Skip to main content
Erschienen in: Gut Pathogens 1/2014

Open Access 01.08.2014 | Research

Clinical pilot study: efficacy of triple antibiotic therapy in Blastocystis positive irritable bowel syndrome patients

verfasst von: Robyn Nagel, Helle Bielefeldt-Ohmann, Rebecca Traub

Erschienen in: Gut Pathogens | Ausgabe 1/2014

Abstract

Background

Blastocystis species are common human enteric parasites. Carriage has been linked to Irritable Bowel Syndrome (IBS). Treatment of Blastocystis spp. with antimicrobials is problematic and insensitive diagnostic methods and re-infection complicate assessment of eradication. We investigated whether triple antibiotic therapy comprising diloxanide furoate, trimethoprim/sulfamethoxazole and secnidazole (TAB) given to diarrhoea-predominant IBS (D-IBS) patients positive for Blastocystis would achieve eradication.

Methods

In a longitudinal, prospective case study 10 D-IBS Blastocystis-positive patients took 14 days of diloxanide furoate 500 mg thrice daily, trimethoprim/sulfamethoxazole 160/80 mg twice daily and secnidazole 400 mg thrice daily. Faecal specimens were collected at baseline, day 15 and 4 weeks after completion of TAB. Specimens were analysed using faecal smear, culture and polymerase chain reaction (PCR) of the 16 SSU rRNA. Patients kept a concurrent clinical diary.

Results

Six (60%) patients cleared Blastocystis spp. after TAB, including three who had failed previous therapy. Subtypes detected were ST3 (60%), ST4 (40%), ST1 (20%) and ST7, 8 (10%); four patients had mixed ST infections. Serum immunoglobulin A (IgA) levels were low in 40% of patients. Higher rates of Blastocystis clearance were observed in patients symptomatic for less than a year (Mann-Whitney, p?=?0.032, 95% confidence) with no associations found with age, previous antibiotic therapy, faecal parasite load, ST, IgA level or clinical improvement.

Conclusions

Clearance of Blastocystis spp. was achieved with TAB in 60% of D-IBS patients, an improvement over conventional monotherapy. Higher clearance rates are needed to facilitate investigation of the relevance of this parasite in clinically heterogenous IBS.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13099-014-0034-0) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare they have no competing interests.

Background

Blastocystis spp.are unicellular, anaerobic enteric parasites found world wide in almost all species of animals [1]. The cystic form survives for days in the environment and likely transmits infection by the faecal-oral route either by direct contact or via contaminated water supplies. Blastocystis spp. are the most common parasite found in human faeces and carriage has been reported to be associated with Irritable Bowel Syndrome (IBS) like symptoms [1]-[3]. IBS is a chronic heterogenous disorder present in up to 10% of the global population characterised by a symptom complex of abdominal pain and variable bowel habit in the absence of gross morphological, histological or inflammatory diagnostic markers [4]. Although IBS does not increase mortality the disease impairs quality of life and costs billions of dollars annually in direct and indirect health care costs [5]. Genetic, environmental, gender and psychological factors are known to be important in the development of the disease but the pathophysiology is unknown. As many as 10% of cases of IBS are recognised to develop after a bout of acute infectious gastroenteritis and this has focused research on microbial interactions with the luminal gut mucosa [6].
Faecal microbiota profiles are reported to be significantly different in IBS patients, most notably in the diarrhoea predominant IBS (D-IBS) subgroup [7]. IBS patients have diminished faecal bacterial diversity with temporal instability, fewer aerobic bacteria and decreased Bifidobacterium spp./Enterobacter spp. ratio [7],[8]. Altered intestinal motility and abdominal hypersensitivity are typically seen in IBS, and many researchers have reported subtle alterations of the enteric immune and nervous system of IBS patients [9]. Increased numbers of mast cells secreting serotonin that are closely aligned to mucosal dendritic cells are consistently observed in the gastrointestinal mucosa of patients with IBS [10]. Changes in cytokine secretion are reported not only with many luminal enteric microbial infections but increased levels of pro-inflammatory cytokines (interleukin (IL)-6, IL-8, and tumour necrosis factor-? (TNF-?)) have been reported in the plasma and peripheral blood mononuclear cells of IBS patients [6]. Many of these cytokines increase epithelial permeability by disrupting the mucosal tight junctions [11]. Proteases are also reported to be elevated in the faeces of IBS patients [12],[13] although it is not clear if these proteases derive from enteric microorganisms, human intestinal cells or both. Blastocystis organisms have been shown to activate IL-8 gene expression in human colonic epithelial cells in vitro[14], increase epithelial permeability [15] and degrade intestinal Immunoglobulin A (IgA) [16], mediated possibly by action of cysteine proteases [17] reported to be present on the cell surface [18].
The prevalence of Blastocystis carriage is increased in all IBS patients but highest in D-IBS patients (73% compared to 27% in healthy control patients) [19], suggesting that this subgroup may provide a more homogenous IBS patient group to study. Seventeen different subtypes (ST) of Blastocystis spp. have been described to date [20] revealing moderate host ST specificity and regional ST variation in humans. Early reports that particular subtypes of Blastocystis may be associated with clinical pathogenicity in humans have not been confirmed [1]. Treatment of Blastocystis spp. with antimicrobials is problematic and insensitive diagnostic methods and possible re-infection complicates assessment of successful eradication [21],[22]. Monotherapy with metronidazole is the most commonly recommended drug and published eradication rates vary from 0-100% [21]. If Blastocystis spp. could be predictably successfully eradicated in symptomatic patients then it may be possible to more easily evaluate whether infection is associated with clinical symptoms. In this pilot study we treated ten patients with diarrhoea-predominant IBS (IBS-D) who were positive for Blastocystis carriage with triple antibiotic therapy (diloxanide furoate, trimethoprim-sulfamethoxazole, secnidazole) and assessed clinical status, ST and clearance rates.

Materials and methods

Study outline

This prospective, longitudinal study was conducted in a rural specialist outpatient clinic (Toowoomba Gastroenterology Clinic). Patients presenting with chronic diarrhea were clinically assessed and patients who were subsequently diagnosed with diarrhoea-predominant IBS (IBS-D) [4] and were positive for Blastocystis carriage were invited to participate in the study. Faecal specimens were collected from study participants at baseline, after two weeks of triple antibiotic therapy and six weeks after completion of antibiotics.

Inclusion protocol

Adult patients presenting to the clinic with chronic diarrhoea from 1/8/11 to 20/02/14 were assessed clinically. Blood tests, including full blood count, serum calcium, thyroid function tests, serum immunoglobulin A (IgA) and coeliac antibody tests were performed. Serum IgA testing was performed using a nephelometry assay using a BNII device (Siemens, Germany). The reference range for a healthy population provided by the commercial laboratory was 1.24-4.16 g/L. Selective IgA deficiency is defined as <50% of the lower range of normal (0.61 g/L). Faecal microscopy for ova, cysts and parasites (including Dientamoeba fragilis trophozoites), culture (for bacterial pathogens including Salmonella sp, Shigella sp, Vibrio cholerae, Campylobacter sp, Aeromonas sp) and PCR analysis for Giardia duodenalis, Clostridium difficile and Entamoebae histolytica (and a 3-day culture for hookworm) were performed by a commercial pathology laboratory to exclude known pathogens and initially detect the presence of Blastocystis spp. All patients proceeded to an upper and lower endoscopy that included gastric antral, duodenal, ileal and colonic biopsies. Ten consecutive, eligible symptomatic patients positive for Blastocystis carriage who had no other cause for symptoms identified and who fulfilled the Rome criteria [4] for D-IBS, namely symptoms of chronic abdominal pain, occurring frequently over the last 3 months, associated with defecation and the passage of predominantly loose stool, were enrolled in the study. All patients who were invited to participate consented to enrolment and completed the study.

Exclusion protocol

Only non-pregnant adults between 18 and 75 years of age were recruited for the study. Patients with a known allergy to sulphur drugs or patients with significant systemic diseases or co-morbidities were excluded.

Study protocol

One baseline faecal specimen was collected from patients prior to 14 days therapy with diloxanide furoate 500 mg three times daily, trimethoprim/sulfamethoxazole 160/800 mg twice daily and secnidazole 400 mg three times daily. Two further faecal samples were taken, the first within 48 hours of ceasing antibiotics and another 4 weeks after ceasing antibiotics. Patients were reviewed clinically at the start of the study, after completion of antibiotics and 6 weeks later. All patients kept a diary which recorded the number of daily bowel movements (number per day), consistency of the stool (scored: 1?=?very hard, 2?=?hard, 3?=?formed, 4?=?loose, 5?=?watery) and general well being (scored 1-10; from poor to excellent). Routine electrolytes, liver function tests and a full blood count were repeated 4 weeks after ceasing the antibiotics.

Diagnostic methods

All samples were run in parallel for the presence of Blastocystis spp. using a simple unstained wet faecal smear, xenic in vitro culture (XIVC) and PCR (confirmed as Blastocystis spp. using DNA sequencing). A patient was considered to be positive if any one of the tests was positive.

Parasitological methods

Fresh unpreserved faecal specimens from patients were examined as a fresh faecal smear under 40× magnification by light microscopy. The load of Blastocystis organisms was classified as light, medium or heavy (<5, 5-10 and >10 organisms per high powered field (O/HPF). Additionally 100 mg of fresh stool was inoculated in Jones’ culture medium [23] and incubated at 37±C for 48 hours. Cultures were examined for the presence of Blastocystis spp. by light microscopy.

Molecular methods

DNA was extracted from the unpreserved faecal samples and pelleted cultures of Blastocystis using the QIAamp DNA Stool Mini Kit (Qiagen, Hilden, Germany) according to the manufacturers instructions, except that following the addition of lysis buffer, the faecal suspension was subjected to three freeze-thaw cycles in liquid nitrogen and a 95±C water bath respectively.
The genomic DNA from stool and cultured Blastocystis specimens from all patients was subjected to PCR analyses. All samples were analysed using the Wong protocol [24] that amplified a 1000 bp of the (SSU) rRNA gene of ST 1-17 of Blastocystis spp. All positive PCR products were subjected to DNA sequencing and phylogenetic analysis to identify the particular ST.
All Blastocystis spp. positive samples were also analysed separately with three specific ST primers (ST1, 3, 4) in order to detect mixed infections. The ST1 specific protocol was developed by Dr CR Stensvold and the ST3 and ST4 protocols were developed in-house. These primers amplify highly ST-specific smaller fragments of the (SSU) rRNA gene and obviate the need for PCR product sequencing. The ST specific primers 1 and 4 were specific for ST when tested against Netski (ST1) and WR1 (ST4) axenic cultures. The STS primer 3 demonstrated mild cross reactivity with ST1 and ST4 and if disparity was found between the Wong primer results and the STS primer result the PCR product was sequenced. The ST specific primers sequences are as follows:

ST1: Forward:TAATACATGAGAAAGTCCTCTGG

Reverse: CTCAATCCAATTGCAAGAC
Annealing temp: 57±C; amplicon size: 336 bp
ST3: Forward: GTAGTTGTGTAATGAATACATTCG
Reverse: GTCCCTTTTAATCTTATTCTCATG
Annealing temp: 62±C; amplicon size: 250 bp
ST4: Forward: GTGATCTTCGGATGACGTGAATC
Reverse: CAGAATTTCACCTCTGACTATTG

Annealing temp: 60±C amplicon size 290 bp

PCR products were run on gels of 1% agarose in 1 x sodium borate (SB)(0.4 gmNaOH/2.25 g Boric acid/1 L H2O) buffer at 200 V in a Biorad electrophoresis system and were purified using a Purelink PCR Purification Kit (Invitrogen, Grand Island, NY, USA) prior to sequencing.
Sequencing was done using an ABI 3130xl Genetic Analyzer (Applied Biosystems) using a Ready Reaction Kit Version 3.1 (PE Applied Biosystems, California, USA). Sequences were then edited and assembled using Finch TV v 1.4.0.
DNA sequences were aligned with 15 reference sequences of Blastocystis SSU rRNA genes sourced from GeneBank (NCBI) representing the established subtypes described from mammals and birds. Distance-based analyses were conducted on readable sequences using Kimura 2-parameter distance estimates and trees were constructed using the Neighbour Joining algorithm using Mega 3.1 [25] software. Preoteromonas lacertae (U37108) was used as an out-group. Bootstrap analyses were conducted using 1000 replicates.

Ethics approval, statistics

The study was approved by the University of Queensland Medical Research Ethics Committee and registered with the Australian and New Zealand Clinical Trials registry (http://​www.​ANZCTR.​org.​au) ACTRN: 12611000918921. The results were analysed using the Mann-Whitney U test (Wizard statistical program).

Results

Clinical characteristics of the study group

Male (1) and female (9) patients with an age range of 28-73 years (median age 49 years) participated in the study. The shortest symptom duration was 3 months and the longest 10 years and the presenting symptoms were chronic diarrhoea and abdominal pain (10/10). Six patients had received prior antibiotic therapy (including metronidazole, norfloxacillin, nitazoxanide) for Blastocystis infection that did not clear either the parasite in the faeces or their symptoms. The remaining patients were naive to therapy. During the period of the study no patient had any organisms other than Blastocystis spp. noted on faecal testing. No patient demonstrated eosinophilia on the peripheral blood film. There were no abnormalities noted in the baseline electrolyte, liver function tests or full blood films blood tests. Five patients recorded a low baseline serum IgA level (<1.24 g/L).

Antibiotic therapy and clinical follow up

All patients completed 14 days of the triple antibiotic therapy (TAB). No serious adverse effects were noted; four patients complained of nausea, two patients of headache and one developed oral thrush and bloating during the antibiotic therapy course. At four weeks post antibiotic therapy repeat electrolytes, liver function tests and full blood count testing remained normal in all patients

Faecal smears, cultures and PCR results

Faecal smear results prior to the commencement of treatment demonstrated a parasite load of <5 O/HPF in five patients, 5 10 O/HPF in four patients and >10 O/HPF in one patient. Five patients had cleared the Blastocystis parasite from faeces on testing with faecal smear, XIVC and PCR at Day 15 and six patients had similarly cleared the organism at 6 weeks. All baseline samples positive to either microscopy or culture were positive on PCR testing using the Wong primers. Four patients had mixed ST infections that were confirmed with the ST-specific primers and subsequent sequencing if indicated. The Blastocystis ST’s detected were ST3 (60%), ST 4 (40%), ST 1 (20%) and ST7 and ST8 (10%). Day 15 and week six samples demonstrated good correlation between faecal smear/XIVC results and PCR testing (Table 1).
Table 1
Blastocystis faecal microscopy, XIVC and PCR results before and after triple antibiotic therapy
Patient identity
Time
Faecal microscopy
PCR-WONG
PCR-ST1 specific
PCR-ST3 specific
PCR-ST4
Clearance
  
Faeces
XIVC
Faeces
XIVC
Faeces
XIVC
Faeces
XIVC
Faeces
XIVC
 
1
Baseline
+
+
ST4*
ST6*
Negative
 
Negative
ST3*
Negative
ST4
 
 
Day 15
Negative
Negative
ST4*
Negative
Negative
Negative
ST3
Negative
Negative
Negative
Yes
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
2
Baseline
+
+
ST7*
Negative
Negative
 
Negative
 
Negative
  
 
Day 15
+
+
+
Negative
      
No
 
Week 6
+
+
ST7*
Negative
Negative
 
Negative
 
Negative
 
No
3
Baseline
+
 
ST1*
 
ST1
 
Negative
 
Negative
  
 
Day 15
+
+
+
 
Negative
 
Negative
 
Negative
 
No
 
Week 6
+
+
Negative
Negative
      
No
4
Baseline
+
+
Negative
ST3*
Negative
Negative
Negative
ST3
Negative
Negative
 
 
Day 15
Negative
Negative
Negative
Negative
      
Yes
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
5
Baseline
+
+
ST4*
Negative
Negative
 
ST3*
 
ST4
  
 
Day 15
Negative
+
Negative
ST6*
Negative
 
ST3*
 
Negative
 
No
 
Week 6
Negative
+
ST4*
Negative
Negative
 
ST3*
 
ST4
 
No
6
Baseline
+
+
ST4*
Negative
Negative
 
Negative
 
ST4
  
 
Day 15
Negative
Negative
ST1*
Negative
Negative
 
Negative
 
Negative
 
Yes
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
7
Baseline
+
Negative
Negative
ST4*
Negative
 
ST3*
 
Negative
  
 
Day 15
Negative
Negative
Negative
Negative
      
Yes
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
8
Baseline
+
+
ST8*
ST8*
Negative
Negative
Negative
Negative
Negative
Negative
 
 
Day 15
+
 
ST8*
 
Negative
 
Negative
 
Negative
 
No
 
Week 6
+
+
ST8*
ST8*
Negative
Negative
Negative
Negative
Negative
Negative
No
9
Baseline
+
+
Negative
ST3*
 
Negative
 
ST3
 
Negative
 
 
Day 15
Negative
 
Negative
       
Yes
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
10
Baseline
+
+
Negative
ST3*
 
Negative
 
ST3
 
Negative
 
 
Day 15
+
+
ST3*
Negative
Negative
 
ST3
 
Negative
 
No
 
Week 6
Negative
Negative
Negative
Negative
      
Yes
XIVC: xenic invitro culture.
Polymerase chain reaction PCR.
*Sequenced.

Clearance and clinical follow up

Overall 60% of patients cleared the organism from their faeces at 6 weeks. Three of these patients had had previous courses of antibiotics. The patient’s were reviewed clinically and their patient diary scored. The average score for the baseline week and week 4 following TAB therapy was calculated taking the weekly mean of the number of daily bowel actions, consistency of stool and general wellbeing. The final clinical score was calculated as the sum of improvement in frequency of bowel habit, consistency and wellbeing from baseline to week four after antibiotic therapy (see Additional file 1). Six patients reported that they felt clinically improved and this correlated with a positive change of greater than 1.0 diary score from baseline to six weeks. However, of these six clinically improved patients only three demonstrated faecal clearance of the organism. In contrast, four patients did not report clinical improvement although two of these patients demonstrated faecal clearance of Blastocystis spp. This data was analysed to assess whether clinical improvement or Blastocystis clearance had any relationship to age of patient, duration of symptoms, naive therapy status, original Blastocystis faecal load, the particular Blastocystis subtype, serum IgA level or clinical outcome. Significantly higher rates of Blastocystis spp. clearance were observed if the patient had been symptomatic for less than a year (Mann-Whitney, p?=?0.032, 95% confidence), but no other associations were found (Table 2).
Table 2
Clinical characteristics and outcome after triple antibiotic therapy
Patient
Age yrs
Duration of symptoms (years)
Previous AB therapy
Blastocystisload in baseline faeces
BlastocystisST’s
SerumIgA g/L
Diary score
Clinical score?>?=1 point
Clearance at 6 weeks
1
35
4
Yes (F,O)
2+
3,4,6
1.51
+4.1
Yes
Yes
2
59
5
No
2+
7
1.12 (L)
+2.4
Yes
No
3
28
1.5
Yes (F)
1+
1
2.1
?0.3
No
No
4
35
0.25
Yes (F,O)
1+
3
2.2
+0.2
No
Yes
5
51
1.5
Yes (F)
1+
3,4,6
2.82
+2.0
Yes
No
6
58
0.5
No
2+
1,4
1.03 (L)
+1.1
Yes
Yes
7
73
0.75
No
1+
3,4
1.22
+4.5
Yes
Yes
8
49
10
Yes (F,O)
3+
8
1.94
+1.3
Yes
No
9
63
1
No
1+
3
0.75 (L)
?0.8
No
Yes
10
36
1
Yes (F)
2+
3
0.98 (L)
?0.6
No
Yes
F = Flagyl.
O = other antibiotics (metronidazole, nitazoxanide, norfloxacillin).
L = low, <1.24 g/L, level defined by laboratory.

Discussion

Successful faecal eradication of Blastocystis spp.occurred in 60% of patients at the end of this study following fourteen days of triple antibiotic therapy using diloxanide furoate, trimethoprim/sulfamethoxazole and secnidazole (TAB). Our previous study [22] used metronidazole as a monotherapy and achieved 0% successful faecal eradication, suggesting that this TAB regimen may be useful to treat Blastocystis infection. Although six patients in our study had already received at least one course of metronidazole therapy previously, three of these patients proceeded to clear the organism after TAB therapy indicating that triple therapy is still a worthwhile alternative in this group. One recent study has reported 100% failure to clear in five patients with Blastocystis ST3 infection treated with TAB combination [26]. However, these five patients had faecal specimens retested 12 months following the TAB therapy and it was not possible to exclude reinfection in this group. Triple antibiotic therapy has been shown to be useful in treating infecting organisms such as Helicobacter pylori and Mycobacterium spp. and it may be necessary to explore combinations of antibiotics and methods of delivery to further improve the clearance rate of Blastocystis infection.
Widely varying rates of eradication (0-100%) of Blastocystis using a variety of antibiotics including ketoconazole, ornidazole, tinidazole, rifaximin and paromomycin have been reported [21] and some of this apparent inconsistency may be due to using insensitive faecal smears as a sole means of diagnosis. Diagnostic methods such as faecal XIVC or PCR have been reported to increase accuracy by 30-50% for detection of Blastocystis spp. [10]. Metronidazole resistance has also been reported [27] and differences in antimicrobial sensitivities to metronidazole and emetine have been demonstrated between axenic ST4 and ST7 cultures [28]. Resistance may therefore contribute to regional variation in eradication rates. A number of antimicrobials have been tested for activity against Blastocystis spp. in vitro, in both axenic and xenic conditions [29]-[31] and this data is a useful starting point for choice of antimicrobial. The Blastocystis organism appears to establish in the anaerobic environment of the human ileum and caecum, thrives in the presence of bacteria, and only in extremely rare circumstances is invasive to the gastrointestinal mucosa [1]. An intrinsic difficulty in treating Blastocystis spp. may be delivering the appropriate drugs to the intraluminal site of infection or understanding how to alter the faecal microbiota to produce an environment that is unsupportive or hostile to the parasite.
Secnidazole, similar to metronidazole, is a 5-imidazole drug substituted at position 1 and has been shown to have significant (although 50% less than metronidazole) inhibitory activity against Blastocystis spp. in vitro tested on axenic cultures [31]. All nitroimidazole drugs are rapidly absorbed from the small bowel, metabolised in the liver and excreted primarily in the urine. They exert their antimicrobial action in an anaerobic intracellular environment after reduction to a toxic metabolite that disrupts DNA linkage [32]. Metronidazole is the most common empiric therapy used against Blastocystis[33] and as many patients that present for therapy have already failed metronidazole we felt it was useful to use a different imidazole particularly as resistance to metronidazole has been shown not to extend across the entire class of 1-sustituted 5-imidazoles [28]. The pharmacodynamics of secnidazole are not as well defined as those of metronidazole but it is known that secnidazole has reduced excretion of parent drug and metabolites in the urine (50% compared to 90% with metronidazole) and therefore may have increased faecal excretion. It also has more than double the excretion half-life of metronidazole at 29 hours [28]. These pharmacologic properties may be an advantage in treating Blastocystis.
Trimethoprim/sulfamethoxazole duotherapy is often proposed as second line therapy for Blastocystis spp. infection and eradication rates of 22-100% have been reported for this combination therapy [21]. These two drugs act synergistically to interfere with the production of dihydrofolate reductase that is required for purine and including DNA synthesis. Blastocystis ST7 does not take up Hthymidine [18],[31], is likely to synthesize purines de novo, and likely to have a dihydrofolate reductase that can be targeted. Trimethoprim is rapidly absorbed from the small bowel [34] is lipophilic with wide tissue distribution and 80% is excreted in the urine unchanged. Trimethoprim therapy alone has been shown to have inhibitory activity similar to secnidazole in vitro[31]. Sulfamethoxazole similarly is rapidly absorbed in the small bowel [34] metabolised in the liver, and over 90% excreted in the urine. They both have a half-life of around 10 hours. Although sulfamethoxazole alone was found to be non-inhibitory in vitro[28] a combination of trimethoprim:sulfamethoxazole (in the ratio of 1:5, Bactrim’) was found to be much less effective at inhibiting Blastocystis growth in axenic cultures than a trimethoprim/sulfamethoxazole 1:2 ratio which may indicate that the synergistic effect of the sulfamethoxazole plays an important role in directly inhibiting Blastocystis spp. growth. Enteric coating this duotherapy compounded in a 1:2 ratio may be a useful therapeutic avenue to explore.
An alternative therapeutic approach would be to use an antimicrobial drug that has already been described to have intraluminal amoebocidal properties. A number of these drugs tested in vitro such as emetine, furazolidone, nitazoxanide, mefloquine, quinicrine, quinine, iodoquinol, clioquinol (Entero-Vioform’) and chloroquine showed moderate to high inhibition on Blastocystis cell growth [28]-[31]. Emetine and clioquinol therapy have serious adverse reactions reported and are not used currently in clinical practice. Other intraluminal drugs such as diloxanide furoate, paromomycin, doxycycline and pyrimethamine showed no inhibitory effect on Blastocystis[29],[30]. Paromomycin is a relatively safe, poorly absorbed aminoglycoside with activity against not only gram-negative but many gram-positive bacteria, including Mycobacterium tuberculosis, and protozoa such as Leishmania spp. [35]. Interestingly two recent studies have reported high Blastocystis clearance rates (77-100%) [26],[36] using paromomycin and if confirmed this would suggest the broad spectrum aminoglycoside is achieving success by an indirect effect on the faecal microbiota. The largest study [36] retrospectively analysed faecal microscopy results on 52 symptomatic Blastocystis carriers treated with paromomycin alone and clearance rates were estimated to be 77%. Rifaximin is an oral broad-spectrum antibiotic derived from rifamycin that is not absorbed systemically (97% excreted in the faeces) [37]. It has been used successfully to treat hepatic encephalopathy, travellers diarrhoea [37] and non-constipated IBS patients [38]. Only one case report of its successful use in Blastocystis infection has been published [39]. Diloxanide furoate was chosen for our study as it had a good safety profile, the drug remains intraluminal until it is hydrolysed to diloxanide, absorbed and subsequently excreted in the urine. It is known to effectively clear intestinal amoebiasis although the mechanism of action is not known [40]. It was not possible to assess whether the addition of diloxanide furoate increased the efficacy of the regimen in this study. The addition of an intraluminal antimicrobial to a therapeutic regimen seems logical and paromomycin may be the most appropriate choice to add to a regimen at the moment. Unfortunately monotherapy is usually a cheaper option (the cost of 14 days of appropriate doses of metronidazole, trimethoprim/sulfamethoxazole, diloxanide furoate, paromomycin, secnidazole and rifaximin being AUS $24, $24, $55, $70, $80, $80 respectively) and the cost of TAB rising to AUS$159. If an effective drug combination could be identified it is likely the costs of a combination therapy would reduce.
This small pilot study was not designed to interrogate specific variables that might be relevant to Blastocystis spp. symptoms, clearance or clinical outcome. The literature is inconsistent but some studies have suggested that particular subtypes, namely ST 1, 3 and 4 of Blastocystis may be pathogenic [1]. In contrast, and in accord with recent studies [22],[26] our patients demonstrated a diverse range of subtypes and no association was seen between subtype and faecal clearance or resolution of symptoms. Although previous studies have reported mixed infection rates of 5-30% [22],[41] almost half of the patients (40%) in this study demonstrated mixed infections. The use of ST specific primers and XIVC techniques that allow ST’s present in small numbers to grow to detectable numbers are factors likely to increase identification of mixed infections.
It is well recognised that many Blastocystis carriers are healthy. Much research has been directed towards identifying genetic characteristics specific to the parasite, such as ST, that may indicate pathogenicity. Some studies have investigated host factors that may allow Blastocystis spp. to cause clinical symptoms. Epidemiological studies conducted in patients with compromised immune function (HIV carriage, chronic renal failure, haematological malignancies with immunosuppressant therapy [42],[43]) have shown increased rates of Blastocystis carriage but no clear consensus has emerged linking carriage to symptoms. IBS [6] and Blastocystis infection [14] have both been linked to changes in inflammatory cytokine expression. The prevalence of single nucleotide gene polymorphisms present in IL-8 (pro-inflammatory) and IL-10 (anti-inflammatory) cytokines is reported to be significantly increased not only in IBS patients but also in IBS patients positive for Blastocystis infection [44] suggesting that factors regulating the immune response in the host are important in expression of clinical disease.
IgA is also an important part of host mucosal immunity. Blastocystis proteases have been reported to cleave human secretory IgA [16]. Interestingly 40% of our group of IBS-D Blastocystis had low serum IgA and this was much higher than the 1.4% (1 in 19) recorded in our healthy adult volunteers negative for Blastocystis spp. (unpublished observations). The levels of serum IgA recorded in our patients are compatible with partial selective IgA deficiency but did not reach <50% of the lower limit of normal defined as selective IgA deficiency. Selective IgA deficiency (prevalence of 0.17% in Australia) is usually asymptomatic, but may be associated with an increased risk of respiratory, urinary and gastrointestinal (including giardiasis) sepsis, and allergic and autoimmune diseases [45]. There was no apparent association between IgA levels and subsequent clearance. Serum IgA levels recorded in children are generally lower than adults [46] and if IgA levels influence the risk of carriage of Blastocystis then children may be more susceptible.
In this paper we describe successful clearance of the Blastocystis organism from faeces after testing at day 15 and week 4 after antibiotics. Blastocystis organisms may be excreted irregularly [47] so it was useful to have had two clear faecal specimens 4 weeks apart to confirm faecal clearance. We used three methods of detecting Blastocystis in the stool, taking care to include XIVC and PCR amplification of Blastocystis DNA as the latter two methods have approximately a 90% sensitivity detection rate compared with only around 50% for microscopy alone [48]. Nevertheless false negative results may occur in a small percent of cases, where complete clearance of Blastocystis spp. in the ileum may not have occurred. It would be useful to be able to distinguish symptomatic and asymptomatic patients positive for Blastocystis faecal carriage. Although Blastocystis spp. are non-invasive organisms serum antibodies against Blastocystis spp. have been reported in humans infected with the parasite [49]. The presence of antibodies often only signifies exposure to that Blastocystis antigen some time in the past. However one study has reported that an antibody reacting to a 29 kDa protein, thought to be a cysteine protease present on the plasma membrane of Blastocystis spp., is more commonly seen in symptomatic patients [50]. If confirmed this finding and similar approaches may allow us to utilise the expression of host response to infection to determine need for therapy.
The only variable that was found to be significant was duration of GIT symptoms and eventual clearance of the parasite. If we assume that the GIT symptoms were related to the Blastocystis carriage this finding may suggest that Blastocystis infection that persists in the host for years may be more difficult to eradicate, particularly as spontaneous clearance has been reported to occur in only 19% of symptomatic patients over 12 months [36].
The patients in this study were a careful selection of D-IBS patients with no other identifiable enteric pathogen or other cause for symptoms. Nevertheless only 50% of patients who reported clinical improvement had cleared the organism. Perhaps reducing parasite load was enough to achieve clinical improvement in these patients. The two patients who reported dramatic clinical improvement (>4 points) both cleared the organism. In contrast 50% of the patients who did not report clinical improvement cleared the organism, suggesting that Blastocystis carriage was not the cause or not the only cause of their D-IBS. IBS is a heterogenous clinical condition and many diseases masquerade as IBS. Over time definitive specific diagnostic tests or therapies that allow us to discriminate other causes of IBS may become available. Although Giardia duodenalis was identified microscopically in the 1700 s acceptance of its pathogenicity only occurred in the last forty years when effective therapy with imidazoles (>90% successful eradication) demonstrated clinical improvement [51]. Although 60% patients cleared in this study clinical cause and effect will be able to be more easily evaluated when we can more reliably and effectively eradicate Blastocystis spp.

Conclusions

Successful faecal eradication of Blastocystis spp. occurred in 60% of D-IBS patients at the end of this study following fourteen days of triple antibiotic therapy using diloxanide furoate, trimethoprim/sulfamethoxazole and secnidazole (TAB). Metronidazole monotherapy achieved 0% successful eradication in our previous study and as some isolates of Blastocystis spp. have also been shown to be resistant to metronidazole, this drug should no longer be recommended as first line therapy. Surprisingly paromomycin, a drug that exhibits little inhibitory activity in vitro, has been reported to have eradication rates above 75%. If these results are confirmed in future studies using sensitive diagnostic markers, paromomycin may prove to be a useful basis for a double or triple antibiotic regimen. Predictable successful eradication (>90%) of Blastocystis spp. will expedite investigation of the clinical significance of Blastocystis infection. No meaningful analysis of clearance and symptoms was possible in this trial but 40% Blastocystis patients demonstrated low serum IgA levels. It may be worth focusing future studies on investigating immune profiles in the host and including host IgA levels.

Author’s contributions

RN carried out the clinical investigations, microbiology and molecular genetic studies, sequence alignment, statistical analysis and drafted the manuscript. RN, HBO and RT conceived the study design, participated in the design and coordination and helped draft the manuscript. All authors read and approved the final manuscript.

Additional file

Acknowledgements

We thank Dr CR Stensvold, Department of Microbiological Diagnostics, Statens Serum Institut, Copenhagen, Denmark for providing ST1 specific primers, and Dr Linda Dunn, Queensland Insititute of Medical Research for donation of axenic cultures of Blastocystis subtypes Netski and WR1. This study was funded by the Royal Australian College of Physicians Murray-Will Fellowship for Rural Physicians (awarded to R. Nagel 2012).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare they have no competing interests.
Literatur
1.
Zurück zum Zitat Tan KS: New insights on classification, identification, and clinical relevance of Blastocystis spp. Clin Microbiol Rev. 2008, 21 (4): 639-665. 10.1128/CMR.00022-08.PubMedCentralCrossRefPubMed Tan KS: New insights on classification, identification, and clinical relevance of Blastocystis spp. Clin Microbiol Rev. 2008, 21 (4): 639-665. 10.1128/CMR.00022-08.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Jimenez-Gonzalez DEM-FW, Reues-Gordillo J, Ramirez-Mirnada M, Arroyo-Escalante S, Romero-Valdovinos M, Stark D, Souza-Saldivar V, Martinez-Hernandez F, Flisser A, Olivo-Diaz A, Maravilla P: Blastocystis infection is associated with irritable bowel syndrome in a Mexican patient population. Parasitol Res. 2012, 110 (3): 1269-1275. 10.1007/s00436-011-2626-7.CrossRefPubMed Jimenez-Gonzalez DEM-FW, Reues-Gordillo J, Ramirez-Mirnada M, Arroyo-Escalante S, Romero-Valdovinos M, Stark D, Souza-Saldivar V, Martinez-Hernandez F, Flisser A, Olivo-Diaz A, Maravilla P: Blastocystis infection is associated with irritable bowel syndrome in a Mexican patient population. Parasitol Res. 2012, 110 (3): 1269-1275. 10.1007/s00436-011-2626-7.CrossRefPubMed
3.
Zurück zum Zitat Dogruman-Al F, Simsek Z, Boorom K, Ekici E, Sahin M, Tuncer C, Kustimur S, Altinbas A: Comparison of methods for detection of Blastocystis infection in routinely submitted stool samples, and also in IBS/IBD Patients in Ankara, Turkey. PLoS One. 2010, 5 (11): e15484-10.1371/journal.pone.0015484.PubMedCentralCrossRefPubMed Dogruman-Al F, Simsek Z, Boorom K, Ekici E, Sahin M, Tuncer C, Kustimur S, Altinbas A: Comparison of methods for detection of Blastocystis infection in routinely submitted stool samples, and also in IBS/IBD Patients in Ankara, Turkey. PLoS One. 2010, 5 (11): e15484-10.1371/journal.pone.0015484.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Drossman DA: Introduction. The Rome Foundation and Rome III. Neurogastroenterol Motil. 2007, 19 (10): 783-786. 10.1111/j.1365-2982.2007.01001.x.CrossRefPubMed Drossman DA: Introduction. The Rome Foundation and Rome III. Neurogastroenterol Motil. 2007, 19 (10): 783-786. 10.1111/j.1365-2982.2007.01001.x.CrossRefPubMed
5.
Zurück zum Zitat Inadomi JMFM, Bjorkman D: The economic impact of irritable bowel syndrome. Aliment Pharmacol Therapeut. 2003, 18 (7): 671-682. 10.1046/j.1365-2036.2003.t01-1-01736.x.CrossRef Inadomi JMFM, Bjorkman D: The economic impact of irritable bowel syndrome. Aliment Pharmacol Therapeut. 2003, 18 (7): 671-682. 10.1046/j.1365-2036.2003.t01-1-01736.x.CrossRef
6.
Zurück zum Zitat Beatty JKBA, Buret AG: Post-infectious irritable bowel syndrome: Mechanistic insights into chronic disturbances following enteric infection. World J Gastroenterol. 2014, 20 (14): 3976-3985. 10.3748/wjg.v20.i14.3976.PubMedCentralCrossRefPubMed Beatty JKBA, Buret AG: Post-infectious irritable bowel syndrome: Mechanistic insights into chronic disturbances following enteric infection. World J Gastroenterol. 2014, 20 (14): 3976-3985. 10.3748/wjg.v20.i14.3976.PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Collins S: A role for the gut microbiota in IBS. Nat Rev Gastroenterol Hepatol. 2014, 11 (8): 497-505. 10.1038/nrgastro.2014.40.CrossRefPubMed Collins S: A role for the gut microbiota in IBS. Nat Rev Gastroenterol Hepatol. 2014, 11 (8): 497-505. 10.1038/nrgastro.2014.40.CrossRefPubMed
8.
Zurück zum Zitat DuPont H: Review article: evidence for the role of gut microbiota in irritable bowel syndorme and its potential influence on therapeutic targets. Aliment Pharmacol Ther. 2014, 39: 1033-1042. 10.1111/apt.12728.CrossRefPubMed DuPont H: Review article: evidence for the role of gut microbiota in irritable bowel syndorme and its potential influence on therapeutic targets. Aliment Pharmacol Ther. 2014, 39: 1033-1042. 10.1111/apt.12728.CrossRefPubMed
9.
Zurück zum Zitat de Salonen A, Vos WM, Palva A: Gastrointestinal microbiota in irritable bowel syndrome:present state and perspectives. Microbiology. 2010, 156: 3205-3215. 10.1099/mic.0.043257-0.CrossRef de Salonen A, Vos WM, Palva A: Gastrointestinal microbiota in irritable bowel syndrome:present state and perspectives. Microbiology. 2010, 156: 3205-3215. 10.1099/mic.0.043257-0.CrossRef
10.
Zurück zum Zitat Matricon JM, Gelot A, Piche T, Dapoigny M, Muller E, Ardid D: Review article:associations between immune activation, intestinal permeability and the irritable bowel syndrome. Aliment Pharmacol Ther. 2012, 36: 1009-1031. 10.1111/apt.12080.CrossRefPubMed Matricon JM, Gelot A, Piche T, Dapoigny M, Muller E, Ardid D: Review article:associations between immune activation, intestinal permeability and the irritable bowel syndrome. Aliment Pharmacol Ther. 2012, 36: 1009-1031. 10.1111/apt.12080.CrossRefPubMed
11.
Zurück zum Zitat LA Bruewer M, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A: Proinflammatory cytokines disrupt epithelial barrier funciton by apoptosis-independent mechanisms. J Immunol. 2003, 171: 6164-6172. 10.4049/jimmunol.171.11.6164.CrossRef LA Bruewer M, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A: Proinflammatory cytokines disrupt epithelial barrier funciton by apoptosis-independent mechanisms. J Immunol. 2003, 171: 6164-6172. 10.4049/jimmunol.171.11.6164.CrossRef
12.
Zurück zum Zitat Steck NMK, Schermann M, Haller D: Bacterial proteases in IBD and IBS. Gut. 2014, 61: 1610-1618. 10.1136/gutjnl-2011-300775.CrossRef Steck NMK, Schermann M, Haller D: Bacterial proteases in IBD and IBS. Gut. 2014, 61: 1610-1618. 10.1136/gutjnl-2011-300775.CrossRef
13.
Zurück zum Zitat Gecse K, Roka R, Ferrier L, Eutamene H, Cartier C, Ait-Belgnaoui A, Rosztoczy A, Izbeki F, Fioramonti J, Wittmann T, Bueno L: Increased faecal serine protease activity in diarrhoeic IBS patients: a colonic luminal factor impairing colonic permeability and sensitivity. Gut. 2008, 57 (5): 591-599. 10.1136/gut.2007.140210.CrossRefPubMed Gecse K, Roka R, Ferrier L, Eutamene H, Cartier C, Ait-Belgnaoui A, Rosztoczy A, Izbeki F, Fioramonti J, Wittmann T, Bueno L: Increased faecal serine protease activity in diarrhoeic IBS patients: a colonic luminal factor impairing colonic permeability and sensitivity. Gut. 2008, 57 (5): 591-599. 10.1136/gut.2007.140210.CrossRefPubMed
14.
Zurück zum Zitat Puthia MK, Lu J, Tan KS: Blastocystis ratti contains cysteine proteases that mediate interleukin-8 response from human intestinal epithelial cells in an NF-kappaB-dependent manner. Eukaryot Cell. 2008, 7 (3): 435-443. 10.1128/EC.00371-07.PubMedCentralCrossRefPubMed Puthia MK, Lu J, Tan KS: Blastocystis ratti contains cysteine proteases that mediate interleukin-8 response from human intestinal epithelial cells in an NF-kappaB-dependent manner. Eukaryot Cell. 2008, 7 (3): 435-443. 10.1128/EC.00371-07.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Puthia MK, Sio SWS, Lu J, Tan KSW: Blastocystis ratti Induces contact-independent apoptosis, F-actin rearrangement, and barrier function disruption in IEC-6 cells. Infect Immun. 2006, 74 (7): 4114-4123. 10.1128/IAI.00328-06.PubMedCentralCrossRefPubMed Puthia MK, Sio SWS, Lu J, Tan KSW: Blastocystis ratti Induces contact-independent apoptosis, F-actin rearrangement, and barrier function disruption in IEC-6 cells. Infect Immun. 2006, 74 (7): 4114-4123. 10.1128/IAI.00328-06.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Puthia MK, Vaithilingam A, Lu J, Tan KS: Degradation of human secretory immunoglobulin A by Blastocystis. Parasitol Res. 2005, 97 (5): 386-389. 10.1007/s00436-005-1461-0.CrossRefPubMed Puthia MK, Vaithilingam A, Lu J, Tan KS: Degradation of human secretory immunoglobulin A by Blastocystis. Parasitol Res. 2005, 97 (5): 386-389. 10.1007/s00436-005-1461-0.CrossRefPubMed
17.
Zurück zum Zitat Mirza H, Tan KS: Blastocystis exhibits inter- and intra-subtype variation in cysteine protease activity. Parasitol Res. 2009, 104 (2): 355-361. 10.1007/s00436-008-1203-1.CrossRefPubMed Mirza H, Tan KS: Blastocystis exhibits inter- and intra-subtype variation in cysteine protease activity. Parasitol Res. 2009, 104 (2): 355-361. 10.1007/s00436-008-1203-1.CrossRefPubMed
18.
Zurück zum Zitat Wu B, Yin J, Texier C, Roussel M, Tan KS: Blastocystis legumain is localized on the cell surface, and specific inhibition of its activity implicates a pro-survival role for the enzyme. J Biol Chem. 2010, 285 (3): 1790-1798. 10.1074/jbc.M109.049064.PubMedCentralCrossRefPubMed Wu B, Yin J, Texier C, Roussel M, Tan KS: Blastocystis legumain is localized on the cell surface, and specific inhibition of its activity implicates a pro-survival role for the enzyme. J Biol Chem. 2010, 285 (3): 1790-1798. 10.1074/jbc.M109.049064.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Yakoob J, Jafri W, Beg MA, Abbas Z, Naz S, Islam M, Khan R: Irritable bowel syndrome: is it associated with genotypes of Blastocystis hominis. Parasitol Res. 2010, 106 (5): 1033-1038. 10.1007/s00436-010-1761-x.CrossRefPubMed Yakoob J, Jafri W, Beg MA, Abbas Z, Naz S, Islam M, Khan R: Irritable bowel syndrome: is it associated with genotypes of Blastocystis hominis. Parasitol Res. 2010, 106 (5): 1033-1038. 10.1007/s00436-010-1761-x.CrossRefPubMed
20.
Zurück zum Zitat Stensvold CR: Comparison of Sequencing (Barcode Region) and Sequence-Tagged-Site PCR for Blastocystis Sub-typing. J Clin Microbiol. 2013, 51: 190-194. 10.1128/JCM.02541-12.PubMedCentralCrossRefPubMed Stensvold CR: Comparison of Sequencing (Barcode Region) and Sequence-Tagged-Site PCR for Blastocystis Sub-typing. J Clin Microbiol. 2013, 51: 190-194. 10.1128/JCM.02541-12.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Stensvold CR, Smith HV, Nagel R, Olsen KE, Traub RJ: Eradication of Blastocystis carriage with antimicrobials: reality or delusion?. J Clin Gastroenterol. 2009, 44 (2): 85-90. 10.1097/MCG.0b013e3181bb86ba.CrossRef Stensvold CR, Smith HV, Nagel R, Olsen KE, Traub RJ: Eradication of Blastocystis carriage with antimicrobials: reality or delusion?. J Clin Gastroenterol. 2009, 44 (2): 85-90. 10.1097/MCG.0b013e3181bb86ba.CrossRef
22.
Zurück zum Zitat Nagel R, Cuttell L, Stensvold CR, Mills PC, Bielefeldt-Ohmann H, Traub RJ: Blastocystis subtypes in symptomatic and asymptomatic family members and pets and response to therapy. Intern Med J. 2012, 42 (11): 1187-1195. 10.1111/j.1445-5994.2011.02626.x.CrossRefPubMed Nagel R, Cuttell L, Stensvold CR, Mills PC, Bielefeldt-Ohmann H, Traub RJ: Blastocystis subtypes in symptomatic and asymptomatic family members and pets and response to therapy. Intern Med J. 2012, 42 (11): 1187-1195. 10.1111/j.1445-5994.2011.02626.x.CrossRefPubMed
23.
Zurück zum Zitat Jones DR: A medium for investigating the breakdown of pectin by bacteria. Nature. 1946, 158 (4018): 625-10.1038/158625b0.CrossRefPubMed Jones DR: A medium for investigating the breakdown of pectin by bacteria. Nature. 1946, 158 (4018): 625-10.1038/158625b0.CrossRefPubMed
24.
Zurück zum Zitat Wong KH, Ng GC, Lin RT, Yoshikawa H, Taylor MB, Tan KS: Predominance of subtype 3 among Blastocystis isolates from a major hospital in Singapore. Parasitol Res. 2008, 102 (4): 663-670. 10.1007/s00436-007-0808-0.CrossRefPubMed Wong KH, Ng GC, Lin RT, Yoshikawa H, Taylor MB, Tan KS: Predominance of subtype 3 among Blastocystis isolates from a major hospital in Singapore. Parasitol Res. 2008, 102 (4): 663-670. 10.1007/s00436-007-0808-0.CrossRefPubMed
25.
Zurück zum Zitat Kumar S, Tamura K, Jakobsen IB, Nei M: MEGA2:Molecular Evolutionary Genetics Analysis software. Bioinformatics. 2001, 17 (12): 1244-1245. 10.1093/bioinformatics/17.12.1244.CrossRefPubMed Kumar S, Tamura K, Jakobsen IB, Nei M: MEGA2:Molecular Evolutionary Genetics Analysis software. Bioinformatics. 2001, 17 (12): 1244-1245. 10.1093/bioinformatics/17.12.1244.CrossRefPubMed
26.
Zurück zum Zitat Roberts T, Ellis J, Harkness J, Marriott D, Starl D: Treatment failure in patients with chronic Blastocystis infection. J Med Microbiol. 2014, 63: 252-257. 10.1099/jmm.0.065508-0.CrossRefPubMed Roberts T, Ellis J, Harkness J, Marriott D, Starl D: Treatment failure in patients with chronic Blastocystis infection. J Med Microbiol. 2014, 63: 252-257. 10.1099/jmm.0.065508-0.CrossRefPubMed
27.
Zurück zum Zitat LA Dunn TK, Vanelle P, Juspin T, Crozet MD, Terme T, Upcroft P, Upcroft JA: Development of metronidazole-resistant lines of Blastocystis spp. Parasitol Res. 2012, 111: 441-450. 10.1007/s00436-012-2860-7.CrossRef LA Dunn TK, Vanelle P, Juspin T, Crozet MD, Terme T, Upcroft P, Upcroft JA: Development of metronidazole-resistant lines of Blastocystis spp. Parasitol Res. 2012, 111: 441-450. 10.1007/s00436-012-2860-7.CrossRef
28.
Zurück zum Zitat Mirza H, Teo JDW, Upcroft J, Tan KSW: A rapid, high-throughput viability assay for Blastocystis spp. reveals metronidazole resistance and extensive subtype-dependent variations in drug susceptibilities. Antimicrob Agents Chemother. 2010, 55 (2): 637-648. 10.1128/AAC.00900-10.PubMedCentralCrossRefPubMed Mirza H, Teo JDW, Upcroft J, Tan KSW: A rapid, high-throughput viability assay for Blastocystis spp. reveals metronidazole resistance and extensive subtype-dependent variations in drug susceptibilities. Antimicrob Agents Chemother. 2010, 55 (2): 637-648. 10.1128/AAC.00900-10.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Zierdt CH, Swan JC, Hosseini J: In vitro response of Blastocystis hominis to antiprotozoal drugs. J Protozool. 1983, 30 (2): 332-334. 10.1111/j.1550-7408.1983.tb02925.x.CrossRefPubMed Zierdt CH, Swan JC, Hosseini J: In vitro response of Blastocystis hominis to antiprotozoal drugs. J Protozool. 1983, 30 (2): 332-334. 10.1111/j.1550-7408.1983.tb02925.x.CrossRefPubMed
30.
Zurück zum Zitat Vdovenko AA, Williams JE: Blastocystis hominis: neutral red supravital staining and its application to in vitro drug sensitivity testing. Parasitol Res. 2000, 86 (7): 573-581. 10.1007/PL00008533.CrossRefPubMed Vdovenko AA, Williams JE: Blastocystis hominis: neutral red supravital staining and its application to in vitro drug sensitivity testing. Parasitol Res. 2000, 86 (7): 573-581. 10.1007/PL00008533.CrossRefPubMed
31.
Zurück zum Zitat Dunn LA, Boreham PF: The in-vitro activity of drugs against Blastocystis hominis. J Antimicrob Chemother. 1991, 27 (4): 507-516. 10.1093/jac/27.4.507.CrossRefPubMed Dunn LA, Boreham PF: The in-vitro activity of drugs against Blastocystis hominis. J Antimicrob Chemother. 1991, 27 (4): 507-516. 10.1093/jac/27.4.507.CrossRefPubMed
32.
Zurück zum Zitat Lamp KCFC, Klutman NE, Lacy MK: Pharmacokinetics and Pharmacodynamics of the Nitroimidazole Antimicrobials. Clin Pharmacokinet. 1999, 36 (5): 353-373. 10.2165/00003088-199936050-00004.CrossRefPubMed Lamp KCFC, Klutman NE, Lacy MK: Pharmacokinetics and Pharmacodynamics of the Nitroimidazole Antimicrobials. Clin Pharmacokinet. 1999, 36 (5): 353-373. 10.2165/00003088-199936050-00004.CrossRefPubMed
33.
Zurück zum Zitat Coyle CMJV, Louis Weiss M, Herbert Tanowitz B: Blastocystis: to treat or not to treat. Clin Prac. 2012, 54: 105-107. Coyle CMJV, Louis Weiss M, Herbert Tanowitz B: Blastocystis: to treat or not to treat. Clin Prac. 2012, 54: 105-107.
34.
Zurück zum Zitat Menardi GGJ: Bioavailability of oral antibiotics in children with short bowel syndrome. J Paediatr Surg. 1984, 19 (1): 84-86. 10.1016/S0022-3468(84)80023-8.CrossRef Menardi GGJ: Bioavailability of oral antibiotics in children with short bowel syndrome. J Paediatr Surg. 1984, 19 (1): 84-86. 10.1016/S0022-3468(84)80023-8.CrossRef
35.
Zurück zum Zitat Davidson RN dBM, Ritmeijer K: Paromomycin. Trans R Soc Trop Med Hyg. 2008, 103: 653-660. 10.1016/j.trstmh.2008.09.008.CrossRefPubMed Davidson RN dBM, Ritmeijer K: Paromomycin. Trans R Soc Trop Med Hyg. 2008, 103: 653-660. 10.1016/j.trstmh.2008.09.008.CrossRefPubMed
36.
Zurück zum Zitat van Hellemond JMN, Koelewijn R, Wismans PJ, Genderen PJ: Is paromomycin the drug of choice for eradication of Blastocystisin adults?. J Infect Chemother. 2013, 19: 545-548. 10.1007/s10156-012-0496-2.CrossRefPubMed van Hellemond JMN, Koelewijn R, Wismans PJ, Genderen PJ: Is paromomycin the drug of choice for eradication of Blastocystisin adults?. J Infect Chemother. 2013, 19: 545-548. 10.1007/s10156-012-0496-2.CrossRefPubMed
37.
Zurück zum Zitat Adachi JA, Rifaximin DH: A novel nonabsorbed rifamycin for gastrointestinal disorders. Clin Infect Dis. 2006, 2006 (42): 541-547. 10.1086/499950.CrossRef Adachi JA, Rifaximin DH: A novel nonabsorbed rifamycin for gastrointestinal disorders. Clin Infect Dis. 2006, 2006 (42): 541-547. 10.1086/499950.CrossRef
38.
Zurück zum Zitat Pimentel M, Lembo A, Chey W, Zakko S, Ringel Y, Yu J, Mareya S, Shaw A, Bortey E, Forbes W: Rifaximin therapy for patients with irritable bowel syndrome wihout constipation. NEJM. 2011, 364: 22-32. 10.1056/NEJMoa1004409.CrossRefPubMed Pimentel M, Lembo A, Chey W, Zakko S, Ringel Y, Yu J, Mareya S, Shaw A, Bortey E, Forbes W: Rifaximin therapy for patients with irritable bowel syndrome wihout constipation. NEJM. 2011, 364: 22-32. 10.1056/NEJMoa1004409.CrossRefPubMed
39.
Zurück zum Zitat Amenta M, Dalle Nogare ER, Colomba C, Prestileo T, Di Lorenzo F, Fundaro S, Colomba A, Ferrieri A: Intestinal protozoa in HIV-infected patients: effect of rifaximin in Cryptosporidium parvum and Blastocystis hominis infections. J Chemother. 1999, 11 (5): 391-395. 10.1179/joc.1999.11.5.391.CrossRefPubMed Amenta M, Dalle Nogare ER, Colomba C, Prestileo T, Di Lorenzo F, Fundaro S, Colomba A, Ferrieri A: Intestinal protozoa in HIV-infected patients: effect of rifaximin in Cryptosporidium parvum and Blastocystis hominis infections. J Chemother. 1999, 11 (5): 391-395. 10.1179/joc.1999.11.5.391.CrossRefPubMed
40.
Zurück zum Zitat Gadkariem EA, Belal F, Abounassif MA, El-Obeid HA, Ibrahim KE: Stability studies on diloxanide furoate: effect of pH, temperature, gastric and intestinal fluids. Il Farmaco. 2004, 59: 323-329. 10.1016/j.farmac.2003.11.015.CrossRefPubMed Gadkariem EA, Belal F, Abounassif MA, El-Obeid HA, Ibrahim KE: Stability studies on diloxanide furoate: effect of pH, temperature, gastric and intestinal fluids. Il Farmaco. 2004, 59: 323-329. 10.1016/j.farmac.2003.11.015.CrossRefPubMed
41.
Zurück zum Zitat Li LH, Zhang XP, Lv S, Zhang L, Yoshikawa H, Wu Z, Steinmann P, Utzinger J, Tong X, Chen S, Zhou X: Cross-sectional surveys and subtype classification of human Blastocystis isolates from four epidemiological settings in China. Parasitol Res. 2007, 102 (1): 83-90. 10.1007/s00436-007-0727-0.CrossRefPubMed Li LH, Zhang XP, Lv S, Zhang L, Yoshikawa H, Wu Z, Steinmann P, Utzinger J, Tong X, Chen S, Zhou X: Cross-sectional surveys and subtype classification of human Blastocystis isolates from four epidemiological settings in China. Parasitol Res. 2007, 102 (1): 83-90. 10.1007/s00436-007-0727-0.CrossRefPubMed
42.
Zurück zum Zitat Poirier P, Wawrzyniak I, Albert A, El Alaoui H, Delbac F, Livrelli V: Development and evaluation of a real-time PCR assay for detection and quantification of blastocystis parasites in human stool samples: prospective study of patients with hematological malignancies. J Clin Microbiol. 2011, 49 (3): 975-983. 10.1128/JCM.01392-10.PubMedCentralCrossRefPubMed Poirier P, Wawrzyniak I, Albert A, El Alaoui H, Delbac F, Livrelli V: Development and evaluation of a real-time PCR assay for detection and quantification of blastocystis parasites in human stool samples: prospective study of patients with hematological malignancies. J Clin Microbiol. 2011, 49 (3): 975-983. 10.1128/JCM.01392-10.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Tasova Y, Sahin B, Koltas S, Paydas S: Clinical significance and frequency of Blastocystis hominis in Turkish patients with hematological malignancy. Acta Med Okayama. 2000, 54 (3): 133-136.PubMed Tasova Y, Sahin B, Koltas S, Paydas S: Clinical significance and frequency of Blastocystis hominis in Turkish patients with hematological malignancy. Acta Med Okayama. 2000, 54 (3): 133-136.PubMed
44.
Zurück zum Zitat Olivo-Diaz A, Romero-Valdovinos M, Gudino-Ramirez A, Reyes-Gordillo J, Jimenez-Gonzalez D, Ramirez-Miranda M, Martinez-Flores W, Martinez-Hernandez F, Flisser A, Maravilla P: Findings related to IL-8 and IL-10 gene polymorphisms in a Mexican patient population with irritable bowel syndrome infected with Blastocystis. Parasitol Res. 2012, 111: 487-491. 10.1007/s00436-012-2830-0.CrossRefPubMed Olivo-Diaz A, Romero-Valdovinos M, Gudino-Ramirez A, Reyes-Gordillo J, Jimenez-Gonzalez D, Ramirez-Miranda M, Martinez-Flores W, Martinez-Hernandez F, Flisser A, Maravilla P: Findings related to IL-8 and IL-10 gene polymorphisms in a Mexican patient population with irritable bowel syndrome infected with Blastocystis. Parasitol Res. 2012, 111: 487-491. 10.1007/s00436-012-2830-0.CrossRefPubMed
45.
Zurück zum Zitat Singh K, Chang C, Gershwin M: IgA deficiency and autoimmunity. Autoimmun Rev. 2014, 13: 163-177. 10.1016/j.autrev.2013.10.005.CrossRefPubMed Singh K, Chang C, Gershwin M: IgA deficiency and autoimmunity. Autoimmun Rev. 2014, 13: 163-177. 10.1016/j.autrev.2013.10.005.CrossRefPubMed
46.
Zurück zum Zitat Stoop JW, Zegers B, Sander P, Ballieux R: Serum immunoglobulin levels in healthy children and adults. Clin exp Immunol. 1969, 4: 101-112.PubMedCentralPubMed Stoop JW, Zegers B, Sander P, Ballieux R: Serum immunoglobulin levels in healthy children and adults. Clin exp Immunol. 1969, 4: 101-112.PubMedCentralPubMed
47.
Zurück zum Zitat Suresh K, Venilla GD, Tan TC, Rohela M: In vivo encystation of Blastocystis hominis. Parasitol Res. 2009, 104: 303-307. 10.1007/s00436-009-1340-1.CrossRef Suresh K, Venilla GD, Tan TC, Rohela M: In vivo encystation of Blastocystis hominis. Parasitol Res. 2009, 104: 303-307. 10.1007/s00436-009-1340-1.CrossRef
48.
Zurück zum Zitat Stensvold CR, Arendrup M, Jespersgaard C, Molbak K, Nielsen H: Detecting Blastocystis using parasitologic and DNA-based methods: a comparative study. Diag Microbiol Inf Dis. 2007, 59: 303-307. 10.1016/j.diagmicrobio.2007.06.003.CrossRef Stensvold CR, Arendrup M, Jespersgaard C, Molbak K, Nielsen H: Detecting Blastocystis using parasitologic and DNA-based methods: a comparative study. Diag Microbiol Inf Dis. 2007, 59: 303-307. 10.1016/j.diagmicrobio.2007.06.003.CrossRef
49.
Zurück zum Zitat Hussain R, Jaferi W, Zuberi A, Baqai R, Abrar N, Ahmed A, Zaman V: Significantly increased IgG2 subclass antibody levels in patients with irritable Bowel Syndrome. Am J Trop Med Hyg. 1997, 56: 301-306.PubMed Hussain R, Jaferi W, Zuberi A, Baqai R, Abrar N, Ahmed A, Zaman V: Significantly increased IgG2 subclass antibody levels in patients with irritable Bowel Syndrome. Am J Trop Med Hyg. 1997, 56: 301-306.PubMed
50.
Zurück zum Zitat Abou Gamra M, Elwakil H, El Deeb H, Khalifa K, Abd Elhafiz H: The potential us of 29 kDa protein as a marker of pathogenicity and diagnosis of symptomatic infections with Blastocystis hominis. Parasitol Res. 2011, 108: 1139-1146. 10.1007/s00436-010-2156-8.CrossRefPubMed Abou Gamra M, Elwakil H, El Deeb H, Khalifa K, Abd Elhafiz H: The potential us of 29 kDa protein as a marker of pathogenicity and diagnosis of symptomatic infections with Blastocystis hominis. Parasitol Res. 2011, 108: 1139-1146. 10.1007/s00436-010-2156-8.CrossRefPubMed
Metadaten
Titel
Clinical pilot study: efficacy of triple antibiotic therapy in Blastocystis positive irritable bowel syndrome patients
verfasst von
Robyn Nagel
Helle Bielefeldt-Ohmann
Rebecca Traub
Publikationsdatum
01.08.2014
Verlag
BioMed Central
Erschienen in
Gut Pathogens / Ausgabe 1/2014
Elektronische ISSN: 1757-4749
DOI
https://doi.org/10.1186/s13099-014-0034-0

Weitere Artikel der Ausgabe 1/2014

Gut Pathogens 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.