 5)
Withdrawal due to AE: 0
Also some minor complaints about the specific taste of soy milk at start. The numbers are not clear
Biruete 2017 Any AE: not reported
Withdrawal due to AE: not reported
None reported
Bliss 1992 Gum Arabic
Any AE: 8/20 (flatulence: 8)
Withdrawal due to AE: unclear
Pectin
Any AE: 0/20
Withdrawal due to AE: unclear
Borges 2018 Any AE: 5/23 (abdominal pain/diarrhoea: 2; constipation: 1; precordial pain: 2)
Withdrawal due to AE: 5
Any AE: 1/23 (abdominal pain: 1; death: 2 (not related to study))
Withdrawal due to AE: 1
Cosola 2021 Any AE: 0/13
Withdrawal due to AE: 0/13
Any AE: 0/13
Withdrawal due to AE: 0/13
de Andrade 2021 Any AE: 0/43
Withdrawal due to AE: 0/43
Any AE: 0/43
Withdrawal due to AE: 0/43
de Araujo 2022 Any AE: 2/40 (abdominal pain/diarrhoea: 2)
Withdrawal due to AE: 2
Any AE: 0/40
Withdrawal due to AE: 0/40
Dehghani 2016 Any AE: 4/denominator not clear
Withdrawal due to AE: 4
Any AE: 3/denominator not clear
Withdrawal due to AE: 3
Ebrahim 2022 Any AE: 0/30
Withdrawals due to AE: 0
Any AE: 0/30
Withdrawals due to AE: 0
Eidi 2018 Any AE: 2/30 (abdominal ascites (acute): 1; vertigo: 1)
Withdrawal due to AE: 2/30
Any AE: 0/29
Withdrawal due to AE: 0/29
Elamin 2017 Any AE: unclear/36
Withdrawal due to AE: unclear/36
Quote: "GA supplements were well tolerated with no significant gastrointestinal side effects (Table 4). One patient discontinued the supplements after one dose because she did not like the texture of the solution."
Comment: these were reported as a mean across the entire 3 prebiotic groups
Esgalhado 2018 Any AE: unclear/19 (unclear deaths but not related to study drugs)
Withdrawal due to AE: unclear
Any AE: unclear/19 (unclear deaths but not related to study drugs)
Withdrawal due to AE: unclear
Withdrawals and dropout reasons are unclear whether attributed to study drug or other reasons
Guida 2014 Any AE: 0/18
Withdrawal due to AE: 0/18
Any AE: 0/12
Withdrawal due to AE: 0/12
Guida 2017 Any AE: 13/22
Withdrawal due to AE: 0/22
Any AE: 0/12
Withdrawal due to AE: 0/12
Quote: "Treatment with synbiotics was well tolerated with no relevant gastrointestinal adverse effect; borborygmi were reported as occasional by most patients (60%), whereas abdominal pain was virtually absent in both groups."
He 2022 Oligosaccharide
Any AE: 0/33
Withdrawal due to AE: 0/33
Maltodextrin
Any AE: 1/33 (peritonitis: 1)
Withdrawal due to AE: 1/33 (peritonitis: 1)
Haghighat 2019 Any AE: 1/50 (headache: 1)
Withdrawal due to AE: 0
Any AE: 3/25 (headache: 1; fatigue: 1; breathing problems: 1)
Withdrawal due to AE: 0
These adverse events in this study were judged by the authors to be unrelated to study interventions
Khosroshahi 2018 Any AE: 5/25 (epigastric pain and vomiting: 1; distention: 2; nausea: 2)
Withdrawal due to AE: 1 (epigastric pain and vomiting: 1)
Any AE: 2/25 (dyspepsia: 1; nausea: 1)
Withdrawal due to AE: 0
 
Kooshki 2019 Any AE: not reported/25
Withdrawal due to AE: not reported
Any AE: not reported/25
Withdrawal due to AE: not reported
Li 2020 Inulin
Any AE: 2/19 (peritonitis: 2)
Withdrawals due to AE: 2
Maltodextrin
Any AE: 3/20 (peritonitis: 2; death: 1 (not related to study drug))
Withdrawal due to AE: 3
Data are not entered into meta‐analysis as these are from both phases of cross‐over
Lim 2021 Any AE: 2/28 (infection: 2)
Withdrawal due to AE: 2/28 (hospitalisation due to infection: 2)
Any AE: 0/28
Withdrawal due to AE: 0/28
One death in each arm but not related to study treatment
Quote: "No significant gastrointestinal side effects were reported or recorded throughout the study in either group."
Liu 2020 Any AE: unclear/25
Withdrawal due to AE: unclear/25
Any AE: unclear/25
Withdrawal due to AE: unclear/25
Three from probiotics and 2 from placebo discontinued but reasons not provided and unclear whether related to adverse effects from study intervention or simply lost.  
Lopes 2018 Any AE: 14/49 (abdominal discomfort: 3; did not fit the food: 5; difficulty consuming product at home: 2; hospitalisation and death (unclear whether related to study): 4)
Withdrawal due to AE: 10 (abdominal discomfort: 3; did not fit the food: 5; difficulty consuming product at home: 2)
Any AE: 13/50 (abdominal discomfort: 4; did not fit the food: 5; difficulty consuming product at home: 4)
Withdrawal due to AE: 13 (abdominal discomfort: 4; did not fit the food: 5; difficulty consuming product at home: 4)
Lydia 2022 Any AE: 9/30 (diarrhoea: 6; nausea: 2; bloating: 1)
Withdrawal due to AE: 2/30 (diarrhoea: 2)
Any AE: 7/30 (diarrhoea: 2; nausea: 3; stomach ache/heartburn: 2)
Withdrawal due to AE: 0/30
Mafi 2018 Any AE: not reported/30
Withdrawal due to AE: 0/30
Any AE: not reported/30
ThreeWithdrawal due to AE: 0/30
Three withdrawals from each group due to personal reasons and not related to the study drug
Mazruei Arani 2019 Any AE: not reported/30
Withdrawals due to AE: not reported/30
Any AE: not reported/30
Withdrawals due to AE: not reported/30
Two withdrawals from probiotic group, 1 withdrawal from placebo group. Reasons not provided and unclear whether related to study or not
Meng 2019 Any AE: 1/37: AE not specified
Withdrawal due to AE: 1/37
Any AE: 0/38
Withdrawal due to AE: 0/38
Miraghajani 2017 Any AE: not reported/24
Withdrawals due to AE: 0/24
Any AE: not reported/24
Withdrawals due to AE: 0/24
 
Miranda Alatriste 2014 Lower dose
Any AE: 0/15
Withdrawal due to AE: 0/16
Higher dose
Any AE: 0/15
Withdrawal due to AE: 0/16
One hospitalised with urinary tract infection in low dose probiotic group, but not specified whether this was related to study drug
One participant did not tolerate the drink, but this was during inclusion criteria and pre‐randomisation
Mirzaeian 2020 Any AE: 0/24
Withdrawal due to AE: 0/24
Any AE: 0/24
Withdrawal due to AE: 0/24
One death in each arm; however, not related to the study intervention
Natarajan 2014 Any AE: unclear/28
Withdrawal due to AE: unclear/28
Any AE: 2/28
Withdrawal due to AE: 2/28 (nausea and vomiting: 2)
Cross‐over numbers and denominators are unclear
One death not related to study intervention: myocardial infarction at home from underlying cardiovascular conditions
Pan 2021 Any AE: 0/58
Withdrawals due to AE: 0/58
Any AE: 0/58
Withdrawals due to AE: 0/58
Other lost to follow‐up not related to study treatment
PREBIOTIC 2022 Any AE: 15/27 (UTI: 9; skin: 2; GI: 1; other: 3)
Serious AE: 9/27 (hospitalised due to GI issue: 4; hospitalised due to skin issue: 2; hospitalised due to other: 3)
Withdrawals due to AE: unclear/27
Any AE: 14/27 (bloodstream infection: 1; respiratory infection: 1; UTI: 6; skin issue: 3; cardiovascular: 1; GI: 2)
Serious AE: 1/27 (hospitalised due to UTI: 1)
Withdrawals due to AE: unclear/27
Withdrawals due to adverse events are unclear. There appear to be several discontinued participants in both arms. Reasons not linked to whether these are due to adverse events or practicalities of the feasibility trial. ITT analysis was undertaken
ProbiotiCKD 2019 Any AE: 0/14
Withdrawal due to AE: 0/14
Any AE: 0/14
Withdrawal due to AE: 0/14
Poesen 2016 Rabinoxylan
Any AE: 1/40 (nausea)
Withdrawal due to AE: 1/40
Maltodextrin
Any AE: 0/40
Withdrawal due to AE: 0/40
Ranganathan 2009 Any AE: 0/16
Withdrawal due to AE: 0/16
Any AE: 0/16
Withdrawal due to AE: 0/16
Three were withdrawn not due to adverse effects from study treatment
The serious AE,MI, was not clear which group
Ramos 2019 Fructo‐oligosaccharide
Any AE: 0/24
Withdrawal due to AE: 0
Maltodextrin
Any AE: 0/26
Withdrawal due to AE: 0
The discontinued patients were not due to GI symptoms or due to the need of antibiotics
Shariaty 2017 Any AE: not reported/18
Withdrawals due to AE: not reported
Any AE: not reported/18
Withdrawals due to AE: not reported
No adverse events reported, possible none
Sirich 2014 Any AE: 0/28
Withdrawal due to AE: 2/28
Any AE: 2/28 (bloating: 1; loose stools: 1)
Withdrawal due to AE: 2/28 (bloating: 1; loose stools: 1)
Soleimani 2017 Any AE: 0/30
Withdrawals due to AE: 0
Any AE: 0/30
Withdrawals due to AE: 0
Soleimani 2019 Any AE: 0/30
Withdrawals due to AE: 0
Any AE: not reported/30
Withdrawals due to AE: 0
Side effects only reported for synbiotic group
Quote: "No side effects were reported following the consumption of synbiotic in diabetic patients undergoing HD throughout the study."
SYNERGY 2014 Any AE: 1/unclear
Withdrawals due to AE: unclear
Any AE: 2/unclear
Withdrawals due to AE: unclear
Cross‐over denominators not reported
Three AE occurred during washout period
SYNERGY II 2021 Any AE: 11/35
Withdrawals due to AE: 0/35
One participant died during the study period for reasons unrelated to the study
Any AE: 7/33
Withdrawals due to AE: 0/33
  Eighteen serious AE in total: 7 in prebiotic (placebo) group, and 11 in synbiotic group. Initial hospitalisation accounted for 17 serious AEs
Synbiotic group: 1 participant died during the study period for reasons unrelated to the study
Viramontes‐Horner 2015 Any AE: 1/22 (diarrhoea: 1)
Withdrawals due to AE: 1
Any AE: 1/20 (diarrhoea: 2)
Withdrawals due to AE: 2
Wang 2015a Any AE: 0/23
Withdrawals due to AE: 0
Any AE: 0/18
Withdrawals due to AE: 0
Two patients in the probiotics group and five patients in the placebo group dropped out of the study because of noncompliance
One patient in the placebo group died due to head injury

AE: adverse event; GI: gastrointestinal; ITT: intention‐to‐treat; MI: myocardial infarction; UTI: urinary tract infection

Data and analyses

Comparison 1. Synbiotic versus synbiotic.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1.1 Patient‐reported outcomes: HRQoL at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
1.1.1 CKD stage G5D with diabetes and hypertension 1 46 Mean Difference (IV, Random, 95% CI) 1.98 [‐11.12, 15.08]

Comparison 2. Synbiotic versus prebiotic.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
2.1 Kidney function: eGFR at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.1.1 CKD stage 3Ga (transplant) 1 34 Mean Difference (IV, Random, 95% CI) ‐3.80 [‐17.98, 10.38]
2.2 Kidney function: serum creatinine at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.2.1 CKD stage G5D 1 42 Mean Difference (IV, Random, 95% CI) ‐0.20 [‐1.50, 1.10]
2.3 Uraemic toxins: urea at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.3.1 CKD stage G5D 1 42 Mean Difference (IV, Random, 95% CI) ‐2.10 [‐13.93, 9.73]
2.4 Uraemic toxins: indoxyl sulfate at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.4.1 CKD stage G5D 1 42 Mean Difference (IV, Random, 95% CI) 128.30 [‐242.77, 499.37]
2.5 Uraemic toxins: p‐cresol sulfate at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.5.1 CKD stage 3Ga (transplant) 1 34 Mean Difference (IV, Random, 95% CI) ‐2.10 [‐3.92, ‐0.28]
2.6 GI function: change in any GI upset or intolerance (prevalence of borborygmi) at 4 weeks 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
2.6.1 CKD stage 3Ga (transplant) 1 34 Risk Ratio (M‐H, Random, 95% CI) 15.26 [0.99, 236.23]
2.7 GI function: change in any GI upset or intolerance (GSRS Total Index at 12 months) 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.7.1 CKD Stage 3 to 4 1 56 Mean Difference (IV, Random, 95% CI) 0.00 [‐0.27, 0.27]
2.8 GI function: faecal pH at 7 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.8.1 CKD stage G5D 1 58 Mean Difference (IV, Random, 95% CI) ‐0.63 [‐1.13, ‐0.13]
2.9 GI function: faecal characteristics (Bristol Stool Chart) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.9.1 CKD stage 3Ga (transplant) 1 34 Mean Difference (IV, Random, 95% CI) ‐0.50 [‐1.15, 0.15]
2.10 GI function: faecal characteristics (Bristol Stool Chart) at 12 months 1   Mean Difference (IV, Random, 95% CI) Subtotals only
2.10.1 CKD stage 3 to 4 1 56 Mean Difference (IV, Random, 95% CI) 0.50 [‐0.18, 1.18]
2.11 Patient‐reported outcomes: HRQoL at 24 weeks 2   Mean Difference (IV, Random, 95% CI) Subtotals only
2.11.1 CKD stage G5D with diabetes and hypertension 2 65 Mean Difference (IV, Random, 95% CI) 6.38 [‐4.88, 17.64]
2.12 Adverse events: any adverse event 5   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
2.12.1 CKD stage G3a (transplant) 2 64 Risk Ratio (M‐H, Random, 95% CI) 15.26 [0.99, 236.23]
2.12.2 CKD stage 3 to 4 1 68 Risk Ratio (M‐H, Random, 95% CI) 1.48 [0.65, 3.36]
2.12.3 CKD stage 5GD 2 147 Risk Ratio (M‐H, Random, 95% CI) 1.10 [0.58, 2.09]
2.13 Withdrawals due to adverse events 4   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
2.13.1 CKD stage 3Ga (transplant) 2 64 Risk Ratio (M‐H, Random, 95% CI) Not estimable
2.13.2 CKD stage G5D 2 147 Risk Ratio (M‐H, Random, 95% CI) 0.78 [0.38, 1.62]

Comparison 3. Prebiotic versus prebiotic.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
3.1 Kidney function: eGFR at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.1.1 CKD stage 3 to 5 (non‐dialysis) 1 50 Mean Difference (IV, Random, 95% CI) 0.00 [‐1.73, 1.73]
3.2 Uraemic toxins: indoxyl sulfate at 6 weeks 2   Mean Difference (IV, Random, 95% CI) Subtotals only
3.2.1 CKD stage G5D with diabetes 2 64 Mean Difference (IV, Random, 95% CI) ‐0.20 [‐1.01, 0.61]
3.3 Uraemic toxins: p‐cresol sulfate at 6 weeks 2   Std. Mean Difference (IV, Random, 95% CI) Subtotals only
3.3.1 CKD stage G5D with diabetes 2 64 Std. Mean Difference (IV, Random, 95% CI) ‐0.04 [‐0.53, 0.45]
3.4 GI function: change in any GI upset or intolerance (burping) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.4.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.17 [‐0.50, 0.84]
3.5 GI function: change in any GI upset or intolerance (cramping) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.5.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐0.17 [‐0.50, 0.16]
3.6 GI function: change in any GI upset or intolerance (distension) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.6.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.33 [‐0.04, 0.70]
3.7 GI function: change in any GI upset or intolerance (flatulence) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.7.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 1.00 [0.25, 1.75]
3.8 GI function: change in any GI upset or intolerance (nausea) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.8.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.00 [‐0.00, 0.00]
3.9 GI function: change in any GI upset or intolerance (reflux) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.9.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.00 [‐0.50, 0.50]
3.10 GI function: change in any GI upset or intolerance (rumblings) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.10.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.50 [‐0.14, 1.14]
3.11 GI function: microbiota composition (Actinobacteria) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.11.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐1.26 [‐4.46, 1.94]
3.12 GI function: microbiota composition (Bacteriodetes) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.12.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 3.23 [‐8.24, 14.70]
3.13 GI function: microbiota composition (Proteobacteria) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.13.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.11 [‐1.61, 1.83]
3.14 GI function: microbiota composition (Firmicutes) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.14.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐2.44 [‐14.19, 9.31]
3.15 GI function: microbiota composition (Synergistetes) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.15.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐0.25 [‐0.89, 0.39]
3.16 GI function: microbiota composition (Verrucomicrobia) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.16.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.96 [‐1.36, 3.28]
3.17 GI function: microbiota composition (faecal acetate) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.17.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐69.91 [‐203.95, 64.13]
3.18 GI function: microbiota composition (faecal propionate) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.18.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐19.35 [‐63.87, 25.17]
3.19 GI function: microbiota composition (faecal butyrate) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.19.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐11.04 [‐39.57, 17.49]
3.20 GI function: microbiota composition (faecal total short‐chain fatty acids) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.20.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐104.71 [‐293.34, 83.92]
3.21 GI function: microbiota composition (faecal indoles) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.21.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 2.71 [‐69.78, 75.20]
3.22 GI function: microbiota composition (faecal p‐cresol) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.22.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 28.84 [‐105.07, 162.75]
3.23 GI function: faecal characteristics (bowel movements) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.23.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐0.27 [‐1.23, 0.69]
3.24 GI function: faecal characteristics (ease of passage) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.24.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) 0.00 [‐0.90, 0.90]
3.25 GI function: faecal characteristics (consistency) at 4 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
3.25.1 CKD stage G5D with diabetes 1 24 Mean Difference (IV, Random, 95% CI) ‐0.19 [‐1.34, 0.96]
3.26 Adverse events: number participants reporting an event 2   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
3.26.1 CKD stage G5D 2 79 Risk Ratio (M‐H, Random, 95% CI) 2.92 [0.10, 88.03]
3.27 Withdrawals due to adverse events 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
3.27.1 CKD stage G5D 1 39 Risk Ratio (M‐H, Random, 95% CI) 0.70 [0.13, 3.75]

Comparison 4. Prebiotic versus probiotic.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
4.1 Dialysis outcomes: peritonitis at 24 weeks 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
4.1.1 CKD stage G5D with diabetes and hypertension 1 47 Risk Ratio (M‐H, Random, 95% CI) Not estimable

Comparison 5. Low versus high dose probiotic.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
5.1 Kidney function: GFR at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
5.1.1 CKD stage G3 to G4 1 30 Mean Difference (IV, Random, 95% CI) ‐0.64 [‐9.51, 8.23]
5.2 Kidney function: serum creatinine at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
5.2.1 CKD stage G3 to G4 1 30 Mean Difference (IV, Random, 95% CI) ‐0.13 [‐0.89, 0.63]
5.3 Uraemic toxins: urea at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
5.3.1 CKD stage G3 to G4 1 30 Mean Difference (IV, Random, 95% CI) 4.57 [‐9.52, 18.66]

Comparison 6. Synbiotic versus placebo.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
6.1 Kidney function: eGFR at 6 or 12 weeks 2   Mean Difference (IV, Random, 95% CI) Subtotals only
6.1.1 CKD stage G3b 2 98 Mean Difference (IV, Random, 95% CI) 1.42 [0.65, 2.20]
6.2 Kidney function: SrCr at 6 or 12 weeks 2 98 Mean Difference (IV, Random, 95% CI) ‐0.57 [‐1.08, ‐0.07]
6.2.1 CKD stage G3b 2 98 Mean Difference (IV, Random, 95% CI) ‐0.57 [‐1.08, ‐0.07]
6.3 Uraemic toxins: urea at 6 or 12 weeks 2 117 Mean Difference (IV, Random, 95% CI) 3.34 [‐15.65, 22.32]
6.3.1 CKD stage G3b 1 75 Mean Difference (IV, Random, 95% CI) ‐3.48 [‐14.50, 7.54]
6.3.2 CKD stage G5D with diabetes and hypertension 1 42 Mean Difference (IV, Random, 95% CI) 17.10 [‐8.80, 43.00]
6.4 GI function: GSRS Item 6 (rumbling) at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
6.4.1 CKD stage 3b 1 23 Mean Difference (IV, Random, 95% CI) ‐0.54 [‐0.77, ‐0.31]
6.5 GI function: GSRS Item 13 (hard stools) at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
6.5.1 CKD stage 3b 1 23 Mean Difference (IV, Random, 95% CI) ‐0.09 [‐0.35, 0.17]
6.6 GI function: abdominal pain at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
6.6.1 CKD stage 3b 1 23 Mean Difference (IV, Random, 95% CI) 0.22 [‐0.02, 0.46]
6.7 GI function: constipation syndrome at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
6.7.1 CKD stage 3b 1 23 Mean Difference (IV, Random, 95% CI) ‐0.17 [‐0.72, 0.38]

Comparison 7. Prebiotic versus placebo or no treatment.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
7.1 Kidney function: serum creatinine at 8 or 12 weeks 3 145 Mean Difference (IV, Random, 95% CI) 0.52 [‐2.39, 3.44]
7.1.1 CKD stage A2 with diabetic nephropathy 1 70 Mean Difference (IV, Random, 95% CI) 4.70 [0.47, 8.93]
7.1.2 CKD stage G5D 2 75 Mean Difference (IV, Random, 95% CI) ‐0.75 [‐3.16, 1.66]
7.2 Uraemic toxins: urea at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.2.1 CKD stage A2 with diabetic nephropathy 1 70 Mean Difference (IV, Random, 95% CI) ‐0.40 [‐0.86, 0.06]
7.3 Uraemic toxins: indoxyl sulfate at 8 weeks 2   Std. Mean Difference (IV, Random, 95% CI) Subtotals only
7.3.1 CKD stage G5D 2 75 Std. Mean Difference (IV, Random, 95% CI) ‐0.14 [‐0.60, 0.31]
7.4 Uraemic toxins: p‐cresyl sulfate at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.4.1 CKD stage G5D 1 44 Mean Difference (IV, Random, 95% CI) ‐1.68 [‐14.30, 10.94]
7.5 GI function: microbiota composition (Facealibacterium) at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.5.1 CKD stage 5GD 1 44 Mean Difference (IV, Random, 95% CI) 2.37 [0.23, 4.51]
7.6 GI function: microbiota composition (Parabacteroides) at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.6.1 CKD stage 5GD 1 44 Mean Difference (IV, Random, 95% CI) 0.22 [0.06, 0.38]
7.7 GI function: microbiota composition (Bifidobacteria) at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.7.1 CKD stage 5GD 1 44 Mean Difference (IV, Random, 95% CI) ‐3.92 [‐9.83, 1.99]
7.8 GI function: microbiota composition (Ruminococcus) at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.8.1 CKD stage 5GD 1 44 Mean Difference (IV, Random, 95% CI) 3.86 [‐0.32, 8.04]
7.9 GI function: microbiota composition (Prevotella) at 8 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
7.9.1 CKD stage 5GD 1 44 Mean Difference (IV, Random, 95% CI) ‐0.43 [‐1.45, 0.59]
7.10 Adverse events: any adverse event (number of participants) 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
7.10.1 CKD stage G5D 1 50 Risk Ratio (M‐H, Random, 95% CI) 2.50 [0.53, 11.70]
7.11 Withdrawals due to adverse events 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
7.11.1 CKD stage G5D 1 50 Risk Ratio (M‐H, Random, 95% CI) 3.00 [0.13, 70.30]

Comparison 8. Probiotic versus placebo or no treatment.

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
8.1 Kidney function: GFR at 8, 12 or 15 weeks 3 128 Mean Difference (IV, Random, 95% CI) 2.73 [‐2.28, 7.75]
8.1.1 CKD stage 1 and type 2 diabetes 1 40 Mean Difference (IV, Random, 95% CI) 12.10 [4.19, 20.01]
8.1.2 CKD stage 3A with diabetes and hypertension 1 28 Mean Difference (IV, Random, 95% CI) 0.40 [‐4.15, 4.95]
8.1.3 CKD stage G5D with diabetes and hypertension 1 60 Mean Difference (IV, Random, 95% CI) 0.02 [‐0.63, 0.67]
8.2 Kidney function: serum creatinine at 8, 12 or 24 weeks 6 303 Mean Difference (IV, Random, 95% CI) ‐0.51 [‐1.06, 0.04]
8.2.1 CKD stage G5D 1 33 Mean Difference (IV, Random, 95% CI) ‐0.70 [‐4.48, 3.08]
8.2.2 Diabetic nephropathy all CKD stages 2 120 Mean Difference (IV, Random, 95% CI) ‐1.19 [‐2.96, 0.57]
8.2.3 CKD stage G5D with diabetes and hypertension 2 110 Mean Difference (IV, Random, 95% CI) 0.08 [‐0.48, 0.64]
8.2.4 CKD stage 1 and type 2 diabetes 1 40 Mean Difference (IV, Random, 95% CI) ‐0.17 [‐0.26, ‐0.08]
8.3 Kidney function: albuminuria at 12 or 24 weeks 4 193 Mean Difference (IV, Random, 95% CI) 0.02 [‐0.08, 0.13]
8.3.1 CKD stage G5D 1 33 Mean Difference (IV, Random, 95% CI) 0.00 [‐0.19, 0.19]
8.3.2 CKD stage G5D with diabetes and hypertension 3 160 Mean Difference (IV, Random, 95% CI) 0.03 [‐0.10, 0.16]
8.4 Kidney function: proteinuria at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
8.4.1 Diabetic nephropathy all CKD stages 1 60 Mean Difference (IV, Random, 95% CI) ‐15.60 [‐34.30, 3.10]
8.5 Uraemic toxins: urea at 12 or 24 weeks 5 263 Mean Difference (IV, Random, 95% CI) ‐1.11 [‐2.58, 0.35]
8.5.1 CKD stage G5D 1 33 Mean Difference (IV, Random, 95% CI) 1.80 [‐9.59, 13.19]
8.5.2 Diabetic nephropathy all CKD stages 2 120 Mean Difference (IV, Random, 95% CI) ‐1.90 [‐5.06, 1.26]
8.5.3 CKD stage G5D with diabetes and hypertension 2 110 Mean Difference (IV, Random, 95% CI) 0.06 [‐5.61, 5.72]
8.6 Uraemic toxins: indoxyl sulfate at 12 or 24 weeks 2 83 Mean Difference (IV, Random, 95% CI) ‐4.24 [‐9.83, 1.35]
8.6.1 CKD stage G5D 1 33 Mean Difference (IV, Random, 95% CI) ‐6.00 [‐15.02, 3.02]
8.6.2 CKD stage G5D with diabetes and hypertension 1 50 Mean Difference (IV, Random, 95% CI) ‐3.14 [‐10.26, 3.98]
8.7 Uraemic toxins: p‐cresyl sulfate at 4, 12 or 24 weeks 3 125 Mean Difference (IV, Random, 95% CI) ‐2.12 [‐5.19, 0.95]
8.7.1 CKD stage G5D 2 75 Mean Difference (IV, Random, 95% CI) ‐1.27 [‐2.31, ‐0.24]
8.7.2 CKD stage G5D with diabetes and hypertension 1 50 Mean Difference (IV, Random, 95% CI) ‐6.21 [‐13.92, 1.50]
8.8 Uraemic toxins: indole‐3‐acetic acid acid at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
8.8.1 CKD stage G5D 1 33 Mean Difference (IV, Random, 95% CI) ‐288.10 [‐464.41, ‐111.79]
8.9 GI function: any GI upset or intolerance (abdominal pain/diarrhoea) at 12 weeks 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
8.9.1 CKD stage G5D 1 33 Risk Ratio (M‐H, Random, 95% CI) 2.12 [0.21, 21.22]
8.10 GI function: any GI upset or intolerance (constipation) at 12 weeks 1   Risk Ratio (M‐H, Random, 95% CI) Subtotals only
8.10.1 CKD stage G5D 1 33 Risk Ratio (M‐H, Random, 95% CI) 3.18 [0.14, 72.75]
8.11 GI function: faecal pH at 12 weeks 1   Mean Difference (IV, Random, 95% CI) Subtotals only
8.11.1 CKD stage G5D 1 20 Mean Difference (IV, Random, 95% CI) ‐0.30 [‐0.83, 0.23]
8.12 Adverse events: any adverse event (number of participants) 4 205 Risk Ratio (M‐H, Random, 95% CI) 1.96 [0.63, 6.12]
8.12.1 CKD stage 1 and type 2 diabetes 1 44 Risk Ratio (M‐H, Random, 95% CI) 0.80 [0.25, 2.59]
8.12.2 CKD stage G5D 2 105 Risk Ratio (M‐H, Random, 95% CI) 4.95 [0.90, 27.12]
8.12.3 CKD stage G5D with diabetes and hypertension 1 56 Risk Ratio (M‐H, Random, 95% CI) 5.00 [0.25, 99.67]
8.13 Withdrawals due to adverse events 4 205 Risk Ratio (M‐H, Random, 95% CI) 4.96 [1.13, 21.77]
8.13.1 CKD stage G5D 2 105 Risk Ratio (M‐H, Random, 95% CI) 4.95 [0.90, 27.12]
8.13.2 CKD stage G5D with diabetes and hypertension 1 56 Risk Ratio (M‐H, Random, 95% CI) 5.00 [0.25, 99.67]
8.13.3 CKD stage 1 and type 2 diabetes 1 44 Risk Ratio (M‐H, Random, 95% CI) Not estimable

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Abbasi 2017.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single centre

  • Inclusion criteria: ≥ 25 years; CKD; microalbuminuria and GFR > 60 mL/min; type 2 DN

  • Exclusion criteria: history of inflammatory bowel disease, infection; liver disease; smoking, rheumatoid arthritis; alcoholism; recent antibiotic therapy; taking multivitamins or omega 3 within 1 month prior to study


Baseline characteristics
  • Number (randomised/analysed): intervention group (22/20); control group (22/20)

  • Mean age ± SD (years): intervention group (56.9 ± 8.1); control group (53.6 ± 7.19)

  • Sex (M/F): 19/21

  • CKD stage: 1 to 2

  • Kidney measurements

    • eGFR (mL/min/1.73 m²): intervention group (71.5 ± 9.5); control group (72.1 ± 9.11)

    • SCr (mg/dL): intervention group (1.01 ± 0.11); control group (1.03 ± 0.16)

  • Comorbidities: diabetic nephropathy; type 2 DM

  • GI status: none reported

Interventions Intervention group
  • Probiotics: soy milk + Lactobacillusplantarum A7 2 x 107 CFU/mL, 200 mL/day

  • Time: 8 weeks


Control group
  • No treatment: soy milk standard, 200 mL/day

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • SCr

  • Proteinuria

  • IL‐18

  • Serum sialic acid

  • TNF‐α

  • High‐sensitivity CRP

  • GFR

  • Serum cholesterol

  • Serum triglyceride

  • LDL

  • HDL

  • Physical activity (MET)

  • Serum sodium

  • Serum potassium

  • Fasting blood sugar

  • Serum genistein

Notes Baseline differences between groups
  • Possible differences in lipid panel, SCr, serum phosphorus, and serum genistein at baseline


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization was done by the use of computer‐generated random numbers"
Allocation concealment (selection bias) Low risk Quote: "Randomization and allocation were concealed from the researcher and volunteers till the analyses were finalised"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The bottles and soy milk were provided by the same soy milk factory. Bottles were identical in shape and probiotics soy milk was indistinguishable in colour, smell, and taste from conventional soy milk"
Quote: "At each study visit, participants received sufficient bottles for a 3‐day period until the next visit, in a double‐blinded design"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "All the tests were performed in a blinded fashion, in pairs (before and after the intervention) at the same time, in the same analytic run, and in random order to reduce systematic error and inter‐assay variability"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition: 9%
ITT analysis not undertaken
Selective reporting (reporting bias) High risk Trial registration or a priori published protocol: Iranian Registry of Clinical Trials IRCT20161027479N2. Missing outcome data for several planned outcomes from the trial registration, including proteinuria and TNF‐alpha
Control for confounding factors Low risk Pre‐treatment period: 2‐week run‐in phase: participants asked to avoid fermented or probiotic food, avoid consumption of any dietary supplements, report changes in medications
Study period: All participants consumed a diet containing 0.8 g/kg protein, 2000 mg sodium, 2000 mg potassium, 1500 mg phosphorus. Obtained using 24‐hour recall food diary
Other bias Unclear risk Conflicts declared: "None declared"
Funding declared: Isfahan University of Medical Sciences

Biruete 2017.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, multicentre, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Countries: Belgium, USA

  • Setting: multicentre (2 sites)

  • Inclusion criteria: ≥ 18 years; HD, 3 times/week for at least 3 months; able to provide 4 faecal samples

  • Exclusion criteria: previous major GI disease or intestinal resection; antibiotic treatment 1 month prior; sustained hypercalcemia; currently taking probiotics or prebiotics


Baseline characteristics
  • Number (randomised/analysed): 13/12

  • Mean age ± SD: 55 ± 10 years

  • Sex (M/F): 6/6

  • CKD stage: G5D

  • Kidney measurements

    • Mean serum albumin ± SD: 3.27 ± 0.25 g/dL

  • Comorbidities: diabetes (46%)

Interventions Intervention group
  • Prebiotic: inulin (females 10 g/day, males 15 g/day) in powder form sachet

  • Time: 4 weeks


Control group
  • Prebiotic: maltodextrin (females 6 g/day, males 9 g/day) in powder form sachet

  • Time: 4 weeks


Washout period
  • 4 weeks

Outcomes Outcomes reported by this study at 4 weeks
  • Faecal characteristics (Bristol Stool Chart)

  • Faecal microbiota (DNA extraction)

  • Faecal dry matter, SCFAs, phenols, indoles

  • Blood sample collection (plasma metabolites)

  • Dietary intake (48‐hour recall food diary)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants were randomized using a simple randomization technique (coin toss)"
Allocation concealment (selection bias) Low risk Quote: "Randomization was performed by a research coordinator so that both researchers and participants were blinded to the treatment allocation"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Randomization was performed by a research coordinator so that both researchers and participants were blinded to the treatment allocation"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Not explicitly clear that the outcome assessors are part of the double‐blinding
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
One death not related to study drug
Attrition = 8%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02718885
Control for confounding factors Unclear risk Pre‐treatment period: no run‐in phase length of time was mentioned. However, patients were excluded if they had been taking antibiotics 1 month prior and could not be currently taking prebiotics or probiotics (length of time unstated)
Study period: not reported
Other bias High risk Conflicts declared: "Beneo GmbH donated Orafti Synergy, but had no input in the study design, data analyses, or manuscript preparation. AB was supported by a Predoctoral Fellowship from CONACyT (Mexico’s Council of Science and Technology), a postdoctoral fellowship NIH‐T32 DK120524, has received honoraria from AMGEN, research grants from Keryx Pharmaceuticals for work unrelated to the present manuscript, and is part of the AUGmeNt workgroup from the Academy of Nutrition and Dietetics. BMK has received consulting fees, research grants, and is a member of the AUGmeNt workgroup from the Academy of Nutrition and Dietetics. KRW has received funding from the National Institutes of Health and the Renal Research Institute."
Funding declared: "This work was supported by a research grant from the Renal Research Institute (C2930) and the Division of Nutritional Sciences at the University of Illinois (USDA Vision 20/20 research program award)"

Bliss 1992.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, single‐blind RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: USA

  • Setting: single centre

  • Inclusion criteria: ≥ 18 years; treated with low protein diet for a minimum 4 months prior

  • Exclusion criteria: on dialysis; liver disease; kidney transplant; pregnant or lactating; active GI bleeding


Baseline characteristics
  • Number (randomised/analysed): 20/16

  • Age range: 20 to 72 years

  • Sex (M/F): 13/7

  • CKD stage: 1 to 3

  • Kidney measurements

    • Mean SCr ± SEM: 0.39 ± 0.07 mol/L

  • Comorbidities: diabetic nephropathy (4); arterial nephrosclerosis (4); glomerular disease (3); amyloid nephropathy (1); FSGS (1); obstructive uropathy (1); polycystic kidney disease (1); scleroderma (1)

Interventions Intervention group
  • Prebiotic: gum Arabic 25 g mixture in 150 mL juice twice/day (total 50 g gum Arabic)

  • Time: 4 weeks


Control group
  • Prebiotic: placebo mixture of pectin 0.5 g in 150 mL juice twice/day (total 1 g pectin)

  • Time: 4 weeks

Outcomes Outcomes reported by this study at 4 weeks
  • Stool fractionation (stool weight and faecal bacterial mass), each 5 days, wet weight and dry weight

  • Nitrogen analyses (nitrogen excretion and serum urea nitrogen)

  • Nutritional assessment and dietary intake (food diary)

  • Adverse events

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "Subjects were randomly divided into two groups during dietary period 1"
Insufficient information provided regarding methodology used to undertake randomisation, unable to form judgement
Allocation concealment (selection bias) Unclear risk No information provided regarding concealment of the allocation
Blinding of participants and personnel (performance bias)
All outcomes High risk "The investigator provided subjects in both groups with the juice mixtures and the subjects were not informed of their content"
Single‐blind study
Blinding of outcome assessment (detection bias)
All outcomes High risk Single‐blind study
Incomplete outcome data (attrition bias)
All outcomes High risk No missing outcome data; however no details about which arm non‐completers were from
Denominators for each group between cross‐over period are unclear
Attrition = 20%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration: not reported
Control for confounding factors Low risk Pre‐treatment period: low protein diet 4 months prior
Study period: continued low‐protein diet
Other bias High risk Conflicts declared: "Supported in part by an individual National Research Award (NR06335), the American Nurses' Foundation 1989 Bristol Myers Fund Scholar Award (to DZB), and the 1989 American Nurses' Foundation/Sigma Theta Tau International Scholar Award (to DZB). Welch's brand frozen fruit juice concentrates and Glad‐Lock freezer bags were generously provided by Welch's Foods, Inc (Westfied, NY) and by First Brands Corporation (Danbury, CT), respectively. The price of Nutriloid Gum Arabic Spray Dry Powder was discounted by TIC Gums, Inc (Belcamp, MD)."
Funding declared: not reported

Borges 2018.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 12 weeks

  • Follow‐up: none reported

Participants Study characteristics
  • Country: Brazil

  • Setting: single centre (HD centre)

  • Inclusion criteria: ≥ 18 years; undergoing maintenance HD for at least 6 months

  • Exclusion criteria: inflammatory diseases; cancer; AIDS; autoimmune disease; smokers; use of a central catheter for HD access; amputated limbs; pregnancy; patients who had used catabolic drugs, antioxidant vitamin supplements pre, pro, and synbiotic and antibiotics in the last 3 months


Baseline characteristics
  • Number (randomised/analysed): intervention group (23/16); control group (23/17)

  • Mean age ± SD (years): intervention group (53.6 ± 11.0); control group (50.3 ± 8.5)

  • Sex (M/F): intervention group (11/5); control group (10/7)

  • CKD stage: G5D

  • Kidney measurements

    • Mean albumin ± SD (g/dL): intervention group (4.2 ± 0.23); control group (4.2 ± 0.22)

    • Mean SCr ± SD (mg/dL): intervention group (14.3 ± 0.8); control group (9.3 ± 1.5)

    • Maintenance HD, time on dialysis (months, range): intervention group (60, 38.2 to 105); control group (36.5, 24.2 to 72)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Probiotic (oral): Streptococcusthermophilus, Lactobacillusacidophilus, and Bifidobacterialongum strains (of 30 billion live bacteria, totalling 90 billion CFU/day), 3 capsules/day

  • Time: 3 months


Control group
  • Placebo (oral): 3 capsules/day

  • Time: 3 months

Outcomes All outcomes reported by this study at 12 weeks
  • Adherence to treatment

  • Adverse events

  • Urea (pre and post‐dialysis)

  • Creatinine

  • Potassium

  • Hb

  • Albumin

  • Globulin

  • CRP

  • IL‐6

  • pH fecalIndoyl sulfate

  • p‐cresyl sulfate

  • Indole‐3‐acetic acid

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The random sequence of treatment (probiotic and placebo) was manually generated for a simple randomization"
Allocation concealment (selection bias) Low risk Quote: "None of the subjects involved in the study had access to the allocation sequence until the end of the statistical analyses"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The participants and the researchers who interviewed and visited the subjects were blinded to the contents of the bottles, which contained probiotic or placebo capsules"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Researchers who interviewed and visited the subjects were blinded to the contents of the bottles, which contained probiotic or placebo capsules. All laboratory measurements were centralized and performed in a blinded manner."
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided (GI issues related to study drug)
Attrition = 30%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial Registration: not reported
A priori published protocol: not reported
Comment: insufficient information provided to permit judgement. Unclear whether pre‐specified outcome data are reported, if any.
Lack of clinically important or relevant outcomes: no issue
Control for confounding factors Unclear risk Pre‐treatment period: none reported
Study period: no mention of control for dietary factors during study
Other bias High risk Funding source
  • 2018: "This study was supported by Coordenacao de Aperfeicoamento de Pessoal de Nivel Superior (CAPES), Comite Francais d’Evaluation de la Cooperation Universitaire avec le Bresil (COFECUB), Conselho Nacional de Desenvolvimento Cientıfico e Tecnologico (CNPq), Fundacao de Amparoa Pesquisa do Estado do Rio de Janeiro (FAPERJ), and Clinical Research Unit‐UPC‐HUAP‐UFF."

  • 2019: "The Heart and Lung Foundation and “Njurfonden” support Peter Stenvinkel’s research. Conselho Nacional de Pesquisa (CNPq), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), and Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) support Denise Mafra’s research. Baxter Novum is the result of a grant from Baxter Healthcare to Karolinska Institutet."


Conflicts of interest
  • 2018: "The authors declare that they have no relevant financial interests"

  • 2019:"Bengt Lindholm is employed by Baxter Healthcare. The other authors do not declare any potential conflicts of interest."

Cosola 2021.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, single‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: 12 weeks (4 weeks past end of treatment)

Participants Study characteristics
  • Country: Italy

  • Setting: single centre (outpatient)

  • Inclusion criteria: 30 to 65 years; CKD stage 3b to 5, non‐dialysis; BMI 18.5 to 29.9; controlled diet

  • Exclusion criteria: type 2 DM; use of antibiotics or probiotics up to 30 days prior to recruitment; chronic GI disorders; systemic inflammatory diseases; suspicion or clinical diagnosis of malignancy; chronic liver disease; treatment with corticosteroids or immunosuppressive drugs; previous acute CVD (MI, stroke); psychiatric conditions reducing the compliance to treatment protocols


Baseline characteristics
  • Number: intervention group (13); control group (10)

  • Mean age ± SD (years): intervention group (51 ± 4.3); control group (51.5 ± 2.8)

  • Sex (M/F): intervention group (7/6); control group (7/3)

  • CKD stage: 3

  • Kidney measurements

    • Mean eGFR ± SD (mL/min): intervention group (31.5 ± 2.8); control group (23.9 ± 3.6)

    • Mean SCr ± SD (mg/dL): intervention group (2.3 ± 0.2); control group (3.2 ± 0.4)

    • Mean serum albumin ± SD (g/dL): intervention group (3.9 ± 0.1); control group (3.9 ± 0.1)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: Lactobacillus casei LC4P1 2.4 x 109, Bifidobacterium animalis BLC1 2.4 x 109, fructo‐oligosaccharides 2.5 g, inulin 2.5 g, and natural antioxidants (a mix of quercetin 0.064 g, reservatrol 0.023 g and proanthocyanidins 0.013 g)

  • Time: 8 weeks


Control Group
  • Placebo: not reported

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • P‐cresyl sulfate

  • Indoxyl sulfate

  • Intestinal permeability

  • GI symptoms (reported in separate paper)

  • BMI

  • eGFR

  • SCr

  • Serum albumin

  • Azotaemia

  • Serum calcium

  • Serum phosphorus

  • Serum potassium

  • Serum sodium

  • CaxP

  • Total serum proteins

  • CRP

  • Urinary creatinine

  • Urinary proteins

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The randomization was performed by an independent researcher by means of software, using gender as a blocking factor"
Allocation concealment (selection bias) Unclear risk No information provided
Blinding of participants and personnel (performance bias)
All outcomes High risk Quote: "The powder formulations were anonymously labelled and were undistinguishable for the patients"
Single‐blind study
Blinding of outcome assessment (detection bias)
All outcomes High risk Single‐blind study
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 0% (in CKD participants)
ITT analysis undertaken in CKD participants
Selective reporting (reporting bias) High risk Trial registration or a priori published protocol: NCT03815786
Outcomes: "TMAO was not evaluated due to technical problems during the execution of the analysis"
Control for confounding factors Unclear risk Pre‐treatment period: Unclear, "controlled diet"
Study period: Unclear
Other bias Unclear risk Conflicts declared: "S.F. is the Chief Executive Officer of Research Center « Sergio Fontana 1900–1982» (Farmalabor); F.M.L.F. is the research and development manager of the Research Center « Sergio Fontana 1900–1982» (Farmalabor). The other authors declare no conflict of interest."
Funding declared: "This study has been funded from the XUANRO4‐NATURE‐Nuovo Approccio Per la Riduzione Delle Tossine Uremiche Renali, REGIONE PUGLIA‐FSC 2007–2013 Ricerca. Intervento 'Cluster Tecnologici Regionali' "

de Andrade 2021.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Brazil

  • Setting: single centre (outpatient clinic)

  • Inclusion criteria: 18 to 80 years; APD, dialysis vintage of at least 3 months; adherent to dialysis treatment

  • Exclusion criteria: currently using prebiotics, probiotics, synbiotics, and antibiotics one month before the beginning of the study; the presence of inflammatory bowel diseases, stomach, or bowel resection; liver cirrhosis; cancer; HIV; peritonitis in the last month; pregnancy; breastfeeding; presenting any GI intolerance after dose adjustments; hospitalised; started antibiotics; underwent kidney transplant once starting study


Baseline characteristics
  • Number (randomised/analysed): 43/26

  • Mean age ± SD: 55 ± 12 years

  • Sex (M/F): 23/43

  • CKD stage: 1 to 5, including G5D

  • Kidney measurements

    • Median SCr (IQR) (mg/dL): 8.3 (6.5 to 11.8)

    • Mean albumin± SD (g/dL): 3.9 ± 0.34

  • Comorbidities: diabetes (85%); diabetic nephropathy (19%); hypertensive nephropathy (4%); GN (8%); polycystic kidney disease (15%)

  • GI status: median (IQR) GSRS score 29 (24–41)

Interventions Intervention group
  • Prebiotic: unripe banana flour (48% resistant starch) 21 g/day. Dose escalation to double/day, after 3 days in the absence of adverse effects

  • Time: 4 weeks


Washout
  • 4 weeks


Control group
  • Placebo: waxy corn starch 12 g/day. Dose escalation to double/day, after 3 days in the absence of adverse effects

  • Time: 4 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Indoxyl sulfate

  • p‐cresyl sulfate

  • Indoxyl 3‐acetic acid

  • High‐sensitivity CRP

  • IL‐6

  • IL‐10

  • TNF‐alpha

  • Serum lipopolysaccharide LPS

  • Dietary intake

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • No available data for meta‐analyses: first phase of crossover not reported as separate data

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The participants were assigned to sequential treatment with UBF and placebo, or vice versa, after a blocked randomization procedure using a random block of 4 participants in a 1:1 ratio."
Allocation concealment (selection bias) Low risk Quote: "Supplements were packaged in identical sachets and identified by the numbers 225 and 653 in a handling pharmacy, under the responsibility of a pharmacist (Magister Pharmacy, Sao Paulo, Brazil)"
Quote: "The list was handed over to another independent researcher who was responsible for separating and allocating supplements to participants"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Supplements were packaged in identical sachets and identified by the numbers 225 and 653 in a handling pharmacy, under the responsibility of a pharmacist (Magister Pharmacy, Sao Paulo, Brazil)"
Quote: "The list was handed over to another independent researcher who was responsible for separating and allocating supplements to participants"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 40%
Reasons for withdrawals not of major concern related to study treatment
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: Brazilian Clinical Trials Registry (www.ensaiosclinicos.gov.br, IDnumber: RBR‐4xxwwj
Control for confounding factors Unclear risk Pre‐treatment period: "The majority of patients had been previously advised by a renal dietician, according to the nutritional guidelines for patients undergoing peritoneal dialysis"
Study period: "During each intervention, the participants were advised to maintain a stable dietary pattern and not to take prebiotics, probiotics, or synbiotics. If necessary, dietary adjustments were advised or reinforced during the follow‐up"
Other bias High risk Conflicts declared: "The authors declare no conflict of interest"
Funding declared: "L.S.d.A., F.A.H.S., N.B.F.P., R.R.T., J.D.d.L. received a scholarship from Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES). SD. Rodrigues receives a scholarship from CAPES—Fundação Araucária and L.C. receives a scholarship from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) (#302765/2017‐4). The production and analysis of UBF were supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) (#13/07914‐8 (FoRC)). Support for this research was provided by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) (#2018/12122‐7)"

de Araujo 2022.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Study duration
  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Brazil

  • Setting: single centre (dialysis clinic)

  • Inclusion criteria: 22 to 69 years; HD

  • Exclusion criteria: taking probiotics; pregnant; kidney transplant patients; GI disorders; cancer; previous GI surgery; paraplegia, tetraplegia or amputations, HIV/AIDs; tumours; autoimmune disease; antibiotics and anti‐inflammatory drugs; > 69 years (morphological and functional changes that come with aging, including lower kidney function); children and adolescents; unable to take probiotics regularly


Baseline characteristics
  • Number (randomised/analysed): intervention group (40/32); control group (40/38)

  • Mean age ± SD (years): intervention group (47 ± 13); control group (49 ± 13)

  • Sex (M/F): intervention group (14/18); control group (19/19)

  • CKD stage: G5D

  • Kidney measurements: not reported

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Lactobacillus Plantarum A87, Lactobacillus rhamnosus, Bifidobacterium bifidum A218 and Bifidobacteriumlongum A101, each 4 x 109 CFU, oral capsule/day

  • Time: 12 weeks


Control group
  • Placebo: oral soybean placebo capsule

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • CRP

  • Cystatin C

  • NGAL

  • Blood glucose

  • BoMI

  • Syndecan‐1

  • INF‐y

  • Calcium

  • Phosphorus

  • PTH

  • Glutamic pyruvic transaminase

  • HCT

  • Hb

  • Potassium

  • Glucose

  • Urea

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Software program Research Randomizer® (www.randomizer.org) was used to randomly assign participants to study groups. Individuals in Group 1 (G1) were administered supplementation with probiotics and subjects in Group 2 (G2) were given placebo."
Allocation concealment (selection bias) Low risk Quote: "The researcher and the participants were blinded for the sequential allocation and the codes assigned to supplementation or placebo"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The supplements and placebo offered to patients had equal organoleptic characteristics... The capsules had the same colour, physical appearance, and flavour"
Insufficient information provided to permit judgement
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 13%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias High risk Conflicts of interest: "The authors have no conflict of interest to declare"
Funding declared: "The authors would like to thank the following institutions for their support: Compounding pharmacies BioPhormula and Galena Farmacêutica for donating probiotics to the study; the authors of the study have no financial ties with the compounding pharmacies. The Ceará Foundation for Scientific and Technological Development (FUNCAP) for funding and encouraging the organization of this trial. The National Council for Scientific and Technological Development (CNPq) for offering performance scholarships to researchers AMCM, EFD e GBSJ."
Industry funding

Dehghani 2016.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 6 weeks

  • Follow‐up: none reported

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (Yazd University Clinic)

  • Inclusion criteria: 35 to 75 years; CKD stage 3 or 4 (GFR 15 mL/min/1.73 m² to 59 mL/min/1.73 m²)

  • Exclusion criteria: pregnancy; antibiotics and lactulose 14 days before the start of the study; alcohol dependence; hepatitis or HIV infection


Baseline characteristics
  • Number (randomised/analysed): intervention group (38/31); control group (37/35)

  • Mean age ± SD (years): intervention group (63.00 ± 6.52); control group (60.00 ± 8.33)

  • Sex (M/F): intervention group (23/8); control group (27/8)

  • CKD stage: G3b

  • Kidney measurements

    • eGFR (mL/min/1.73 m²): intervention group (41.35 ± 15.74); control group (41.40 ± 16.91)

    • SCr (mg/dL): intervention group (2.00 ± 0.70); control group (2.15 ± 1.02)

    • CrCl (mL/min /1.73 m²): intervention group (28.24 ± 13.32); control group (28.24 ± 13.32)

    • BUN (mg/dL): intervention group (40.80 ± 22.11); control group (37.22 ± 21.95)

    • Serum uric acid (mg/dL): intervention group (5.89 ± 1.70); control group (5.30 ± 1.00)

  • Comorbidities: DM (35%); hypertension (55%); heart disease (6%)

  • GI status: 'gastrointestinal diseases' 4%

Interventions Intervention group
  • Synbiotic: 2 Familact capsules/day (Zist Takhmir, Tehran, Iran), 500 mg (containing 7 strains of probiotics: Lactobacilluscasei, Lactobacillusacidophilus, Lactobacillusbulgarigus, Lactobacillusrhamnosus, Bifidobacteriumbreve, Bifidobacterium longum, Streptococcusthermophilus, and prebiotic fructo‐oligosaccharides), after the meal

  • Time: 6 weeks


Control group
  • Placebo 500 mg, 2 capsules/day

  • Time: 6 weeks

Outcomes All outcomes reported by this study at 6 weeks
  • BUN

  • SCr

  • Serum uric acid

  • CrCl

  • GFR

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "This study was a randomized ... clinical trial"
Allocation concealment (selection bias) Unclear risk Quote: No information regarding the concealment of allocation to treatment groups
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "For blinding, placebo was produced in similar colour and appearance of the supplement capsules, as well as the shape of packaging (Zist Takhmir, Tehran, Iran). To differentiate the two types of capsules, a small code was written on the box of the capsules (‘A’ or ‘B’), and neither the patients nor the person delivering the capsule to the patients were informed of the codes and type of supplement capsules."
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information provided
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 13%
ITT analysis not/undertaken
Quote: "Attrition criteria were the use of antibiotics and lactulose during the study and starting treatment with haemodialysis."
Selective reporting (reporting bias) High risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: none reported
Study period: no control for dietary factors reported
Other bias High risk Funding source: not reported
Conflicts of interest: "None declared"

Ebrahim 2022.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, single‐blind RCT


Time frame
  • August 2018 to December 2019

  • Treatment: 14 weeks

  • Follow‐up: 14 weeks of treatment

Participants Study characteristics
  • Country: South Africa

  • Setting: single centre (pre‐dialysis clinic)

  • Inclusion criteria: ≥ 18 years; CKD stage 3 to 5, GFR < 60 mL/min/1.73 m²

  • Exclusion criteria: taking antibiotics, prebiotics or probiotics 4 weeks prior; inflammatory bowel disease; bowel malignancy; previous colorectal surgery (or any other serious bowel disorder); pregnancy; DM; coeliac disease; HIV disease; malignant hypertension; crescentic glomerular nephritis; on immunosuppressant medications,; expected to start immediate dialysis


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/23); control group (29/22)

  • Mean age ± SD (years): intervention group (40.6 ± 11.4); control group (41.3 ± 12.0)

  • Sex (M/F): intervention group (11/19); control group (14/15)

  • CKD stage: stage 3 to 5 non‐dialysis

  • Kidney measurements

    • Median eGFR, IQR (mL/min/1.73 m²): stage 3 (19, 32.2); stage 4 (16, 27.1); stage 5 (24,40.7)

  • Comorbidities: polycystic (3); hypertension (29); glomerular disease (13)

  • GI status: not reported

Interventions Intervention group
  • Prebiotic + diet control

    • Prebiotic: ß‐glucan prebiotic fibre powder added to meals 13.5 g,day

    • Diet control: simplified diet based on natural, healthy food with the avoidance of take‐aways, salt and processed foods rich in additives, and protein restriction of 0.8 g/kg

  • Time: 14 weeks


Control group
  • No treatment + diet control as above

  • Time: 14 weeks

Outcomes Outcomes reported by this study at 14 weeks
  • Urea

  • Creatinine

  • eGFR

  • Total cholesterol

  • LDL cholesterol

  • HDL cholesterol

  • Triglycerides

  • Total and free indoxyl sulfate

  • Total and free p‐cresyl sulfate

  • Total and free p‐cresyl glucuronide

  • Total and free indole‐3‐acetic acid

  • CRP

  • Potassium

  • Phosphate

  • Body weight

  • BMI

  • Mid‐upper arm circumference

  • Energy

  • Protein

  • Total fat

  • Saturated fat

  • Diet fibre

  • Sodium

  • Microbial compositions of general relative abundance

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants (were) randomized to the prebiotic supplement or the diet control group using a simple computer‐generated randomized list with an equal allocation ratio"
Allocation concealment (selection bias) High risk Quote: "Sequentially numbered sealed opaque envelopes were used to assign group allocation"
Single‐blind study, blinded to the investigators
Blinding of participants and personnel (performance bias)
All outcomes High risk Quote: "The principal investigator (PI) was blinded to the treatment"
Single‐blind study, blinded to the investigators
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 24%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: PACTR202002892187265
Control for confounding factors Low risk Pre‐treatment period: "All participants had a 4 week run‐in period on diet only before being randomized to the prebiotic supplement or the diet control group"
Study period: Co‐intervention was diet control
Other bias Low risk Conflicts declared: "The authors declare no conflict of interest"
Funding declared: "This study received funding from the Early Researcher Fund (Stellenbosch University), Thuthuka National Research Fund (Thuthuka grant number: 129901) and the Harry Crossley Foundation"

Eidi 2018.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, single‐centre, triple‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past 4 weeks treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (hospital dialysis ward)

  • Inclusion criteria: ≥ 20 years ; kidney failure undergoing chronic maintenance HD for at least 3 months; may have diabetic nephropathy, hypertensive nephropathy, nephrotic syndrome, GN

  • Exclusion criteria: history of smoking; PD or previous kidney transplant; lactation or pregnancy; drug history including antibiotics, prebiotics, probiotics, herbal drugs, psychedelic drugs


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/21); control group (29/21)

  • Mean age ± SD (years): intervention group (57.05 ± 13.95); control group (59.67 ± 15.04)

  • Sex (M/F): intervention group (15/6); control group (17/4)

  • CKD stage: G5D

  • Kidney measurements

    • Albumin (g/dL): intervention group (4.17 ± .57); control group (4.32 ±.80)

    • SCr (mg/dL): intervention group (8.67 ± 1.92); control group (8.28 ± 3.04)

    • Uric acid (mg/dL): intervention group (6.83 ± 1.24); control group (7.11 ± 1.47)

    • Other CKD biomarkers: hypertensive nephropathy most common cause of kidney failure

  • Comorbidities

    • Diabetes: intervention group (23.8%); control group (38.1%)

    • Hypertension: intervention group (42.9%); control group (38.1%)

    • GN: intervention group (0%); control group (4.8%)

  • GI status: dietary fibre intake (g/day): intervention group (11.33 ± 7.33); control group (11.50 ± 6.61)

Interventions Intervention group
  • Probiotic: 1 capsule/day containing 1.6 x 107 CFU of LactobacillusRhamnosus from yogurt and cheese of different farms located in the suburbs Heris after meal

  • Time: 28 days


Control group
  • Placebo: 1 capsule/day infant formula after a meal

  • Time: 28 days

Outcomes Outcomes reported by this study at 4 weeks
  • Serum phenol

  • Serum p‐cresol

  • Anthropometric measurements: weight using calibrated Seca scale with a precision of 0.1 kg

  • Energy intake: 3 days food recall was used and analysed with Nutritionist 4 software

  • FGIDs: symptoms in the oesophagus, stomach and intestines, using 93 items questionnaire based on ROM III

  • Anthropometric measurements, BMI

  • Macronutrient intake: proteins, fats and carbohydrates, 3 days food recall was used and analysed with Nutritionist 4 software

  • FGIDs: symptoms in the gall bladder and pancreas, rectum and anal canal, using 93 items questionnaire based on ROM III

  • Intestinal (stool) microbiome, bacteroidetes: real‐time PCR of DNA extracted from stool sample

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "Triple ‐blind randomized placebo‐controlled trial with an allocation ratio of (1:1)"
Quote: "The eligible participants were randomly assigned to Lactobacillus Rhamnosus as probiotics or placebo groups by a randomization procedure and subject were matched in each arms regarding to age, sex and history of haemodialysis duration."
Allocation concealment (selection bias) Unclear risk No information regarding the concealment of allocation to treatment groups. Insufficient information provided within abstract to permit judgement.
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Study states to be triple‐blind however no information regarding the methods use to keep participants or study personnel blinded treatment groups
Insufficient information provided within abstract to permit judgement
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Study states to be triple‐blind however no information regarding the methods use to keep outcome assessors blinded treatment groups.
Insufficient information provided within abstract to permit judgement.
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = unclear
ITT analysis unclear
Serious issues regarding ITT population, number of participants received intervention, whether ITT analysis was correctly performed. Numbers do not match.
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: IRCT201504182017N21. Study registration planned a 4‐arm trial: (1) Probiotic; Lactobacillus rhamnosus (16 Billion CFU) one capsule per day for 28 days; (2) Prebiotic; Inulin (10 grams) one sachet per day for 28 days; (3) Synbiotic; Lactobacillus Rhamnosus (16 Billion CFU, one capsule) with Inulin (10 grams, one sachet) per day for 28 days; (4) Corn starch (10 grams, one sachet) with Infant formula (one capsule) per day, for 28 days. Query on why this was not undertaken
Lack of clinically important or relevant outcomes: no issue
Control for confounding factors Unclear risk Pre‐treatment period: Nothing reported
Study period: "All the participants were asked not to change their usual dietary intakes, recommended vitamin or minerals supplements, physical activity, medication or traditional medicine as adjuvant therapy over the course of the study." "Food intake was assessed using three 24‐h dietary recalls undertaken on separate days (one haemodialysis weekday, one non‐haemodialysis weekday and one non‐haemodialysis weekend day) using a visual aid photo album of real foods before and after treatment. Energy and nutrient intake were analyzed using Nutritionist IV (Axxya Systems, Stafford, TX)."
Other bias Low risk Conflicts declared
  • "Authors of this study declare no conflict of interest"


Funding declared
  • "No sources of funding"

Elamin 2017.

Study characteristics
Methods Study design
  • Parallel, 3‐arm, open‐label RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Saudi Arabia

  • Setting: single centre (Nephrology clinic)

  • Inclusion criteria: ≥ 18 years; CKD stages 3B or 4

  • Exclusion criteria: malignancy; liver disease; intestinal resection; inflammatory bowel disease; recent antibiotic therapy; recent gum Arabic use


Baseline characteristics
  • Number (randomised/analysed): intervention group 1 (12/12); intervention group 2 (12/9); intervention group 3 (12/9)

  • Mean age ± SD (years): intervention group 1 (48 ± 12); intervention group 2 (52 ± 18); intervention group 3 50 ± 17)

  • Sex (M/F): intervention group 1 (8/4); intervention group 2 (6/3); intervention group 3 (6/3)

  • CKD stage: 3B (15); 4 (15)

  • Kidney measurements

    • eGFR (mL/min/1.73m²): intervention group 1 (26 ± 10); intervention group 2 (33 ± 11); intervention group 3 (29 ± 9)

    • BUN (mmol/L): intervention group 1 (14 ± 6); intervention group 2 (12 ± 5); intervention group 3 (14 ± 4)

  • Comorbidities: Immunosuppression for lupus nephritis and glomerular disease: intervention group (33.3%); intervention group 2 (22.2%); intervention group 3 (22.2%)

  • GI status: not reported

Interventions Intervention group 1
  • Prebiotic: gum Arabic 10 g instantly soluble granules in glass of water or juice, once/day

  • Time: 4 weeks


Intervention group 2
  • Prebiotic: gum Arabic 20 g instantly soluble granules in glass of water or juice, once/day

  • Time: 4 weeks


Intervention group 3
  • Prebiotic: gum Arabic 40 g instantly soluble granules in glass of water or juice, once/day

  • Time: 4 weeks

Outcomes All outcomes reported by this study at 4 weeks
  • GFR

  • Creatinine

  • BUN

  • Sodium

  • Potassium

  • Bicarbonate

  • Uric acid

  • Calcium

  • Phosphorus

  • Magnesium

  • PTH

  • Hb

  • Indoxyl sulfate

  • Urine volume

  • Creatinine excretion

  • Urea excretion

  • Protein excretion

  • CrCl

  • Urea clearance

  • Serum CRP

  • Serum IL‐1

  • Serum IL‐2

  • Serum IL‐4

  • Serum IL‐5

  • Serum IL‐6

  • Serum IL‐10

  • Serum IL‐12

  • Serum IL‐13

  • Serum IFN

  • Serum TNF

  • Urinary TGF

  • GI symptoms: abdominal pain; abdominal distention; flatulence; nausea; diarrhoea; constipation

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article/abstract

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "According to recruitment order, we divided patients into three equal groups using computer‐generated random number lists"
Allocation concealment (selection bias) Unclear risk No information regarding the concealment of allocation to treatment groups
Blinding of participants and personnel (performance bias)
All outcomes High risk Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Open‐label study
Incomplete outcome data (attrition bias)
All outcomes Unclear risk All participants were accounted for with reasons for withdrawals provided
Attrition = 25%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias Unclear risk Conflicts declared: "The authors declared no conflicts of interest"
Funding declared: "This clinical trial was funded by a generous grant from King Abdul‐Aziz City for Science and Technology (KACST) in Saudi Arabia (no. 35‐300). The authors would like to acknowledge their gratitude to Professor Yacine Badjah and the Chair of Advanced Materials Research at King Saud University in Riyadh for their kind assistance in performing the liquid chromatography assay."

Esgalhado 2018.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: none reported past treatment

Participants Study characteristics
  • Country: Brazil

  • Setting: single centre (Renal clinic)

  • Inclusion criteria: ≥ 18 years; undergoing maintenance HD3 times/week for at least 6 months; arteriovenous fistula for vascular access in upper limb

  • Exclusion criteria: autoimmune, infectious or neoplastic diseases; HIV; taking catabolic drugs, antioxidants or vitamin supplements, pre‐, pro‐, synbiotic and antibiotic in the last 3 months before this study or pregnant; regularly practiced physical exercise


Baseline characteristics
  • Number (randomised/analysed): intervention group (19/15); control group (19/16)

  • Mean age ± SD (years): intervention group (56.0 ± 7.5); control group (53.5 ± 11.5)

  • Sex (M/F): intervention group (7/8); control group (11/5)

  • CKD stage: G5D

  • Kidney measurements

    • Albumin (g/dL): intervention group (3.8 ± 0.3); control group (3.7 ± 0.3)

    • SCr (mg/dL): intervention group (8.4 ± 2.3); control group (8.1 ± 2.3)

    • Time on dialysis (months): intervention group 50.0 ± 36.5); 4control group (4.3 ± 26.4)

  • Comorbidities: not reported

  • GI status: dietary fibre intake (g/day): intervention group (18.5 ± 7.1); control group (18.8 ± 8.3)

Interventions Intervention group
  • Prebiotic: resistant starch 16 g, once/day (cookie on dialysis days, powder sachet in a drink on non‐dialysis days)

  • Time: 4 weeks


Control group
  • Placebo: (manioc flour) sachet in a drink, once daily

  • Time: 4 weeks


Cross‐over
  • Four weeks of washout of no treatment

Outcomes All outcomes reported by this study at 12 weeks
  • RNA

  • Nuclear factor‐κB

  • Nuclear factor erythroid‐derived 2‐like 2

  • NAD (P)H: quinone oxidoreductase‐1

  • Vascular cell adhesion molecule

  • Kelch‐like ECH‐associated protein 1

  • Heme oxygenase‐1

  • Plasma levels

  • Uraemic toxin plasma levels after resistant starch supplementation

  • CRP (mg L−1)

  • IL‐6

  • Thiobarbituric acid reactive substances

  • Indoxyl sulphate

  • P=cresyl sulphate

  • Indole‐acetic‐acid

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "An independent investigator, with no involvement in the study, randomized and allocated the patients in two groups. The randomization and the allocation were created using the statistical RStudio software version 1.1463, and the allocation of the patients to different groups was performed using a statistical tool that generates random samples without replacement."
Allocation concealment (selection bias) Low risk Quote: "An independent investigator, with no involvement in the study, randomized and allocated the patients in two groups. The randomization and the allocation were created using the statistical RStudio software version 1.1463, and the allocation of the patients to different groups was performed using a statistical tool that generates random samples without replacement."
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Quote: "The study investigators were blinded until the end of the supplementations"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Quote: "The study investigators were blinded until the end of the supplementations"
Incomplete outcome data (attrition bias)
All outcomes Unclear risk All participants were accounted for with reasons for withdrawals provided
Attrition = 28%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial Registration or a priori published protocol: NCT02706808
Control for confounding factors Unclear risk Pre‐treatment period: excluded if had been taking catabolic drugs, antioxidants or vitamin supplements, pre‐, pro‐, synbiotic and antibiotic in the last 3 months before this study
Study period: none reported
Other bias High risk Conflicts declared: "There are no conflicts to declare"
Funding declared: "The resistant starch from Hi‐Maize® 260 used for supplementation was kindly provided by Ingredion, USA.This study was supported in parts by grants from the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)"

Guida 2014.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Italy

  • Setting: single centre (Nutritional Unit, University Hospital)

  • Inclusion criteria: ≥ 18 years; eGFR 15 to 60 mL/min

  • Exclusion criteria: kidney transplant; severe infections; diabetes; malignancy; history of food intolerance; autoimmune disorders; severe malnutrition; clinical conditions requiring artificial feeding


Baseline characteristics
  • Number: intervention group (18); control group (12)

  • Mean age ± SD (years): intervention group (57.0 ± 14.0); control group (63.2 ± 11.1)

  • Sex (M/F): intervention group (14/4); control group (12/0)

  • CKD stage: 2 and 3

  • Kidney measurements

    • Median eGFR, 25th to 75th percentiles (mL/min/1.73 m²): intervention group (25.5, 19.4 to 43.3); control group (32.8, 26.3 to 45.4)

    • Median albumin, 25th to 75th percentiles (mg/dL): intervention group (5.0, 4.4 to 5.0); control group (4.5, 4.0 to 5.0)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: 5 g powder of lyophilised bacteria (5 x 109Lactobacillusplantarum, 2 x 109Lactobacilluscasei subsp. rhamnosus and 2 x 109Lactobacillus gasseri, 1 x 109Bifidobacteriuminfantis and 1 x 109Bifidobactetriumlongum, 1 x 109Lactobacillusacidophilus, 1 x 109Lactobacillussalivarius and 1 x 109Lactobacillussporogenes and 5 x 109Streptococcusthermophilus), prebiotics inulin 2.2 g, and tapioca‐resistant starch 1.3 g per day

  • Time: 4 weeks


Control group
  • Prebiotic: tapioca‐resistant starch, 5 g powder/day (placebo is actually a prebiotic)

  • Time: 4 weeks

Outcomes Outcomes reported by this study at 4 weeks
  • Decrease in total plasma p‐cresol concentration

  • Improvement of any of the following GI symptoms: defecation frequency or ease, stool shape, upper or lower abdominal pain, and frequency of borborygmi or flatus

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients were allocated to the two study groups by simple randomization using a computer‐generated random binary list"
Allocation concealment (selection bias) Low risk Quote: "The medical doctor performing the first visit was responsible for enrolment and randomized group assignment. Neither the patients nor the medical doctors performing further patient evaluation were aware of group assignment."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Neither the patients nor the medical doctors performing further patient evaluation were aware of group assignment"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Neither the patients nor the medical doctors performing further patient evaluation were aware of group assignment"
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for
Attrition = 0%
ITT analysis undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02008331
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: all participants remained on their standard diet and drug regimen during the study
Other bias Unclear risk Conflicts declared: not reported
Funding declared: "Sincere thanks to Dr. A. Caramelli (CadiGroup. Rome. Italy) for providing placebo and Probinul neutroÒ. There was no external funding for the present study."
Potentially industry‐funded intervention

Guida 2017.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind RCT


Time frame
  • Treatment: 4 weeks

  • Follow‐up: none reported

Participants Study characteristics
  • Country: Italy

  • Setting: single centre (Nutrition Unit of the Federico II University of Naples)

  • Inclusion criteria: ≥ 18 years; kidney transplant recipients, vintage > 12 months, stable graft function (SCr < 2.5 mg/dL in the last 3 months); no episode of acute rejection or infection in the last 3 months

  • Exclusion criteria: diarrhoea; diabetes; malignancy; pregnancy; food intolerance; autoimmune disorders; severe malnutrition; clinical conditions requiring artificial feeding


Baseline characteristics
  • Number (randomised/analysed): intervention group (22/20); control group (12/12)

  • Mean age ± SD (years): intervention group (54.0 ± 8.9); control group (47.3 ± 8.5)

  • Sex (M/F): intervention group (16/6); control group (12/0)

  • CKD stage: G3a, kidney transplant

  • Kidney measurements

    • eGFR ± SD (mL/min/1.73 m²): intervention group (50.6 ± 17.6); control group (58.5 ± 24.0)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: lyophilised bacteria (5 x 109, Lactobacillusplantarum 2 x 109, Lactobacilluscasei subsp. rhamnosus and 2 x 109, Lactobacillusgasseri, 1 x 109Bifidobacteriuminfantis and 1 x 109Bifidobacteriumlongum, 1 x 109Lactobacillusacidophilus,1 x 109Lactobacillussalivarius and 1 x 109Lactobacillussporogenes and 5 x 109Streptococcusthermophilus), prebiotic inulin 2.2 g, and 1.3 g of tapioca‐resistant starch 5 g powder packets to be dissolved in water 3 times/day far from meals

  • Time: 4 weeks


Control group
  • Prebiotic: cellulose is a polysaccharide: placebo powder was comparable in colour, texture and taste to the synbiotic and contained only cellulose (the placebo is actually a prebiotic)

  • Time: 4 weeks


Co‐interventions or additional treatments
  • Current medications

    • Corticosteroids: intervention group (83%); control group (91%)

    • Cyclosporin: intervention group (67%); control group (73%)

    • Tacrolimus: intervention group (33%); control group (9%)

    • Mycophenolic acid: intervention group (83%); control group (36%)

    • Everolimus: intervention group (0%); control group (18%)

Outcomes Outcomes reported by this study at 4 weeks
  • Stool characteristics: Bristol Stool Chart

  • Borborygmi

  • Defecation frequency

  • Ease of defecation

  • Upper and inferior abdominal pain

  • Decrease in total plasma p‐cresol concentration

  • Total cholesterol

  • HDL

  • Triglycerides

  • Glucose

  • Albumin

  • Daily intake of: carbohydrates, protein, phenylalanine, tyrosine, fibre, protein/fibre ratio

  • Lipids

  • Body weight

  • BMI

  • Waist circumference

  • eGFR

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Uneven randomization was chosen to encourage patient participation"
Quote: "Using a computer‐generated random binary list, they were allocated to one of 2 arms"
Allocation concealment (selection bias) Unclear risk Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Quote: "This was a single‐centre, parallel‐group, 2:1 synbiotic to placebo randomized and double‐blinded study"
Methods used to ensure this blinding were not provided
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 8%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial Registration or a priori published protocol: NCT02179229
Control for confounding factors Low risk Pre‐treatment period: "At the time of enrolment, patients were already taking the standard diet that we recommend to all kidney transplant recipients who attend our nutrition unit for consultation. This Mediterranean pattern diet is based on foods rich in monounsaturated fatty acids, such as olive oil—the primary source of fat in a Mediterranean diet—or in complex carbohydrates and fibres such as whole grains, fruit, vegetables, and legumes."
Study period: "Both study groups remained on a standard diet and drug regimen during the study." "In accordance with current nutritional guidelines for kidney transplanted patients [35–37], our dietary plan fits an energy intake higher than 25/kcal/kg/ideal body weight/day, with 55% of carbohydrates and total fat not exceeding 30% of calories (fatty acids < 10% of calories and dietary cholesterol limited to 300 mg/day). In addition; protein intake is restricted to 0.8 g/kg of ideal body weight/day. Our diet includes both insoluble and soluble fibres in a ratio of about 3 to 1 because of the high content of insoluble fibres in whole grains and vegetables. Patients are left free to drink water as they need and the average volume of water intake ranges between 1.5 and 2.0 L/day. Diet composition was determined by expert dietitians who interviewed each patient using a detailed food frequency questionnaire that includes 130 foods and beverages [38]. This questionnaire inquires about amount and frequency of foods and beverages consumed everyday by the patient. Data on nutrient composition of the different foods were obtained using the tables of the Italian National Institute of Nutrition, Souci’s Food Composition and Nutrition Tables, and the European Institute of Oncology as reported elsewhere [33,39]. Data were analyzed with an Excel‐based computer program."
Other bias High risk Conflicts declared: not reported
Funding declared: not reported

Haghighat 2019.

Study characteristics
Methods Study design
  • Parallel, 3‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 12 weeks

  • Follow‐up 24 weeks

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (Dialysis centre)

  • Inclusion criteria: 30 to 65 years; undergoing HD for at least 3 months prior, have arteriovenous fistula

  • Exclusion criteria: kidney transplant recipient; kidney transplant candidate likely to receive; inflammatory and infectious diseases; malignancy; chronic liver disease; use of a central catheter for HD access; amputated limbs; pregnancy; using steroidal and/or nonsteroidal anti‐inflammatory drugs and antibiotics, antioxidant, and/or anti‐inflammatory supplements (e.g., vitamin E, vitamin C, and omega‐3 fatty acids), pre‐, pro‐, and synbiotic and other forms of probiotics (including probiotic yogurt, kefir, and other fermented foods) within 3 months of study commencement


Baseline characteristics
  • Number (randomised/analysed): intervention group 1 (25/23); intervention group 2 (25/23); control group (25/19)

  • Mean age ± SD (years): intervention group 1 (48.04 ± 10.11); intervention group 2 (46.21 ± 11.49); control group (45.47 ± 10.76)

  • Sex (M/F): intervention group 1 (12/11); intervention group 2 (12/11); control group (10/9)

  • CKD stage: G5D

  • Kidney measurements

    • SCr ± SD (mg/dL): intervention group 1 (7.11 ± 2.84); intervention group 2 (60.2 ± 2.40); control group (6.33 ± 3.36)

    • Albumin ± SD (g/dL): intervention group 1 (4.03 ± 0.55); intervention group 2 (4.02 ± 0.52); control group (4.19 ± 0.49)

  • Comorbidities

    • Diabetes: intervention group 1 (69.6%); intervention group 2 (65.2%); control group (63.2%)

    • Hypertension: intervention group 1 (78.3%); intervention group 2 (82.6%); control group (63.2%)

  • GI status

    • GSRS ± SD: intervention group 1 (10.95 ± 3.67); intervention group 2 (11.13 ± 2.76); control group (11.26 ± 3.31)

Interventions Intervention group 1
  • Synbiotic 1

    • Prebiotics: 5 g fructo‐oligosaccharides, 5 g galacto‐oligosaccharides, 5 g inulin

    • Probiotics: 5 g of probiotic powder (Lactobacillusacidophilus T16, Bifodobacteriumbifidum BIA‐6, Bifidobacteriumlactis BIA‐6, and Bifidobacteriumlongum LAF‐5, all 2.7 x 107 CFU

  • Time: 12 weeks


Intervention group 2
  • Synbiotic 2

    • Prebiotics: 15 g of maltodextrin powder sachets

    • Probiotics: 5 g of probiotic powder (Lactobacillusacidophilus T16, Bifodobacteriumbifidum BIA‐6, Bifidobacteriumlactis BIA‐6, and Bifidobacteriumlongum LAF‐5, all 2.7 x 107 CFU

  • Time: 12 weeks


Control group
  • Prebiotic: 20 g of maltodextrin powder as the 'placebo' in sachets but was in fact prebiotic

  • Time: 12 weeks


Co‐interventions or additional treatments
  • EPO and IV iron as a regular treatment in accordance with regional HD guidelines

Outcomes All outcomes recorded at 12 weeks
  • Soluble intercellular adhesion molecule type 1s

  • Soluble vascular cell adhesion molecule type

  • Cytokeratin

  • Uric acid

  • Phosphorus

  • BUN

  • SCr

  • Phosphate

  • Calcium

  • Albumin

  • Brain‐derived neurotrophic factor

  • Hospital anxiety and depression score

  • Energy intake (macros and micros)

  • High‐sensitivity CRP

  • IL‐6

  • Anti‐HSP70

  • Endotoxin

  • Beck anxiety and depression indexes

  • HRQoL

  • Treatment adherence (phone call and stool sample tests)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "A research assistant from Tak Gen Zist Pharmaceutical Company, who was not otherwise involved in the study, performed the randomization. Patients were randomly assigned to one of three study groups following a block randomization procedure with stratified patients in each block based on sex and age. Randomization assignment was carried out using computer‐generated random numbers."
Allocation concealment (selection bias) Low risk Quote: "Supplement packaging was done in Tak Gen Zist Pharmaceutical Company"
Quote: "A nutritionist at the haemodialysis centre, who was not a member of the core study team and not aware of random sequences, conducted the randomized allocation sequence, enrolling participants and allocating them to interventions. Randomization and allocation were hidden from the researchers and patients until the final analyses were completed."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Randomization and allocation were hidden from the researchers and patients until the final analyses were completed. Sachets (synbiotic, probiotic and placebo) were pre‐packaged according to the randomization code by Tak Gen Zist Pharmaceutical Company, Tehran, Iran."
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Randomization and allocation were hidden from the researchers and patients until the final analyses were completed. Sachets (synbiotic, probiotic and placebo) were pre‐packaged according to the randomization code by Tak Gen Zist Pharmaceutical Company, Tehran, Iran."
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 13%
ITT not undertaken; analysis based on completers N = 65
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: IRCT2017041233393N1
Control for confounding factors Low risk Pre‐treatment period: 3 months prior: no steroidal and/or nonsteroidal anti‐inflammatory drugs and antibiotics, antioxidant, and/or anti‐inflammatory supplements (e.g., vitamin E, vitamin C, and omega‐3 fatty acids), pre‐, pro‐, and synbiotic and other forms of probiotics (including probiotic yogurt, kefir, and other fermented foods)
Study period: "Throughout the intervention, patients were requested to maintain stable dietary intakes, physical activity, medications, and not to consume any supplements other than the one provided to them by this trial"
Other bias High risk Conflicts declared: "The authors declare that they have no conflicts of interest"
Funding declared: Packaging provided by industry; "Supplement packaging was done in Tak Gen Zist Pharmaceutical Company"; "This study was supported by grants from Vice Chancellor of Research, Ahvaz University of Medical Sciences (Ahvaz, Iran)"

Haghighat 2019 (second comparison).

Study characteristics
Methods  
Participants  
Interventions  
Outcomes  
Notes This study has been split into 2 for the meta‐analysis in this review. We needed to add a third arm under the same comparison: Synbiotic versus Prebiotic

He 2022.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 2017 to 2020

  • Treatment: 12 weeks

  • Follow‐up: none past treatment periods, 36 weeks in total, including washout

Participants Study characteristics
  • Country: China

  • Setting: single centre (Dialysis clinic)

  • Inclusion criteria: ≥ 18 years; CAPD for a minimum 3 months

  • Exclusion criteria: diabetic nephropathy,; pregnant; antibiotic use within previous month; uric acid‐lowering medication; GI disorders (irritable bowel syndrome, Crohn’s disease, ulcerative colitis); severe malnourishment (SGA > 15)


Baseline characteristics
  • Number (randomised/analysed): 33/16

  • Mean age ± SD: 37.67 ± 11.65 years

  • Sex (M/F): 10/6

  • CKD stage: G5D

  • Kidney measurements

    • Median SCr (IQR) (μmol/L): 1109.50 (907.00 to 1221.50)

    • Mean serum albumin ± SD (g/dL): 40.96 ± 3.68

  • Comorbidities: none reported

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: inulin and oligofructose, type and dose not described

  • Time: 12 weeks

  • Number: 33


Washout
  • Time: 12 weeks


Control Group
  • Prebiotic: maltodextrin (placebo is prebiotic) in water, 10 g, per day

  • Time: 12 weeks

  • Number: 33

Outcomes Outcomes reported by this study at 12 weeks
  • Total energy intake

  • Gut microbial compositions

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • No available data for meta‐analyses: first phase of cross‐over not reported as separate data

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The random allocation sequence was generated by an investigator without involvement in subject recruitment. Computer‐generated randomization numbers were obtained from the tool on the website http://tools.medsci.cn/rand/getNewNum to set the treatment order for eligible participants."
Allocation concealment (selection bias) Unclear risk Quote: Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "All participants and investigators (except the research designer) were blinded to intervention allocation and data analysis"
Quote: "The placebo was identical to the prebiotics in appearance, packaging and schedule of administration"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "All participants and investigators (except the research designer) were blinded to intervention allocation and data analysis"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 52%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: ChiCTR‐INR‐17013739
Control for confounding factors Low risk Pre‐treatment period: "The first intervention period was preceded by a 2‐week run‐in phase during which patients were instructed to maintain their habitual diet and physical activity"
Study period: "Dietary intake data were collected via 3‐day food records completed by the participants in real time and reviewed by nutritionists during each intervention period"
Other bias Low risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "This work was supported by National Natural Science Foundation of China [grant numbers: 81673161]"

Khosroshahi 2018.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • February to September 2017

  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (Hospital)

  • Inclusion criteria: ≥ 18 years; maintenance HD 3 times/week for at least 6 months

  • Exclusion criteria: diabetes; GI diseases; active inflammatory disorders; infections; malignancies; changes in dialysis planning or pattern; have received antibiotics prior to the enrolment


Baseline characteristics
  • Number (randomised/analysed): intervention group (25/23); control group (25/21)

  • Mean age ± SD (years): intervention group (53.17 ± 10.15); control group (57.90 ± 13.34)

  • Sex (M/F): intervention group (14/11); control group (15/10)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (mg/dL): intervention group (8.51 ± 2.05); control group (8.83 ± 2.45)

    • Mean serum albumin ± SD (g/dL): intervention group (4.46 ± 0.42); control group (4.54 ± 0.44)

    • Causes of kidney failure: hypertensive nephrosclerosis (28); GN(8); chronic interstitial nephropathy (6); polycystic kidney disease (1)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: dietary fibre: 25 g high amylose maize resistant starch type 2 (HAM‐RS2) cracker

  • Time: 8 weeks


Control group
  • Placebo: 25 g waxy corn starch cracker

  • Time: 8 weeks

Outcomes All outcomes measured at 8 weeks
  • P‐cresol sulfate

  • Indoxyl sulfate

  • high sensitivity CRP

  • GI symptoms

  • SCr

  • KDQoL

  • Serum urea

  • Serum albumin

  • Calcium

  • Phosphorus

  • Cholesterol

  • Triglycerides

  • HDL

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients were ... randomly allocated to two groups: Intervention (HAM‐RS2) and comparison (placebo) using a randomization list generated by Randlist software (version 1.2), resulting 25 patients in each group"
Allocation concealment (selection bias) Unclear risk Quote: "The crackers had the same rectangular shape, weight (60 g) and calories"
Insufficient information regarding allocation concealment, but probably okay
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "As a double‐blind study, the patients and clinical investigators were blinded from knowing the per‐patient treatment regimen"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "As a double‐blind study, the patients and clinical investigators were blinded from knowing the per‐patient treatment regimen"
Probably includes outcome assessors
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 16%
ITT analysis not undertaken, based on completers N = 44
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: IRCT2016062628644N1
Control for confounding factors Unclear risk Pre‐treatment period: none
Study period: not reported
Other bias Unclear risk Conflicts declared: “The authors declare that they have no competing interests”
Funding declared: "No one"; "High fibre diet and placebo were prepared as cracker by Araspar Benis Inc. (Tabriz, Iran)"

Kooshki 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up:none past 8 weeks treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (dialysis clinic, university hospital)

  • Inclusion criteria: ≥ 18 years; receiving HD 3 times/week

  • Exclusion criteria: overt inflammatory and infectious diseases, including hepatitis; receiving Nigella sativa oil, omega‐3 fatty acid, L‐carnitine, vitamin E and/or C supplements, or steroidal and/or nonsteroidal anti‐inflammatory drugs


Baseline characteristics
  • Number (randomised/analysed): intervention group (25/23); control group (25/23)

  • Mean age ± SD (years): intervention group (62.92 ± 16.80); control group (62.83 ± 16.62)

  • Sex (M/F): intervention group (10/13); control group (11/12)

  • CKD stage: G5D

  • Kidney measurements

    • eGFR (mL/min/1.73 m²): not reported

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: lactol probiotic tablet, which contains Lactobacilluscoagulans and fructo‐oligosaccharides, 100 mg/day

  • Time: 8 weeks


Control group
  • Placebo: oral tablet containing farin, once/day

  • Time: 8 weeks


Co‐interventions or additional treatments
  • Subjects in both groups were instructed not to change their dietary habits, physical activities or drug regimens

Outcomes Outcomes reported by this study at 8 weeks
  • High‐sensitive CRP

  • Malondialdehyde

  • Daily food and energy intake (food diary)

  • Compliance

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The subjects were randomly allocated to either the synbiotic or placebo group using blocked randomization"
Allocation concealment (selection bias) Unclear risk Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Insufficient information provided to permit judgement
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Unclear risk All participants were accounted for with reasons for withdrawals provided
Attrition = 8%
ITTt analysis not appear to be undertaken
Quote: "Of the 50 randomized subjects, data from 2 participants from the synbiotic group and 2 from the placebo group were excluded due to lack of patient cooperation in the follow up period"
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Low risk Pre‐treatment period: participants could not be consuming the following prior to study start: Nigella sativa oil, omega‐3 fatty acid, L‐carnitine, vitamin E and/or C supplements, or steroidal and/or nonsteroidal anti‐inflammatory drugs
Study period: subjects in both groups were instructed not to otherwise change their dietary habits, physical activities or drug regimens
Other bias Low risk Conflicts declared: "The authors affirm that they have no conflict of interest to declare"
Funding declared: "None"

Li 2020.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Study duration
  • Treatment: 24 weeks

  • Follow‐up: none past 24 weeks treatment

Participants Study characteristics
  • Country: China

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 18 to 65 years; CAPD for at least 3 months

  • Exclusion criteria: diabetes; antibiotics; radiation to bowel or large bowel resection; irritable bowel syndrome; Crohn's disease; ulcerative colitis; severely malnourished (SGA > 15)


Baseline characteristics
  • Number (randomised/analysed 1st period/analysed 2nd period): 21/18/15

  • Median age, IQR (years): 30.88 (27.81 to 46.63)

  • Sex (M/F): 9/6

  • CKD stage: G5D

  • Kidney measurements

    • Median kidney failure duration (IQR) (months): 23.01 (16.04 to 57.73)

    • Median dialysis duration (IQR) (months): 16.47 (11.93 to 50.60)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: 50:50 mixture of long‐chain inulin and oligofructose, 10 g /ay

  • Time: 12 weeks


Washout
  • Time: 12 weeks


Control group
  • Prebiotic: maltodextrin, 10 g/day (placebo is actually a prebiotic)

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • eGFR

  • Serum albumin

  • SCr

  • Gut microbiome

  • Fecal indole and p‐cresol

  • Indole‐producing bacteria

  • P‐cresol‐producing bacteria

  • Indoxyl sulfate

  • p‐Cresol sulfate

  • Fecal pH

  • 24‐hour urine excretion

  • Dialysis removal of indoxyl sulfate and p‐Cresol sulfate

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • No available data for meta‐analyses: first phase of crossover not reported as separate data

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The intervention order was randomly assigned using a computer‐based, pre generated intervention order"
Allocation concealment (selection bias) Low risk Quote: "This allocation was concealed to patients, investigators, and data analysts other than the research designer (CY)"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The placebo ... was identical to the prebiotic ... in appearance and packaging and schedule of administration. To maintain overall quality and legitimacy, un‐blinding occurred only in exceptional circumstances when knowledge of the actual treatment was essential for further management"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "The placebo ... was identical to the prebiotic ... in appearance and packaging and schedule of administration. To maintain overall quality and legitimacy, un‐blinding occurred only in exceptional circumstances when knowledge of the actual treatment was essential for further management"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Deaths were not related to study drugs
Attrition = 29%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: ChiCTR‐INR‐17013739
Control for confounding factors Unclear risk Pre‐treatment period: "There was a 2‐wk run‐in period during which participants were instructed to maintain their habitual diet and physical activity and not take prebiotic‐ or probiotic‐containing foods, drinks, and supplements"
Study period: not reported
Other bias Low risk Conflicts declared: "The authors report no conflicts of interest"
Funding declared: "This project was supported by the National Natural Science Foundation of China (no. 81673161) and the Fundamental Research Funds for the Central Universities (HUST: 2016YXMS222). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript."

Lim 2021.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, , double‐blind, placebo‐controlled RCT


Study duration
  • 24 weeks treatment

  • Follow‐up: none past 24 weeks treatment

Participants Study characteristics
  • Country: China

  • Setting: single‐centre (Dialysis centre)

  • Inclusion criteria: ≥ 20 years; receiving stable HD 3 times/week for at least 6 months

  • Exclusion criteria: inflammatory diseases; cancer, autoimmune disease; decompensated liver cirrhosis; use of a central catheter for HD access; amputated limbs; pregnancy; had used catabolic drugs; antioxidant vitamin supplements, prebiotics, probiotics, synbiotics, and antibiotics in the last 3 months before starting study


Baseline characteristics
  • Number (randomised/analysed): intervention group (28/25); control group (28/25)

  • Mean age ± SD (years): intervention group (61.50 ± 10.30); control group (56.28 ± 12.36)

  • Sex (M/F): intervention group (10/15); control group (10/15)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (mg/dL): intervention group (11.22 ± 2.21); control group (10.50 ± 1.91)

    • Mean albumin ± SD (g/dL): intervention group (4.12 ± 0.23); control group (4.14 ± 0.27)

  • Comorbidities: hypertension (33); diabetes (25); atherosclerotic vascular disease (18)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Lactococcuslactis subspecies 6 g (Lactis LL358, Lactobaccillussalivarius LS159, and Lactobaccilluspentosus LPE588), 100 billion; 1 x 1011 CFU, 2 sachets/day

  • Time: 24 weeks


Control group
  • Placebo: powder sachet, 2 sachets/day

  • Time: 24 weeks

Outcomes Outcomes reported by this study at 24 weeks
  • Triglycerides

  • HDL cholesterol

  • Indoxyl sulfate

  • P‐cresyl sulfate

  • CRP

  • IL‐6

  • TNF‐alpha

  • Ferritin

  • Lipopolysaccharide binding protein

  • Soluble CD14

  • HRQoL

  • Urea

  • Creatinine

  • Uric Acid

  • Albumin

  • Phosphorus

  • Total CO2

  • Glucose

  • Total cholesterol

  • Anthropometric parameters

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization assignment was performed by the use of computer‐generated block randomization, and the block size was 4"
Allocation concealment (selection bias) Low risk Quote: "Professor Hsueh Fang Wang generated the randomization sequence. Participants, other research staff, and outcome assessors were all blinded to the group assignment process"
Quote: "To maintain the conditions of a double‐blind parallel study, the statistician was not aware of the allocation of participants to each intervention. Code breaking was performed after the analysis was completed"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The sachets with probiotics or placebo were indistinguishable in appearance"
Quote: "To maintain the conditions of a double‐blind parallel study, the statistician was not aware of the allocation of participants to each intervention. Code breaking was performed after the analysis was completed"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Participants, other research staff, and outcome assessors were all blinded to the group assignment process"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 11%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT03066921
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "Participants’ diet, dialysis schedule, and the medications they used did not change during the study. A food frequency questionnaire was used to confirm unchanged diet habits"
Other bias High risk Conflicts declared: "The authors declare that they have no relevant financial interests"
Funding declared: "This study was supported by the Ministry of Science and Technology (grant no. MOST 104‐2622‐E‐241‐001‐CC3). The authors are thankful to the New Bellus Enterprise Co., Ltd. (Tainan, Taiwan) for providing us with a sample of Lactobacillus and for sponsoring our study"
Industry funding

Liu 2020.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 9 December 2016 to 31 May 2017

  • Treatment: 24 weeks

  • Follow‐up: none past 24 weeks of treatment

Participants Study characteristics
  • Country: China

  • Setting: single‐centre (University hospital dialysis centre)

  • Inclusion criteria: 18 to 70 years; CKD stage 5, currently receiving HD treatment > 3 months

  • Exclusion criteria: intolerance to whole milk and dairy products; pregnant; kidney transplant; severe infections; severe cardiac diseases and liver diseases; malignancy; autoimmune disorders; severe malnutrition consumed pre‐or probiotics or had antibiotic therapy within 1 month of study commencement; diagnosed irritable bowel syndrome, Crohn’s disease, or ulcerative colitis; receiving or have received bowel radiation or had large bowel resection


Baseline characteristics
  • Number (randomised/analysed): intervention group (25/22); control group (25/23)

  • Mean age ± SD (years): intervention group (49 ± 9); control group (48 ± 11)

  • Sex (M/F): intervention group (13/12); control group (15/10)

  • CKD stage: G5D

  • Kidney measurements

    • Mean serum albumin ± SD (g/L): intervention group (39.02 ± 3.59); control group (39.25 ± 3.09)

  • Comorbidities: type 2 DM (16); glomerular disease (22); hypertension (3); nephrotic syndrome (1); IgA nephropathy (4); polycystic kidney disease (4)

  • GI status: median GSRS IQR: intervention group (5.0, 4.0 to 7.0); control group (5.0, 2.5 to 7.0)

Interventions Intervention group
  • Probiotic: Bifidobacteriumlongum NQ1501 2.2 × 109 CFU, Lactobacillusacidophilus YIT2004 0.53 × 109 CFU, Enterococcifaecalis YIT0072 1.1 × 109 CFU, powder capsules 210 mg with water, twice/day

  • Time: 24 weeks


Control group
  • Placebo: pre‐gelatinised starch and lactose powder oral capsules, twice/day

  • Time: 24 weeks

Outcomes Outcomes reported by this study at 24 weeks
  • Fecal microbiota profile

  • Serum and faecal metabolic profiles

  • High‐sensitive CRP

  • IL‐6

  • TNF‐alpha

  • Serum endotoxin

  • Serum albumin

  • Endothelial activation markers (soluble intercellular adhesion molecule (ICAM)‐1 and E‐selectin)

  • HRQoL: patient‐reported health SF‐36 and GSRS

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants were randomly assigned in a 1:1 ratio to either the probiotics or placebo group for 6 months. A computer‐generated random binary list was produced"
Allocation concealment (selection bias) Low risk Quote: "A computer‐generated random binary list was produced by an external statistical consultant and maintained by a member of the department staff who did not deal with patients."
Quote: "Neither the patients nor the clinicians performing patient evaluations were aware of group assignments"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Neither the patients nor the clinicians performing patient evaluations were aware of group assignments. Placebo capsules were comparable in appearance, texture, and taste to the probiotics"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Neither the patients nor the clinicians performing patient evaluations were aware of group assignments"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for but reasons for withdrawals were not provided
Attrition = 10%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02929225
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "Both study groups remained on their standard diet and drug regimen during the study"
Other bias High risk Conflicts declared: "None declared"
Funding declared: "The probiotics and placebo capsules were obtained from Sine Pharmaceutical Co., Ltd, Shanghai, China"
Industry funding: "This study was supported by Shannxi Provincial Science and Technology Foundation (Grant no. 2017ZDXM‐SF‐057) and the National Natural Science Foundation of China (Grant no. 81570670)"

Lopes 2018.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, single‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 7 weeks

  • Follow‐up: none reported

Participants Study characteristics
  • Country: Brazil

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: ≥ 18 years; undergoing HD 3 to 4 times/week for at least 3 months

  • Exclusion criteria: auditory deficiency; autoimmune disease; hepatitis B and C; newly implanted catheters; haemodynamic instability; lactose intolerance or milk discomfort


Baseline characteristics
  • Number (randomised/analysed): intervention group (49/29); control group (50/29)

  • Mean age ± SD (years): intervention group (63.17 ± 11.16); control group (63.03 ± 10.77)

  • Sex (M/F): intervention group (17/12); control group (21/8)

  • CKD stage: G5D

  • Kidney measurements

    • Median albumin, IQR (g/dL): intervention group (4.00, 3.71 to 4.09); control group (4.00, 3.60 to 4.10)

    • Mean SCr ± SD (mg/dL): intervention group (8.48 ± 2.84); control group (8.71 ± 3.32)

  • Comorbidities: diabetes (45%); hypertensive nephrosclerosis (22%); GN (5%); polycystic kidney disease (4%)

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: 100 mL pasteurised (unfermented) milk with probiotic Bifidobacteriumlongum BL‐G301 7.4 x 108 CFU/100 mL, and 40 g of extruded sorghum flakes (prebiotic) per day

  • Time: 7 weeks


Control group
  • Prebiotic: 100 mL pasteurised (unfermented) milk and 40 g of flakes of extruded corn per day

  • Time: 7 weeks


Co‐interventions or additional treatments
  • Not reported


Follow‐up details
  • Not reported

Outcomes Outcomes reported by this study at 7 weeks
  • Food consumption (3‐day food recall diary)

  • Albumin

  • Urea

  • Potassium

  • Phosphorus

  • Calcium

  • Hb

  • HCT

  • Iron

  • Alkaline phosphatase

  • Indoxyl sulfate

  • P‐cresol sulfate

  • Indole 3‐acetic acid

  • Faecal hydrogen potential

  • Faecal composition

  • Organic acids

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "...subjects were randomly distributed into two groups..."
Insufficient information provided on methods how random sequence was generated
Allocation concealment (selection bias) Unclear risk Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes High risk Single‐blind study
Blinding of outcome assessment (detection bias)
All outcomes High risk Single‐blind study
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Some concern for reasons for withdrawals
Attrition = 41%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: www.ensaiosclinicos.gov.br RBR‐2d9ny6
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias Unclear risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "The authors thank the Foundation for Research Support of Minas Gerais (FAPEMIG CDS‐APQ‐01683‐15, Brazil), Coordination for the Improvement of Higher Education Personnel (CAPES, Brazil), National Counsel of Technological and Scientific Development (CNPq, Brazil) and the Embrapa Maize and Sorghum (Brazil) for granting of financial support for undergraduate research and scholarships."

Lydia 2022.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • August to December 2020

  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Indonesia

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: ≥ 18 years; HD twice/week for 5 hours, for at least 3 months; patients with GI complaints (difficulty defecating, faeces with hard consistency, or a bowel movement frequency of fewer than 3 times/week)

  • Exclusion criteria: history of malignancy; chemotherapy or radiotherapy; autoimmune disorders or receiving immunosuppressants; underwent gut resection; Crohn’s disease or ulcerative colitis; HD schedule was altered; consuming prebiotics/probiotics/synbiotics; suffering from infection or consuming antibiotics


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/27); control group (30/30)

  • Mean age ± SD (years): intervention group (51.23 ± 13.57); control group (52.33 ± 11.29)

  • Sex (M/F): intervention group (10/20); control group (11/19)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (mg/dL): intervention group ( 11.86 ± 3.46); control group (12.35 ± 3.7)

    • Mean albumin ± SD (g/dL): intervention group (3.90 ± 0.28); control group ( 3.87 ± 0.4)

  • Comorbidities: hypertension (50); diabetes (19); heart failure (10); coronary heart disease (6); stroke (4)

  • GI status

    • Median Patient Assessment of Constipation Symptoms, IQR: intervention group (8, 5 to 11.25); control group (6, 4 to 9.5)

    • Median Patient Assessment of Constipation Quality of Life, IQR: intervention group (17.5, 14 to 24); control group (18, 13 to 26.25)

Interventions Intervention group
  • Synbiotic: Lactobacillusacidophilus and Bifidobacteriumlongum 5x109 CFU and fructo‐oligosaccharides 60 mg, 2 capsules/day

  • Time: 8 weeks


Control group
  • Placebo: Saccharum lactis oral capsules, 2/day

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • Indoxyl sulfate

  • Constipation‐related symptoms

  • QoL

  • Compliance measured at 4 weeks

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "This study was a double‐blinded randomized controlled clinical trial"
Insufficient information provided on randomization methods
Allocation concealment (selection bias) Low risk Quote: "A third party (pharmacist) randomized the subjects into two study groups using a computer randomizer"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Synbiotics and placebo were both packed on an identical clear gastroenteric coated capsule. Each subject was given one plastic pot containing either intervention or placebo drugs; each identical pot contained 60 capsules. A white paper with information on drug use and administration was placed on the cover of the pot."
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "The drugs were distributed by a third party. None of the patients, researchers, or physicians in charge were aware of the treatment groups"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 5%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT04527640
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "Food intake was assessed before and after the intervention was administered using 24‐hour food recall and was carried out by a nutritionist"
Other bias Low risk Conflicts declared: "The authors have no conflicts of interest to declare"
Funding declared: "The authors received no funding support for this research"

Mafi 2018.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 45 to 85 years; diabetic nephropathy (defined as diabetic kidney disease with proteinuria, with or without the elevation of SCr) with proteinuria level > 0.3 g/24 hours

  • Exclusion criteria: history of active infection within 3 months; intake of probiotic; and/or synbiotic supplements within 3 months; history of hospital admission within 3 months; malignancy and/or liver cirrhosis


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/30); control group (30/30)

  • Mean age ± SD (years): intervention group (58.9 ± 8.8); control group (60.9 ± 4.4)

  • Sex (M/F): not reported

  • CKD stage: undefined

  • Kidney measurements: not reported, inclusion criteria only proteinuria level > 0.3 g/24 hours

  • Comorbidities: type 1 DM (4); type 2 DM (56)

  • GI status: not reported

Interventions Intervention group
  • Probiotics:Lactobacillusacidophilus ZT‐L1 8 × 109 CFU day−1, Bifidobacteriumbifidum ZT‐B1 8 × 109 CFU day−1, Lactobacillusreuteri ZT‐Lre 8 × 109 CFU day−1, and Lactobacillusfermentum ZT‐L3 2 × 109 CFU, per day

  • Time: 12 weeks


Control group
  • Placebo: starch tablet/day

  • Time: 12 weeks

Outcomes Outcomes measured by this study at 12 weeks
  • Insulin resistance (measured by homeostasis model of assessment‐insulin resistance (HOMA‐IR))

  • Food and energy intake (3‐day food recall diary)

  • Metabolic profiles

  • Biomarkers of inflammation and oxidative stress

  • SCr

  • BUN

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization was carried out using computer‐generated random numbers by a trained staff member at the internal clinic"
Allocation concealment (selection bias) Unclear risk Unclear information, insufficient to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Study states to be double‐blind but provides no further methods on how this was ensured
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Unclear information, insufficient to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided (not related to study drugs)
Attrition = 10%
ITT analysis undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: IRCT2017061134458N1
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias High risk Conflicts declared: "There are no conflicts to declare"
Funding declared: "The present study was supported by a grant from the Vice‐Chancellor for Research, KaUMS, Iran (96043). We are grateful to thank LactoCare®, Zisttakhmir Company, in Tehran that provided probiotic capsules for the present study."

Mazruei Arani 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • April to December 2017

  • Treatment: 12 weeks

  • Follow‐up none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 45 to 85 years; diabetic nephropathy with a proteinuria level > 0.3 g/24 hours. Diabetic nephropathy defined as "diabetic renal disease with proteinuria, with or without elevation of serum creatinine levels"

  • Exclusion criteria: history of active infection within 3 months; the intake of probiotic and/or synbiotic supplements within 3 months; and malignancy and/or liver cirrhosis


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/30); control group (30/30)

  • Mean age ± SD (years): intervention group (62.7 ± 9.1); control group (60.3 ± 8.5)

  • Sex (M/F): not reported

  • CKD stage: undefined

  • Kidney measurements: not reported

  • Comorbidities: DM (60)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: honey containing a viable and heat‐resistant probiotic Bacillus coagulans T4 (IBRC‐N10791) 108 CFU/g, 25 g/day

  • Time: 12 weeks


Control group
  • Placebo: control honey, 25 g/day

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Parameters of insulin metabolism

  • Fasting plasma glucose

  • Lipid profiles

  • Biomarkers of inflammation

  • Oxidative stress

  • SCr

  • BUN

  • Micro and macro nutrient intake (3‐day food recall diary)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "Patients with DN were randomly divided into two groups"
Insufficient information provided as to how randomization methods were undertaken
Allocation concealment (selection bias) Low risk Quote: "Randomization was done using a random number table by one of the investigators who had no clinical involvement in the study"
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Study states to be double‐blind but insufficient information provided regarding methods to permit judgement
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information provided regarding methods to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for; however, reasons for withdrawals are not provided
Attrition = 5%
ITT analysis undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: IRCT201705035623N115
Control for confounding factors Unclear risk Pre‐treatment period: no intake of probiotic and/or synbiotic supplements within 3 months prior to study start
Study period: not reported
Other bias Low risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "The present study was supported by a grant from the Vice‐chancellor for Research, KAUMS, and Iran. Special thanks to Gaz Sekkeh Company for providing probiotic and control honey."

Meng 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, open‐label RCT


Time frame
  • 2016 to 2017

  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: China

  • Setting: single centre (Hospital)

  • Inclusion criteria: ≥ 18 years; early type 2 diabetic nephropathy defined as UACR 2.5 to 20 mg/mmol (men) or 3.5 to 20 mg/mmol (women)

  • Exclusion criteria: did not have type 2 diabetes, macroalbuminuria or overt proteinuria; recent infectious diseases; surgery; malignancy; liver cirrhosis


Baseline characteristics
  • Number (randomised/analysed): intervention group (37/34); control group (38/36)

  • Mean age ± SD (years): intervention group (62.85 ± 9.3); control group (61 ± 9.5)

  • Sex (M/F): intervention group (18/16); control group (21/15)

  • CKD stage: A2

  • Kidney measurements

    • Mean SCr ± SD (μmol/L): intervention group (62.4 ± 16.7); control group (63.1 ± 16.1)

    • Mean albumin ± SD (g/L): intervention group (45.9 ± 3.0); control group (46.3 ± 3.3)

  • Comorbidities: diabetes (75); hypertension (48); cerebral apoplexy (5); coronary heart disease (26)

  • GI status: not reported

Interventions Intervention group
  • Prebiotics: high‐resistant starch, low‐protein flour, 50 g instead of a common staple of equal quality at lunch and dinner each day. The lack of protein is supplemented with high‐quality protein eggs, poultry meat, and dairy products. According to the previous measurement result, the total daily intake was 17.41 g/day

  • Time: 12 weeks


Control group
  • No treatment: patients followed protein restriction diet daily with a common staple per day

  • Time: 12 weeks


Co‐interventions or additional treatments
  • "During the 12‐week intervention, all patients with DN received low‐salt, low‐fat, and low‐protein dietary pattern and were given basic treatment such as glucose‐lowering, lipid‐lowering, and blood pressure–lowering treatment. But, patients were requested not to change their medicine and insulin treatment programs arbitrarily. All participants received dietary guidance from dietitians and completed 3‐day food records."

Outcomes Outcomes reported by this study at 12 weeks
  • Fasting blood glucose

  • HbA1c

  • Cholesterol

  • Triglycerides

  • Albumin

  • Prealbumin

  • Serum uric acid

  • Beta2‐microglobulin

  • BUN

  • Creatinine

  • UACR

  • Superoxide dismutase

  • Malondialdehyde

  • IL‐6

  • TNF‐alpha

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The patients were randomly divided into intervention group ... or control group ... by using computer‐generated 1:1 randomization sequences"
Allocation concealment (selection bias) High risk Quote: "Delivery of the intervention and assessment of outcomes were not blinded to the treatment assignment"
Blinding of participants and personnel (performance bias)
All outcomes High risk Quote: "Delivery of the intervention and assessment of outcomes were not blinded to the treatment assignment"
Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Quote: "Delivery of the intervention and assessment of outcomes were not blinded to the treatment assignment"
Open‐label study
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 7%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "During the 12‐week intervention, all patients with DN received low‐salt, low‐fat, and low‐protein dietary pattern and were given basic treatment such as glucose‐lowering, lipid‐lowering, and blood pressure–lowering treatment. But, patients were requested not to change their medicine and insulin treatment programs arbitrarily. All participants received dietary guidance from dietitians and completed 3‐day food records."
Other bias Low risk Conflicts declared: "The authors declare that they have no relevant financial interests"
Funding declared: "This work was supported by the Medical Health and Science Development Project of Shandong Province (grant number: 2016ws0428)"

Miraghajani 2017.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind RCT


Time frame
  • November 2013 to February 2014

  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: ≥ 18 years; CKD stages 1 and 2 defined as fasting blood glucose > 126 mg/dL, hypoglycaemic agents or insulin intake, proteinuria > 300 mg/day, GFR > 90 mL/min

  • Exclusion criteria: changing the dosage of medications; allergy or intolerance to soy milk or avoidance of soy milk consumption; smoking; alcoholism; recent antibiotic therapy; use of supplements containing vitamins and minerals; any medical condition such as inflammatory bowel disease, infection, liver disease, and rheumatoid arthritis


Baseline characteristics
  • Number (randomised/analysed): intervention group (24/20); control group (24/20)

  • Mean age ± SD (years): intervention group (56.90 ± 1.81); control group (53.60 ± 1.60)

  • Sex (M/F): 22/26

  • CKD stage: CKD stage 1 and 2 with diabetic nephropathy

  • Kidney measurements: not reported

  • Comorbidities: diabetes (21); hypertension (34)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Lactobacillus plantarum A7, 2 x 107 CFU/mL in soy milk, 200 mL/day

  • Time: 8 weeks


Control Group
  • Control: standard soy milk, 200 mL/day

  • Time: 8 weeks


Co‐interventions or additional treatments
  • "All participants received individualized dietary counselling aimed at achieving a daily energy and restricting dietary protein, sodium, and potassium intake"

Outcomes Outcomes reported by this study at 8 weeks
  • Malondialdehyde

  • 8‐iso‐PGF2a

  • Glutathione

  • Oxidized glutathione

  • Total antioxidant capacity

  • Glutathione peroxidase

  • Glutathione reductase

  • Neutrophil gelatinase‐associated lipocalin

  • Cytokine receptor‐soluble tumor necrosis factor receptor 1

  • cystatin C

  • inflammatory adipokine—Progranulin

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients ... were randomized (into) intervention arms using a simple random allocation sequence (www.randomization.com)"
Allocation concealment (selection bias) Low risk Quote: "Concealed envelopes with consecutive numbers were locked up in a drawer and withdrawn in numerical order by the main author"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Patients and investigators, including those assessing outcomes and performing analyses, were masked to group assignment"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Patients and investigators, including those assessing outcomes and performing analyses, were masked to group assignment"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 17%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02704884
Control for confounding factors Low risk Pre‐treatment period: 2‐week, single‐masked, run‐in period
Study period: "All participants received individualized dietary counselling aimed at achieving a daily energy and restricting dietary protein, sodium, and potassium intake"
Other bias Low risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "The authors appreciate the financial support provided by the Research Council of the Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran"

Miranda Alatriste 2014.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, open‐label RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Mexico

  • Setting: single‐centre (Research clinic)

  • Inclusion criteria: 18 to 65 years; CKD stage 3 or 4, GFR 59 to 15 mL/min/1.73 m²

  • Exclusion criteria: KRT; DM; lupus erythematosus; kidney transplant; intolerance to whole milk and dairy products


Baseline characteristics
  • Number (randomised/analysed): intervention group 1 (16/15); intervention group 2 (15/15)

  • Mean age ± SD (years): intervention group 1 (43.8 ± 14.44); intervention group 2 (39.13 ± 16.36)

  • Sex (M/F): intervention group 1 (8/7); intervention group 2 (8/7)

  • CKD stage: G3 to G4

  • Kidney measurements

    • Mean GFR ± SD (mL/min/1.73 m²): intervention group 1 (30.66 ± 12.18); intervention group 2 (30.74 ± 11.71)

    • Mean SCr ± SD (mg/dL): intervention group 1 (2.44 ± 0.79); intervention group 2 (2.52 ± 1.01)

  • Comorbidities: hypertension (27)

  • GI status: not reported

Interventions Intervention group 1
  • Probiotics: fermented dairy drink containing Lactobacillus casei Shirota 8 x 109 CFU in 80 mL/day

  • Time: 8 weeks


Intervention group 2
  • Probiotics: fermented dairy drink containing Lactobacillus casei Shirota 16 x 109 CFU,in 160 mL/day

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • Urea

  • SCr

  • GFR

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "The present study is a ... simple randomized clinical trial"
Insufficient details provided
Allocation concealment (selection bias) Low risk Quote: "The size, color, flavor and physical aspects of the bottles were the same for both groups"
Blinding of participants and personnel (performance bias)
All outcomes High risk Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Open‐label study
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 3%
ITT analysis not undertaken
Selective reporting (reporting bias) High risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias High risk Conflicts declared: mot reported
Funding declared: not reported

Mirzaeian 2020.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • October 2015 to December 2016

  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Dialysis centre)

  • Inclusion criteria: any age; HD 3 times/week

  • Exclusion criteria: pregnant; lactating; history of active cancers; history of severe chronic conditions (pulmonary, cardiovascular, hepatic); alcohol or drug addiction; lack of GI disorders; HIV; psychiatric problems; severe oedema; infection 4 weeks prior; use of synbiotics 4 weeks prior; immunosuppressants; anticoagulants


Baseline characteristics
  • Number (randomised/analysed): intervention group 1 (24/21); intervention group 2 (24/21)

  • Mean age ± SD (years): intervention group 1 (58.30 ± 11.3); intervention group 2 (69.74 ± 42.87)

  • Sex (M/F): intervention group 1 (14/7); intervention group 2 (16/5)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (mg/dL): intervention group 1 (8.5 ± 2.3); intervention group 2 (8.4 ± 2.4)

    • Mean serum albumin ± SD (g/dL): intervention group 1 (4.1 ± 0.5); intervention group 2 (3.9 ± 0.9)

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group 1
  • Synbiotic: Lactobacillus casei 3.5 x 109 CFU, Lactobacillus acidophilus 1.5 x 109 CFU, Lactobacillus rhamnosus 3 x 109 CFU, Lactobacillus bulgaricus 3.5 x 108 CFU, Bifidobacterium breve 5 x 109 CFU, Bifidobacterium longum 1 x 1010 CFU, and Streptococcus thermophiles (3.5 x 108 CFU), fructo‐oligosaccharide lactose (dose not provided), magnesium stearate, and talc as filling materials, total 500 mg of synbiotic, 2 capsules/day

  • Time: 8 weeks


Intervention group 2
  • Prebiotic: placebo oral capsules of maltodextrin 500 mg/day

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • Indoxyl sulfate and phenol

  • Liver enzymes

  • BUN

  • Creatinine

  • High‐sensitivity CRP

  • PTH

  • Albumin

  • BP

  • Alanine aminotransferase

  • Alkaline phosphatase

  • Aspartate aminotransferase

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients were randomly allocated to block 1 or 2 with the use of stratified block randomization according to sex, age (± 5 years), and duration of HD (± 6 months)."
Allocation concealment (selection bias) Low risk Quote: "The synbiotic and placebo capsules were identical in size, color, odor, and packaging."
Quote: "Patients and investigators were blinded as to the arms of the study."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "To conduct the study in a double‐blinded nature, the placebo, containing maltodextrin, and synbiotic capsules were encoded as A and B, respectively, by a third person, and patients and investigators were blinded as to the arms of the study."
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "To conduct the study in a double‐blinded nature, the placebo, containing maltodextrin, and synbiotic capsules were encoded as A and B, respectively, by a third person, and patients and investigators were blinded as to the arms of the study."
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 13%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: "Before the start of the study, researchers introduced enriched dairy products with probiotics to all patients. All patients were asked not to consume foods such as yogurt, cheese, and kefir, which probably contain probiotic strains"
Study period: not reported
Other bias High risk Conflicts declared: not reported
Funding declared: "The authors acknowledge the Zist Takhmir Company, Iran, for gifting the synbiotic supplements."

Natarajan 2014.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • April 2011

  • 24 weeks treatment

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: USA

  • Setting: single‐centre (Dialysis centre)

  • Inclusion criteria: 18 to 80 years; CKD 5D receiving HD

  • Exclusion criteria: pregnant; HIV/AIDs; liver disease; dependency to substances and alcohol,;anticoagulants; social conditions or medical debilitating disease/disorder


Baseline characteristics
  • Number (randomised/analysed): 28/22

  • Mean age (range) (years): 54 (29 to 79)

  • Sex (M/F): 6/16

  • CKD stage: G5D

  • Kidney measurements: not reported

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Streptococcus thermophilus KB19, Lactobacillus acidophilus KB 27, and Bifidobacteriumlongum KB 31 3 x 1010 CFU; 2 capsules, 3 times/day

  • Time: 24 weeks


Washout
  • 8 weeks


Control group
  • Prebiotic: placebo capsules 1:1 blend of cream of wheat and psyllium husk

  • Time: 24 weeks


Co‐interventions or additional treatments
  • Not reported

Outcomes Outcomes reported by this study at 8 weeks
  • QoL

  • Urea

  • Creatinine

  • Haematological values

  • Hepatic function

  • p‐cresol sulfate

  • Serum pentosidine

  • Beta‐2 microglobulin

  • Necrosis factor‐kappa‐B

  • sCKD30

  • Uric acid

  • Free indole acetic acid

  • Free hippuric acid

  • 3‐carboxyl‐4‐methyl‐5‐propyl‐2‐furan‐propanoic acid

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • Cross‐over data not reported separately for first phase of study, no meta‐analysis

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "Patient was examined, randomly assigned to either treatment or control group"
Allocation concealment (selection bias) Unclear risk No information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Study states to be double‐blind but no further information provided on how this was undertaken
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 21%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT01450709
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias High risk Conflicts declared: "The authors declare that they have no conflict of interests regarding the publication of this paper"
Funding declared: "Kibow Biotech, Inc., a privately owned biotechnology company focused on probiotics, financed this clinical investigation at the Downstate Medical Center through 2009 Qualifying Therapeutic Discovery Project (QTDP) award, a US government special grant program to support promising and emerging technologies. Part of the data was also obtained in Kibow’s own fully equipped research laboratories"

Pan 2021.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, open‐label, placebo‐controlled RCT


Time frame
  • March 2017 to February 2018

  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: China

  • Setting: single centre (Dialysis clinic)

  • Inclusion criteria: 18 to 75 years; PD for at least 3 months

  • Exclusion criteria: advanced malignant disease; a history of drug or alcohol abuse; intolerance to probiotic supplements; more than 2 episodes of peritonitis within the last year; active infectious disease, or uncontrolled autoimmune disease such as systemic lupus erythematosus


Baseline characteristics
  • Number (randomised/analysed): intervention group (58/50); control group (58/48)

  • Mean age ± SD (years): intervention group (50.92 ± 17.60); control group (49.31 ± 13.13)

  • Sex (M/F): intervention group (28/22); control group (28/20)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (µmol/L): intervention group (902.82 ± 248.99); control group (955.90 ± 248.16)

  • Comorbidities: glomerular disease (37); diabetic kidney disease (32); hypertension (16); polycystic kidney disease (11); obstructive nephropathy (2); CVD (36)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Bifidobacteriumlongum, Lactobacillusbulgaricus, and Streptococcusthermophilus 1 x 109 CFU; 2 capsules, 3 times/day

  • Time: 8 weeks


Control group
  • Prebiotic: oral capsules maltodextrin (placebo is actually a prebiotic), dose not specified

  • Time: 8 weeks

Outcomes Outcomes reported by this study at 8 weeks
  • BMI

  • Upper arm circumference

  • HDL cholesterol

  • High‐sensitivity CRP

  • IL‐6

  • Left calf circumference

  • LDL cholesterol

  • Triceps skinfold thickness

  • HRQoL

  • Fat mass

  • Albumin

  • Hb

  • Triglycerides

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Eligible patients undergoing PD were randomized in a 1:1 ratio to one of two parallel groups (Randomization assignment was conducted using computer‐generated random numbers)"
Allocation concealment (selection bias) Unclear risk Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes High risk Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Open‐label study
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 16%
ITT analysis not undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "Medications known to affect the gut microbiome (such as proton pump inhibitor phosphate binders) were not used during the study"
Other bias High risk Conflicts declared: "The authors declare that they have no relevant financial interests"
Funding declared: "This study was not financially supported by any funding source"; "The probiotic supplements were produced by the Shanghai Xinyi pharmaceutical factory (S10950032, Shanghai, China)"
Industry funding of intervention

Poesen 2016.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Study duration
  • May 2014 to March 2015

  • Treatment: 4 weeks

  • Follow‐up: none past study treatment

Participants Study characteristics
  • Country: Belgium

  • Setting: single‐centre (Hospital outpatient clinic)

  • Inclusion criteria: ≥ 18 years; eGFR 15 to 45 mL/min/1.73 m²

  • Exclusion criteria: GI disease (i.e. inflammatory bowel disease, malignancy); previous colorectal surgery; insulin‐dependent DM; use of antibiotics, prebiotics or probiotics in the past 4 weeks


Baseline characteristics
  • Number (randomised/analysed): 40/40

  • Mean age ± SD (years): 70 ± 6

  • Sex (M/F): 28/12

  • CKD stage: G3b

  • Kidney measurements

    • Median SCr (IQR) (mg/dL): 1.98 (1.60 to 2.18)

    • Median eGFR (IQR) (mL/min/1.73 m²): 33 (27 to 38)

    • Median 24 hour proteinuria (IQR) (g): 0.161 (0.078 to 0.498)

  • Comorbidities: vascular disease (18); glomerular disease (8); diabetes (7)

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: rabinoxylan oligosaccharides obtained from commercial wheat bran, 20 g/day, dissolved in 200 mL water

  • Time: 4 weeks


Washout
  • 4 weeks


Control Group
  • Prebiotic: maltodextrin, an oligosaccharide obtained by enzymatic hydrolysis of potato starch with total digestion in the small intestine (placebo is actually prebiotic), approximately 13.4 g/day, dissolved in 200 mL water

  • Time: 4 weeks


Co‐interventions or additional treatments
  • None reported

Outcomes Outcomes reported by study at 4 weeks
  • GI tolerance (5‐level Likert scale)

  • Stool frequency and consistency (Bristol Stool Chart)

  • Creatinine

  • CRP

  • Alanine aminotransferase

  • Potassium

  • Urea

  • Fasting blood glucose

  • Insulin

  • P‐cresyl sulfate

  • P‐cresyl glucuronide

  • Indoxyl sulfate

  • Trimethylamine N‐oxide

  • Phenylacetylglutamine

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization was performed by the sealed envelope system, in which the study nurse randomly opened a preformed envelope containing the allocated treatment regimen."
Allocation concealment (selection bias) Low risk Quote: "Patients were randomized by the study nurse, blinded from both the investigator and study participant. Randomization was performed by the sealed envelope system, in which the study nurse randomly opened a preformed envelope containing the allocated treatment regimen. Patients were allocated to sequential treatment with AXOS and placebo, or vice versa, with a wash‐out period between both intervention periods.
Quote: "Treatment with AXOS or placebo was offered to patients in identical vials and boxes. Each box was also labelled with a numerical code, unique to treatment allocation and again blinded from both the investigator and study participant, as an additional measure to allow review of the correct treatment allocation by the study nurse."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Treatment with AXOS or placebo was offered to patients in identical vials and boxes. Each box was also labelled with a numerical code, unique to treatment allocation and again blinded from both the investigator and study participant, as an additional measure to allow review of the correct treatment allocation by the study nurse."
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Quote: "... treatment allocation and again blinded from both the investigator and study participant, as an additional measure to allow review of the correct treatment allocation by the study nurse."
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 3%
ITT analysis undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02141815
Control for confounding factors Unclear risk Pre‐treatment period: use of antibiotics, prebiotics or probiotics in the past 4 weeks was not allowed
Study period: not reported
Other bias High risk Conflicts declared: "The authors have declared that no competing interests exist."
Funding declared: "RP is the recipient of a Ph.D. fellowship of the Research Foundation ‐ Flanders (FWO) (grant 11E9813N) (www.fwo.be). Part of the research has been funded by the Research Foundation ‐ Flanders (FWO) (grant G077514N) (www.fwo.be). JAD and KV are jointly chair holders of the W.K. Kellogg Chair in Cereal Science and Nutrition at KU Leuven (www.wkkf.org). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript."

PREBIOTIC 2022.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 22 October 2020 to 11 May 2021

  • Treatment: 7 weeks

  • Follow‐up: none past 7 weeks treatment

Participants Study characteristics
  • Country: Australia

  • Setting: single‐centre (Kidney transplant centre)

  • Inclusion criteria: ≥ 18 years; kidney transplant recipient

  • Exclusion criteria: radiation to the bowel; large bowel resection; medically diagnosed and active inflammatory bowel disease


Baseline characteristics
  • Number (randomised/analysed): intervention group (27/27); control group (29/29)

  • Mean age ± SD (years): intervention group (52.9 ± 12.1); control group (53.4 ± 11.7)

  • Sex (M/F): intervention group (17/10); control group (18/11)

  • CKD stage: kidney transplant

  • Kidney measurements: not reported

  • Comorbidities: glomerular disease (21); genetic kidney disease (6); diabetic kidney disease (13); reflux nephropathy (6); renovascular disease (4)

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: green banana–resistant starch, 7.5 g/day titrated to 15 g/day after 2 weeks

  • Time: 7 weeks


Control group
  • Placebo: waxy corn starch, 7.5 g/day titrated to 15 g/day after 2 weeks

  • Time: 7 weeks

Outcomes Outcomes reported by this study at 7 weeks
  • Tolerability and adherence

  • Adverse effects

  • GSRS

  • Faecal microbiota composition

  • Serum indoxyl sulfate

  • Serum p‐cresyl sulfate

  • QoL: EQ‐5D

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants were randomly assigned in a 1:1 ratio to receive either prebiotic or placebo"
Quote: "The randomization schedule was prepared by a researcher not involved with treatment allocation and involved stratification factors of age (,65 years, $65 years) and sex"
Allocation concealment (selection bias) Low risk Quote: "The randomization schedule was prepared by a researcher not involved with treatment allocation and involved stratification factors of age (<65 years, >65 years) and sex"
Quote: "A blinded allocation list was maintained in an Excel spreadsheet on a secure server in a folder not accessible to trial staff involved in recruitment"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "... a matched, identical placebo ... "
Quote: "A blinded allocation list was maintained in an Excel spreadsheet on a secure server in a folder not accessible to trial staff involved in recruitment. Participants, caregivers, treating physicians and surgeons, laboratory staff, and members of the study team were blinded to the treatment."
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "A blinded allocation list was maintained in an Excel spreadsheet on a secure server in a folder not accessible to trial staff involved in recruitment. Participants, caregivers, treating physicians and surgeons, laboratory staff, and members of the study team were blinded to the treatment."
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 71%
ITT analysis undertaken for originally randomized 56
High attrition rate but does not appear to be due to adverse effects of study treatment
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: ACTRN12618001057279
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: not reported
Other bias Low risk Conflicts declared: "Samuel Chan is supported by the Australian National Health and Medical Research Council (NHMRC) Postgraduate Scholarship, the Microba recipient grant, the Metro South Research Support Scheme, and the Royal Australasian College of Physicians NHMRC Research Excellence top‐up award. Furthermore, Dr Chan is a current recipient of the 2018 Sir Gustav Nossal NHMRC Postgraduate Scholarship award. Carmel Hawley is the recipient of research grants paid to her institution from Baxter Healthcare and Fresenius Medical Care and from Otsuka, Janssen and GlaxoSmithKLline for trial steering committee activities, paid to her institution. David Johnson has received consultancy fees, research grants, speaker’s honoraria and travel sponsorships from Baxter Healthcare and Fresenius Medical Care. He has received consultancy fees from Astra Zeneca and travel sponsorships from Amgen. He is a current recipient of an Australian NHMRC Practitioner Fellowship. Nicole Isbel has received consultancy fees and speaker’s honoraria from Alexion Pharmaceuticals, Novo Nordisk and Amgen. The remaining authors have no conflicts of interest to declare with respect to the context and scope of this manuscript."
Funding declared: "This study was funded by the Metro South Research Support Scheme Project Grant"

ProbiotiCKD 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, open‐label, placebo‐controlled RCT


Time frame
  • January 2016 to March 2017

  • Treatment: 15 weeks

  • Follow‐up: none past 15 weeks treatment

Participants Study characteristics
  • Country: Italy

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: ≥ 18 years; CKD stage 3A; Caucasian; eGFR 60 to 45 mL/min/1.73 m²

  • Exclusion criteria: Montreal classification criteria for inflammatory bowel diseases positive; malabsorption; autoimmune systemic diseases; cancer; kidney transplant recipients; pregnant; current or recent antibiotic therapy or immunosuppressant drugs


Baseline characteristics
  • Number (randomised/analysed): intervention group (14/14); control group (14/14)

  • Mean age ± SD (years): intervention group (61.3 ± 5.2); control group (58.2 ± 6.2)

  • Sex (M/F): intervention group (9/5); control group (6/8)

  • CKD stage: 3A

  • Kidney measurements

    • Mean SCr ± SD (mg/dL): intervention group (1.78 ± 0.4); control group (1.8 ± 0.3)

    • Mean eGFR ± SD (mL/min/1.73m²): intervention group (48.4 ± 7.4); control group (49.3 ± 5.8)

    • Mean albumin ± SD (g/dL): intervention group (4.16 ± 0.2); control group (4.05 ± 0.1)

  • Comorbidities: hypertension (28); diabetes (6)

  • GI status: none, no altered gut microbiota

Interventions Intervention group
  • Week 1

    • 0.377 g of Enterococcusfaecium UBEF‐41, Lactobacillusacidophilus (LA‐14) and Saccharomycescerevisiae subspecies Boulardii (MTCC‐5375); 1 capsule, 3 times/day

  • Weeks 2 to 3

    • 0.455 g of Bifidobacteriumbrevis (BB03), Bifidobacteriumbifidum (BB06), Bifidobacteriumlongum (BL05); 1 capsule, 3 times/day

    • 0.455 g of Lactobacillusrhamnosus (HN‐001), Lactobacillusrhamnosus (LR‐32) and Lactobacillusacidophilus (LA14); 1 capsule; 3 times/day

  • Weeks 4 to 15

    • 0.455 g of Bifidobacteriumbrevis (BB03), Bifidobacteriumbifidum (BB06), Bifidobacteriumlongum (BL05); 1 capsule, twice/day

    • 0.455 g of Lactobacillusrhamnosus (HN‐001), Lactobacillusrhamnosus (LR‐32) and Lactobacillusacidophilus (LA14); 1 capsule, twice/day

  • Time: 15 weeks


Control group
  • Placebo: oral capsules of placebo powder matching the regime above

  • Time: 15 weeks

Outcomes Outcomes reported by this study at 15 weeks
  • Urinary indican and 3‐MI concentration

  • Faecal microbiota composition: Lactobacillales and Bifidobacteria concentrations

  • CRP

  • Ferritin

  • eGFR

  • Lipid profile

  • Calcium

  • Beta‐2‐microglobulin

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients were randomized by a computer‐generated random number list to receive either placebo or probiotics"
Allocation concealment (selection bias) Low risk Quote: "Placebo capsules were prepared ad hoc by a galenic local pharmacy to look exactly as probiotic capsules. Even placebo plastic containers looked exactly as those of probiotics."
Blinding of participants and personnel (performance bias)
All outcomes High risk Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Open‐label study
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for, no withdrawals
Attrition = 0%
ITT analysis undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Low risk Pre‐treatment period: "To standardize baseline conditions, any prebiotic or probiotic supplement had to be suspended at least 1 month before enrolment"; "Enrolled subjects had to adhere to a protein dietary intake ranging 0.7–1 g/ kg/day, also assuring a daily consumption of two pieces of fruit (apple or pear) and 200 g of double‐boiled leafy green vegetables (the double boiling was used to discharge vegetables’ potassium content)"
Study period: dietary recall
Other bias High risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "Probiotics were kindly provided by Bromatech Ltd, Viale Premuda, 46–20129 Milano"
Industry funding

Ramos 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • May 2015 to October 2016

  • Treatment: 12 weeks

  • Follow‐up: none reported

Participants Study characteristics
  • Country: Brazil

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 18 to 80 years; non‐dialysis dependent CKD; eGFR < 45 mL/min/1.73 m²

  • Exclusion criteria: DM; chronic liver disease; auto‐immune disease (i.e. systemic lupus erythematosus; rheumatoid arthritis); congestive heart failure (Stages III/ IV); HIV; current malignancy; bowel diseases (i.e. inflammatory bowel diseases; celiac disease); cognitive limitations; current smokers and using medications including phosphate binders; immunosuppressants; anti‐inflammatories; antibiotics; laxatives; prebiotics; probiotics or synbiotics 3 months preceding baseline assessment


Baseline characteristics
  • Number (randomised/analysed): intervention group 1 (24/23); intervention group 2 (26/23)

  • Mean age ± SD (years): intervention group 1 (62.2 ± 11.3); intervention group 2 (52.8 ± 16.1)

  • Sex (M/F): intervention group 1 (13/11); intervention group 2 (14/12)

  • CKD stage: 3b (7); 4 (31); 5 (12)

  • Kidney measurements

    • Median SCr, IQR (mg/dL): intervention group 1 (2.87, 2.31 to 3.55); intervention group 2 (2.85, 2.35 to 3.64)

    • Mean albumin ± SD (g/dL): intervention group 1 (4.4 ± 0.4); intervention group 2 (4.3 ± 0.3)

    • Mean eGFR ± SD (mL/min/1.73 m²): intervention group 1 (21.1 ± 8.5); intervention group 2 ( 21.6 ± 6.8)

  • Comorbidities: hypertensive nephropathy (15); glomerulonephritis (6); polycystic kidney disease (3); other causes (6); unknown causes (20)

  • GI status: constipation (19); GSRS score 10‐point scale (average 5.0)

Interventions Intervention group 1
  • Prebiotic: short‐chain fructo‐oligosaccharide powder sachet, 12 g/day

  • Time: 12 weeks (dose escalation for first 9 days)


Intervention group 2
  • Prebiotic: maltodextrin powder sachet, 12 g/day

  • Time: 12 weeks (dose escalation for first 9 days)

Outcomes Outcomes reported by this study at 12 weeks
  • P‐cresol sulfate

  • Indoxyl sulfate

  • Indole 3‐acetic acid

  • Zonulin

  • Epidermal growth factor

  • glucagon‐like peptide GLP‐2

  • High‐sensitive CRP

  • IL‐6

  • Urea nitrogen

  • Creatinine

  • eGFR

  • Proteinuria

  • Albumin

  • Glucose

  • Homeostatic model assessment‐insulin resistance

  • Lipid profile

  • GI symptoms (Rome III Criteria and GSRS)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • Most outcomes reported as median (inter‐quartile range) and unable to contribute to meta‐analysis

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants were randomly assigned to placebo or intervention group (blinded as A or B). The randomization was computer generated (ratio of 1:1, blocks of six) by an independent investigator, and stratified by gender and eGFR (from 45.0 to 22.5 mL/min/1.73 m²)."
Allocation concealment (selection bias) Low risk Quote: "Both supplements were distributed in powdered form with the same texture and color, in identical sachets identified by the letters A and B, packaged off‐site by a food‐grade manufacturer (HileIndustria de Alimentos Ltda, Santa Catarina, Brazil) and labelled by an independent investigator."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Both supplements were distributed in powdered form with the same texture and color, in identical sachets identified by the letters A and B, packaged off‐site by a food‐grade manufacturer (HileIndstria de Alimentos Ltda, Santa Catarina, Brazil) and labelled by an independent investigator."
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 8%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT02364869
Control for confounding factors Low risk Pre‐treatment period: no use of phosphate binders, immunosuppressants, antiinflammatories, antibiotics, laxatives, prebiotics, probiotics or synbiotics 3 months preceding baseline assessment
Study period: "All participants had been previously advised by a renal dietitian to keep a diet composed by 0.6–0.8 g/kg/day of protein, 30–35 kcal/kg/day of energy, restricted in sodium and controlled in potassium if necessary. During the follow‐up, the participants were encouraged to maintain stable protein and fiber intake, and not to use laxatives, other prebiotics and/or probiotics."
Other bias Low risk Conflicts declared: "None declared"
Funding declared: "Conselho Nacional de Desenvolvimento Cientıfico e Tecnologico (CNPq, Brazil); Fundacao de Amparo a` Pesquisa do Estado de Sao Paulo (FAPESP, Brazil); Coordenacao de Aperfeicoamento de Pessoal de Nıvel Superior (CAPES, Brazil); Hospital do Rim–Fundacao Oswaldo Ramos (Sao Paulo, Brazil)"

Ranganathan 2009.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT

  • Pilot study


Time frame
  • July 2007 to August 2008

  • Treatment: 12 weeks

  • Follow‐up: none past 12 weeks treatment

Participants Study characteristics
  • Country: Canada

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 18 to 75 years; CKD stage 3 or 4; SCr > 2.5 mg/dL

  • Exclusion criteria: pregnant; antibiotics; anticoagulants; active dependency on drugs or alcohol; HIV/AIDS/liver disease; any medical, psychiatric, debilitating disease/disorder or social condition


Baseline characteristics
  • Number (randomised/analysed): 16/13

  • Mean age ± SD (years): 54 ± 8.8

  • Sex (M/F): 9/4

  • CKD stage: 3 and 4

  • Kidney measurements: not reported

  • Comorbidities: hypertension (11); hyperlipidaemia (1); type 2 DM (1); IgA nephritis (1); polycystic kidney disease (1)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: 1 oral capsule of Lactobacillusacidophilus KB31, Bifidobacteriumlongum KB35, and Streptococcusthermophilus KB27, total of 1.5 x 1010 CFU; 2 capsules 3 times/day

  • Time: 12 weeks


Washout
  • None


Control group
  • Placebo: oral capsule placebo powder; 2 capsules 3 times/day

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • SCr

  • Uric acid

  • BUN

  • CRP

  • Faecal microbiological composition

  • QoL

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

  • Pilot


Available data
  • No available data for meta‐analyses: first phase of cross‐over not reported as separate data

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization sequence was generated by alternative patient sequential methodology"
Allocation concealment (selection bias) Unclear risk Insufficient information provided to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The placebo was composed of wheat germ plus Psyllium husks. It was also matched in color, size and enteric coating identical to the interventional product"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information provided to permit judgement
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 19%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol
Control for confounding factors Unclear risk Pre‐treatment period
  • Not reported


Study period
  • Dietary recall

Other bias High risk Conflicts declared
  • "N.R. has disclosed that he is the Senior Vice‐President of research and development at Kibow Biotech, Inc. P.R. has disclosed that she is the Vice‐President for clinical and regulatory affairs, Kibow Biotech, Inc. N.R. and P.R. have both disclosed that they hold a substantial combined business interest in Kibow Biotech, Inc. R.D. has disclosed that he is an employee of Kibow Biotech, Inc. E.A.F. has disclosed that he serves, without compensation, as Chair of Kibow Biotech’s Scientific Advisory Board, and that his Renal Division currently receives research funding for a clinical trial of Kibow Probiotics. P.T. and V.R. have disclosed that they have not received any compensation from Kibow Biotech for conducting this pilot‐scale clinical study. All peer reviewers receive honoraria from CMRO for their review work. Peer Reviewer 1 has disclosed that he/she is a scientific consultant on clinical trials for Jamieson Laboratories Inc. Peer Reviewer 2 has disclosed that he/she has no relevant financial relationships.


Funding declared
  • "Funding for this Canadian study was provided by Gelda Scientific, Inc, Mississauga, Ontario, Canada (in exchange for rights to distribute the product in Canada). Kibow Biotech has funded the publication of this article."

Shariaty 2017.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 23 August 2014 to 22 November 2014

  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: multi‐centre (5 hospital dialysis centres)

  • Inclusion criteria: ≥ 17 years; undergoing HD 3 times/week, positive CRP

  • Exclusion criteria: active bleeding, surgery in past 3 months; blood disorders; anaemia due to iron; folate, or vitamin B12 deficiencies; active infections; immune system disorders; malignancies; alcoholic; treatment with antibiotics, corticosteroids;HIV


Baseline characteristics
  • Number (randomised/analysed): intervention group (18/17); control group (18/17)

  • Age range (years): 47 to 60

  • Sex (M/F): 20/16

  • CKD stage: G5D

  • Kidney measurements: not reported

  • Comorbidities: not reported

  • GI status: not reported

Interventions Intervention group
  • Probiotics: Lactobacillus acidophilus 3 × 1010 CFU, Lactobacillus casei 3 × 109 CFU, Lactobacillus rhamnosus 7 × 109 CFU, Lactobacillus bulgaricus 5 × 108 CFU, Bifidobacterium breve 2 × 1010 CFU, Bifidobacterium longum 1 × 109 CFU, Streptococcus thermophilus 3 × 108 CFU; oral tablets of 500 mg/day

  • Time: 12 weeks


Control group
  • Placebo: oral capsule "that was similar to the probiotic supplements and contained starch" per day

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Hb

  • CRP

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • Study does not report outcomes of interest for this review

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Convenience sampling and simple randomly allocation was used to designate the patients to intervention or control groups. The patients were randomly allocated to Groups A and B (n = 18 for each) using two sets of random numbers with codes 1 to 36 and were assigned probiotic capsules in one group and placebos in the other."
Allocation concealment (selection bias) Unclear risk No information provided
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Quote: "The study was double blinded, so that neither the patients nor the evaluators had any information about either of the two groups. Indeed the participants did not know which groups they belonged to (probiotic or placebo group)"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Quote: "The study was double blinded, so that neither the patients nor the evaluators had any information about either of the two groups. Indeed the participants did not know which groups they belonged to (probiotic or placebo group)"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 6%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: Iranian Registry 2013072710325N2
Control for confounding factors Unclear risk Pre‐treatment period: not reported
Study period: "Participants’ diet, levels of activity, and medications used did not change during the study and all the patients received the necessary training on these subjects"; "Both groups received daily folic acid supplements and monthly vitamin B12 supplements and received no other additional supplements"
Other bias Low risk Conflicts declared: "There are no conflicts of interest"
Funding declared: "This study was financed by Golestan Medical Sciences University. This study recorded and approved by the Iranian Registry of Clinical Trial Code: 2013072710325N2"

Sirich 2014.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, single‐blind, placebo‐controlled RCT


Time frame
  • October 2010 to May 2013

  • Treatment: 6 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: USA

  • Setting: multicentre (6 sites)

  • Inclusion criteria: ≥ 18 years; HD

  • Exclusion criteria: residual urea clearance > 2 mL/min or if they reported significant urine production; active GI disease; use of antibiotics in 4 weeks prior; poor dialysis attendance


Baseline characteristics
  • Number (randomised/analysed): intervention group (28/20); control group (28/20)

  • Mean age ± SD (years): intervention group (54 ± 14); control group (58 ± 13)

  • Sex (M/F): intervention group (11/9); control group (13/7)

  • CKD stage: G5D

  • Kidney measurements

    • Mean albumin ± SD (g/dL): intervention group (4.0 ± 0.3); control group (3.9 ± 0.3)

  • Comorbidities: diabetes (18)

  • GI status: not reported

Interventions Intervention group
  • Prebiotic: high‐amylose corn starch (Hi‐maize 260), composed of approximately 40% digestible starch and 60% resistant starch; 15 g/day

  • Time: 6 weeks


Control group
  • Prebiotic: waxy corn starch (high‐amylopectin starch); 15 g/day

  • Time: 6 weeks

Outcomes Outcomes reported by this study at 6 weeks
  • Indoxyl sulfate

  • p‐Cresol sulfate

  • Body weight

  • Urea nitrogen

  • Albumin by bromocresol purple

  • Albumin by nephelometry

  • Prealbumin

  • CRP

  • Phosphate

  • KDQoL‐36 score

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Permuted‐block randomization was utilized to assign patients to fibre or control starch"
Allocation concealment (selection bias) Unclear risk No information provided
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Fiber and control supplements were provided as white powder in identical sachets"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information provided
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 29%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: ClinicalTrials.gov NCT0118627
Control for confounding factors Unclear risk Pre‐treatment period: participants must not have used antibiotics in the 4 weeks prior to starting study treatment
Study period: not reported
Other bias High risk Conflicts declared: "None."
Funding declared: "Supplements were provided ... by Ingredion Incorporated (Bridgewater, NJ)"

Soleimani 2017.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 29 February to 5 March 2016

  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Hospital dialysis unit)

  • Inclusion criteria: 18 to 80 years; HD for at least 1 year; diabetes

  • Exclusion criteria: pregnant; intestinal diseases; probiotic, prebiotic, antioxidant, anti‐inflammatory, antibiotics or immunosuppressive medications within 3 months before enrolment in the study


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/30); control group (30/30)

  • Mean age ± SD (years): intervention group (54.0 ± 16.0); control group (59.4 ± 16.0)

  • Sex (M/F): intervention group (20/10); control group (20/10)

  • CKD stage: G5D

  • Kidney measurements

    • Mean eGFR ± SD (mL/min/1.73 m²): intervention group (2.49 ± 1.15); control group (2.22 ± 0.86)

    • Mean albumin ± SD (g/dL): intervention group (4.3 ± 0.4); control group (4.0 ± 0.4)

    • Mean SCr ± SD (mg/dL): intervention group (7.4 ± 3.1); control group (7.8 ± 3.0)

  • Comorbidities: type 1 diabetes (6); type 2 diabetes (54); hypertension (58); cancer (4)

  • GI status: not reported

Interventions Intervention group
  • Probiotic: Lactobacillus acidophilus, Lactobacillus casei, and Bifidobactiera bifidum 2 x 109 CFU/g; oral capsule/day

  • Time: 12 weeks


Control Group
  • Placebo: oral capsule of 'placebo' undefined

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Fasting plasma glucose

  • Insulin

  • Homeostasis model of assessment–estimated insulin resistance

  • Homeostasis model of assessment–estimated beta‐cell function

  • Quantitative insulin sensitivity check index

  • HbA1c

  • Triglycerides

  • VLDL cholesterol

  • Total cholesterol

  • LDL cholesterol

  • HDL cholesterol

  • High‐sensitivity CRP

  • Nitric oxide

  • Total antioxidant capacity

  • Total glutathione

  • Malondialdehyde

  • Albumin

  • Total iron binding capacity

  • Sodium

  • Potassium

  • GFR

  • SCr

  • BUN

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Randomization assignment was conducted using computer‐generated random numbers"
Allocation concealment (selection bias) Low risk Quote: "Randomization and allocation were hidden from the researchers and patients until the final analyses were completed"
Quote: "The randomized allocation sequence, enrolling participants and allocating them to interventions, was conducted by a trained nutritionist at the dialysis clinic"
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Randomization and allocation were hidden from the researchers and patients until the final analyses were completed"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 8%
ITT undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: http://www.irct.ir: IRCT201601025623N69
Control for confounding factors Low risk Pre‐treatment period: "Taking probiotic supplements and other forms of probiotics (including probiotic yogurt, kefir, and other fermented foods), taking prebiotic, antioxidant, and/or anti‐inflammatory supplements (e.g., vitamin E, vitamin C, and omega‐3 fatty acids), or taking antibiotics and immunosuppressive medications within 3 months before enrolment in the study"
Study period: "Patients were requested not to change their routine physical activity, and not to take any anti‐inflammatory medications and supplements that might affect their nutritional status during the 12‐week intervention"
Other bias High risk Conflicts declared: "All the authors declared no competing interests"
Funding declared: "Probiotic supplements were produced by Tak Gen Zist Pharmaceutical Company (Tehran, Iran)"; "The present study was supported by a grant from the Vice‐chancellor for Research, KUMS, and Iran"

Soleimani 2019.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • November 2017 to February 2018

  • Treatment: 12 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Dialysis centre)

  • Inclusion criteria: 18 to 80 years; HD; diabetes

  • Exclusion criteria: pregnant; synbiotics, antioxidant, anti‐inflammatory within the last 3 months; patients requiring medications adjustment during study; recently been diagnosed with type 1 DM or type 2 DM


Baseline characteristics
  • Number (randomised/analysed): intervention group (30/30); control group (30/30)

  • Mean age ± SD (years): intervention group (62.8 ± 12.7); control group (62.8 ± 14.8)

  • Sex (M/F): intervention group (21/9); control group (21/9)

  • CKD stage: G5D

  • Kidney measurements: not reported

  • Comorbidities: type 1 DM (4); type 2 DM (56); hypertension (unclear)

  • GI status: not reported

Interventions Intervention group
  • Synbiotic: Lactobacillus acidophilus, Lactobacillus casei, and Bifidobacterium bifidum 2 × 109 CFU, and 0.8 g of inulin per day

  • Time: 12 weeks


Control group
  • Placebo: corn starch oral capsule

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Fasting plasma glucose

  • Insulin

  • Homeostasis model of assessment–estimated insulin resistance

  • Quantitative insulin sensitivity check index

  • HbA1c

  • Triglycerides

  • VLDL cholesterol

  • Total cholesterol

  • LDL cholesterol

  • HDL cholesterol

  • Total‐/HDL‐cholesterol ratio

  • High‐sensitivity CRP

  • Nitric oxide

  • Total antioxidant capacity

  • Total glutathione

  • Malondialdehyde

  • Malnutrition via SGA score (20‐point scale)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • No data available for meta‐analysis on our primary and secondary outcomes

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants were randomized using computer‐generated random numbers."
Allocation concealment (selection bias) Low risk Quote: "Randomization and allocation were concealed from both researchers and patients until the completion of final analyses. The randomized allocation sequence, enrolling participants, and allocating them to interventions were conducted by a trained nutritionist in dialysis clinic."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "Randomization and allocation were concealed from both researchers and patients until the completion of final analyses."
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information to permit judgement
Incomplete outcome data (attrition bias)
All outcomes Low risk All participants were accounted for with reasons for withdrawals provided
Attrition = 0%
ITT analysis undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: http://www.irct.ir: IRCT2017090133941N17
Control for confounding factors Low risk Pre‐treatment period: No probiotic, synbiotic, antioxidant, anti‐inflammatory supplements 3 months prior to enrolment.
Study period: "Patients were requested not to change their routine physical activity or usual diets throughout the study and not take any anti‐inflammatory and antioxidant medications or supplements during the 12‐week intervention which might affect the results of the study."
Other bias High risk Conflicts declared: "The authors declare that they have no conflict of interest"
Funding declared: "Synbiotic supplements and placebos (corn starch) were produced by Tak Gen Zist Pharmaceutical Company, Tehran, Iran"; "The research grant provided by Research Deputy of Kashan University of Medical Sciences (KAUMS)"

SYNERGY 2014.

Study characteristics
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • 5 January 2013 to 12 November 2013

  • Treatment: 6 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Australia

  • Setting: single centre (Dialysis centre)

  • Inclusion criteria: ≥ 18 years; non‐dialysed CKD stage 4 or 5, eGFR 10 to 30 mL/min/1.73 m²

  • Exclusion criteria: previous kidney transplant; bowel radiation or large bowel resection; prebiotics, probiotics or antibiotics 1 month prior; irritable bowel syndrome; Crohn disease; ulcerative colitis; likely to receive a transplant or progress to dialysis within 6 months; severely malnourished (SGA: C); clinically significant change to immunosuppressant dose within 6 months


Baseline characteristics
  • Number (randomised/analysed phase 1/analysed phase 2): 37/37/31

  • Mean age ± SD: 69 ± 10 years

  • Sex (M/F): 21/16

  • CKD stage: 3 to 5

  • Kidney measurements

    • Mean GFR ± SD: 24 ± 8 mL/min/1.7 3m²

    • Median proteinuria (IQR) (mg/24 hours): 318 (165 to 1600)

    • Median albuminuria (IQR) (mg/24 hours): 107 (20 to 1100)

  • Comorbidities: glomerular diseases (5); hypertension (37); diabetic nephropathy (14); hyperlipidaemia (29)

  • GI status: not reported

Interventions Intervention group
  • Synbiotic

    • Prebiotic high–molecular weight inulin (inulin high performance), fructo‐oligosaccharides, and galacto‐oligosaccharides; 15 g powder sachet/day (dose escalation)

    • Probiotic Lactobacillus, Bifidobacteria, and Streptococcus, 90 billion CFU; 2 oral capsules/day

  • Time: 6 weeks


Washout
  • 4 weeks


Control group
  • Prebiotic: maltodextrin powder sachet/day (dose escalation) and maltodextrin 2 oral capsules/day (placebo is actually a prebiotic)

  • Time: 6 weeks

Outcomes Outcomes reported by this study at 6 weeks
  • Total indoxyl sulfate

  • Total p‐cresol sulfate

  • Free indoxyl sulfate

  • Free p‐cresol sulfate

  • GFR

  • SCr

  • Kidney injury molecule‐1

  • Proteinuria

  • Albuminuria

  • IL‐1β

  • IL‐6

  • IL‐10

  • TNF‐α

  • F2‐isoprostanes

  • Glutathione peroxidase

  • Endotoxins

  • Patient‐reported health: physical and mental composite score

  • GI Symptom Score

  • Energy

  • Protein

  • Fibre

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article; abstract


Available data
  • No data available for meta‐analyses: first phase of cross‐over not reported separately. Author contacted to request data

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Participants underwent ... randomization in a 1:1 ratio to either synbiotic supplements or placebo"
Quote: "A computer–generated blocked randomization list with blocks of size 2 was produced"
Allocation concealment (selection bias) Low risk Quote: "A computer–generated blocked randomization list with blocks of size 2 was produced by an external statistical consultant and maintained on a secure server not accessible to those recruiting for the trial. Random allocation was performed by telephoning the list custodian, who reported the next numbered supplement kit in the list. Supplement packaging was done offsite."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The synbiotic therapy (prebiotic powder and probiotic capsule) and the identically matched placebo (maltodextrin powder and capsule)"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Insufficient information to permit judgement.
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 16%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: Australian New Zealand Clinical Trials Registry: ACTRN12613000493741
Control for confounding factors Low risk Pre‐treatment period: "Participants underwent a 2‐week run–in period followed by randomization"; "All participants underwent face‐to‐face dietary education and counselling with a qualified dietitian to achieve evidence–based guideline–recommended targets during the first week of run‐in"
Study period: "Throughout the intervention, patients were encouraged to maintain stable dietary intakes, with a focus on stabilizing protein and fibre intake. Participants were also provided with a standard evening meal preceding their overnight fast before each blood collection."
Other bias Low risk Conflicts declared: "None."
Funding declared: "This study was funded through a project grant from the Princess Alexandra Private Practice Trust Fund (PPTF). M.R. is a recipient of the Princess Alexandra PPTF Postgraduate Scholarship. D.W.J. is supported by a Queensland Government Health and Medical Research (HMR) Health Research Fellowship. K.L.C. is supported by a Queensland Government HMR Health Research Fellowship and a Lions Senior Medical Research Fellowship."

SYNERGY II 2021.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • April 2017 to August 2018

  • Treatment: 12 months

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Australia

  • Setting: single‐centre (Outpatient clinic)

  • Inclusion criteria: ≥ 18 years; non‐dialysed CKD stage 3 or 4, eGFR 15 to 60 mL/min/1.73 m²

  • Exclusion criteria: anticipated dialysis commencement within 12 months or anticipated death within 6 months; non‐English speaking or unable to give informed consent; a clinically significant change in immunosuppressant dose within 6 months; receiving or had received radiation to the bowel or had a large bowel resection; consumed pre‐, probiotic, or antibiotic therapy within 1 month of study commencement; medically diagnosed and active irritable bowel syndrome, Crohn’s disease, or ulcerative colitis; cirrhotic liver disease; or severely malnourished (SGA: C)


Baseline characteristics
  • Number (randomised/analysed): intervention group (35/28); control group (33/28)

  • Median age (IQR) (years): intervention group (72, 66 to 76); control group 69, 56 to 73()

  • Sex (M/F): intervention group (23/12); control group (22/11)

  • CKD stage: 3 to 4

  • Kidney measurements

    • Median eGFR (IQR) (mL/min/1.73 m²): intervention group (31.5, 26.0 to 37.0); control group (36.0, 29.0 to 44.0)

    • Median creatinine (IQR) (mumol/L): intervention group (168, 135 to 217); control group (163, 131 to 191)

  • Comorbidities: hypertension (52); diabetes (30); hyperlipidemia (19); CVD (26)

Interventions Intervention group
  • Synbiotic

    • Probiotic Lactobacillus, Bifidobacteria, and Streptococcus, 4.5 x 1011 CFU/day

    • Prebiotic high‐resistant starch fibre supplement (Hi‐Maize 260, 50% resistant starch; Ingredion); 20 g powder sachet/day

  • Time: 12 months


Control group
  • Prebiotic: maltodextrin powder sachet/day (dose escalation) and waxy maize powder/day (placebo is actually a prebiotic)

  • Time: 12 months

Outcomes Outcomes reported by this study at 3, 6, 9, 12 months
  • GI symptoms (GSRS)

  • Stool frequency (patient‐reported/day)

  • Stool consistency (Bristol Stool Form Scale)

  • Treatment adherence

  • Dietary intake

  • Adverse events

  • QoL (Assessment of Quality of Life Questionnaire)

  • Global longitudinal strain

  • Left ventricular mass index

  • Ejection fraction

  • SCr

  • eGFR

  • Free and protein‐bound serum concentrations of serum indoxyl sulfate and p‐cresyl sulfate

  • Stool microbiota analysis (DNA extraction sequencing)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "The randomization was completed by a computer‐generated random number (1:1 ratio, blocks of 4, stratified by CKD stage and presence of diabetes)"
Allocation concealment (selection bias) Unclear risk Quote: "randomization ... was carried out by an independent investigator not involved in the recruitment or implementation of the study protocol"
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk Quote: "a ... double‐blind ... trial"
Comment: study states to be double‐blind but no detailed description of how these methods were undertaken
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Quote: "a ... double‐blind ... trial"
Comment: study states to be double‐blind but no detailed description of how these methods were undertaken
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawal provided
Attrition = 18%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or an a priori published protocol: ANZCTR (ACTRN12617000324314)
All outcomes planned in the methods were reported in the results
Control for confounding factors Low risk Quote: "Throughout the intervention, participants were provided individualized and tailored dietary advice in line with best practice guidelines and were encouraged to maintain a stable dietary intake, focusing on not altering protein and fiber intake."
Other bias Low risk Conflicts declared: "The authors declare no conflict of interest."
Funding declared: "This study was funded through project grants from the Princess Alexandra Research Foundation; Wishlist Research Grant Scheme, Study Education and Research Trust Fund, Sunshine Coast; and BEAT‐CKD via Australasian Kidney Trial Network."

Viramontes‐Horner 2015.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Mexico

  • Setting: multi‐centre (2 public hospitals)

  • Inclusion criteria: ≥ 18 years; HD for at least 3 months with arteriovenous fistula

  • Exclusion criteria: taking probiotics, omega‐3 fatty acids, pentoxifylline, immunosuppressants, nonsteroidal anti‐inflammatory drugs; cancer; decompensated heart failure; chronic liver diseases; intestinal malabsorption; active infections; AIDs; filters reuse; kidney transplant antecedent


Baseline characteristics
  • Number (randomised/analysed): intervention group (22/14); control group (20/15)

  • Mean age ± SD (years): intervention group (40.6 ± 17.1); control group (39.0 ± 16.0)

  • Sex (M/F): intervention group (16/6); control group (16/4)

  • CKD stage: G5D

  • Kidney measurements

    • Median SCr, IQR (mg/dL): intervention group (9.8, 7.5 to 12.9); control group (11.4, 10.2 to 13.2)

    • Mean albumin ± SD (g/dL): intervention group (3.7 ± 0.3); control group (3.7 ± 0.2)

  • Comorbidities: diabetes (7); hypertension (3); uric acid nephropathy (1); renal polycystosis (3);

  • GI status: GI issues present at baseline

Interventions Intervention group
  • Synbiotic: Lactobacillus acidophilus, Bifidobacteriumlactis Bi‐07 11 x 106 CFU, 2.31 g inulin, 1.5 g of omega‐3 fatty acids (eicosapentaenoic and docosahexaenoic acids), and vitamins undefined dose (complex B, folic acid, ascorbic acid, and vitamin E) per day

  • Time: 8 weeks


Control group
  • Placebo: oral capsule undefined per day

  • Time: 8 weeks


Cointerventions
  • Nutritional counselling for both groups

Outcomes Outcomes reported by this study at 8 weeks
  • Albumin

  • Glucose

  • Total cholesterol

  • Triglycerides

  • HDL cholesterol

  • LDL cholesterol

  • Phosphorus

  • Potassium

  • Sodium

  • Calcium

  • Urea

  • BUN

  • Creatinine

  • CRP

  • TNF‐α

  • IL‐6

  • Energy

  • Carbohydrate

  • Lipid

  • Protein

  • Fibre

  • Dry weight

  • Height

  • Dry BMI

  • Triceps skinfold

  • Subscapular skinfold

  • Mid‐arm muscle area

  • Mid‐arm fat area

  • Overhydration

  • Total body water

  • Extracellular/intracellular water index

  • Lean tissue mass

  • Fat tissue

  • Body cell mass

  • Nutritional status (SGA)

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "Patients were randomly allocated ..."
Insufficient information to permit judgement
Allocation concealment (selection bias) Unclear risk Insufficient information to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "The placebo had identical color, size, and flavor to the interventional product"
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Every month, tolerability and safety of the symbiotic gel were evaluated by direct interview by the same experienced dietician (D.V.H.), who was blinded to the intervention treatment"
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 17%
ITT analysis not undertaken
Selective reporting (reporting bias) High risk Trial registration or a priori published protocol: not reported
Control for confounding factors Low risk Pre‐treatment period: participants must not have been taking probiotics, omega‐3 fatty acids, pentoxifylline, immunosuppressants, nonsteroidal anti‐inflammatory drugs prior to start
Study period: nutritional counselling provided as a co‐intervention throughout study to maintain similar and regular nutrient intakes
Other bias Low risk Conflicts declared: "Special acknowledgment is made to chemists of Central Laboratory and nurses of the haemodialysis units for all the support given during the study. FMC works for Nutrimentos Inteligentes, S.A. de C.V. Nevertheless, her participation in the present study consisted in provide methodological and statistical advisory. During all her participation, she did her assigned activities completely blinded. Although D.V.H., BSc, started working for Nutrimentos Inteligentes, S.A. de C.V. after the conclusion of the study, during the fieldwork and the statistical analysis, she also did all her tasks totally blinded."
Funding declared: "The present study was supported from CONACYT (Consejo Nacional de Ciencia y Tecnologıa) (supportive number: 174193) and Nutrimentos Inteligentes, S.A. de C.V."

Wang 2015a.

Study characteristics
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT


Time frame
  • July 2011 to June 2012

  • Treatment: 24 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: China

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: ≥ 18 years; PD for at least 1 month; eGFR < 15 mL/min

  • Exclusion criteria: active infectious conditions within the previous 30 days; pregnancy; autoimmune diseases; on immunosuppressive agents or the consumption of antibiotics within 30 days prior to enrolment


Baseline characteristics
  • Number (randomised/analysed): intervention group (23/21); control group (24/18)

  • Median age, IQR (years): intervention group (51, 39 to 57); control group (53.5, 43 to 59)

  • Sex (M/F): intervention group (10/11); control group (8/10)

  • CKD stage: G5D

  • Kidney measurements

    • Median SCr, IQR (mg/dL): intervention group (12.23, 8.80 to 13.82); control group (12.88, 11.29 to 14.29)

    • Median albumin, IQR (mg/dL): intervention group ( 3.7, 3.6 to 3.9); control group (3.8, 3.5 to 4)

  • Comorbidities: diabetes (8); hypertension (32); coronary artery disease (8); chronic hepatitis B (3); chronic hepatitis C (1)

  • GI status: not reported

Interventions Intervention group
  • Probiotics: Bifidobacteriumbifidum A218, Bifidobacteriumcatenulatum A302, Bifidobacteriumlongum A101, Lactobacillusplantarum A87, total of 4 x 109 CFU/day

  • Time: 24 weeks


Controlgroup
  • Prebiotic: maltodextrin oral capsule; dose not reported

  • Time: 24 weeks

Outcomes Outcomes reported by this study at 24 weeks
  • Serum endotoxin

  • TNF‐α

  • IFN‐gamma

  • IL‐5

  • IL‐6

  • IL‐10

  • IL‐17

  • Peritonitis

  • Cardiovascular events

  • Residual CrCl

  • Residual urea clearance

  • Residual kidney function

  • BUN

  • Creatinine

  • Uric acid

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Quote: "Patients were randomised into probiotic or control groups using a random‐number table sequence."
Allocation concealment (selection bias) Low risk Quote: "Randomisation was performed using sequential numbers generated at the computer centre of China Medical University Hospital. The allocations were contained in opaque, sequentially numbered, sealed envelopes."
Blinding of participants and personnel (performance bias)
All outcomes Low risk Quote: "While the placebo group received similar capsules".
Quote: "Both investigators and patients were blind as to the assignment."
Blinding of outcome assessment (detection bias)
All outcomes Low risk Quote: "Both investigators and patients were blind as to the assignment."
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were accounted for with reasons for withdrawals provided
Attrition = 17%
ITT analysis not undertaken
Selective reporting (reporting bias) Low risk Trial registration or a priori published protocol: NCT01391468
Control for confounding factors Low risk Pre‐treatment period: No immunosuppressive agents or the consumption of antibiotics within 30 days prior to enrolment
Study period: "Patients were advised not to change their dietary habit during the study period"
Other bias Low risk Conflicts declared: "The authors state that they have no conflict of interest"
Funding declared: "This study is supported in part by Taiwan Ministry of Health and Welfare Clinical Trial and Research Center of Excellence (MOHW103‐TDU‐B‐212‐113002), the research laboratory of pediatrics, Children’s Hospital of China Medical University, and China Medical University Hospital (grant numbers DMR‐101‐016 and DMR‐103‐013)"

Xie 2015a.

Study characteristics
Methods Study design
  • Parallel, 3‐arm, open‐label, placebo‐controlled RCT


Time frame
  • April 2013 to August 2014

  • Treatment: 6 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: China

  • Setting: single‐centre (3 hospital dialysis centres)

  • Inclusion criteria: age range not specified; maintenance HD 3 times/week

  • Exclusion criteria: complications with acute inflammation; trauma; GI disorders; liver diseases; cancers; mental retardation within recent 3 months; taking supplementation of herbs, antioxidants, vitamins/minerals, or fish oils


Baseline characteristics
  • Number: intervention group 1 (41); intervention group 2 (39); control group (44)

  • Mean age ± SD (years): intervention group 1 (53.7 ± 14.2); intervention group 2 (51.7 ± 15.7); control group (53.1 ± 13.2)

  • Sex (M/F): intervention group 1 (24/17); intervention group 2 (18/20); control group (26/18)

  • CKD stage: G5D

  • Kidney measurements

    • Mean SCr ± SD (mmol/L): intervention group 1 (912.4 ± 268.9); intervention group 2 (1013.5 ± 284.1); control group (948.6 ± 272.4)

    • Mean albumin ± SD (g/L): intervention group 1 (35.6 ± 1.4); intervention group 2 (34.8 ± 1.2); control group (35.2 ± 1.6)

  • Comorbidities: chronic GN (69); diabetic nephropathy (39); hypertensive nephropathy (11); polycystic kidney disease (5)

  • GI status: not reported

Interventions Intervention group 1
  • Prebiotic: undefined water‐soluble fibre (fermentable rate > 75%) added to rice; 10 g/day

  • Time: 6 weeks


Intervention group 2
  • Prebiotic: undefined water‐soluble fibre (fermentable rate > 75%) added to rice; 20 g/day

  • Time: 6 weeks


Control Group
  • Placebo: placebo starch added to rice; unstated dose/day

  • Time: 6 weeks


Co‐interventions
  • Regular diet: caloric intake 35 kcal/kg/BW; protein intake 1 to 1.2 g/kg/BW, fats < 35%; odium and potassium restriction

Outcomes Outcomes reported by this study at 6 weeks
  • Energy

  • Protein

  • Carbohydrate

  • Fats

  • Dietary fibre without added

  • Calcium

  • Weight

  • Mid‐arm circumference

  • Triceps skinfold thickness

  • BMI

  • Triglycerides

  • Total cholesterol

  • LDL

  • HDL

  • Total cholesterol:HDL ratio

  • Superoxide dismutase

  • Total antioxidant capacity

  • Malondialdehyde

  • Glutathione peroxidase

  • TNF‐α

  • IL‐6

  • IL‐8

  • High‐sensitivity CRP

Notes Baseline differences between groups
  • None


Publication type
  • Full‐text article


Available data
  • Data or outcomes are not reported in a useable way; however, do not include data from this study in the meta‐analyses as the prebiotic intervention is only described as a 'water soluble fibre' with no description of its contents

Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Quote: "This 6‐week randomized controlled trial was conducted"
Insufficient information to permit judgement.
Allocation concealment (selection bias) Unclear risk Insufficient information to permit judgement
Blinding of participants and personnel (performance bias)
All outcomes High risk Open‐label study
Blinding of outcome assessment (detection bias)
All outcomes High risk Open‐label study
Incomplete outcome data (attrition bias)
All outcomes High risk All participants were not accounted for, and completers or withdrawals were not reported
Attrition = not reported
ITT analysis unclear if undertaken
Selective reporting (reporting bias) Unclear risk Trial registration or a priori published protocol: not reported
Control for confounding factors Low risk Pre‐treatment period: "This study consisted of one week on regular diet to establish baseline dietary intake"
Study period: Regular diet: "caloric intake 35 kcal/kg/bw, protein intake 1‐1.2 g/kg/bw, fats < 35%, and with sodium and potassium restriction"
Other bias Low risk Conflicts declared: "None"
Funding declared: "This study was supported by Shanghai Municipal Education Commission Scientific Research Fund (14ZZ043)"

AIDs: acquired immunodeficiency disorder; APD: automated peritoneal dialysis; BMI: body mass index; BP: blood pressure; BW: body weight (in kilograms); BUN: blood urea nitrogen; CAPD: continuous ambulatory peritoneal dialysis; CaxP: calcium‐phosphorus product; CFU: colony forming units; CKD: chronic kidney disease; CrCl: creatinine clearance; CRP: C‐reactive protein; CVD: cardiovascular disease; DM: diabetes mellitus; eGFR: estimated glomerular filtration rate; EPO: erythropoietin; EQ‐5D: EuroQol‐5‐Dimension; FGID: functional gastrointestinal disorders; GI: gastrointestinal; Hb: haemoglobin; HbA1c: glycated Hb; GN: glomerulonephritis; GSRS: Gastrointestinal Symptom Rating Scale; HCY: haematocrit; HD: haemodialysis; HDL: high‐density lipoprotein; HIV: human immunodeficiency virus; HRQoL: health‐related quality of life; IFN: interferon; IQR: interquartile range; IL: interleukin; ITT; intention‐to‐treat; IV: intravenous; KDQoL: Kidney Disease Quality of Life; KRT: kidney replacement therapy; LDL: low‐density lipoprotein; M/F: males/females; MET: metabolic equivalent; MI: myocardial infarction; NGAL: neutrophil gelatinase‐associated lipocalin; PTH: parathyroid hormone; RCT: randomised controlled trial; SCr: serum creatinine; SD: standard deviation; SEM: standard error of the mean; SF: short‐form; SGA: subjective global assessment; TGF: transforming growth factor; TNF: tumour necrosis factor; UACR: urinary albumin‐creatinine ratio; VLDL: very low density lipoprotein

Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion
Afaghi 2016 Wrong intervention: the intervention is multi‐vitamin and multi‐mineral powder that contains 2 probiotics and 1 prebiotic, but these are not the majority of the dose, and there are too many other ingredients to be able to ascertain any effect.

Group A: ISOWHEY oral nutritional supplement
Whey protein concentrate (whey protein, soy lecithin), Litesse® (polydextrose), xylitol, maltodextrin, medium chain triglycerides, calcium carbonate (calcium), cream flavour, vanilla flavour, whey protein isolate, magnesium oxide heavy (magnesium), potassium citrate (potassium), glutamine, xanthan gum, seagreens (Ascophyllum nodosum seaweed), enzyme mix, ascorbic acid (vitamin C), ferrous fumarate (iron), Lactobacillus acidophilus, chromium picolinate (chromium), zinc oxide (zinc), d‐alpha tocopheryl succinate (vitamin E), nicotinamide (vitamin B3), potassium iodide (iodine), manganese sulfate monohydrate (manganese), vitamin A acetate (vitamin A), Bifidobacterium lactis, biotin, cupric sulfate pentahydrate (copper), folic acid (vitamin B9), selenomethionine (selenium), pyridoxine hydrochloride (vitamin B6), calcium d‐pantothenate (vitamin B5), riboflavin (vitamin B2), thiamine hydrochloride (vitamin B1), cholecalciferol (vitamin D3), cyanocobalamin (vitamin B12), sodium. Contains milk and soy.

Group B: BCAA (branded‐chain amino acid) oral nutritional supplement
BCAA (L‐Leucine, L‐Isoleucine, L‐Valine), Citrulline Malate, L‐Glutamine, Minerals (Magnesium Phosphate, Calcium Phosphate, Sodium Chloride, Potassium Chloride), Flavour, Citric Acid, Sucralose, Vitamins (Calcium Ascorbate, Vitamin B6, Vitamin B12), Colour (Beta Carotene or Anthocyanins)

Group C: routine diet without any supplements
Cruz‐Mora 2014 Wrong intervention: blended with nutritional counselling, albeit in both groups; however, nutritional counselling directly related to diet and too similar to taking dietary supplement probiotics, so it wouldn't be possible to ascertain what has an effect
Eguchi 2011 Wrong population: liver transplantation
Ferrara 2009 Wrong population: girls with recurrent UTI, not CKD population
Firouzi 2015a Wrong population: type 2 diabetes
"Exclusion criteria include subjects who … have any acute or chronic disease other than diabetes, hyperlipidemia, and hypertension."
Fitschen 2015 Wrong intervention: not pre or probiotics
Fortes 2020 Wrong population: not CKD
Grat 2017 Wrong population: liver transplantation
Hassan 2017 Wrong intervention: not a prebiotic or probiotic treatment
Jiang 2021a Wrong population: not entirely CKD study population
Lin 2021 Wrong intervention: Chinese Herbal Medicine
Orr 2016 Wrong population: liver transplantation
Pavan 2016 Wrong study design and intervention: quasi‐observational study with mixed controls
PrePro 2018 Wrong population: liver transplantation
ProLow CKD 2022 Wrong intervention: the primary intervention is a low‐protein diet compared to no‐protein control diet. Probiotics and prebiotics are mixed into these treatment arms, however there are too many variables and interventions to be able to ascertain a causal pathway for efficacy.
Rayes 2002 Wrong population: liver transplantation
Rayes 2002a Wrong population: post‐op major abdominal surgery
Rayes 2005 Wrong population: liver transplantation
Saxena 2022 Wrong intervention: not including enzobiotics
Tayebi‐Khosroshahi 2016 Wrong intervention: not synbiotic, prebiotic or probiotic intervention treatment
Wang 2018e Wrong intervention: not pre or probiotics

CKD: chronic kidney disease; RCT: randomised controlled trial; UTI: urinary tract infection

Characteristics of studies awaiting classification [ordered by study ID]

Chen 2019b.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None reported

Mady 2018.

Methods Study design
  • Parallel, 2‐arm, single‐centre, placebo‐controlled trial

  • Randomisation unclear from the abstract

  • Blinding unclear from abstract


Study duration
  • Treatment period: not reported in the abstract

  • Follow‐up: not reported in the abstract

  • Study conducted on 92 kidney failure patients on regular HD from January 2017 to March 2017. Patients were divided into two groups: the intervention group (50 patients) received probiotics regimen containing 5 strains for 6 weeks, while the control group (42 patients) received placebo for the same period. Indoxyl sulphate using ELISA was measured before and after intervention

Participants Study characteristics
  • Country: Iran

  • Setting: unclear from abstract

  • Inclusion criteria: adults; regular HD

  • Exclusion criteria: not reported in the abstract


Baseline characteristics
  • Number: 92

Interventions Intervention group
  • Probiotics (5 strains)

  • Time: 6 weeks


Control group
  • Placebo

  • Time: 6 weeks

Outcomes Outcomes reported
  • Indoxyl sulphate

Notes Publication type
  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Marks 2010.

Methods Study design
  • Parallel, 2‐arm RCT

  • Blinding unclear from abstract


Time frame
  • Treatment period: 4 months

  • Follow‐up: 4 months, 5 months post‐transplant

Participants Study characteristics
  • Country: USA

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: undergoing kidney transplantation

  • Exclusion criteria: not reported in the abstract


Baseline characteristics
  • Number: intervention group (21); control group (22)

Interventions Intervention group
  • Probiotics: Bifidobacterium lactis, B. bifidum, B. longum, Lactobacillus acidophilus, L. rhamnosus, and L. paracasei, total of 50 billion CFU; 2 capsules/day

  • Time: taken before transplant surgery, continued for 4 months


Control group
  • Placebo: 2 capsules/day

  • Time: taken before transplant surgery, continued for 4 months

Outcomes Outcomes reported
  • Immunosuppression‐associated diarrhoea

  • Adverse events

Notes Publication type
  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available


Attrition Notes
  • Probiotic completers: 19/21

  • Placebo completers: 16/22


Baseline differences between groups
  • None

Ogawa 2020.

Methods Study design
  • Parallel, double‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 24 weeks

  • Follow‐up: unclear from abstract

Participants Study characteristics
  • Country: Japan

  • Setting: multi‐centre (dialysis centres)

  • Inclusion criteria: patients receiving HD

  • Exclusion criteria: not reported in the abstract


Baseline characteristics
  • Number: intervention group (37); control group (36)

Interventions Intervention group
  • Probiotic: combination of Streptococcus faecalis T‐110 2 mg, Clostridium butyricum TOA 10 mg, and Bacillusmesentericus TOA 10 mg; 6 tablets, 3 times/day

  • Time: 24 weeks


Control group
  • Placebo: oral capsules matching

  • Time: 24 weeks

Outcomes Outcomes reported
  • CRP to albumin ratio

  • Gut microbiota composition (analysed by a 16s rRNA gene‐based sequencing)

Notes Publication type
  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Prado 2020.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None reported

Ranganathan 2019a.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Soliman 2018.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Takayama 2003.

Methods Study design
  • Randomisation: unclear from the abstract

  • Parallel, 2‐arm study

  • Unclear blinding


Study duration
  • Treatment: 5 weeks

  • Follow‐up: unclear from abstract

Participants Study characteristics
  • Country: Japan

  • Setting: single‐centre (Hospital dialysis unit)

  • Inclusion criteria: HD patients

  • Exclusion criteria: unclear from the abstract


Baseline characteristics
  • Number: intervention group (11); control group (11)

Interventions Intervention group
  • Probiotic: Bifidobacterium longum, dose not reported in abstract, within gastro‐resistant seamless capsule

  • Time: 5 weeks


Control group
  • Prebiotic: Bifidobacterium longum, dose not reported in abstract, in powder formulation

  • Time: 5 weeks

Outcomes Outcomes reported
  • Indoxyl sulfate

Notes Publication type
  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

TCTR20220317007.

Methods Study design
  • Parallel RCT

  • Treatment: not reported

  • Follow‐up: 12 weeks

  • Recruitment status: completed

Participants Study characteristics
  • Country: Thailand

  • Inclusion criteria: males and females > 20 years with CKD and stable eGFR < 20 mL/min/1.73 m2

  • Exclusion criteria: anticipated kidney replacement therapy within 3 months; active malignancy; active infection; active autoimmune disease; immunosuppressive state; pregnancy; obstructive or reversible kidney disease

  • Planned enrolment: 40

Interventions Intervention group
  • Chitosan 500 mg/capsule; 1 capsule once/day after breakfast


Control group
  • Placebo: starch in capsules with the same appearance as chitosan capsule

Outcomes Planned outcomes
  • Indoxyl sulfate at 4 and 12 weeks

  • eGFR at 4 and 12 weeks

Notes Abstract‐only publication
Funding: not reported

Wu 2012.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Younes 2006.

Methods Study design
  • Cross‐over, 2‐arm, unclear blinding, placebo‐controlled RCT


Time frame
  • Treatment: 5 weeks

  • Follow‐up: unclear from abstract

Participants Study characteristics
  • Country: France

  • Setting: single‐centre

  • Inclusion criteria: chronic kidney failure on a moderated restrictive protein diet

  • Exclusion criteria: unclear from the abstract


Baseline characteristics
  • Number: 9

Interventions Intervention group
  • Prebiotic: fermentable carbohydrate 40 g/day + moderated restrictive protein diet 0.8 g/kg/day

  • Time: 5 weeks


Control group
  • No treatment: nothing + moderated restrictive protein diet 0.8 g/kg/day

  • Time: 5 weeks

Outcomes Outcomes reported
  • Uremia

  • Plasma urea

  • Stool sodium excretion

  • Urinary sodium excretion

Notes Publication type
  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

Zhang 2019a.

Methods  
Participants  
Interventions  
Outcomes  
Notes Publication type
  • Unable to locate copy of abstract

  • Abstract only, not eligible according to inclusion criteria

  • Unable to assess full methods and quality assessment

  • Include in an update if full article and data are published


Trial registration details
  • None available

CFU: colony‐forming units; CKD: chronic kidney disease; CRP: C‐reactive protein; HD: haemodialysis; RCT: randomised controlled trial

Characteristics of ongoing studies [ordered by study ID]

ChiCTR2200061930.

Study name Effects of non‐protein energy supplements and exercise on protein‐energy wasting and gut microbes in maintenance hemodialysis patients
Methods Study design
  • Parallel RCT

  • Recruitment: pending

Participants Study characteristics
  • Country: China

  • Inclusion criteria: aged 18 to 80 yearsl meet the diagnosis of protein‐energy depletion, that is, SGA ≤ 5 points; regular HD for 4 hours 3 times/week for more than 3 months; not receiving any nutritional support within 3 months, did not take prebiotics, probiotics and synbiotics and other foods that regulate intestinal flora; no antibiotics, hormonal drugs and immunosuppressants have been used in the past half a month; oral intake is acceptable; willing to cooperate and accept the intervention, the subjects signed the written informed consent

  • Exclusion criteria: intolerant to oral non‐protein energy supplements and exercise; amputation, severe angina pectoris, arrhythmia, and severe dyspnea; infectious diseases such as tuberculosis, liver disease, and syphilis; serious wasting diseases such as infection, malignant tumor, heart failure, peptic ulcer; dementia, schizophrenia and other mental illnesses; need surgical treatment; antibiotics, hormonal drugs and immunosuppressants were used during the experiment; those with incomplete information

  • Planned enrolment: 36 (12 per group)

Interventions Group 1
  • Conventional treatment


Group 2
  • Oral non‐protein calorie supplements


Group 3
  • Oral non‐protein calorie supplements and exercise

Outcomes Planned outcomes
  • SGA

  • 6‐meter walk

  • 3‐meter timed‐up‐and‐go test

  • Creatinine

  • CRP

  • Albumin

  • Phosphorus

  • Prealbumin

  • Potassium

  • Hb

  • Lymphocyte

  • Hand gripping strength

  • Triceps skinfold thickness

  • Body composition

Starting date Date of first enrolment: 1 August 2022
Contact information Chunrong Tang
18 Zhongshan 2nd Road, Baise, Guangxi
+86 18778676788
yyfyxt@163.com
Affiliated Hospital of Youjiang Medical University for Nationalities
Notes Funding: Foundation for High‐level Talents of Affiliated Hospital of Youjiang Medical University for Nationalities (No. R202011705)

ChiCTR2200064821.

Study name Clinical effect and mechanism of modified Shenling Baizhu powder on peritoneal dialysis
Methods Study design
  • Parallel RCT

  • Currently recruiting

Participants Study characteristics
  • Country: China

  • Inclusion criteria: males and females 18 to 70 years; undergoing CAPD and had stable dialysis for more than 6 months; syndrome differentiation standard of spleen deficiency and blood stasis syndrome in Chinese medicine; met the diagnostic criteria of functional GI diseases; agreed to join the study and signed the informed consent form

  • Exclusion criteria: receiving HD treatment simultaneously; severe primary diseases such as active malignant tumours, decompensated cirrhosis or hematopoietic system; abdominal infection occurred within 3 months before the collection of indicators; severe arrhythmias, severe heart failure, or MI or cerebrovascular events in the 3 months prior to inclusion

  • Planned enrolment: 43 per group

Interventions Intervention group
  • Modified Shenling Baizhu Powder


Control group
  • Clostridium Butyricum and Bifidobacterium Capsule

Outcomes Planned outcomes
  • Pepsinogen I

  • Pepsinogen II

  • Gastrin

  • Motilin

  • Somatostatin

  • Cholecystokinin

  • Vasoactive intestinal peptide

  • Pancreatic polypeptide

  • Intestinal flora;

  • Albumin

  • Leptin

  • IL 10

  • IL 18

Starting date Date of first enrolment: 1 November 2022
Contact information Luan Min
1 Qianjiang Road, Xinzhan District, Hefei, Anhui
+86 18040010378
1782406861@qq.com
Anhui University of Chinese Medicine
Notes Funding: Key Project of Natural Science Research in Anhui Universities

ChiCTR2300070381.

Study name Effects of dietary fiber‐added diets on protein‐binding toxoids and microinflammatory states in patients with chronic kidney disease
Methods Study design
  • Parallel RCT

  • Recruitment: pending

Participants Study characteristics
  • Country: China

  • Inclusion criteria: males and females > 18 years; eGFR = 44 mL/min/1.73 m2; accepting of dietary nutrition therapy; no cognitive and mental abnormalities; able to cooperate; volunteer to participate in this research and sign an informed consent form

  • Exclusion criteria: diagnosis with AKI; after active GI disease or GI surgery; serious primary diseases such as heart, brain, lung, liver, hematopoietic system; malignant tumours, tuberculosis and other acute infectious diseases; using or have received corticosteroids or immunosuppressive drugs and nonsteroidal anti‐inflammatory drugs in the past 3 months, and the course of treatment is more than one week; participating in other drug‐based clinical trials

  • Planned enrolment: 26 per group

Interventions Intervention group
  • Conventional low‐salt, high‐quality, low‐protein diet (0.6 to 0.8 g/kg/day, 30 to 35 kcal/kg/day)

  • After enrolment, all patients were enrolled in one low‐protein cake with a raw weight of 40 to 50 g/day, supplemented with 10 grams of dietary fibre inulin


Control group
  • Conventional low‐salt, high‐quality, low‐protein diet (0.6 to 0.8 g/kg/day, 30 to 35 kcal/kg/day)

  • After enrolment, all patients were fed a low‐protein cake weighing 40 to 50 g/day, with 10 grams of sucrose added as a placebo

Outcomes Planned outcomes
  • Serum p‐cresol sulfate

  • Assessment of inflammatory states: IL‐6 and hypersensitivity CRP

  • Evaluation of GI symptoms

  • Laboratory indicators: routine indicators include SCr, BUN, albumin, prealbumin, transferrin, blood lipids, blood electrolytes, and Hb

  • Adherence assessment: package recycling and 3‐day dietary records

  • Anthropometric measurements: height, weekly weight, upper arm muscle circumference, triceps skin fold thickness, waist circumference

  • QoL level

Starting date First enrolment: 1 November 2018
Contact information Yao Xu
453 Stadium Road, West Lake District, Hangzhou City, Zhejiang Province
13018868203
1002591331@qq.com
Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University
Notes Funding: Medical Scientific Research Foundation of All appendix sections must Zhejiang province

ChiCTR2300072339.

Study name Probiotics improve immune metabolism in maintenance hemodialysis patients by regulating intestinal flora
Methods Study design
  • Quasi RCT

  • Currently recruiting

Participants Study characteristics
  • Country: China

  • Inclusion criteria: males and females aged 18 to 70 years; stable; can eat normally and take care of themselves; no antibiotics, hormones and immunosuppressant drugs were used within 1 month before enrolment; no food and drugs containing prebiotics and probiotics have been used in the past half a year; knew the content of this study, voluntarily participated in it and signed the informed consent

  • Exclusion criteria: acute and chronic infection; unable to eat normally, need enteral and parenteral nutrition intervention; DM; complicated with immune system diseases, tumours, chronic diseases of digestive tract; previous history of cholecystectomy or colectomy

  • Planned recruitment: 30 per group

Interventions Intervention group
  • Take probiotics; diet instruction


Control group
  • Diet instruction


Healthy control group
  • Nothing

Outcomes Planned outcomes
  • TMA, TMAO

  • Lymphocyte subpopulation 16S V3‐V4;

Starting date First enrolment: 18 October 2022
Contact information Yan Lv
No. 99, Longcheng street, Taiyuan, Shanxi Province
+86 138 3414 8662
lvyanrcx@126.com
Shanxi Bethune hospital,Shanxi Academy of Medical Sciences
Notes Funding: Four “Batches” Innovation Project of Invigorating Medical through Science and Technology ofShanxi Province

CTRI/2022/02/040296.

Study name A study to assess the effect of probiotic in prevention of progression of kidney disease in patients with chronic kidney disease
Methods Study design
  • Parallel, placebo‐controlled RCT

  • Treatment: 6 months

  • Not yet recruiting

Participants Study characteristics
  • Country: India

  • Inclusion criteria: either gender 18 to 65 years; diagnosed with CKD and have been stable for at least 3 months; SCr ≥ 2.5 mg/dL at the time of screening; ability to comprehend the full nature and purpose of the study, including possible risks and adverse events; ability to co‐operate with the Investigator and to comply with the requirements of the entire study; willing to sign the written informed consent prior to inclusion in the study at the time of screening

  • Exclusion criteria: pregnant or nursing women; antibiotic treatment at the time of screening or within 14 days before screening; history of positive serologic test for HIV, hepatitis B surface antigen or Hepatitis C in the last 6 months; active dependency on controlled substances and alcohol; administration of an investigational drug either currently or within 30 days of screening; active addictive drug or alcohol use or dependence that, in the opinion of the investigator, would interfere with adherence to study requirements or assessment; any illness or condition that in the opinion of the investigator may affect the safety of the participant or the evaluation of any study endpoint; pre‐existing cardiac or pulmonary or hepatic or neurological co‐morbidities which in the judgement of the Nephrologist would interfere with the study; social conditions or medical debilitating disease/disorder, which, in the judgement of the investigator, would interfere with or serve as a contraindication to adherence to the study protocol or ability to give informed consent or affect overall prognosis of the subject

Interventions Intervention group
  • Renadyl: 1 capsule twice/day immediately after food for 6 months


Control group
  • No treatment

Outcomes Planned outcomes
  • Uremic toxins plasma levels: changes in indoxyl sulfate, p‐cresyl sulfate and indole‐3‐ acetic acid from baseline

  • Changes in kidney function tests: Serum urea, SCr and uric acid from baseline.

  • QoL: SF 8 Questionnaire score

  • Inflammatory biomarkers: high‐sensitivity CRP; IL‐6; TNF‐alpha

  • Biomarkers of oxidative stress: NO, MDA, GSH

  • eGFR

  • Evaluation of safety of study drugs

  • Timepoint: every 30 days for 6 months after enrolment

Starting date Planned date: 18 February 2022
Contact information Dr C Prabhakar Reddy
Clinical Pharmacology and Therapeutics NIMS Punjagutta Hyderabad Punjagutta Hyderabad 500082 500082 Mahbubnagar, TELANGANA India
7416512888
cptnims@gmail.com
Nizams Institute of Medical Sciences
Notes Funding: Nizams Institute of Medical Sciencess for infrastructure support

CTRI/2022/11/047767.

Study name Global phase 2 clinical trial to evaluate safety and efficacy of US‐APR2020 in subjects with chronic kidney disease stage IV
Methods Study design
  • Parallel, placebo‐controlled RCT

  • Treatment: 180 days

  • Open to recruitment

Participants Study characteristics
  • Country: India

  • Inclusion criteria" 18 to 80 years; CKD stage IV with declining kidney function for a period more than 6 months; SCr > 2.0 mg/mL; adherence to low protein diet of 0.6 to 0.8 g/kg/day based on subject response and on advisory

  • Exclusion criteria: on probiotic supplements in the past 3 months; pregnancy, breast feeding or females of child‐bearing potential who are unwilling to use a reliable form of contraception; immunosuppressant medications therapy specific to immune mediated kidney diseases; HIV/AIDs; underweight (BMI ≤ 18.5); infection that requires oral antibiotic administration at the time of randomisation or close to that visit; GI disease (irritable bowel syndrome or anal fissures or anal fistulas or perianal abscesses or perianal infections or diverticular diseases or colitis or colon polyps) at the time of screening and randomisation, or within 6 months prior to the randomisation visit (visit 2); internal prosthesis including orthopedics, neurosurgery, heart valves, vascular stents < 2 years post ‐surgery procedure; biological/tissue grafts or prosthesis or implant; on anticoagulant medicines, including Vit K antagonists (e.g., warfarin, coumadin) or the new class of oral anti‐coagulants approved by FDA in the last 10 years; on PD; AKI; mental conditions or medically debilitating disease/disorder other than CKD, which, in the judgement of the investigator, would interfere with or serve as a contraindication to adherence to the study protocol or affect the ability to give informed consent or affect overall prognosis of the patient

  • Planned enrolment: 630

Interventions Intervention group
  • US‐APR2020: Live BioTherapeutic Product containing a mix of Streptococcus thermophilus (KB19), Lactobacillus acidophilus (KB27) and Bifidobacterium longum (KB31)

  • Duration: 180 days. 1 capsule in the morning and 1 capsule in the evening after meal


Control group
  • Placebo: blend of cream of corn and cream of rice with magnesium stearate (for flow‐properties)

  • Duration: 180 days. 1 capsule in the morning and 1 capsule in the evening after meal

Outcomes Planned outcomes
  • Presence of adverse events in less than 10% of the study population, as a measure of safety

  • Arresting the decline of eGFR as per NKF‐USFDA guidelines

  • Changes in basic blood uremic metabolic markers

  • Changes in complete blood count and hematology parameters

  • Changes in C‐CRP

  • QoL

Starting date Planned first enrolment: 30 November 2022
Contact information Mr Rajendra M Sawant
5th and 6th floor, B‐wing, Centaur House, Near Hotel Grand Hyatt, Santacruz East, Mumbai 400055, India 400055 Mumbai (Suburban), MAHARASHTRA India
91‐22‐66499154
drzarapkar@lifesan.in
LifeSan Clinical Research, division of Centaur Pharmaceuticals Pvt. Ltd.
Notes Funding: Kibow Biotech, Inc 4781 West Chester Pike, Newtown Square, PA 19073, USA Tel: 888‐271‐2560 Email: info@kibowbiotech.com; CRO LifeSan Clinical Research a division of Centaur Pharmaceuticals Pvt Ltd

Ghoreishy 2022.

Study name Effect of daily consumption of probiotic yoghurt on albumin to creatinine ratio, eGFR and metabolic parameters in patients with type 2 diabetes with microalbuminuria: study protocol for a randomised controlled clinical trial
Methods Study design
  • Parallel, 2‐arm RCT

  • Unclear blinding

  • Treatment: 8 weeks

Participants Study characteristics
  • Country: Iran

  • Setting: single‐centre (Hospital dialysis unit)

  • Inclusion criteria: 20 to 70 years; diabetic nephropathy with microalbuminuria (ACR 30 to 299 mg/g, eGFR ≥ 30 mL/min/1.73 m²); BMI < 40 kg/m²; have a fixed plan for medication use during the last 3 months

  • Exclusion criteria: history of chronic diseases; inflammatory bowel disease, irritable bowel disease, liver disease, rheumatoid arthritis, other renal disorders except for diabetic nephropathy; history of smoking and alcohol use; history of taking antibiotics and nonsteroidal anti‐inflammatory drugs during the last month; taking omega 3, vitamins E and C supplements during the last 3 months; consuming probiotic supplements or foods containing probiotics during the last 3months; pregnant or lactating women, individuals with lactose intolerance or those planning to get pregnant in the next 3months; have a weight change (± 3 kg) during the study; change the dose of medications during the study; on a specific diet throughout the study

  • Planned enrolment: 60

Interventions Intervention group
  • Probiotic yoghurt 300 g/day: containing Lactobacillusacidophilus and Bifidobacteriumlactis 106 CFU/g

  • Time: 8 weeks


Control group
  • Plain yoghurt 300 g/day

  • Time: 8 weeks

Outcomes Planned outcomes
  • Albumin to creatinine ratio

  • eGFR

  • HbA1c

  • Fasting blood sugar

  • High sensitivity CRP

  • Total cholesterol

  • Triglyceride

  • LDL

  • HDL

Starting date 11 November 2021
Contact information Dr Ahmad Esmaillzadeh
Email: a‐esmaillzadeh@tums.ac.ir
Notes Trial registration or a priori published protocol
  • Iranian Registry of Clinical Trials: IRCT20201125049491N1

  • A priori published protocol: https://bmjopen.bmj.com/content/bmjopen/12/3/e056110.full.pdf


Publication type
  • Full‐text article ‐ published protocol


Available data
  • No available data at this stage

Headley 2020.

Study name The effects of 16‐weeks of prebiotic supplementation and aerobic exercise training on inflammatory markers, oxidative stress, uraemic toxins, and the microbiota in pre‐dialysis kidney patients: a randomized controlled trial‐protocol paper
Methods Study design
  • Parallel, 4‐arm, double‐blind, placebo‐controlled RCT

  • Treatment: 16 weeks

  • Follow‐up: none past treatment

  • Recruitment status: unknown

Participants Study characteristics
  • Country: USA

  • Setting: single‐centre (Hospital)

  • Inclusion criteria: 30 to 75 years; CKD stage 3 to 4, eGFR 15 to 59 mL/min/1.73 m²; must be capable of complying with and following the study protocol (diet and exercise)

  • Exclusion criteria: kidney transplant; currently in a structured exercise program; on antibiotic therapy within the last month; on a probiotic or prebiotic supplement within the last month; GI disorder that prohibits the use of resistant starch (i.e. high‐amylose corn starch, which resists digestion); HIV positive; gastric by‐pass surgery; Clostridium difficile; marijuana user; lupus; rheumatoid arthritis; hepatitis C; post‐traumatic stress disorder; deep vein thrombosis; pancreatitis

  • Planned enrolment: 60

Interventions Intervention group 1
  • Exercise


Intervention group 2
  • Resistant starch


Intervention group 3
  • Exercise and resistant starch


Control group
  • Placebo starch


Time: 16 weeks
Outcomes Planned outcomes
  • High‐sensitivity CRP

  • TNF‐alpha

  • IL‐6

  • IL‐10

  • Monocyte chemoattractant protein‐1

  • Vascular function: pulse wave velocity

  • Microbiome composition

  • BP

Starting date September 2018 (ongoing)
Contact information Samuel A Headley, PhD
Springfield College, USA
Notes Trial registration or a priori published protocol
  • NCT03689569 and Headley 2020 published protocol

  • 9/28/2018, retrospectively registered


Publication type
  • Full‐text article ‐ published protocol


Available data
  • No available data at this stage

IRCT20100223003408N5.

Study name Effect of synbiotic dessert fortified with vitamin D and calcium on nutritional indices, inflammation, oxidative stress, malnutrition and quality of life in hemodialysis patients
Methods Study design
  • Parallel, 2‐arm RCT

  • Patients and investigators blinded

  • Treatment: 8 weeks

  • Recruitment complete

Participants Study characteristics
  • Country: Iran

  • Setting: multicenter (Namazi and Shahid Faghihi hospitals)

  • Inclusion criteria: males and females 20 to 85 years; HD > 6 months; KT/V > 1.2; on special regimens for dialysis patients; not have any changes in their received EPO for a month

  • Exclusion criteria: diabetes; cancer; malignity; autoimmune diseases; sensitivity to dairy products; kidney transplant; vitamin D level > 70

  • Planned enrolment: 42

Interventions Intervention group
  • Fortified dessert: 50 g/day for 8 weeks in addition to conventional medical treatment of HD patients


Control group
  • Ordinary dessert: 50 g/day for 8 weeks in addition to conventional medical treatment of HD patients

Outcomes Planned outcomes
  • Malondialdehyde

  • Malnutrition‐inflammation score

  • Subjective global assessment

  • QoL

  • Vitamin D

  • Total antioxidant capacity

  • high‐sensitivity CRP

  • Complete blood count

  • Sodium

  • Potassium

  • Phosphorous

  • Calcium

  • Albumin

  • Creatinine

  • Urea

  • Ferritin

Starting date Starting date
  • Not reported


Other dates
  • Registration date: 2019‐04‐06, 1398/01/17

  • Registration timing: retrospective

  • Last update: 2019‐04‐06, 1398/01/17

  • Update count: 0

  • Registration Date: 2019‐04‐06, 1398/01/17

Contact information Maryam Ekramzadeh
Shiraz University of Medical Sciences, Nutrition Department
Iran (Islamic Republic of)
+98 917 317 3891
ekramzadeh@sums.ac.ir
Notes IRCT registration number: IRCT20100223003408N5
Status: recruitment complete

IRCT20131013014994N7.

Study name The effect of synbiotic supplementation on plasma levels of Advanced Glycation End Products and cardiovascular risk factors in hemodialysis patients: a double‐blind clinical trial
Methods Study design
  • Parallel, double‐blind, placebo‐controlled RCT

  • Recruitment status: pending

  • Treatment: 12 weeks

Participants Study characteristics
  • Country: Iran

  • Inclusion criteria: males and females, any age; perform HD treatment at least twice/week for a maximum of 4 hours each time for at least 6 months; no pregnancy or breastfeeding; no immune system defects; no history of active cancers; no history of severe chronic diseases and acute medical conditions such as lung disease, CVD, liver disease and acute pancreatitis; no addiction to alcohol or drugs; lack of severe digestive disorders and diseases, HIV disease, mental problems; ability to drink at least 200 mL water/day; life expectancy and survival for at least 3 months; confirmation of written consent

  • Exclusion criteria: severe oedema; had infection 4 weeks ago; took synbiotics, probiotics, prebiotics, or antibiotics during the 4 weeks before the study; candidates for kidney and organ transplantation or PD during 3 months of study; taking immunosuppressive drugs or anticoagulants and chemotherapy drugs; sensitivity to complementary compounds; unwillingness to participate in the study; lack of complete answers to questions; report side effects from taking synbiotics

  • Planned enrolment: 34

Interventions Intervention group 1
  • Synbiotic: 2 capsules/day, after lunch and dinner. Each synbiotic capsule (under the brand name GeriLact; Bio Fermentation Company, Tehran, Iran) contains Lactobacillus rhamnosus, Lactobacillus casei, lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus fermentum, Lactobacillus plantarus and Lactobacillus plantarum at a dose of 109 CFU as a probiotic and 21 mg of fructooligosaccharide as a prebiotic


Control group 2
  • In patients undergoing hemodialysis in the control group, two placebo capsules daily (Bio Fermentation Company, Tehran, Iran) after lunch and dinner (each capsule contains 350 mg of inulin, maltodextrin and all excipients in the synbiotic product except the active ingredient) Will receive. Synbiotic and placebo capsules will be the same size, color, odor and packaging.

Outcomes Planned outcomes
  • Plasma levels of advanced glycation end products

  • Plasma fibrinogen levels

  • Fasting blood sugar and HgA1C

  • GI function (GSRS)

  • Plasma homocysteine

Starting date Date of enrolment: 22 May 2022
Contact information Hadi Abdollahzad
Isaar Square, Faculty of Nutrition Sciences and Food Industry 6719851552 kermanshah Iran (Islamic Republic of)
+98 83 3710 2008
abdollahzad@kums.ac.ir
Kermanshah University of Medical Sciences
Notes Funding: Kermanshah University of Medical Sciences

IRCT20160206026390N11.

Study name Evaluation of the efficacy of probiotic supplementation on bio‐markers of inflammation and oxidative stress in paediatric patients under treatment with haemodialysis: randomized double blind clinical trial
Methods Study design
  • Parallel, 2‐arm, double‐blind, RCT

  • Treatment: not reported

  • Recruitment complete

Participants Study characteristics
  • Country: Iran

  • Setting: single centre (Doctor Sheikh hospital)

  • Inclusion criteria: children on HD; 2 weeks after discontinuing antibiotics, resolving acute inflammatory settings, and discontinuing probiotics

  • Exclusion criteria: chronic inflammatory setting (e.g. GN or nephrotic syndrome); received antibiotic for pulmonary infection or gastroenteritis; candidate for transplantation receiving immunosuppressive drugs, especially steroids; receiving probiotics for any reason

  • Planned enrolment: 20

Interventions Intervention group
  • Probiotic contain following strains: Lactobacillus casei, L. acidophilus, L. rhamnosus, L. bulgaricus, Bifidobacterium breve, Bifidobacterium longum, Streptococcus thermophilus in the count of 109 CFU in 500 mg tablet from Zist Takhmir. Because these Bacteria remain in the intestine and not absorb, nothing uptake during dialysis


Control group
  • Placebo

Outcomes Planned outcomes
  • Total antioxidant capacity

  • Nuclear factor‐kappa B

  • Vascular cell adhesion molecule ‐ 1

Starting date Starting date
  • Not reported


Other dates
  • Registration date: 2019‐06‐10, 1398/03/20

  • Registration timing: retrospective

  • Last update: 2019‐06‐10, 1398/03/20

  • Update count: 0

  • Registration date: 2019‐06‐10, 1398/03/20

Contact information Ehsan Ghaedi
Ahwaz Jundishapoor, University of Medical Sciences
Iran (Islamic Republic of)
+98 61 3374 3285
ghaedi.e@ajums.ac.ir
Notes Funding: Mashhad University of Medical Sciences
IRCT registration number: IRCT20160206026390N11

IRCT20230608058421N1.

Study name The effect of probiotics on patients with end‐stage renal failure
Methods Study design
  • Parallel, double‐blind, placebo‐controlled RCT

  • September 2023 to May 2024

  • Treatment: 3 months

  • Recruitment status: pending

Participants Study characteristics
  • Country: Iran

  • Inclusion criteria: male and females, 18 and 60 years; CKD with albuminuria or creatinine drop; 3 months have passed since the mentioned disorders; patients should be under the same dialysis machines and similar filters

  • Exclusion criteria: history of kidney transplant; any possible cause other than CKD for creatinine drop; other causes of metabolic acidosis such as diarrhea, heart failure or shock; metformin use; taking BP‐lowering medications due to the possibility of acidosis

  • Planned enrolment: 50

Interventions Intervention group
  • Probiotics: advised to take probiotic capsules orally every day after lunch for 3months. The medicine used will be Cap Lactocare 109 CFU (Cap Lactocare)


Control group
  • Placebo: advised to take placebo capsules orally every day after lunch for 3 months

Outcomes Planned outcomes
  • Arterial blood pH

  • Arterial blood HCO3

  • GFR

  • SCr

Starting date Date of 1st enrolment: 23 August 2023
Contact information Melika Gholizadeh
South Kargar 1333635445 Tehran Iran (Islamic Republic of)
+98 21 5541 9005
Melika_g1368@yahoo.com
Shahid Beheshti University of Medical Science
Notes Funding: Shahid Beheshti University of Medical Sciences

KCT0007834.

Study name The effect of carboneceous oral adsorbent and synbiotics on the gut microbiome and muscle health in chronic kidney disease patients
Methods Study design
  • Parallel RCT

  • Treatment duration: not reported

  • Recruitment status: not yet recruiting

Participants Study characteristics
  • Country: South Korea

  • Inclusion criteria: males and females ≥ 20 years; all cause of CKD patients with SCr between 2.0 and 5.0 mg/dL; measured or anticipated change of eGFR < 6 mL/min/1.73 m2; naive to oral carboneceous adsorbent (AST‐120), prebiotics, probiotics, synbiotics, and postbiotics 4 weeks prior to the screening; written informed consents

  • Exclusion criteria: BMI > 30 or < 18.5 kg/m2; HbA1c > 10%; spot urine protein/creatinine ratio > 5,000 mg/g; GI transit disorder or uncontrolled constipation; irritable bowel syndrome, Crohn's disease, or ulcerative colitis; severe malnutrition determined by the investigator; taking immunosuppressants due to kidney transplantation or other diseases; history of radiation therapy to the area including colons or underwent the colectomy; expected to receive dialysis or kidney transplantation within 6 months after the enrolment; received antibiotics treatment 1 month prior to the screening; active infection; malignancies (except for malignancies which did not recur more than 5 years after the completion of treatment or regular follow up without any treatment after complete remission); pregnant or lactating woman, or woman who did not agree with the contraception during the study period; participation in another clinical study; researcher’s decision regarding inappropriateness of enrollment in the study

  • Planned enrolment: 180

Interventions Intervention group 1
  • AST‐120 (Kremezin®, HK inno.N, Corp): 6 g/day ingestion


Intervention group 2
  • AST‐120 (Kremezin®, HK inno.N, Corp): 6 g/day ingestion

  • Synbiotics (Nutine xylobiotics, HK inno.N, Corp): 1 pcak/day


Control group
  • No treatment


Co‐interventions
  • Conventional treatment for dialysis‐independent CKD patients

Outcomes Planned outcomes
  • Change of gut microbiome

  • Gait speed

  • Uraemic toxins in the serum (indoxyl sulfate, p‐cresyl sulfate)

  • Markers of inflammation and oxidative stress (TNF‐ a, IL‐6, myostatin, malondialdehyde)

  • Physical activity

  • QoL

Starting date Planned date of first enrolment: 2 January 2023
Contact information Chun Soo Lim
20, Boramae‐ro 5‐gil, Dongjak‐gu
+82‐2‐870‐2215
cslimjy@snu.ac.kr
Seoul Metropolitan Government Seoul National University Boramae Medical Center
Notes Funding: HK inno.N

Mitrovic 2023.

Study name The impact of synbiotic treatment on the levels of gut‐derived uraemic toxins, inflammation, and gut microbiome of chronic kidney disease patients‐ A randomized trial
Methods Study design
  • Parallel, 2‐arm, double‐blind, placebo‐controlled RCT

  • Treatment: 12 weeks


Time frame
  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Serbia

  • Setting: single‐centre (Hospital dialysis clinic)

  • Inclusion criteria: ≥ 18 years; CKD non‐dialysis, eGFR 15 to 45 mL/min/1.73 m²

  • Exclusion criteria: kidney transplantation; inflammatory bowel disease; previous intestinal resection; radiotherapy or course of antibiotic, probiotic and immunosuppressive therapy 2 weeks prior to and during the study


Baseline characteristics
  • Number: intervention group (22); control group (22)

  • Mean age ± SD (years): intervention group (69 ± 10); control group (69 ± 8)

  • Sex (M/F): 18/26

  • CKD stage: 3 to 5 non‐dialysis

  • Kidney measurements

    • Mean eGFR ± SD (mL/min.1.73 m²): intervention group (26.5 ± 6.5); control group (31.3 ± 7.3)

  • Comorbidities: hypertension (21); DM (10)

  • GI status: not reported

Interventions Intervention group
  • Synbiotic

  • Time: 12 weeks


Control group
  • Placebo

  • Time: 12 weeks

Outcomes Outcomes reported by this study at 12 weeks
  • Total serum indoxyl sulfate

  • p‐cresyl sulfate

  • Trimethylamine N‐oxide

  • stool microbiome

  • IL‐6

  • High sensitivity CRP

  • eGFR

  • Albuminuria

  • Diet

  • GI symptom dynamics

  • Safety

Starting date Unpublished manuscript
  • This study is to be included when a peer‐reviewed journal article is published

Contact information Corresponding author: Miloš Mitrović
Address: Dunavski Kej 13 11000 Belgrade
Telephone: +381637603510
E‐mail: dr.milosh.mitrovic@gmail.com
Notes Publication type
  • Unpublished manuscript

  • This study to be included when a peer reviewed journal article becomes published


Funding
  • "The work of the authors is supported by Serbian Society of Nephrology under grant number 3110/17. We would like to thank Pharma S.d.o.o. for the donation of synbiotic and placebo material"

NCT03770611.

Study name Effect of prebiotics and/or probiotics on uremic toxins and inflammation markers in peritoneal dialysis patients
Methods Study design
  • Parallel, 4‐arm, triple‐blind (patients, investigators, care providers) RCT

  • Treatment: 3 months

  • Recruitment status: unknown

Participants Study characteristics
  • Country: Mexico

  • Setting: single centre

  • Inclusion criteria: males and females 18 to 80 years; APD > 3 months; signed informed consent

  • Exclusion criteria: kidney failure of inflammatory cause (lupus, vasculitis, collagenopathies); intake of probiotics, prebiotics or fibre in the last 3 months; use of anti‐inflammatory drugs or nutritional supplements (immunosuppressants, pentoxifylline, NSAIDs, omega‐3); treated with antibiotics or sevelamer; treated with research drugs or participants in any clinical trial; peritonitis or active infection 2 weeks prior the study; any medical condition affecting intestinal absorption (inflammatory bowel disease, short bowel syndrome, bariatric surgery) or severe dysmotility; severe malnutrition; previous kidney transplantation; serious diseases altering the final outcomes of the study: decompensated heart failure, chronic liver disease, cancer, AIDS

  • Planned enrollment: 112

Interventions Intervention group 1
  • Probiotic: Bacillus coagulans, Bacillus subtilis, Bifidobacterium (B) bifidum, B. breve, B. longum, Lactobacillus (L) acidophilus, L. brevis, L. casei, L. helveticus, L. Paracasei, L plantarum, L. rhamnosus, L. salivarus, Lactococcus lactis, Pediococcus acidilactici, Pediococcus parvulus, Weisella confusa, Weisella paramesenteroides; 2 x 108 CFU probiotic bacteria + prebiotic placebo/day for 3 months


Intervention group 2
  • Prebiotic: agave inulin; 20 g of prebiotic fibre + probiotic placebo/day for 3 months


Intervention group 3
  • Synbiotic: combination of the probiotic product + the prebiotic fibre; 2 x 108 CFU probiotic bacteria + 20 g of prebiotic fibre/day for 3 months


Control group
  • Placebo: combination of probiotic placebo and prebiotic fibre placebo. Placebo will consist of maltodextrin for both cases; probiotic and prebiotic placebo/day for 3 months

Outcomes Planned outcomes
  • Change of uraemic toxins from basal to 1 and 3 months

    • P‐cresyl sulphate and indoxyl sulphate

  • Change of uraemic toxins from basal to 1 and 3 months

    • Serum endotoxin

  • Change in gut microbiota composition from basal to 1 and 3 months

    • Faecal bacterial composition

  • Change in GI symptoms from basal to 1 and 3 months

    • Appetite and frequency and severity of GI symptoms (nausea, vomiting, bloating, diarrhoea, constipation)

  • Change of inflammatory cytokines from basal to 1 and 3 months

    • IL‐6, IL‐10 and TNF‐alpha

  • Change of inflammatory cytokines from basal to 1 and 3 months

    • CRP

Starting date Planned starting date
  • 7 January 2019


Other dates
  • Estimated study completion date: August 2020

  • Estimated primary completion date: December 2019 (final data collection date for primary outcome measure)

Contact information Alfonso Martín Cueto Manzano
Unidad de Investigacion Medica en Enfermedades Renales
+52 (33) 38097269
a_cueto_manzano@hotmail.com
Fabiola Martín‐del‐Campo, MSc
+52 (33) 10711190
fabi_mc@hotmail.com
Notes Funding: Unidad de Investigacion Medica en Enfermedades Renales

NCT03789708.

Study name Effects of gum arabic supplementation in hemodialysis patients
Methods Study design
  • Parallel, 4‐arm, double‐blind RCT

  • Treatment: 4 weeks

  • Recruitment status: unknown

Participants Study characteristics
  • Country: Egypt

  • Setting: single centre

  • Inclusion criteria: males and females ≥ 18 years; on regular HD

  • Exclusion criteria: chronic liver disease; malignant condition; inflammatory bowel disease; history of bowel resection; long‐term antibiotic therapy; pregnancy or lactation; current use of immunosuppressive medication

  • Planned enrolment: 80

Interventions Intervention group 1
  • Gum Arabic (100% Acacia Senegal): 10 g/day for 4 weeks

    • Gum Arabic is provided in the form of easily soluble granules. Participants are asked to dissolve it in water or juice and drink it


Intervention group 2
  • Gum Arabic (100% Acacia Senegal): 20 g/day for 4 weeks


Intervention group 3
  • Gum Arabic (100% Acacia Senegal): 40 g/day for 4 weeks


Control group
  • Placebo: 5 g of maltodextrin/day for 4 weeks

    • Maltodextrin is an easily digested polysaccharide provided in the form of a soluble whitish powder that has no taste or odour. Participants are asked to dissolve it in water or juice and drink it

Outcomes Planned outcomes
  • Highly sensitive CRP at 4 weeks

  • Total antioxidant capacity at 4 weeks

  • Blood urea level at 4 weeks

  • Serum calcium at 4 weeks

  • Serum phosphorus at 4 weeks

  • Serum uric acid at 4 weeks

Starting date Starting date
  • 1 November 2018


Other dates
  • Estimated study completion date: June 1, 2019

  • Estimated primary completion date: March 1, 2019 (final data collection date for primary outcome measure)

Contact information Sarra Elamin, MD
Consultant Nephrologist
University of Khartoum
Notes Funding: University of Khartoum

NCT03924089.

Study name Oral nutritional supplement on nutritional and functional status, and biomarkers in malnourished hemodialysis patients (RENACARE)
Methods Study design
  • Parallel, 3‐arm, double‐blind RCT

  • Recruitment period: 12 months

  • Study duration: 6 months

  • Recruitment status: recruiting

Participants Study characteristics
  • Country: Spain

  • Setting: multicentre (3 sites; Hospital Regional Universitario de Málaga, Hospital San Cecilio, Granada, Hospital Rey Juan Carlos, Móstoles, Madrid)

  • Inclusion criteria:: males and females ≥ 18 years undergoing HD 3 days/week > 6 months; at least one of these caloric malnutrition criteria: a) involuntary weight loss > 5% in 3 months or > 10% in 6 months; b) serum albumin < 3.5 g/dL or prealbumin < 28 mg/dL; c) BMI < 23 kg/m2; d) muscular mass loss > 5% in 3 months or > 10% in 6 months; written informed consent

  • Exclusion criteria: type 1 DM or type 2 DM with HbA1c > 9%; unstable dry weight; limb amputation; significant oedema; active malignancy; hospital admissions in the last 3 months; acute GI disease in the 2 weeks before the inclusion; Gastrectomy, gastroparesis or abnormal gastric emptying; acute heart failure grade IV; severe hepatic insufficiency; alcohol or other drug abuse; enrolled in another research study at inclusion; pregnant no informed consent; received any oral nutritional supplement in the 4 weeks before the inclusion; receiving enteral tube feeding; galactosemia, fructosemia, or requirement of a no fibre diet; allergy or hypersensitivity to any ingredient of the oral nutritional supplement; ongoing treatment with glucocorticoids; received any oral fatty acids omega‐3 supplement in the 4 weeks before the inclusion; received intra‐dialytic parenteral nutrition in the 3 months before the inclusion; received any probiotics or prebiotics (not as part of the diet) in the 3 months before the inclusion; anaemia (Hb < 10 g/dL) or Epoetin resistance

  • Planned enrollment: 120 patients

Interventions Intervention group 1
  • Oral nutritional supplement with probiotics

    • Oral nutritional supplement specifically developed for undernourished HD patients enriched with functional nutrients (extra virgin olive oil, omega‐3 fatty acids, whey protein, antioxidants and carnitine)

    • Probiotics

  • Physical activity recommendations


Intervention group 2
  • Oral nutritional supplement without probiotics

    • Oral nutritional supplement specifically developed for undernourished HD patients enriched with functional nutrients (extra virgin olive oil, omega‐3 fatty acids, whey protein, antioxidants and carnitine)

  • Physical activity recommendations


Control group
  • Dietary recommendations

    • Individualized dietary recommendations

  • Physical activity recommendations

Outcomes Planned outcomes
  • Change in weight

  • Change in fat‐free body mass

  • Change in serum albumin

  • Change in serum prealbumin

  • Change in handgrip strength

  • Change in the score of the "Barthel" test

  • Change in HADS

  • Changes in plasma levels of high‐sensitivity CRP

  • Changes in plasma levels of 8‐iso‐prostaglandin F2 α

Starting date Start date
  • Actual study start date: 15 April 2019


Other dates
  • Estimated primary completion date: October 2020 (final data collection date for primary outcome measure)

Contact information Gabriel Olveira, MD, PhD
Instituto de Investigación biomédica de Málaga
0034951290343
gabrielm.olveira.sspa@juntadeandalucia.es
Notes Funding: Fundación Pública Andaluza para la Investigación de Málaga en Biomedicina y Salud

NCT05183737.

Study name Effects of microencapsulated propolis and turmeric in patients with chronic kidney disease
Methods Study design
  • Parallel, double‐blind, placebo‐controlled RCT

  • Treatment: 12 weeks

  • Recruitment status: active, not recruiting

Participants Study characteristics
  • Country: Brazil

  • Inclusion criteria: stage 5 CKD (GFR < 15 mL/min); HD > 6 months; arteriovenous fistula as vascular access

  • Exclusion criteria: pregnant women; smokers; using antibiotics in the last 3 months; using antioxidant supplements; habitual intake of propolis, curcumin and turmeric; autoimmune and infectious diseases, cancer, liver and AIDS (Acquired Immunodeficiency Syndrome)

  • Planned enrolment: 34

Interventions Intervention group
  • Turmeric and propolis: microcapsules containing 0.250 mg of turmeric 95% curcumin and 0.250 mg of green propolis


Control group
  • Placebo: microcapsules containing arabic gum and cornstarch with the same weight and characteristics as the intervention microcapsules

Outcomes Planned outcomes
  • Bomarkers related to inflammation and antioxidant capacity, such as: erythroid nuclear transcription factor 2 and nuclear factor Kappa B

  • Inflammatory cytokines (IL‐6, TNF‐α)

  • CRP

Starting date Starting date: 7 March 2022
Contact information Denise Mafra, PhD
Universidade Federal Fluminense
Notes Funding: Universidade Federal Fluminense

NCT05336305.

Study name Polydextrose for patients with chronic kidney disease
Methods Study design
  • Parallel RCT

  • Treatment: 2 months

  • Recruitment status: completed

Participants Study characteristics
  • Country: Brazil

  • Inclusion criteria: males and females, 18 to 65 years; HD > 6 months

  • Exclusion criteria: pregnant; smokers; using antibiotics in the last 3 months; autoimmune diseases; clinical diagnosis of infectious diseases; cancer; AIDS

  • Planned enrolment: 40

Interventions Intervention group
  • Polydextrose: 12g/day for 2 months


Control group
  • Placebo: corn starch daily for 2 months

Outcomes Planned outcomes
  • Change constipation status: stool frequency and Bristol Stool Form

  • Change in cytokines plasma levels

Starting date Starting date: 30 March 2021
Contact information Denise Mafra, Universidade Federal Fluminense
Notes Funding: Universidade Federal Fluminense

NCT05359094.

Study name Probiotic supplements in chronic kidney disease
Methods Study design
  • Parallel RCT

  • Treatment: not reported

  • Follow‐up: 24 weeks

  • Recruitment status: enrolling by invitation

Participants Study characteristics
  • Country: China

  • Inclusion criteria: males and females > 20 years; CKD stage 2‐3a; can cooperate with the research plan for 3 visits and retain samples (blood, urine)

  • Exclusion criteria: use of other probiotics during the study; active infectious diseases in the past month; used antibiotics within the past 1 month or during the study; pregnant or breastfeeding; obstructive nephropathy within the past month; polycystic kidney disease; AKI within the past 3 months; GI bleeding within the past 3 months; malignancy; severe CVD in the past 3 months, such as coronary artery disease, myocardial ischemia, NYHA class IV myocardial failure, cerebrovascular disease, or peripheral artery disease

  • Planned enrolment: 80

Interventions Intervention group
  • Probiotics (Pediococcus acidilactici GKA4) supplement with a plant‐dominant low‐protein diet


Control group
  • Placebo supplement with a plant‐dominant low‐protein diet

Outcomes Planned outcomes
  • eGFR

  • BUN

  • Electrolytes

  • Uremic toxins: indoxyl sulfates and trimethylamine N‐oxide levels

  • Sugar test: fasting sugar and HbA1C

  • Nutrition status: albumin, TIBC, TFS and uric acid

  • Inflammation makers: ALT, AST

  • Lipid analysis: total cholesterol, triglyceride, HDL and LDL

  • Complete blood count

  • Urine analysis: bacteria, protein and MCP‐1

  • 24‐hour Dietary recall

  • Safety and compliance: GI symptoms measured by questionnaire

  • Grip strength

Starting date Starting date: 1 June 2022
Contact information Cheng‐Hsu Chen, MDPHD
Division of Nephrology in Taichung Veterans General Hospital
Notes Funding: Taichung Veterans General Hospital

NCT05540431.

Study name Evaluation of protective effect of activated charcoal and probiotic against progression of chronic kidney disease
Methods Study design
  • Parallel, open‐label RCT

  • Treatment: 6 weeks

  • Recruitment status: not yet recruiting

Participants Study design
  • Country: Iraq

  • Inclusion criteria: males and females ≥ 18 years with CKD

  • Exclusion criteria: < 18 years; inability or rejection to take activated charcoal or probiotic; clinically unstable; pregnant; unlikely to adhere to study procedure (eg. due to cognitive limitations, severe psychiatric disorder or alcoholism)

  • Planned enrolment: 60

Interventions Intervention group 1
  • Oral activated charcoal: standard care plus activated charcoal capsule (charconut) 3 timesday for 6 weeks


Intervention group 2
  • Oral probiotic: standard care plus probiotic tablet twice/day for 6 weeks


Control group
  • Standard care for 6 weeks

Outcomes Planned outcomes
  • BUN

  • Indol sulfate

  • SCr

  • Nutritional status: serum albumin

Starting date Planned starting date: 20 September 2023
Contact information Waleed Khaild Rahman Kareem Al‐kabi, Kufa University
Notes Funding: not reported

NCT05674981.

Study name To evaluate the beneficial effect of probiotics on DKD patients and the role of gut microbiota modulation
Methods Study design
  • Parallel, double‐blind RCT

  • Treatment: not reported

  • Follow‐up: 3 and 6 months

  • Recruitment status: recruiting

Participants Study design
  • Country: China

  • Inclusion criteria: 25 and 80 years; type 2 diabetes and stable medication for 3 months; HbA1c before meals between 7% and 10%; stage 1‐3a diabetic nephropathies (eGFR > 45 mL/min); microalbuminuria estimated between 30 to 300 mg/day

  • Exclusion criteria: type I DM; inflammatory bowel disease, liver disease, liver cirrhosis, systemic lupus erythematosus, malignancy, and high BP; hypoglycemic coma, diabetic ketoacidosis, hyperosmolar non‐ketotic diabetic coma, or DM acute complications; acute infection medical history in past 3 month; fasting blood glucose > 13.3 mmol/L; GPT> 100U/L (2.5 times than usual situation); vulnerable population (including breeding or pregnant women, prisoner, aboriginal, disabled population); smoker or alcoholic; taking antibiotics in past 1 month; stably taking probiotics supplements in past 1 months (yogurt or dairy products were excluded); taking immunosuppressive drug, angiotensin‐converting enzyme inhibitors, or angiotensin receptor blockers in past 3 months

  • Planned enrolment: 50

Interventions Intervention group
  • Probiotic: 2‐strain probiotic supplement includes Lactobacillus reuteri ADR‐1 (alive) and Lactobacillus rhamnosus GM‐020 ( alive)


Control group
  • Placebo: Same additives to probiotic group but replace probiotics with corn starch and maltodextrin

Outcomes Planned outcomes
  • Change in cystatin‐C

  • Change in BMI

  • Change in waist and hip circumference

  • Change in BP

  • Change in fasting plasma glucose

  • Change in serum inulin

  • Change in HbA1c

  • Change in homeostatic model assessment for inulin resistance

  • Change in quantitative insulin sensitivity check index

  • Change in glycated albumin

  • Change in creatinine

  • Change in BUN

  • Change in potassium

  • Change in urine protein/albumin

  • Change in urine microalbuminuria

  • Change in urine acid

  • Change in GFR: Cockcroft and Gault formula

  • Change in eGFR

  • Change in blood lipid‐related index: triglycerides, total cholesterol, VLDL, LDL, HDL

  • Change in cytokine index

  • Change in total iron‐binding capacity

  • Change in short‐chain fatty acids

  • Change in trimethylamine N‐oxide

  • Change in International Physical Activity Questionnaire

  • Change in gut microbiota

Starting date Starting date: 24 April 2023
Contact information Yi‐Sun Yang, PhD
Chung Shan Medical University
Notes Funding: GenMont Biotech Incorporation

NCT05724511.

Study name The effect of probiotics on depression syndrome and risk factors of cardiovascular disease in hemodialysis patients
Methods Study design
  • Parallel, open‐label RCT

  • Treatment: 3 months

  • Recruitment status: completed

Participants Study design
  • Country: Taiwan

  • Inclusion criteria: ≥ 20 years undergoing HD(at least 3 months), 3 times/week, and URR ≥ 65%

  • Exclusion criteria: allergic to the components of the intervention drug, low tolerance of milk or dairy products; diagnosis of dementia, delirium, bipolar disorder, schizophrenia, or liver failure; Beck Depression Inventory Chinese version 2.0 score higher than 14, or has significant suicide risk during study; obsessive‐compulsive disorder, schizoid personality disorder, schizotypal personality, Paranoid‐Antisocial Personality Disorder, histrionic personality disorder; addiction to alcohol or drugs, terminal cancer, severe infection, heart failure, central venous catheter in the past 6 months; taking antibiotics, anti‐oxidant vitamin supplements, probiotics, prebiotics, antidepressants, anti‐anxiety medicine, yoghurt and the products in the past 3 months; pregnant or lactating

  • Planned/actual enrolment: 70/55

Interventions Intervention group
  • MIYAIRI 588: 1g/package with meal, and 3 packages/day for 3 months


Control group
  • No intervention

Outcomes Planned outcomes
  • Change in depression score (Beck Depression Inventory Score (Chinese version 2.0))

Starting date Starting date: 27 February 2023
Contact information Pei‐Yu Wu
China Medical University Hospital
Notes Funding: China Medical University Hospital

NCT05835648.

Study name Effect of dietary fiber on non‐dialysis patients with chronic kidney disease
Methods Study design
  • Parallel, open‐label RCT

  • Treatment: 3 months

  • Follow‐up: 9 months

  • Recruitment status: not yet recruiting

Participants Study characteristics
  • Country: China

  • Inclusion criteria: males and females, ≥ 18 years; clinically confirmed as stage 3‐5 CKD, endogenous CrCl < 60mL/min according to Cockcroft‐Gault formula; generally in good condition, have a good understanding of their own illness and physical condition, have self‐awareness, and can communicate well with others; Hb 100 to 130g/L; in a non‐dialysis state; dietary fiber intake < 25 g/day; voluntarily join the study, understand the significance of the test and the indicators to be measured, and sign the informed consent

  • Exclusion criteria: unwilling to join the study; severe infection: fever, cough and sputum, pharyngeal pain, abdominal pain, diarrhea, carbuncle and other skin and soft tissue infection and other clinical manifestations, blood routine white blood cell count beyond the normal range (10×109/L); severe CVD: including chronic cardiac insufficiency grade 3 or above and various arrhythmias; obvious hypoproteinemia: albumin < 30 g/L; corrective measures for anaemia (blood transfusion, EPO, HIF‐PHI, iron supplement) have been applied in the past 3 months; tumour evidence: certain tumours have been detected or clinical manifestations and tumour markers suggest the possibility of tumor; pregnant; recent history of heavy blood loss

  • Planned enrolment: 56

Interventions Intervention group
  • Nutrasumma: on the basis of daily diet, dietary fibre supplement (Nutrasumma) was given once/day, 1 strip each time, before meals for 3 months


Control group
  • No intervention

Outcomes Planned outcomes
  • Change in Hb

  • Change in GFR

Starting date Planned starting date: 30 April 2023
Contact information Zunsong Wang
Qianfoshan Hospital
Notes Funding: not reported

RBR‐3WRNF.

Study name Effect of fibers on intestinal transit and inflammation of patients with pre‐dialytic chronic renal disease
Methods Study design
  • Parallel, 2‐arm, single‐blind, placebo‐controlled RCT


Time frame
  • Treatment: 8 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: Brazil

  • Setting: single centre (Hospital)

  • Inclusion criteria: ≥ 18 years; CKD stage 5 non‐dialysis; without gastroenterological pathologies

  • Exclusion criteria: cancer; use continuous laxatives; pregnant


Baseline characteristics
  • Number: intervention group (31); control group (31)

Interventions Intervention group
  • Prebiotics

  • Time: 8 weeks


Control group
  • Placebo

  • Time: 8 weeks

Outcomes Planned outcomes
  • Change in GI symptoms: Bristol Stool Chart

  • Lipid profile

  • Inflammatory markers

Starting date Unclear
Contact information Marina Nogueira Berbel Bufarah
Address: Rua Paulo Bruder, 412 Residencial Parque Laguna 2City: Botucatu / Brazil Zip code: 18615‐412
Phone: +55‐14‐997402119
Email: mnberbel@fmb.unesp.br
Affiliation: Faculdade de Medicina de Botucatu ‐ Unesp
Notes Trial registration
  • https://ensaiosclinicos.gov.br/rg/RBR‐3wrnrf


Funding
  • Faculdade de Medicina de Botucatu ‐ Unesp

ReSPECKD 2022.

Study name A randomized double‐blind cross‐over trial to study the effects of resistant starch prebiotic in chronic kidney disease (ReSPECKD)
Methods Study design
  • Cross‐over, 2‐arm, double‐blind, placebo‐controlled RCT


Study duration
  • Treatment: 10 weeks

  • Follow‐up: none past treatment

Participants Study characteristics
  • Country: USA

  • Setting: single centre (Hospital)

  • Inclusion criteria: ≥ 18 years; CKD stages 1 to 5 non‐dialysis

  • Exclusion criteria: allergy to study treatment; kidney transplant; antibiotics; bowel diseases; bowel cancer; GI surgery


Baseline characteristics
  • Number: 18

Interventions Intervention group
  • Prebiotic

  • Time: 10 weeks


Control group
  • Placebo

  • Time: 10 weeks

Outcomes Planned outcomes
  • GI symptoms

  • QoL

  • Microbiota composition

  • Indoxyl sulfate

  • P‐cresol sulphate

  • Dietary intake

Starting date September 2021
Contact information Dylan Mackay
Email: dylan.mackay@umanitoba.ca
Address: Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
Notes Publication type
  • A priori published protocol

ACR: albumin to creatinine ratio; AIDS: Acquired Immunodeficiency Syndrome; AKI: acute kidney injury; APD: automated peritoneal dialysis; BMI: body mass index; BP: blood pressure; BUN: blood urea nitrogen; CKD: chronic kidney disease; CRP: C‐reactive protein; CVD: cardiovascular disease; DM: diabetes mellitus; eGFR: estimated glomerular filtration rate; EPO: erythropoietin; GI: gastrointestinal; GN: glomerulonephritis; GSRC: gastrointestinal symptom rating scale; HADS: Hospital Anxiety and Depression Scale; Hb: haemoglobin; HbA1C: haemoglobin A1; HDL: high‐density lipoprotein; IL: interleukin; LDL: low‐density lipoprotein; M/F: males/females; KT/V: dialyser clearance time/volume; MI: myocardial infarction; QoL: quality of life; RCT: randomised controlled trial; SCr: serum creatinine; SD: standard deviation; TNF: tumor‐necrosis factor; VLDL: very low density lipoprotein

Differences between protocol and review

Abstracts were not eligible studies in this review due to not being peer reviewed and a lack of available detail to assess quality.

Contributions of authors

  1. Draft the protocol: TC, RK, JC, CH, MH, DJ, ATP, AT, GW

  2. Study selection: TC, RK

  3. Extract data from studies: TC, RK

  4. Enter data into RevMan: TC, RK

  5. Carry out the analysis: TC, RK, ATP

  6. Interpret the analysis: TC, RK, ATP

  7. Draft the final review: TC, RK, SC, JC, CH, MH, DJ, ATP, AT, GW

  8. Disagreement resolution: SC

  9. Update the review: TC

Sources of support

Internal sources

  • No sources of support provided

External sources

  • BEAT‐CKD Funding Grant 1092957, Australia

    TC and RK are employed under funding from this grant.

Declarations of interest

  • TC: none known

  • RK: none known

  • SC: none known

  • JC: none known

  • Carmel Hawley has received fees paid to her institution from Janssen and GlaxoSmithKline; Advisory Board fees paid to her from Otsuka; Research Grants to her institution from Otsuka, Shire, Fresenius, and Baxter; none of these are related to the current study. In addition, she has received grants paid to her institution from the Polycystic Kidney Disease foundation of Australia and from Otsuka for work that is related to the current study

  • MH: none known

  • David Johnson has received consultancy fees, research grants, speaker's honoraria and travel sponsorships from Baxter Healthcare and Fresenius Medical Care. He has received consultancy fees from AstraZeneca and AWAK, and travel sponsorships from Amgen

  • ATP: none known

  • AT: none known

  • GW: none known

New

References

References to studies included in this review

Abbasi 2017 {published data only}

  1. Abbasi B, Ghiasvand R, Mirlohi M. Kidney function improvement by soy milk containing lactobacillus plantarum A7 in type 2 diabetic patients with nephropathy: a double-blinded randomized controlled trial. Iranian Journal of Kidney Diseases 2017;11(1):36-43. [MEDLINE: ] [PubMed] [Google Scholar]
  2. Abbasi B, Mirlohi M, Daniali M, Ghiasvand R. Effects of probiotic soy milk on lipid panel in type 2 diabetic patients with nephropathy: a double-blind randomized clinical trial. Progress in Nutrition 2018;20:70-8. [EMBASE: 2001649336] [Google Scholar]

Biruete 2017 {published data only}

  1. Biruete A, Cross TL, Allen JM, Kistler BM, Loor H, Evenepoel P, et al. Effect of dietary inulin supplementation on the gut microbiota composition and derived metabolites of individuals undergoing hemodialysis: a pilot study. Journal of Renal Nutrition 2021;31(5):512-22. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Biruete A, Kistler B, Allen J, Bauer L, Fahey G, Swanson K, et al. Effect of inulin supplementation on mineral metabolism and short-chain fatty acid excretion in hemodialysis patients [abstract no: MP434]. Nephrology Dialysis Transplantation 2017;32(Suppl 3):iii588-9. [EMBASE: 617289888] [Google Scholar]

Bliss 1992 {published data only}

  1. Bliss DZ, Stein TP, Schleifer CR, Settle RG. Supplementation with gum arabic fiber increases fecal nitrogen excretion and lowers serum urea nitrogen concentration in chronic renal failure patients consuming a low-protein diet. American Journal of Clinical Nutrition 1996;63(3):392-8. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Bliss DZ. Effect of a gum Arabic supplement on the nitrogen excretion and serum urea nitrogen concentration of chronic renal failure patients on a low protein diet [PhD thesis]. University of Pennsylvania 1992:1-206. [CINAHL: 109870911]

Borges 2018 {published data only}

  1. Borges NA, Carmo FL, Stockler-Pinto MB, Brito JS, Dolenga CJ, Ferreira DC, et al. Probiotic supplementation in chronic kidney disease: a double-blind, randomized, placebo-controlled trial. Journal of Renal Nutrition 2018;28(1):28-36. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Borges NA, Stenvinkel P, Bergman P, Qureshi AR, Lindholm B, Moraes C, et al. Effects of probiotic supplementation on trimethylamine-N-oxide plasma levels in hemodialysis patients: a pilot study. Probiotics & Antimicrobial Proteins 2019;11(2):648-54. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Mafra D, Borges NA, Moraes C, Stockler-Pinto MB, Bergman P, Stenvinkel P. Effects of probiotic supplementation on trimethylamine-N-oxide plasma levels in chronic kidney disease patients [abstract no: TH-PO764]. Journal of the American Society of Nephrology 2016;27(Abstract Suppl):270A. [EMBASE: 641135082] [Google Scholar]
  4. Mafra D, Borges NA, Nakau L, Dolenga C, Bergman P, Stenvinkel P. Effects of probiotic supplementation on uremic toxins levels in non-dialysis CKD patients [abstract no: MP431]. Nephrology Dialysis Transplantation 2017;32(Suppl 3):iii587-8. [EMBASE: 617289825] [Google Scholar]
  5. Mafra D, Borges NA, Stockler-Pinto MB, Fouque D, Barros AF. Does a probiotic supplementation alter the indoxyl sulfate levels in non-dialysis chronic kidney disease patients? A randomized placebo-controlled clinical trial [abstract no: FR-PO862]. Journal of the American Society of Nephrology 2015;26(Abstract Suppl):563A. [EMBASE: 641102246] [Google Scholar]

Cosola 2021 {published data only}

  1. Cosola C, Rocchetti MT, di Bari I, Acquaviva PM, Maranzano V, Corciulo S, et al. An innovative synbiotic formulation decreases free serum indoxyl sulfate, small intestine permeability and ameliorates gastrointestinal symptoms in a randomized pilot trial in stage IIIb-IV CKD patients. Toxins 2021;13(5):334. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

de Andrade 2021 {published data only}

  1. Andrade LS, Sarda FA, Pereira NB, Teixeira RR, Rodrigues SD, Lima JD, et al. Effect of unripe banana flour on gut-derived uremic toxins in individuals undergoing peritoneal dialysis: a randomized, double-blind, placebo-controlled, crossover trial. Nutrients 2021;13(2):646. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

de Araujo 2022 {published data only}

  1. De Araujo EM, Meneses GC, Martins AM, De Francesco Daher E, Da Silva Junior GB. Effect of probiotic supplementation on endothelial injury and inflammation biomarkers among patients with chronic kidney diseases on hemodialysis [abstract no: MO584]. Nephrology Dialysis Transplantation 2022;37(Suppl 3):i429. [DOI: 10.1093/ndt/gfac074.029] [DOI] [Google Scholar]
  2. Araujo EM, Meneses GC, Carioca AA, Martins AM, Daher EF, da Silva Junior GB. Use of probiotics in patients with chronic kidney disease on hemodialysis: a randomized clinical trial. Jornal Brasileiro de Nefrologia 2022;45(2):152-61. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Dehghani 2016 {published data only}

  1. Dehghani H, Heidari F, Mozaffari-Khosravi H, Nouri-Majelan N, Dehghani A. Synbiotic supplementations for azotemia in patients with chronic kidney disease: a randomized controlled trial. Iranian Journal of Kidney Diseases 2016;10(6):351-7. [MEDLINE: ] [PubMed] [Google Scholar]

Ebrahim 2022 {published data only}

  1. Ebrahim Z, Proost S, Tito RY, Raes J, Glorieux G, Moosa MR, et al. The effect of s-glucan prebiotic on kidney function, uremic toxins and gut microbiome in stage 3 to 5 chronic kidney disease (CKD) predialysis participants: a randomized controlled trial. Nutrients 2022;14(4):805. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Eidi 2018 {published data only}

  1. Eidi F, Poor-Reza GF, Ostadrahimi A, Dalili N, Samadian F, Barzegari A. Effect of Lactobacillus rhamnosus on serum uremic toxins (phenol and P-cresol) in hemodialysis patients: A double blind randomized clinical trial. Clinical Nutrition ESPEN 2018;28:158-64. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Gholi FP, Dalili N, Eidi F, Ostadrahimi A. Effect of lactobacillus rhamnosus on serum uremic toxins in hemodialysis patients [abstract no: SP705]. Nephrology Dialysis Transplantation 2017;32(Suppl 3):iii375. [EMBASE: 617290088] [Google Scholar]

Elamin 2017 {published data only}

  1. Elamin S, Alkhawaja MJ, Bukhamsin AY, Idris MA, Abdelrahman MM, Abutaleb NK, et al. Gum Arabic reduces C-reactive protein in chronic kidney disease patients without affecting urea or indoxyl sulfate levels. International Journal of Nephrology 2017;2017:9501470. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Esgalhado 2018 {published data only}

  1. Azevedo R, Esgalhado M, Kemp JA, Regis B, Cardozo LF, Nakao LS, et al. Resistant starch supplementation effects on plasma indole 3-acetic acid and aryl hydrocarbon receptor mRNA expression in hemodialysis patients: Randomized, double blind and controlled clinical trial [Efeitos da suplementacao de amido resistente no acido indol-3-acetico plasmatico e na expressao do mRNA do receptor aril-hidrocarboneto em pacientesemhemodialise:ensaioclinicorandomizado,duplo-cegoecontrolado]. Jornal Brasileiro de Nefrologia 2020;42(3):273-9. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Esgalhado M, Kemp JA, Azevedo R, Paiva BR, Stockler-Pinto MB, Dolenga CJ, et al. Could resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food & Function 2018;9(12):6508-16. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Esgalhado M, Kemp JA, Paiva BR, Brito JS, Cardozo LF, Azevedo R, et al. Resistant starch type-2 enriched cookies modulate uremic toxins and inflammation in hemodialysis patients: a randomized, double-blind, crossover and placebo-controlled trial. Food & Function 2020;11(3):2617-25. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  4. Kemp JA, Dos Santos HF, Jesus HE, Esgalhado M, Paiva BR, Azevedo R, et al. Resistant starch type-2 supplementation does not decrease trimethylamine n-oxide (TMAO) plasma level in hemodialysis patients. Journal of the American Nutrition Association 2022;41(8):788-95. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  5. Kemp JA, Regis de Paiva B, Fragoso Dos Santos H, Emiliano de Jesus H, Craven H, Z Ijaz U, et al. The impact of enriched resistant starch type-2 cookies on the gut microbiome in hemodialysis patients: a randomized controlled trial. Molecular Nutrition & Food Research 2021;65(19):e2100374. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  6. Mafra D, Ann Kemp J, Esgalhado M, Paiva B, De Jesus HE, Santos HF. Effects of resistant starch (RS) type 2 cookies on gut microbiota profile in hemodialysis (HD) patients [abstract no: PO2051]. Journal of the American Society of Nephrology 2020;31(Abstract Suppl):629-30. [EMBASE: 633701738] [Google Scholar]
  7. Mafra D, Ann Kemp J, Santos HF, De Jesus HE, Esgalhado M, Paiva B, et al. High-amylose resistant starch (RS) cookies supplementation does not decrease trimethylamine n-oxide (TMAO) plasma level in hemodialysis (HD) patients [abstract no: PO2039]. Journal of the American Society of Nephrology 2020;31(Abstract Suppl):626-7. [EMBASE: 633701431] [Google Scholar]
  8. Mafra D, Esgalhado M, Macedo RD, Kemp J, Paiva B, Nakao LS, et al. Resistant starch supplementation reduces indoxyl sulfate levels in hemodialysis patients: a randomized, double-blind, crossover, placebo-controlled study [abstract no: FR-OR127]. Journal of the American Society of Nephrology 2018;29(Abstract Suppl):72. [EMBASE: 633737357] [Google Scholar]
  9. Mafra D, Esgalhado M, Stockler-Pinto MB, Borges NA, Cardozo LF, Paiva B, et al. Effects of resistant starch supplementation on inflammatory and oxidative stress status in hemodialysis patients: a pilot randomized, double-blind, placebo-controlled clinical trial [abstract no: SA-PO149]. Journal of the American Society of Nephrology 2017;28(Abstract Suppl):716. [EMBASE: 633701761] [Google Scholar]
  10. Mafra D, Macedo RD, Cardozo LF, Borges NA, Nakao LS, Jardim MZ, et al. Effect of resistant starch supplementation on the indole-3-acetic acid levels and aryl hydrocarbon receptor expression in hemodialysis patients [abstract no: FR-PO545]. Journal of the American Society of Nephrology 2018;29(Abstract Suppl):561. [EMBASE: 633736002] [Google Scholar]
  11. Mafra D, Paiva B, Esgalhado M, Borges NA, Kemp JA, Cardozo LF, et al. Resistant starch supplementation attenuates inflammation in hemodialysis patients [abstract no: SA-PO818]. Journal of the American Society of Nephrology 2019;30(Abstract Suppl):974. [EMBASE: 633769632] [Google Scholar]
  12. Paiva BR, Esgalhado M, Borges NA, Kemp JA, Alves G, Leite PE, et al. Resistant starch supplementation attenuates inflammation in hemodialysis patients: a pilot study. International Urology & Nephrology 2020;52(3):549-55. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Guida 2014 {published data only}

  1. Guida B, Germano R, Trio R, Russo D, Memoli B, Grumetto L, et al. Effect of short-term synbiotic treatment on plasma p-cresol levels in patients with chronic renal failure: a randomized clinical trial. Nutrition Metabolism & Cardiovascular Diseases 2014;24(9):1043-9. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Guida 2017 {published data only}

  1. Guida B, Cataldi M, Memoli A, Trio R, di Maro M, Grumetto L, et al. Effect of a short-course treatment with synbiotics on plasma p-cresol concentration in kidney transplant recipients. Journal of the American College of Nutrition 2017;36(7):586-91. [PMID: ] [DOI] [PubMed] [Google Scholar]

Haghighat 2019 {published data only}

  1. Haghighat N, Mohammadshahi M, Shayanpour S, Haghighizadeh MH, Rahmdel S, Rajaei M. The effect of synbiotic and probiotic supplementation on mental health parameters in patients undergoing hemodialysis: A double-blind, randomized, placebo-controlled trial. Indian Journal of Nephrology 2021;31(2):149-56. [EMBASE: 634933481] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Haghighat N, Mohammadshahi M, Shayanpour S, Haghighizadeh MH. Effect of synbiotic and probiotic supplementation on serum levels of endothelial cell adhesion molecules in hemodialysis patients: a randomized control study. Probiotics & Antimicrobial Proteins 2019;11(4):1210-8. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Haghighat N, Mohammadshahi M, Shayanpour S, Haghighizadeh MH. Effects of synbiotics and probiotics supplementation on serum levels of endotoxin, heat shock protein 70 antibodies and inflammatory markers in hemodialysis patients: a randomized double-blinded controlled trial. Probiotics & Antimicrobial Proteins 2020;12(1):144-51. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  4. Haghighat N, Rajabi S, Mohammadshahi M. Effect of synbiotic and probiotic supplementation on serum brain-derived neurotrophic factor level, depression and anxiety symptoms in hemodialysis patients: a randomized, double-blinded, clinical trial. Nutritional Neuroscience 2019;24(6):490-9. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  5. Haghighat N, Rajabi S, Mohammadshahi M. Effect of synbiotic and probiotic supplementation on serum brain-derived neurotrophic factor level, depression and anxiety symptoms in hemodialysis patients: a randomized, double-blinded, clinical trial. Nutritional Neuroscience 2021;24(6):490-9. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Haghighat 2019 (second comparison) {published data only}

He 2022 {published data only}

  1. He S, Xiong Q, Tian C, Li L, Zhao J, Lin X, et al. Inulin-type prebiotics reduce serum uric acid levels via gut microbiota modulation: a randomized, controlled crossover trial in peritoneal dialysis patients. European Journal of Nutrition 2022;61(2):665-77. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Khosroshahi 2018 {published data only}

  1. Khosroshahi HT, Abedi B, Ghojazadeh M, Samadi A, Jouyban A. Effects of fermentable high fiber diet supplementation on gut derived and conventional nitrogenous product in patients on maintenance hemodialysis: a randomized controlled trial. Nutrition & Metabolism 2019;16:18. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Laffin MR, Tayebi Khosroshahi H, Park H, Laffin LJ, Madsen K, Kafil HS, et al. Amylose resistant starch (HAM-RS2) supplementation increases the proportion of Faecalibacterium bacteria in end-stage renal disease patients: microbial analysis from a randomized placebo-controlled trial. Hemodialysis International 2019;23(3):343-7. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Tayebi Khosroshahi H, Vaziri ND, Abedi B, Asl BH, Ghojazadeh M, Jing W, et al. Effect of high amylose resistant starch (HAM-RS2) supplementation on biomarkers of inflammation and oxidative stress in hemodialysis patients: a randomized clinical trial. Hemodialysis International 2018;22(4):492-500. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Kooshki 2019 {published data only}

  1. Kooshki A, Tofighiyan T, Miri M. A synbiotic supplement for inflammation and oxidative stress and lipid abnormalities in hemodialysis patients. Hemodialysis International 2019;23(2):254-60. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Li 2020 {published data only}

  1. Li L, Xiong Q, Zhao J, Lin X, He S, Wu N, et al. Inulin-type fructan intervention restricts the increase in gut microbiome-generated indole in patients with peritoneal dialysis: a randomized crossover study [Erratum in: Am J Clin Nutr. 2022 Jun 7;115(6):1659]. American Journal of Clinical Nutrition 2020;111(5):1087-99. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Xiong Q, Li L, Xiao Y, He S, Zhao J, Lin X, et al. The effect of inulin-type fructans on plasma trimethylamine n-oxide levels in peritoneal dialysis patients: a randomized crossover trial. Molecular Nutrition & Food Research 2023;67(9):e2200531. [DOI: 10.1002/mnfr.202200531] [DOI] [PubMed] [Google Scholar]

Lim 2021 {published data only}

  1. Lim PS, Wang HF, Lee MC, Chiu LS, Wu MY, Chang WC, et al. The efficacy of lactobacillus-containing probiotic supplementation in hemodialysis patients: a randomized, double-blind, placebo-controlled trial. Journal of Renal Nutrition 2021;31(2):189-98. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Liu 2020 {published data only}

  1. Liu S, Liu H, Chen L, Liang SS, Shi K, Meng W, et al. Effect of probiotics on the intestinal microbiota of hemodialysis patients: a randomized trial. European Journal of Nutrition 2020;59(8):3755-66. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Lopes 2018 {published data only}

  1. Lopes RC, Theodoro JM, da Silva BP, Queiroz VA, Castro Moreira ME, Mantovani HC, et al. Synbiotic meal decreases uremic toxins in hemodialysis individuals: a placebo-controlled trial. Food Research International 2019;116:241-8. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Lopes RC, Lima SL, da Silva BP, Toledo RC, Moreira ME, Anunciacao PC, et al. Evaluation of the health benefits of consumption of extruded tannin sorghum with unfermented probiotic milk in individuals with chronic kidney disease. Food Research International 2018;107:629-38. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Lydia 2022 {published data only}

  1. Lydia A, Indra TA, Rizka A, Abdullah M. The effects of synbiotics on indoxyl sulphate level, constipation, and quality of life associated with constipation in chronic haemodialysis patients: a randomized controlled trial. BMC Nephrology 2022;23(1):259. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Mafi 2018 {published data only}

  1. Mafi A, Namazi G, Soleimani A, Bahmani F, Aghadavod E, Asemi Z. Metabolic and genetic response to probiotics supplementation in patients with diabetic nephropathy: a randomized, double-blind, placebo-controlled trial [Expression of Concern in: Food Funct. 2022 Apr 4;13(7):4229]. Food & Function 2018;9(9):4763-70. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Mazruei Arani 2019 {published data only}

  1. Mazruei Arani N, Emam-Djomeh Z, Tavakolipour H, Sharafati-Chaleshtori R, Soleimani A, Asemi Z. The effects of probiotic honey consumption on metabolic status in patients with diabetic nephropathy: a randomized, double-blind, controlled trial. Probiotics & Antimicrobial Proteins 2019;11(4):1195-201. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Meng 2019 {published data only}

  1. Meng Y, Bai H, Yu Q, Yan J, Zhao L, Wang S, et al. High-resistant starch, low-protein flour intervention on patients with early type 2 diabetic nephropathy: a randomized trial. Journal of Renal Nutrition 2019;29(5):386-93. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Miraghajani 2017 {published data only}

  1. Miraghajani M, Zaghian N, Dehkohneh A, Mirlohi M, Ghiasvand R. Probiotic soy milk consumption and renal function among type 2 diabetic patients with nephropathy: a randomized controlled clinical trial. Probiotics & Antimicrobial Proteins 2019;11(1):124-32. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Miraghajani M, Zaghian N, Mirlohi M, Feizi A, Ghiasvand R. Impact of probiotic soy milk consumption on oxidative stress among type 2 diabetic kidney disease patients: a randomized controlled clinical trial. Journal of Renal Nutrition 2017;27(5):317-24. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Miranda Alatriste 2014 {published data only}

  1. Miranda Alatriste PV, Urbina AR, Gomez Espinosa CO, Espinosa Cuevas ML. Effect of probiotics on human blood urea levels in patients with chronic renal failure. Nutricion Hospitalaria 2014;29(3):582-90. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Mirzaeian 2020 {published data only}

  1. Mirzaeian S, Saraf-Bank S, Entezari MH, Hekmatdoost A, Feizi A, Atapour A. Effects of synbiotic supplementation on microbiota-derived protein-bound uremic toxins, systemic inflammation, and biochemical parameters in patients on hemodialysis: A double-blind, placebo-controlled, randomized clinical trial. Nutrition 2020;73:110713. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Natarajan 2014 {published data only}

  1. Natarajan R, Pechenyak B, Vyas U, Ranganathan P, Weinberg A, Liang P, et al. Randomized controlled trial of strain-specific probiotic formulation (Renadyl) in dialysis patients. BioMed Research International 2014;2014:568571. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Pan 2021 {published data only}

  1. Pan Y, Yang L, Dai B, Lin B, Lin S, Lin E. Effects of probiotics on malnutrition and health-related quality of life in patients undergoing peritoneal dialysis: a randomized controlled trial. Journal of Renal Nutrition 2021;31(2):199-205. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Poesen 2016 {published data only}

  1. Poesen R, Evenepoel P, Loor H, Delcour JA, Courtin CM, Kuypers D, et al. The influence of prebiotic arabinoxylan oligosaccharides on microbiota derived uremic retention solutes in patients with chronic kidney disease: a randomized controlled trial. PLoS ONE {Electronic Resource] 2016;11(4):e0153893. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

PREBIOTIC 2022 {published data only}

  1. Chan S, Hawley CM, Pascoe EM, Cao C, Campbell KL, Campbell SB, et al. PREBIOTIC: a study protocol of a randomised controlled trial to assess prebiotic supplementation in kidney transplant recipients for preventing infections and gastrointestinal upset — a feasibility study. Pilot & Feasibility Studies 2023;9(1):11. [DOI: 10.1186/s40814-023-01236-y] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Chan S, Hawley CM, Pascoe EM, Cao C, Campbell SB, Campbell KL, et al. Prebiotic supplementation in kidney transplant recipients for preventing infections and gastrointestinal upset: a randomized controlled feasibility study. Journal of Renal Nutrition 2022;32(6):718-25. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Chan S, Hawley CM, Pascoe EM, Cao CZ, Campbell SB, Campbell KL, et al. Prebiotic supplementation in kidney transplant recipients for preventing infections and gastrointestinal upset: a randomized controlled feasibility study. American Journal of Transplantation 2022;22:783. [DOI: 10.1111/ajt.17073] [DOI] [PubMed] [Google Scholar]

ProbiotiCKD 2019 {published data only}

  1. Simeoni M, Cianfrone P, Capria M, Deodato F, Citraro M, Cerantonio A, et al. ProbiotiCKD: A monocentric, randomized, openlabel, placebo-controlled study to reveal and correct gut dysbiosis in early CKD stages [abstract no: SP367]. Nephrology Dialysis Transplantation 2018;33(Suppl 1):i469-70. [EMBASE: 622605912] [Google Scholar]
  2. Simeoni M, Citraro ML, Cerantonio A, Deodato F, Provenzano M, Cianfrone P, et al. An open-label, randomized, placebo-controlled study on the effectiveness of a novel probiotics administration protocol (ProbiotiCKD) in patients with mild renal insufficiency (stage 3a of CKD) [Erratum in: Eur J Nutr. 2018 58(5):2157]. European Journal of Nutrition 2019;58(5):2145-56. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Simeoni M, Citraro ML, Cerantonio A, Deodato F, Provenzano M, Cianfrone P, et al. Correction to: An open-label, randomized, placebo-controlled study on the effectiveness of a novel probiotics administration protocol (ProbiotiCKD) in patients with mild renal insufficiency (stage 3a of CKD) (European Journal of Nutrition, (2019), 58, 5, (2145-2156), 10.1007/s00394-018-1785-z). European Journal of Nutrition 2019;58(5):2157. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Ramos 2019 {published data only}

  1. Armani R, Ishikawa C, Hong V, Bortolotto LA, Cassiolatto JL, Klassen A, et al. Effect of frutooligosaccharide on endothelial function in CKD patients: a randomized controlled trial [abstract no: SAT-206]. Kidney International Reports 2019;4(7 Suppl):S93. [EMBASE: 2002179674] [Google Scholar]
  2. Armani RG, Carvalho AB, Ramos CI, Hong V, Bortolotto LA, Cassiolato JL, et al. Effect of fructooligosaccharide on endothelial function in CKD patients: a randomized controlled trial. Nephrology Dialysis Transplantation 2021;37(1):85-91. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  3. Armani RG, Ramos CI, Cuppari L, Canziani ME. Effect of fructooligosaccharide on endothelium function in CKD patients: a randomized controlled trial [abstract no: TH-PO1138]. Journal of the American Society of Nephrology 2018;29(Abstract Suppl):417. [EMBASE: 633736090] [Google Scholar]
  4. Ramos CI, Armani RG, Canziani MEF, Dalboni MA, Dolenga CJR, Nakao LS, et al. Effect of prebiotic (fructooligosaccharide) on uremic toxins of chronic kidney disease patients: a randomized controlled trial. Nephrology Dialysis Transplantation 2019;34(11):1876-84. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  5. Ramos CI, Armani RG, Nakao LS, Canziani MEF, Campbell KL, Cuppari L. Effect of fructooligosaccharide on microbiota-derived uremic toxins in predialysis patients: a randomized controlled trial [abstract no: SA-PO161]. Journal of the American Society of Nephrology 2017;28(Abstract Suppl):720. [EMBASE: 633702127] [Google Scholar]

Ranganathan 2009 {published data only}

  1. Ranganathan N, Friedman EA, Tam P, Rao V, Ranganathan P, Dheer R. Probiotic dietary supplementation in patients with stage 3 and 4 chronic kidney disease: a 6-month pilot scale trial in Canada. Current Medical Research & Opinion 2009;25(8):1919-30. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]
  2. Ranganathan N, Friedman EA, Tam PY, Rao V, Ranganathan PL. Dietary supplementation with a probiotic formulation (Kibow biotics) on CKD III and IV patients- a short term pilot scale study in Canada [abstract no: SA-PO2755]. Journal of the American Society of Nephrology 2009;20(Abstract Suppl):741A. [Google Scholar]
  3. Ranganathan N, Ranganathan P, Friedman EA, Joseph A, Delano B, Goldfarb DS, et al. Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease. Advances in Therapy 2010;27(9):634-47. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Shariaty 2017 {published data only}

  1. Shariaty Z, Mahmoodi Shan GR, Farajollahi M, Amerian M, Behnam PN. The effects of probiotic supplement on hemoglobin in chronic renal failure patients under hemodialysis: a randomized clinical trial. Journal of Research in Medical Sciences 2017;22:74. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Sirich 2014 {published data only}

  1. Sirich TL, Plummer N, Hostetter TH, Meyer T. Increasing dietary fiber reduces plasma levels of colon-derived uremic solutes in hemodialysis patients [abstract no: FR-PO793]. Journal of the American Society of Nephrology 2013;24(Abstract Suppl):545A. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Sirich TL, Plummer NS, Gardner CD, Hostetter TH, Meyer TW. Effect of increasing dietary fiber on plasma levels of colon-derived solutes in hemodialysis patients. Clinical Journal of the American Society of Nephrology: CJASN 2014;9(9):1603-10. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Soleimani 2017 {published data only}

  1. Soleimani A, Zarrati Mojarrad M, Bahmani F, Taghizadeh M, Ramezani M, Tajabadi-Ebrahimi M, et al. Probiotic supplementation in diabetic hemodialysis patients has beneficial metabolic effects. Kidney International 2017;91(2):435-42. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Soleimani 2019 {published data only}

  1. Soleimani A, Motamedzadeh A, Zarrati Mojarrad M, Bahmani F, Amirani E, Ostadmohammadi V, et al. The effects of synbiotic supplementation on metabolic status in diabetic patients undergoing hemodialysis: a randomized, double-blinded, placebo-controlled trial. Probiotics & Antimicrobial Proteins 2019;11(4):1248-56. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

SYNERGY 2014 {published data only}

  1. Rossi M, Johnson D, Pascoe E, Coombes J, Forbes J, Szeto CC, et al. Synbiotic therapy in pre-dialysis: a randomised controlled trial [abstract no: SAO029]. Nephrology Dialysis Transplantation 2015;30(Suppl 3):iii38. [EMBASE: 72206363] [Google Scholar]
  2. Rossi M, Johnson DW, Morrison M, Pascoe E, Coombes JS, Forbes JM, et al. SYNbiotics Easing Renal failure by improving Gut microbiologY (SYNERGY): a protocol of placebo-controlled randomised cross-over trial. BMC Nephrology 2014;15:106. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Rossi M, Johnson DW, Morrison M, Pascoe EM, Coombes JS, Forbes JM, et al. Synbiotics easing renal failure by improving gut microbiology (SYNERGY): a randomized trial. Clinical Journal of the American Society of Nephrology: CJASN 2016;11(2):223-31. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Rossi M, Johnson DW, Xu H, Carrero JJ, Pascoe E, French C, et al. Dietary protein-fiber ratio associates with circulating levels of indoxyl sulfate and p-cresyl sulfate in chronic kidney disease patients. Nutrition Metabolism & Cardiovascular Diseases 2015;25(9):860-5. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

SYNERGY II 2021 {published data only}ACTRN12617000324314

  1. McFarlane C, Krishnasamy R, Stanton T, Savill E, Snelson M, Mihala G, et al. Synbiotics easing renal failure by improving gut microbiology II (SYNERGY II): a feasibility randomized controlled trial. Nutrients 2021;13(12):4481. [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Viramontes‐Horner 2015 {published data only}

  1. Viramontes-Horner D, Marquez-Sandoval F, Martin-del-Campo F, Vizmanos-Lamotte B, Sandoval-Rodriguez A, Armendariz-Borunda J, et al. Effect of a symbiotic gel (Lactobacillus acidophilus + Bifidobacterium lactis + inulin) on presence and severity of gastrointestinal symptoms in hemodialysis patients. Journal of Renal Nutrition 2015;25(3):284-91. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Wang 2015a {published data only}

  1. Wang IK, Wu YY, Yang YF, Ting IW, Lin CC, Yen TH, et al. The effect of probiotics on serum levels of cytokine and endotoxin in peritoneal dialysis patients: a randomised, double-blind, placebo-controlled trial. Beneficial Microbes 2015;6(4):423-30. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Xie 2015a {published data only}

  1. Xie LM, Ge YY, Huang X, Zhang Y, Li JX. Effects of fermentable dietary fiber supplementation on oxidative and inflammatory status in hemodialysis patients. International Journal of Clinical & Experimental Medicine 2015;8(1):1363-9. [EMBASE: 602272251] [PMC free article] [PubMed] [Google Scholar]

References to studies excluded from this review

Afaghi 2016 {published data only}

  1. Afaghi E, Tayebi A, Ebadi A, Sobhani V, Einollahi B, Tayebi M. The effect of BCAA and ISO-WHEY oral nutritional supplements on dialysis adequacy. Nephrourology Monthly 2016;8(6):e34993. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Cruz‐Mora 2014 {published data only}

  1. Cruz-Mora J, Martinez-Hernandez NE, Martin del Campo-Lopez F, Viramontes-Horner D, Vizmanos-Lamotte B, Munoz-Valle JF, et al. Effects of a symbiotic on gut microbiota in Mexican patients with end-stage renal disease. Journal of Renal Nutrition 2014;24(5):330-5. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Eguchi 2011 {published data only}

  1. Eguchi S, Takatsuki M, Hidaka M, Soyama A, Ichikawa T, Kanematsu T. Perioperative synbiotic treatment to prevent infectious complications in patients after elective living donor liver transplantation: a prospective randomized study. American Journal of Surgery 2011;201(4):498-502. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Ferrara 2009 {published data only}

  1. Ferrara P, Romaniello L, Vitelli O, Gatto A, Serva M, Cataldi L. Cranberry juice for the prevention of recurrent urinary tract infections: a randomized controlled trial in children. Scandinavian Journal of Urology & Nephrology 2009;43(5):369-72. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Firouzi 2015a {published data only}

  1. Firouzi S, Mohd-Yusof BN, Majid HA, Ismail A, Kamaruddin NA. Effect of microbial cell preparation on renal profile and liver function among type 2 diabetics: a randomized controlled trial. BMC Complementary & Alternative Medicine 2015;15:433. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Fitschen 2015 {published data only}

  1. Fitschen PJ, Kistler B, Wu PT, Chung HR, Jeong JH, Phillips S, et al. Effects of intradialytic whey protein supplementation on body composition in non-malnourished hemodialysis patients [abstract no: SA-PO543]. Journal of the American Society of Nephrology 2012;23(Abstract Suppl):763A. [Google Scholar]
  2. Tomayko E, Wu PT, Chung HR, Fitschen P, Kistler B, Yudell B, et al. Intradialytic protein supplementation attenuates dialysis-associated inflammation and reduces co-morbid disease risk [abstract no: SA-OR401]. Journal of the American Society of Nephrology 2011;22(Abstract Suppl):96A. [Google Scholar]
  3. Tomayko E, Yudell B, Jeanes E, Kistler B, Fitschen P, Jeong JH, et al. Intradialytic protein supplementation improves co-morbid disease risk in hemodialysis patients [abstract]. FASEB Journal 2012;26(Suppl 1):na. [EMBASE: 70854879] [Google Scholar]
  4. Tomayko EJ, Kistler BM, Fitschen PJ, Wilund KR. Intradialytic protein supplementation reduces inflammation and improves physical function in maintenance hemodialysis patients. Journal of Renal Nutrition 2015;25(3):276-83. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Fortes 2020 {published data only}

  1. Fortes PM, Teles Filho RV, Azevedo LH, Queiroz VC, da Costa PS. Inflammatory cytokines and lipid profile in children and adolescents with nephrotic syndrome receiving L. Plantarum: a randomized, controlled feasibility trial. Revista Da Associacao Medica Brasileira 2020;66(11):1487-92. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Grat 2017 {published data only}

  1. Grat M, Krawczyk M, Wronka K, Lewandowski Z, Grat K, Krasnodebski M, et al. Impact of pre-transplant use of probiotics on allograft function after liver transplantation: post-hoc analysis of a randomized controlled trial [abstract]. HPB 2018;20(Suppl 2):S798. [EMBASE: 2001142904] [Google Scholar]
  2. Grat M, Wronka KM, Lewandowski Z, Grat K, Krasnodebski M, Stypulkowski J, et al. Effects of continuous use of probiotics before liver transplantation: a randomized, double-blind, placebo-controlled trial. Clinical Nutrition 2017;36(6):1530-9. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Hassan 2017 {published data only}

  1. Hassan K, Armaly Z, Hassan D, Khayr M, Rubinchik I, Khazim K, et al. Does whey protein supplementation affect systemic inflammation in hypoalbuminemic peritoneal dialysis patients? [abstract no: SP496]. Nephrology Dialysis Transplantation 2017;32(Suppl 3):iii295-6. [EMBASE: 617291379] [Google Scholar]

Jiang 2021a {published data only}

  1. Jiang H, Zhang Y, Xu D, Wang Q. Probiotics ameliorates glycemic control of patients with diabetic nephropathy: a randomized clinical study. Journal of Clinical Laboratory Analysis 2021;35(4):e23650. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Liu S, Jiang H. Probiotic dietary supplementation in hemodialysis patients: a double-blind,randomized, placebo-controlled trial [abstract no: FR-OR128]. Journal of the American Society of Nephrology 2018;29(Abstract Suppl):73. [EMBASE: 633737360] [Google Scholar]

Lin 2021 {published data only}

  1. Lin W, Jiang C, Yu H, Wang L, Li J, Liu X, et al. The effects of fushen granule on the composition and function of the gut microbiota during peritoneal dialysis-related peritonitis. Phytomedicine 2021;86:153561. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Orr 2016 {published data only}

  1. Orr DW, Myint H, Murphy R. Probiotic supplementation after very low calorie diet does not aid improvement of the metabolic syndrome or maintenance of weight loss post liver transplant. A randomised double-blind placebo controlled trial [abstract no: 214]. Hepatology 2016;64(1 Suppl 1):113-4A. [EMBASE: 612594308] [Google Scholar]

Pavan 2016 {published data only}

  1. Pavan M. Influence of prebiotic and probiotic supplementation on the progression of chronic kidney disease. Minerva Urologica e Nefrologica 2016;68(2):222-6. [MEDLINE: ] [PubMed] [Google Scholar]

PrePro 2018 {published data only}

  1. Mallick S, Kathirvel M, Thillai M, Sethi P, Durairaj MS, Nair K, et al. PrePro Trial: randomized double-blind placebo controlled trial to analyze the effect of synbiotics on infectious complications following living donor liver transplantion [CTRI no. - CTRI/2017/09/009869]. HPB 2018;20(Suppl 2):S283-4. [EMBASE: 2000962392] [Google Scholar]
  2. Mallick S, Mathew JS, Binoj S, Unnikrishnan G, Menon RN, Dinesh B, et al. PrePro trial: a randomized double blind placebo controlled trial to analyze the effect of synbiotics on infectious complications following living donor liver transplant (LDLT). [CTRI/2017/09/009869]. Transplantation 2018;102(5 Suppl 1):63. [EMBASE: 622538666] [Google Scholar]

ProLow CKD 2022 {published data only}

  1. De Mauri A, Carrera D, Bagnati M, Rolla R, Vidali M, Chiarinotti D, et al. Probiotics-supplemented low-protein diet for microbiota modulation in patients with advanced chronic kidney disease (ProLowCKD): results from a placebo-controlled randomized trial. Nutrients 2022;14(8):1637. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Rayes 2002 {published data only}

  1. Rayes N, Seehofer D, Hansen S, Boucsein K, Muller AR, Serke S, et al. Early enteral supply of lactobacillus and fiber versus selective bowel decontamination: a controlled trial in liver transplant recipients. Transplantation 2002;74(1):123-7. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Rayes 2002a {published data only}

  1. Rayes N, Seehofer D, Muller AR, Hansen S, Bengmark S, Neuhaus P. Influence of probiotics and fibre on the incidence of bacterial infections following major abdominal surgery - results of a prospective trial [Einfluss von Probiotika und Ballaststoffen auf die Inzidenz bakterieller Infektionen nach viszeralchirurgischen Eingriffen - Ergebnisse einer prospektiven Studie]. Zeitschrift fur Gastroenterologie 2002;40(10):869-76. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Rayes 2005 {published data only}

  1. Rayes N, Seehofer D, Theruvath T, Schiller RA, Langrehr JM, Jonas S, et al. Supply of pre- and probiotics reduces bacterial infection rates after liver transplantation--a randomized, double-blind trial. American Journal of Transplantation 2005;5(1):125-30. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Saxena 2022 {published data only}

  1. Saxena A, Jacob CK, Sreenivasa S, Veerappan I, Mahaldar AR, Gupta A, et al. A prospective, double-blind, randomized, placebo-controlled interventional study to evaluate the safety and efficacy of enzobiotics in pre-dialysis CKD patients [abstract no: PO2628]. Journal of the American Society of Nephrology 2020;31(Abstract Suppl):B5. [EMBASE: 633696629] [Google Scholar]
  2. Saxena A, Srinivasa S, Veerappan I, Jacob C, Mahaldar A, Gupta A, et al. Enzobiotics-a novel therapy for the elimination of uremic toxins in patients with CKD (EETOX Study): a multicenter double-blind randomized controlled trial. Nutrients 2022;14(18):3804. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Tayebi‐Khosroshahi 2016 {published data only}

  1. Tayebi-Khosroshahi H, Habibzadeh A, Niknafs B, Ghotaslou R, Yeganeh Sefidan F, Ghojazadeh M, et al. The effect of lactulose supplementation on fecal microflora of patients with chronic kidney disease; a randomized clinical trial. Journal of Renal Injury Prevention 2016;5(3):162-7. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Wang 2018e {published data only}

  1. Wang M, Liu H. Dietary advanced glycation end products restriction effects on intestinal bacterial flora and microinflammation state in maintenance hemodialysis patients [abstract no: SA-PO1013]. Journal of the American Society of Nephrology 2019;30(Abstract Suppl):1027. [EMBASE: 633768911] [Google Scholar]
  2. Wang M. Dietary advanced glycation end products restriction effects on intestinal bacterial flora and microinflammation state in maintenance hemodialysis patients [abstract no: SP664]. Nephrology Dialysis Transplantation 2018;33(Suppl 1):i569-70. [EMBASE: 622605572] [Google Scholar]

References to studies awaiting assessment

Chen 2019b {published data only}

  1. Chen X, Chen S, Huang L, Mao Z. Probiotics decreased concentrations of indoxylsulphate in PD patients [abstract no: SA-PO954]. Journal of the American Society of Nephrology 2019;30(Abstract Suppl):1011. [EMBASE: 633767865] [Google Scholar]

Mady 2018 {published data only}

  1. Mady G, Sarhan I, Shawki S, Halim A, Mahanna N, Abdallah M. Effect of probiotics on serum indoxyl sulphate in hemodialysis patients [abstract]. Hemodialysis International 2018;22(1):A15. [EMBASE: 620701234] [Google Scholar]

Marks 2010 {published data only}

  1. Marks WH, Spinelli TY, Olmstead SF. Probiotics to reduce immunosuppression-associated diarrhea following kidney transplantation - a prospective double-blinded randomized placebo-controlled trial [abstract no: 96]. American Journal of Transplantation 2010;10(Suppl 4):68. [EMBASE: 70463457] [Google Scholar]

Ogawa 2020 {published data only}

  1. Ogawa T, Hosoda Y, Horimoto A, Nishizawa Y, Omae K, Nagano N. Probiotic bio-three decreases serum phosphate levels with involvement of bacteria in gut microbioma in hemodialysis patients: A prospective, randomized, double-blind, placebo-contolled study [abstract no: SO055]. Nephrology Dialysis Transplantation 2020;35(Suppl 3):iii61. [EMBASE: 633421401] [Google Scholar]

Prado 2020 {published data only}

  1. Prado L, Aguilar K, Arellano J, Mariscal L, Campos I, Tuz F, et al. Lipid profile improvement in chronic kidney disease patients using a symbiotic supplement with agave inulin, Lactobacillus rhamnosus and Bifidobacterium longum. A post-hoc analysis from a randomized controlled trial [abstract no: 21]. Blood Purification 2020;49(1-2):243. [EMBASE: 631196086] [Google Scholar]

Ranganathan 2019a {published data only}

  1. Ranganathan N, Vyas UN, Ranganathan P, Irvin A, Weinberg AD. A double-blind, randomized, placebo-controlled with an open-label rollover extension phase 2/3 clinical trial to evaluate safety and efficacy of US-APR2020 in subjects with CKD stage IV [abstract no: PUB036]. Journal of the American Society of Nephrology 2020;31(Abstract Suppl):801. [EMBASE: 633696783] [Google Scholar]
  2. Ranganathan N, Vyas UN, Ranganathan P, Irvin A, Weinberg AD. Kibow multisite hope study dialysis randomized clinical trial protocol: a unique double-blind placebo-controlled cross-over design using renadyl standard-care therapy (n=100, 5 sites in the United States) [abstract no: PUB134]. Journal of the American Society of Nephrology 2019;30(Abstract Suppl):1108. [EMBASE: 633771192] [Google Scholar]
  3. Ranganathan N, Vyas UN, Ranganathan P, Irvin A, Weinberg AD. Kibow multisite hope study-CKD IV randomized clinical trial protocol: a unique double-blind placebo-controlled cross-over design using renadyl with standard-of-care therapy (n=500-600, 20-25 sites in the United States) [abstract no: FR-PO339]. Journal of the American Society of Nephrology 2019;30(Abstract Suppl):524-5. [EMBASE: 633768784] [Google Scholar]

Soliman 2018 {published data only}

  1. Soliman Ahmed Y, Ibrahim Sarhaan E, Shaker Mehanna N, Saeed Hassan M, Abd-El Nasier Abd-El Gawad M, Nagdy Madbouli N. The effect of synbiotics on serum indoxyl sulfate in maintenance haemodialysis patients [abstract]. QJM 2018;111(Suppl 1):i84-5. [EMBASE: 631035999] [Google Scholar]
  2. Soliman Y, Sarhan I, Mahanna N, Saeed M, Abd-ElNasier M, Nagdy N. The effect of synbiotic on serum indoxyl sulfate in maintenance hemodialysis patients [abstract]. Hemodialysis International 2018;22(1):A15. [EMBASE: 620701232] [Google Scholar]

Takayama 2003 {published data only}

  1. Takayama F, Enomoto A, Aoyama I. Oral administration of bifidobacteria in gastro-resistant seamless capsule reduces serum levels of indoxyl sulfate in patients on hemodialysis [abstract no: T297]. Nephrology Dialysis Transplantation 2002;17(Suppl 1):271. [CENTRAL: CN-00644305] [Google Scholar]
  2. Takayama F, Taki K, Niwa T. Bifidobacterium in gastro-resistant seamless capsule reduces serum levels of indoxyl sulfate in patients on hemodialysis. American Journal of Kidney Diseases 2003;41(3 Suppl 1):S142-5. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

TCTR20220317007 {published data only}

  1. Amelioration of uremic toxin indoxyl sulfate by oral chito-oligosaccharide in predialysis patients: a randomized controlled trial. https://trialsearch.who.int/Trial2.aspx?TrialID=TCTR20220317007 (date posted 1 March 2022). [DOI] [PubMed]
  2. Sirisuksakun C, Thimachai P, Tasanavipas P, Siriwattanasit N, Inkong P, Varothai N, et al. Amelioration of uremic toxin indoxyl sulfate by oral chito-oligosaccharide in predialysis patients: a randomized controlled trial [abstract no: SA-PO901]. Journal of the American Society of Nephrology 2022;33:852. [Google Scholar]

Wu 2012 {published data only}

  1. Wu AB, Wang MC, Chang YT, Tseng CC. Probiotics decrease the incidence of gram negative peritonitis in patients on peritoneal dialysis [abstract no: SA-PO864]. Journal of the American Society of Nephrology 2012;23(Abstract Suppl):840A. [Google Scholar]

Younes 2006 {published data only}

  1. Younes H, Egret N, Hadj-Abdelkader M, Remesy C, Demigne C, Gueret C, et al. Fermentable carbohydrate supplementation alters nitrogen excretion in chronic renal failure. Journal of Renal Nutrition 2006;16(1):67-74. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Zhang 2019a {published data only}

  1. Zhang Q. Effect of probiotic on gut microbiota in peritoneal dialysis patient with protein-energy wasting: 1 year follow up of a randomised controlled trial [abstract no: P0917]. European Journal of Immunology 2019;49(Suppl 3):1127-8. [EMBASE: 631545578] [Google Scholar]

References to ongoing studies

ChiCTR2200061930 {published data only}

  1. Effects of non-protein energy supplements and exercise on protein-energy wasting and gut microbes in maintenance hemodialysis patients. https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2200061930 (date posted 11 July 2022).

ChiCTR2200064821 {published data only}

  1. Clinical effect and mechanism of modified Shenling Baizhu powder on peritoneal dialysis. https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2200064821 (date posted 19 October 2022).

ChiCTR2300070381 {published data only}

  1. Effects of dietary fiber-added diets on protein-binding toxoids and microinflammatory states in patients with chronic kidney disease. https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2300070381 (date posted 11 April 2023).

ChiCTR2300072339 {published data only}

  1. Probiotics improve immune metabolism in maintenance hemodialysis patients by regulating intestinal flora. https://trialsearch.who.int/Trial2.aspx?TrialID=ChiCTR2300072339 (date posted 9 June 2023).

CTRI/2022/02/040296 {published data only}

  1. A study to assess the effect of probiotic in prevention of progression of kidney disease in patients with chronic kidney disease [A controlled randomized study to evaluate the effect of probiotic on uremic solutes,inflammatory status and oxidative stress in chronic kidney disease subjects - PRO-URE-CKD]. https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2022/02/040296 (date posted 15 February 2022).

CTRI/2022/11/047767 {published data only}

  1. Global phase 2 clinical trial to evaluate safety and efficacy of US-APR2020 in subjects with chronic kidney disease stage IV. https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2022/11/047767 (date posted 29 November 2022).

Ghoreishy 2022 {published data only}

  1. Ghoreishy SM, Shirzad N, Nakhjavani M, Esteghamati A, Djafarian K, Esmaillzadeh A. Effect of daily consumption of probiotic yoghurt on albumin to creatinine ratio, eGFR and metabolic parameters in patients with type 2 diabetes with microalbuminuria: study protocol for a randomised controlled clinical trial. BMJ Open 2022;12(3):e056110. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Headley 2020 {published data only}

  1. Headley SA, Chapman DJ, Germain MJ, Evans EE, Hutchinson J, Madsen KL, et al. The effects of 16-weeks of prebiotic supplementation and aerobic exercise training on inflammatory markers, oxidative stress, uremic toxins, and the microbiota in pre-dialysis kidney patients: a randomized controlled trial-protocol paper. BMC Nephrology 2020;21(1):517. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

IRCT20100223003408N5 {published data only}

  1. Ekramzadeh M, Azad F, Hamidianshirazi M, Mazloomi SM, Shafiee M. Efficacy of fortified synbiotic dessert on malnutrition and oxidative stress outcomes in hemodialysis patients: a randomized double-blind controlled trial [abstract]. American Journal of Kidney Diseases 2023;81(4):S113. [DOI: 10.1053/j.ajkd.2023.01.390] [DOI] [Google Scholar]
  2. Ekramzadeh M. Synbiotic dessert fortified with vitamin D and dalcium in hemodialysis patients [Effect of synbiotic dessert fortified with vitamin d and calcium on nutritional indices, inflammation, oxidative stress, malnutrition and quality of life in hemodialysis patients]. en.irct.ir/trial/35600 (date posted 6 April 2019).

IRCT20131013014994N7 {published data only}

  1. The effect of synbiotic supplementation on plasma levels of Advanced Glycation End Products and cardiovascular risk factors in hemodialysis patients: a double-blind clinical trial. https://trialsearch.who.int/Trial2.aspx?TrialID=IRCT20131013014994N7 (date posted 27 April 2022). [DOI] [PMC free article] [PubMed]

IRCT20160206026390N11 {published data only}

  1. Ghaedi E. Probiotics and children under hemodialysis treatment [Evaluation of the efficacy of probiotic supplementation on bio-markers of inflammation and oxidative stress in pediatric patients under treatment with hemodialysis: randomized double blind clinical trial]. en.irct.ir/trial/35899 (date posted 10 June 2019).

IRCT20230608058421N1 {published data only}

  1. The effect of probiotics on patients with end-stage renal failure. https://trialsearch.who.int/Trial2.aspx?TrialID=IRCT20230608058421N1 (date posted 29 June 2023).

KCT0007834 {published data only}

  1. The effect of carboneceous oral adsorbent and synbiotics on the gut microbiome and muscle health in chronic kidney disease patients. https://trialsearch.who.int/Trial2.aspx?TrialID=KCT0007834 (date posted 21 October 2022).

Mitrovic 2023 {published data only}

  1. Mitrovic M, Stankovic-Popovic V, Tolinacki M, Golic N, Sokovic Bajic S, Veljovic K, et al. The impact of synbiotic treatment on the levels of gut-derived uremic toxins, inflammation, and gut microbiome of chronic kidney disease patients- a randomized trial. Journal of Renal Nutrition 2023;33(2):278-88. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

NCT03770611 {published data only}

  1. Cueto-Manzano A. Effect of prebiotics and/or probiotics on uremic toxins and inflammation markers in peritoneal dialysis patients [Effect of a nutritional supplement of probiotics and/or prebiotics vs placebo on serum concentrations of uremic toxins and inflammatory cytokines in automated peritoneal dialysis patients]. www.clinicaltrials.gov/show/NCT03770611 (date posted 10 December 2018).

NCT03789708 {published data only}

  1. Elamin S. Effects of gum arabic supplementation in hemodialysis patients [Study of some pharmacological and biochemical effects of gum arabic supplementation to hemodilaysis patients]. www.clinicaltrials.gov/show/NCT03789708 (date posted 31 December 2018).

NCT03924089 {published data only}

  1. Olveira G. Oral nutritional supplement on nutritional and functional status, and biomarkers in malnourished hemodialysis patients (RENACARE) [Effect of an oral nutritional supplement on nutritional and functional status, biological markers (inflammation and oxidative stress, intestinal microbiota, circulating microRNA And its target genes) in malnourished hemodialysis patients]. clinicaltrials.gov/show/NCT03924089 (date posted 23 April 2019).

NCT05183737 {published data only}

  1. Effects of microencapsulated propolis and turmeric in patients with chronic kidney disease. https://clinicaltrials.gov/show/NCT05183737 (date posted 11 January 2022).

NCT05336305 {published data only}

  1. Polydextrose for patients with chronic kidney disease. https://clinicaltrials.gov/show/NCT05336305 (date posted 20 April 2022).

NCT05359094 {published data only}

  1. Probiotic supplements in chronic kidney disease. https://clinicaltrials.gov/show/NCT05359094 (date posted 3 May 2022).

NCT05540431 {published data only}

  1. Evaluation of protective effect of activated charcoal and probiotic against progression of chronic kidney disease. https://clinicaltrials.gov/show/NCT05540431 (date posted 14 September 2022).

NCT05674981 {published data only}

  1. To evaluate the beneficial effect of probiotics on DKD patients and the role of gut microbiota modulation. https://clinicaltrials.gov/show/NCT05674981 (date posted 9 January 2023).

NCT05724511 {published data only}

  1. The effect of probiotics on depression syndrome and risk factors of cardiovascular disease in hemodialysis patients. https://clinicaltrials.gov/show/NCT05724511 (date posted 13 February 2023).

NCT05835648 {published data only}

  1. Effect of dietary fiber on non-dialysis patients with chronic kidney disease. https://clinicaltrials.gov/show/NCT05835648 (date posted 28 April 2023).

RBR‐3WRNF {published data only}

  1. Moraes da Silva A. Effect of prebiotics on intestinal transit and inflammation of patients with pre-dialytic chronic renal disease. www.ensaiosclinicos.gov.br/rg/RBR-3wrnrf/ (date posted 13 December 2018).

ReSPECKD 2022 {published data only}

  1. Shamloo M, Mollard R, Wang H, Kingra K, Tangri N, MacKay D. A randomized double-blind cross-over trial to study the effects of resistant starch prebiotic in chronic kidney disease (ReSPECKD). Trials [Electronic Resource] 2022;23(1):72. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Additional references

Aron‐Wisnewsky 2016

  1. Aron-Wisnewsky J, Clement K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nature Reviews Nephrology 2016;12(3):169-81. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Azad 2018

  1. Azad MA, Sarker M, Li T, Yin J. Probiotic species in the modulation of gut microbiota: an overview. BioMed Research International 2018;2018:9478630. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Balshem 2011

  1. Balshem H, Helfand M, Schünemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64(4):401-6. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Beerepoot 2016

  1. Beerepoot M, Geerlings S. Non-antibiotic prophylaxis for urinary tract infections. Pathogens 2016;5(2):36. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Bromberg 2015

  1. Bromberg JS, Fricke WF, Brinkman CC, Simon T, Mongodin EF. Microbiota - implications for immunity and transplantation. Nature Reviews Nephrology 2015;11(6):342-53. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Cao 2022

  1. Cao C, Zhu H, Yao Y, Zeng R. Gut dysbiosis and kidney diseases. Frontiers in Medicine 2022;3(9):829349. [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Chan 2020

  1. Chan S, Cao C, Pascoe EM, Johnson DW, Shah A, Holtman GA, et al. Patient-reported gastrointestinal symptoms and the association with quality of life following kidney transplantation. Kidney International Reports 2020;6(1):138-45. [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Chen 2022

  1. Chen C, Wang J, Li J, Zhang W, Ou S. Probiotics, prebiotics, and synbiotics for patients on dialysis: a systematic review and meta-analysis of randomized controlled trials. Journal of Renal Nutrition 2023;33(1):129-39. [DOI: ] [DOI] [PubMed] [Google Scholar]

Cremon 2018

  1. Cremon C, Barbaro MR, Ventura M, Barbara G. Pre- and probiotic overview. Current Opinion in Pharmacology 2018;43:87-92. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Davani‐Davari 2019

  1. Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, Masoumi SJ, et al. Prebiotics: definition, types, sources, mechanisms, and clinical applications. Foods 2019;8(3):92. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

FAO/WHO 2002

  1. Joint FAO/WHO Working Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food London, Ontario, Canada, April 30 and May 1, 2002. Guidelines for the evaluation of probiotics in food. www.who.int/foodsafety/fs_management/en/probiotic_guidelines.pdf (last accessed 7 May 2020).

GBD 2017

  1. GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017 [Erratum in: Lancet. 2019 Jun 22;393(10190):e44; PMID: 31232379] [Erratum in: Lancet. 2018 Nov 17;392(10160):2170; PMID: 31329658]. Lancet 2018;392(10159):1736-88. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Gibson 2017

  1. Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, et al. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nature Reviews. Gastroenterology & Hepatology 2017;14(8):491-502. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

GRADE 2008

  1. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008;336(7650):924-6. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

GRADE 2011

  1. Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. Journal of Clinical Epidemiology 2011;64(4):383-94. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Higgins 2003

  1. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327(7414):557-60. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Higgins 2022

  1. Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, et al. Cochrane Handbook for Systematic Reviews of Interventions version 6.3 (updated February 2022). Cochrane, 2022. Available from www.training.cochrane.org/handbook.

Hill 2016

  1. Hill NR, Fatoba ST, Oke JL, Hirst JA, O'Callaghan CA, Lasserson DS, et al. Global prevalence of chronic kidney disease - a systematic review and meta-analysis. PLoS ONE [Electronic Resource] 2016;11(7):e0158765. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Jha 2013

  1. Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives [Erratum in: Lancet. 2013 Jul 20;382(9888):208]. Lancet 2013;382(9888):260-72. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Kato 2008

  1. Kato S, Chmielewski M, Honda H, Pecoits-Filho R, Matsuo S, Yuzawa Y, et al. Aspects of immune dysfunction in end-stage renal disease. Clinical Journal of The American Society of Nephrology: CJASN 2008;3(5):1526-33. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

KDIGO 2013

  1. Stevens PE, Levin A. Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Annals of Internal Medicine 2013;158(11):825-30. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Lehto 2018

  1. Lehto M, Groop PH. The gut-kidney axis: putative interconnections between gastrointestinal and renal disorders. Frontiers in Endocrinology 2018;9:553. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Lewis 1997

  1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scandinavian Journal of Gastroenterology 1997;32(9):920-4. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Luyckx 2018

  1. Luyckx VA, Tonelli M, Stanifer JW. The global burden of kidney disease and the sustainable development goals. Bulletin of the World Health Organization 2018;96(6):414-22D. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Mafra 2019

  1. Mafra D, Borges N, Alvarenga L, Esgalhado M, Cardozo L, Lindholm B, et al. Dietary components that may influence the disturbed gut microbiota in chronic kidney disease. Nutrients 2019;11(3):496. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

March 2020

  1. March DS, Jones AW, Bishop NC, Burton JO. The efficacy of prebiotic, probiotic, and synbiotic supplementation in modulating gut-derived circulatory particles associated with cardiovascular disease in individuals receiving dialysis: a systematic review and meta-analysis of randomized controlled trials. Journal of Renal Nutrition 2020;30(4):347-59. [PMID: ] [DOI] [PubMed] [Google Scholar]

McFarlane 2019

  1. McFarlane C, Ramos CI, Johnson DW, Campbell KL. Prebiotic, probiotic, and synbiotic supplementation in chronic kidney disease: a systematic review and meta-analysis. Journal of Renal Nutrition 2019;9(3):209-20. [PMID: ] [DOI] [PubMed] [Google Scholar]

Pan 2018

  1. Pan W, Kang Y. Gut microbiota and chronic kidney disease: implications for novel mechanistic insights and therapeutic strategies. International Urology & Nephrology 2018;50(2):289-99. [MEDLINE: ] [DOI] [PubMed] [Google Scholar]

Sampai‐Maia 2016

  1. Sampaio-Maia B, Simões-Silva L, Pestana M, Araujo R, Soares-Silva IJ. The role of the gut microbiome on chronic kidney disease. Advances in Applied Microbiology 2016;96:65-94. [PMID: ] [DOI] [PubMed] [Google Scholar]

Schünemann 2022a

  1. Schünemann HJ, Higgins JP, Vist GE, Glasziou P, Akl EA, Skoetz N, et al. Chapter 14: Completing ‘Summary of findings’ tables and grading the certainty of the evidence. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (editors). Cochrane Handbook for Systematic Reviews of Interventions version 6.3 (updated February 2022). Cochrane, 2022. www.training.cochrane.org/handbook.

Schünemann 2022b

  1. Schünemann HJ, Vist GE, Higgins JP, Santesso N, Deeks JJ, Glasziou P, et al. Chapter 15: Interpreting results and drawing conclusions. In: Higgins JP, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (editors). Cochrane Handbook for Systematic Reviews of Interventions version 6.3 (updated February 2022). Cochrane, 2022. Available from www.training.cochrane.org/handbook.

SONG 2017

  1. SONG Initiative. The SONG Handbook Version 1.0. www.songinitiative.org/reports-and-publications/ 2017.

Valdes 2018

  1. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ 2018;361:k2179. [MEDLINE: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

Zheng 2020

  1. Zheng JH, Guo J, Wang Q, Wang L, Wang Y, Zhang F, et al. Probiotics, prebiotics, and synbiotics for the improvement of metabolic profiles in patients with chronic kidney disease: A systematic review and meta-analysis of randomized controlled trials. Critical Reviews in Food Science & Nutrition 2020;61(4):577–98. [PMID: ] [DOI] [PubMed] [Google Scholar]

References to other published versions of this review

Cooper 2020

  1. Cooper TE, Khalid R, Craig JC, Hawley CM, Howell M, Johnson DW, et al. Synbiotics, prebiotics and probiotics for people with chronic kidney disease. Cochrane Database of Systematic Reviews 2020, Issue 5. Art. No: CD013631. [DOI: 10.1002/14651858.CD013631] [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Cochrane Database of Systematic Reviews are provided here courtesy of Wiley

RESOURCES