Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Systematic review and meta-analysis of traditional Chinese medicine in the treatment of constipation-predominant irritable bowel syndrome

  • Dan-yan Li ,

    Contributed equally to this work with: Dan-yan Li, Yun-kai Dai

    Roles Writing – original draft

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Yun-kai Dai ,

    Contributed equally to this work with: Dan-yan Li, Yun-kai Dai

    Roles Supervision, Writing – review & editing

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Yun-zhan Zhang,

    Roles Investigation

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Meng-xin Huang,

    Roles Methodology

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Ru-liu Li,

    Roles Supervision

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Jia Ou-yang,

    Roles Visualization

    Affiliation Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China

  • Wei-jing Chen,

    Roles Data curation, Supervision

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

  • Ling Hu

    Roles Supervision

    drhuling@163.com

    Affiliation Institute of Gastroenterology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China

Abstract

Aim

This meta-analysis analyzed the efficacy and safety of traditional Chinese medicine (TCM) for the treatment of irritable bowel syndrome with constipation (IBS-C).

Methods

We searched seven electronic databases for randomized controlled trials investigating the efficacy of TCM in the treatment of IBS-C. The search period was from inception to June 1, 2017. Eligible RCTs compared TCM with cisapride and mosapride. Article quality was evaluated with the Cochrane Risk Bias Tool in the Cochrane Handbook by two independent reviewers. Begg’s test was performed to evaluate publication bias. Review Manager 5.3 and Stata 12.0 were used for analyses.

Results

Eleven eligible studies comprising a total of 906 participants were identified. In the primary outcome, TCM showed significant improvement in overall clinical efficacy compared with cisapride and mosapride (odds ratio [OR] = 4.00; 95% confidence interval [CI]: 2.74,5.84; P < 0.00001). In terms of secondary outcomes, TCM significantly alleviated abdominal pain (OR = 5.69; 95% CI: 2.35, 13.78; P = 0.0001), defecation frequency (OR = 4.38; 95% CI: 1.93, 9.93. P = 0.0004), and stool form (OR = 4.96; 95% CI: 2.11, 11.65; P = 0.0002) in the treatment group as compared to the control group. A lower recurrence rate was associated with TCM as compared to cisapride and mosapride (OR = 0.15; 95% CI: 0.08, 0.27; P < 0.00001). No adverse effects were observed during TCM treatment.

Conclusions

TCM showed greater improvement in terms of clinical efficacy in the treatment of IBS-C than cisapride and mosapride, although it was not possible to draw a definitive conclusion due to the small sample size, high risk, and low quality of the studies. Large multi-center and long-term high-quality randomized control trials are needed.

1. Introduction

Irritable bowel syndrome (IBS) is one of the most common functional bowel disorders, which is characterized by recurrent abdominal pain or discomfort with accompanying disturbances in defecation that cannot be attributed to any demonstrable biochemical, structural, or anatomical abnormality [1, 2]. Thus, the diagnosis of IBS is based on symptoms, according to which the disorder is classified as IBS-C (IBS with pain or discomfort and predominant constipation), IBS-D (IBS with diarrhea), IBS-M (mixed IBS), or IBS-U (unsubtyped) [3, 4]. Epidemiologic studies have reported a high prevalence of IBS ranging from 5%–22% in various countries [5, 6], 2.9%–15.6% in Asian countries [7], and 0.82%–11.5% in China [8, 9]. Although IBS causes no physical injury to patients, it reduces their quality of life and consumes a significant amount of medical resources due to its chronicity and frequency of symptoms [10].

Drugs that promote gastrointestinal motility are the main treatment for IBS-C; for example, the 5-hydroxytryptamine (serotonin; 5-HT) agonists cisapride and mosapride provide effective relief of the predominant symptoms. However, long-term use of these drugs is associated with a risk of cardiovascular events [11]. Many IBS patients seek alternative treatments such as traditional Chinese medicine (TCM) [1214], in which treatment is based on syndrome differentiation. TCM represent one aspect of Chinese medical philosophy that is characterized by its emphasis on maintaining and restoring balance[15].

A series of systematic reviews evaluating the effectiveness of TCM for treating IBS-D treatment have been published [1620]. However, there is little information on the efficacy of TCM for the treatment of IBS-C. To address this issue, we carried out a meta-analysis to evaluate the safety and efficacy of TCM for IBS-C treatment.

2. Methods

2.1. Data sources and search strategy

Seven electronic databases including PubMed, Springer, EMBASE, CNKI(China National Knowledge Infrastructure), CBM (Chinese Biomedicine Database), WanFang, and VIP (Chinese Scientific Journals Database) were searched for randomized controlled trials (RCTs) evaluating the effects of TCM and gastrointestinal motility drugs on the clinical outcome of IBS-C. The search period was from inception to June 1, 2017. The following key words were used for our search: “Traditional Chinese medicine”, “irritable bowel syndrome”, “IBS”, “Constipation Type of Irritable Bowel Syndrome”, “IBS-C”, “clinical research”, and “clinical trial”.

2.2. Selection criteria

Two reviewers (D.-Y.L. and Y.-K.D.) independently reviewed the full-text versions of all the articles retrieved in the literature search to identify eligible studies. Conflicts in study selection were resolved by a third reviewer (J.-O.Y). Inclusion criteria were as follows: (1) IBS-C was definitively diagnosed according to Rome II[21], III[22], or IV criteria[23]; (2) clinical trial; (3) treatment groups used only traditional Chinese medicine (decoction, granules, capsule or tablet), while control groups used mosapride or cisapride (capsule or tablet); (4) randomized controlled trial (RCT); (5) duration of the treatment was at least 4 weeks for all groups; and (6) published in any language. Exclusion criteria were as follows: (1) not IBS-C; (2) IBS-D, IBS-M, IBS-U; (3) duplicate publication; (4) included subjects with severe enteric disease or who developed a malignancy, heart failure, or renal failure during the study period, and (5) publication was a commentary, editorial, reviews or case report.

2.3 Efficacy evaluation index

The indicator of primary outcome was overall clinical efficacy; secondary outcome indicators were symptom improvement rate, recurrence rate, and adverse reaction. According to the TCM Illness Diagnosis Affect Standard and Guidance Principle of Clinical Study on New Drug of Traditional Chinese Medicine [24, 25], efficacy evaluation was divided into four grades: cured, markedly effective, effective, and ineffective. The nimodipine method [24] was used to assess changes in the efficacy index, which was calculated with the formula [(pre-treatment symptom score − symptom score after treatment / pre-treatment symptom score)] × 100%.

2.4 Literature quality evaluation

The Cochrane Risk Bias Tool in the Cochrane Handbook for Systematic Reviews of Interventions was used to assess the quality of included studies, which supplemented by Jadad score[26]. Evaluation of methodological quality was also assessed by two independent reviewers, and a third reviewer was consulted when there were discrepancies.

2.5. Data extraction

Two reviewers (D.-Y.L. and Y.-K.D.) used a standardized data extraction form to independently extract data from studies that met the inclusion criteria, including (1) general information, including name of the first author, publication year, sample size, age, disease duration, trial duration, and intervention; (2) details of the study design, including descriptions of randomization methods, blinding, allocation concealment, and bias prevention; (3) clinical outcomes at the end of the treatment, number of withdrawals or dropouts, and follow-up periods; and (4) adverse events during treatment. Extracted data were cross-checked by the two reviewers. In cases of uncertainty about eligibility, a third reviewer was consulted.

2.6. Statistical analysis

Outcomes for which data could not be compared directly across studies were synthesized qualitatively, as were outcomes for which insufficient data were reported across studies. Outcomes for which sufficient, equivalent data were reported across studies were meta -analyzed using Forest plots generated with RevMan 5.3[27]. Enumerated data were analyzed using OR or relative risk (RR) and 95% CI. The χ2 test and the inconsistency index statistic (I2) for heterogeneity were conducted [28]. If substantial heterogeneity existed (I2> 50% or P< 0.05), a random effect model was applied. If there was no observed heterogeneity, fixed effect models were chosen [29]. A sensitivity analysis was done to explore potential sources of heterogeneity. Begg’s test was performed to evaluate publication bias [30]. Review Manager 5.3 and Stata 12.0 were used for analyses.

3. Results

3.1 Study description

The literature search identified 1062 studies. The detailed screening process is shown in Fig 1(Fig 1 Flow diagram of literature search). Based on the inclusion and exclusion criteria, 11 RCTs [3141] were ultimately included in the meta-analysis, comprising 906 participants (494 in the trial group and 412 in the control group) ranging in age from 17 to 72 years old; in two studies [32, 40] the participants’ ages were not mentioned. Disease duration was between 3 months and 12 years; three articles [35, 36, 38] did not mention the duration. The course of treatment was between 4 and 8 weeks. Ten of the treatment groups used a Chinese medicine decoction which refers to the liquid formulation that is obtained by boiling pieces or coarse grains of herbs in water or soaking the crude medicine in boiling water followed by filtration [42].while one used granules [35]. In the control group, four articles used cisapride [3134] and seven articles used mosapride [3541]. All selected RCTs reported overall clinical efficacy at the end of the treatment, and three [3133] reported recurrence rate and follow-up periods. The characteristics of the studies are listed in Table 1.

thumbnail
Table 1. Characteristics of the studies included in the meta-analysis.

https://doi.org/10.1371/journal.pone.0189491.t001

3.2 Methodological quality

All included studies were randomized studies with comparable baselines. Two studies [35, 39] used random number tables: one [40] according to the principle of minimum distribution of unbalanced index, and one [32] as a simple random table. The remaining seven studies did not mention a specific method. Ten studies did not mention double blinding or allocation concealment; only one [35] mentioned the use of sealed envelopes although a non-blinded approach was adopted. One trial [40] reported patient dropout, but an Intention-to-Treat (ITT) analysis was not performed. For other types of bias, seven studies [32, 3537, 3941] were rated as unclear risk due to the lack of information on age, disease duration, and dosage of Chinese medicine. This meta-analysis was considered as having a high risk of bias (Fig 2 Methodological quality assessment of the risk of bias).A description of the evaluation of methodological quality of the 11 trials can be found in Table 2.

thumbnail
Fig 2. Methodological quality assessment of the risk of bias.

https://doi.org/10.1371/journal.pone.0189491.g002

thumbnail
Table 2. Evaluation of methodological quality of the included studies.

https://doi.org/10.1371/journal.pone.0189491.t002

3.3 Primary outcome: Overall clinical efficacy

Overall clinical efficacy was evaluated in 11 trials. The study included 906 patients; 494 were assigned to treatment groups, whereas 412 were assigned to control groups. TCM significantly improved overall clinical efficacy as compared to cisapride and mosapride (OR = 4.00; 95% CI: 2.74, 5.84; P < 0.00001) (Fig 3 Forest plot of primary outcomes (fixed effect model). No heterogeneity was observed (χ2 = 7.52, P = 0.68, I2 = 0%). Funnel plot analysis demonstrated evidence of publication bias (Begg’s test, P = 0.013) (Fig 4 Begg’s funnel plot analysis of primary outcomes).

thumbnail
Fig 3. Forest plot of primary outcomes (fixed effect model).

https://doi.org/10.1371/journal.pone.0189491.g003

thumbnail
Fig 4. Begg’s funnel plot analysis of primary outcomes.

https://doi.org/10.1371/journal.pone.0189491.g004

3.4 Secondary outcomes

Abdominal pain.

Five of the 11 studies [32, 35, 36, 39, 40] reported the outcome of abdominal pain improvement; three of these [32, 35, 36] used symptom scores, although the scoring criteria were inconsistent. The remaining two studies [39, 40], in which evaluation was based on efficacy, were incorporated into the meta-analysis. TCM resulted in greater abdominal pain alleviation relative to controls (OR = 5.69; 95% CI: 2.35, 13.78; P = 0.0001), with no statistical heterogeneity (χ2 = 0.05, P = 0.82, I2 = 0%) (Fig 5 Forest plot of improvements in abdominal pain (fixed effects model)).

thumbnail
Fig 5. Forest plot of improvements in abdominal pain (fixed effects model).

https://doi.org/10.1371/journal.pone.0189491.g005

Defecation frequency.

Three studies [32, 39, 40] reported an alleviation in defecation frequency. One [32] used symptom scores to assess the alleviation whereas the other two [39, 40] evaluated the efficacy and were therefore included in our analysis. TCM significantly alleviated defecation frequency as compared to control groups (OR = 4.38; 95% CI: 1.93, 9.93. P = 0.0004). There was no apparent heterogeneity (χ2 = 0.99, P = 0.32, I2 = 0%) (Fig 6 Forest plot of the improvement in defecation frequency (fixed effects model)).

thumbnail
Fig 6. Forest plot of the improvement in defecation frequency (fixed effects model).

https://doi.org/10.1371/journal.pone.0189491.g006

Stool form.

Four studies [32, 36, 39, 40] reported an alleviation in stool form according to symptoms. Two [39, 40] used the same criteria and were included in the analysis. TCM alleviated stool form relative to the control group (OR = 4.96; 95% CI: 2.11, 11.65; P = 0.0002); there was no heterogeneity observed (χ2 = 0.02, P = 0.89, I2 = 0%) (Fig 7 Forest plot of the improvement in stool form (fixed effects model)). There were limited descriptions in the literature of other symptoms such as abnormal bowel movements, abdominal tenderness [32], incomplete evacuation, fatigue [36] or anorexia [40]; this information was therefore only useful for a qualitative analysis.

thumbnail
Fig 7. Forest plot of the improvement in stool form (fixed effects model).

https://doi.org/10.1371/journal.pone.0189491.g007

3.5 Recurrence rate and adverse events

Among included studies, three trials [3133] reported recurrence rate during a 6-month follow-up after the treatment course. The fixed-effects model showed that TCM had a significantly lower recurrence rate than cisapride and mosapride (OR = 0.15; 95% CI: 0.08, 0.27; P < 0.00001). There was no observed heterogeneity (I2 = 0%, P = 0.57) (Fig 8 Forest plot of recurrence rate (fixed effects model)). Summary of meta-analysis outcomes can be found in Table 3.

thumbnail
Fig 8. Forest plot of recurrence rate (fixed effects model).

https://doi.org/10.1371/journal.pone.0189491.g008

Two studies [32, 40] mentioned adverse events in the control group. One patient withdrew from the study after reporting dizziness and dry mouth [40], and two cases of low abdominal pain, three cases of mild diarrhea, and one case of bowel movements were also reported [32]. No adverse effects were observed during TCM treatment.

Discussion

This meta-analysis investigated the efficacy of TCM in the treatment of IBS-C as compared to gastrointestinal motility drugs. The results showed that TCM produced greater alleviation in IBS-C symptoms than cisapride and mosapride. Data from only two trials were included in the analysis of secondary outcomes, and the analysis showed that TCM alleviated abdominal pain (OR = 5.69), defecation frequency (OR = 4.38) and stool form (OR = 4.96) as compared to the control group. Furthermore, patients treated with TCM had a significantly lower recurrence rate than controls (OR = 0.15), with no adverse events. For instance, in one study [35] patients were aware of which treatment they received, whereas another study [31] excluded one patient due to adverse effects but an ITT analysis was not performed, potentially leading to incomplete outcome data. Therefore, although TCM appears to be superior to gastrointestinal motility drugs for the treatment of IBS-C, the efficacy of traditional Chinese medicine requires further study.

The pathophysiology of IBS is not fully understood; it is thought to involve many factors, including gastrointestinal dysmotility [43], visceral hypersensitivity [44, 45], intestinal mucosa activation [4], increased intestinal permeability [46], inflammation, epigenetics, and genetics [47]. Many studies have reported that TCM is effective for IBS-C treatment. Ma Zhi Jiang Zhuo was shown to relieve constipation symptoms in a rat model of IBS-C, possibly by increasing the rate of small intestine propulsion and reducing 5-HT level in the colon [48]. Maziren pills increased visceral hypersensitivity by decreasing 5-HT levels in IBS-C rats [49]. Clinical trials have shown that flavored Maren pills improved abdominal symptoms and constipation in IBS-C by modulating the balance between pro- and anti-inflammatory cytokines—i.e., by decreasing interleukin (IL)-6 and increasing IL-10 levels [50]. On the other hand, clinical data have shown that liver-discharging, qi-regulating, and intestine-moistening therapies can relieve abdominal pain and improve constipation symptoms, possibly by reducing plasma vasoactive intestinal peptide (VIP) and somatostatin (SS) levels [51]. Kuanzhongjieyutang can improve IBS symptoms and patients’ quality of life by reducing plasma levels of VIP, SS, and 5-HT and modulating the brain-gut axis, thereby decreasing visceral hypersensitivity and improving intestinal motor function [52]. Ballast capsules can improve IBS symptoms by increasing motilin and decreasing endothelin levels [53]. Additional studies of these herbal formulae are needed in order to clarify the mechanisms underlying these effects.

There were several limitations to this meta-analysis. First, the course of treatment was between 4 and 8 weeks, and three trials [3133] reported a 6-month follow-up, which was not sufficiently long to evaluate the long-term efficacy of TCM for IBS-C in all included studies. Second, only three studies [33, 37, 39] reported safety evaluation. In one, routine blood, urine, and stool tests were performed and liver and kidney function were evaluated before and after treatment [33]. In the other two studies, enteroscopy [37] and electrocardiography [39] were performed in addition to these tests. Results from all of the tests were normal. The safety of TCM must be confirmed in future studies. Third, although the results showed that disease recurrence rate in patients treated by TCM was relatively low, this may be unreliable due to the limited number of studies that were suitable for analysis [3133]. Fourth, dropouts were reported in one study [40], but ITT analysis was not performed, which could lead to bias in evaluating the clinical efficacy of the intervention. Finally, our meta-analysis included published studies that were conducted in China and published in Chinese, only, and some of the unpublished data were not available for various reasons. In general, the studies had a high risk of bias and low methodological quality. Whether the effect size of TCM would be similar in large-scale trials remains to be determined.

Conclusions

IBM-C patients treated with TCM showed greater improvement than those treated with cisapride or mosapride. However, it is difficult to draw definitive conclusions from our meta-analysis due to the small sample size, high heterogeneity, and low quality of the included studies. Large, multi-center, long-term, and high-quality RCTs are needed to confirm whether TCM is a viable alternative to conventional drugs for the treatment of IBS-C.

Supporting information

Acknowledgments

This research was supported by National Natural Science Foundation of China, No.81373563; Innovation team to foster scientific research projects of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine (2016) No.7, No.2016KYTD07; Construction of high-level university of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine (2016) No.64; National Natural Science Foundation of China, No.81774238.

References

  1. 1. Chang JY, Talley NJ. Current and emerging therapies in irritable bowel syndrome: from pathophysiology to treatment. Trends in pharmacological sciences. 2010; 31:326–334. pmid:20554042
  2. 2. Longstreth GF, Thompson WG, Chey WD, Houghton LA, Mearin F, Spiller RC. Functional bowel disorders. Gastroenterology. 2006; 130:1480–1491. pmid:16678561
  3. 3. Krogius-Kurikka L, Lyra A, Malinen E, Aarnikunnas J, Tuimala J, Paulin L, et al. Microbial community analysis reveals high level phylogenetic alterations in the overall gastrointestinal microbiota of diarrhoea-predominant irritable bowel syndrome sufferers. BMC gastroenterology. 2009; 9:95. pmid:20015409
  4. 4. Camilleri M, Lasch K, Zhou W. Irritable bowel syndrome: methods, mechanisms, and pathophysiology. The confluence of increased permeability, inflammation, and pain in irritable bowel syndrome. American journal of physiology Gastrointestinal and liver physiology. 2012; 303:G775–785. pmid:22837345
  5. 5. Lovell RM, Ford AC. Global prevalence of and risk factors for irritable bowel syndrome: a meta-analysis. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2012; 10:712–721.
  6. 6. Grundmann O, Yoon SL. Irritable bowel syndrome: epidemiology, diagnosis and treatment: an update for health-care practitioners. Journal of gastroenterology and hepatology. 2010; 25:691–699. pmid:20074154
  7. 7. Gwee KA, Bak YT, Ghoshal UC, Gonlachanvit S, Lee OY, Fock KM, et al. Asian consensus on irritable bowel syndrome. Journal of gastroenterology and hepatology. 2010; 25:1189–1205. pmid:20594245
  8. 8. Pan G, Lu S, Ke M, Han S, Guo H, Fang X. An epidemiologic study of irritable bowel syndrome in Beijing—a stratified randomized study by clustering sampling. Zhonghua Liu Xing Bing Xue Za Zhi. 2000; 21:26–29. pmid:11860753
  9. 9. Xiong LS, Chen MH, Chen HX, Xu AG, Wang WA, Hu PJ. A population-based epidemiologic study of irritable bowel syndrome in Guangdong province. Zhonghua Yi Xue Za Zhi. 2004;84:278–281. pmid:15059507
  10. 10. Everhart JE, Ruhl CE. Burden of digestive diseases in the United States part I: overall and upper gastrointestinal diseases. Gastroenterology. 2009; 136:376–386. pmid:19124023
  11. 11. Brandt LJ, Chey WD, Foxx-Orenstein AE, Schiller LR, Schoenfeld PS, Spiegel BM, et al. An evidence-based position statement on the management of irritable bowel syndrome. The American journal of gastroenterology. 2009;104 Suppl 1:S1–35.
  12. 12. Hussain Z, Quigley EM. Systematic review: Complementary and alternative medicine in the irritable bowel syndrome. Alimentary pharmacology & therapeutics. 2006; 23:465–471.
  13. 13. Liu JP, Yang M, Liu YX, Wei M, Grimsgaard S. Herbal medicines for treatment of irritable bowel syndrome. The Cochrane database of systematic reviews. 2006: Cd004116. pmid:16437473
  14. 14. Shi J, Tong Y, Shen JG, Li HX. Effectiveness and safety of herbal medicines in the treatment of irritable bowel syndrome: a systematic review. World journal of gastroenterology. 2008; 14:454–462. pmid:18200670
  15. 15. Yuan R, Lin Y. Traditional Chinese medicine: an approach to scientific proof and clinical validation. Pharmacology & Therapeutics. 2000;86(2): 191–198.
  16. 16. Chinese Society of Digestive Diseases and Chinese Association of Chinese Medicine. Consensus on the diagnosis and treatment of functional dyspepsia irritable bowel syndrome by Traditional Chinese Medicine. China J Trad Chin Med Pharm 2010; 25:1062–1065.
  17. 17. Leung WK, Wu JC, Liang SM, Chan LS, Chan FK, Xie H, et al. Treatment of diarrhea-predominant irritable bowel syndrome with traditional Chinese herbal medicine: a randomized placebo-controlled trial. The American journal of gastroenterology. 2006;101:1574–1580. pmid:16863563
  18. 18. Sung JJ, Leung WK, Ching JY, Lao L, Zhang G, Wu JC, et al. Agreements among traditional Chinese medicine practitioners in the diagnosis and treatment of irritable bowel syndrome. Alimentary pharmacology & therapeutics. 2004; 20:1205–1210.
  19. 19. Xiao Y, Liu Y, Huang S, Sun X, Tang Y, Cheng J, et al. The efficacy of Shugan Jianpi Zhixie therapy for diarrhea-predominant irritable bowel syndrome: a meta-analysis of randomized, double-blind, placebo-controlled trials. PloS one. 2015;10:e0122397. pmid:25853241
  20. 20. Li Q, Yang GY, Liu JP. Syndrome differentiation in chinese herbal medicine for irritable bowel syndrome: a literature review of randomized trials. Evidence-based complementary and alternative medicine: eCAM. 2013;2013:232147.
  21. 21. Drossman DA.The functional gastrointestinal disorders and the Rome II process. Gut, 1999,45 Suppl 2:II 1–II 5.
  22. 22. Drossman DA.The functional gastrointestinal disorders and the Rome III process.Gastroenterology, 2006, 130(5):1377–1390. pmid:16678553
  23. 23. Schmulson MJ, Drossman DA. What Is New in Rome IV. J Neurogastroenterol Motil,2017; 23 (2): 151–163. pmid:28274109
  24. 24. National TCM Authority.TCM Illness Diagnosis Effect Standard. Nanjing:Nanjing University Press.1994:11.
  25. 25. Zheng XY. Chinese herbal medicine new medicine clinical research guiding principle [M]. Beijing: China Medical Science Press. 2002:124–129.
  26. 26. Jadad A. R., Moore R. A., Carroll D. et al., “Assessing the quality of reports of randomized clinical trials: is blinding necessary?”Controlled Clinical Trials. 1996;17(1):1–12. pmid:8721797
  27. 27. Yue C1, Kang P, Pei F.Comparison of Direct Anterior and Lateral Approaches in Total Hip Arthroplasty: A Systematic Review and Meta-Analysis (PRISMA).Medicine (Baltimore).2015;94(50):e2126,1–10.
  28. 28. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ (Clinical research ed). 2003; 327:557–560.
  29. 29. DerSimonian R, Laird N.Meta-analysis in clinical trials. Control Clin Trials. 1986;7(3):177–188. pmid:3802833
  30. 30. Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias.Biometrics 1994;50(4): 1088–10101. pmid:7786990
  31. 31. Cai W. Clinical Observation of Liver-Discharging and Stomach-Harmonizing Therapy for Constipation-Predominant Irritable Bowel Syndrome in 60 Cases, Shanghai Journal of Traditional Chinese Medicine.2005,39(5):14–15.
  32. 32. Chen XG, Hu XY, Zhang WP, Xu SH, Chen ZZ, Qiu XS. Clinical research on Yigi II recipe in treating Constipation-Predominant Irritable Bowel Syndrome of children. ShanDong Journal of Traditional Chinese Medical.2005,24(9):526–528.
  33. 33. Wu JH. Clinical Observation of Tonifying Qi, Strengthening Spleen and regulating Qi Therapy for Constipation-Predominant Irritable Bowel Syndrome in 85 Cases. Guangming Journal of Chinese Medicine,2006,21(4):34–35.
  34. 34. Wang CL.Treatment of Constipation-Predominant Irritable Bowel Syndrome by “Yangxue Tongfu Decoction” in 50 Cases. Jiangxi Journal of Traditional Chinese Medicine,2008,39(3):21.
  35. 35. Li HY, Zhang YQ. A randomized controlled trial of Traditional Chinese Medicine compound granules with Regulating Qi and Moistening the Intestines for Constipation-Predominant Irritable Bowel Syndrome. Zhejiang Journal of Traditional Chinese Medicine.2011,46(11):788–789.
  36. 36. Liao J,Xiong J. A Clinical Observation of Sini Powder and Liumo Decoction for Constipation-Predominant Irritable Bowel Syndrome. Journal of Sichuan of Traditional Chinese Medicine. 2011,29(9): 59–60.
  37. 37. Qian CJ. The Clinical Study of Supplemented Fluid-increasing Decoction in Treating Syndrome of intestinal dryness due to Yin deficiency of Irritable Bowel Syndrome[D]. Nanjing:Nanjing Chinese Medical University;2011.
  38. 38. Chen J, Ren NX, Tan DY. 20 Cases of Constipation-Predominant Irritable Bowel Syndrome treated with therapy of Replenishing Qi to invigorate the Spleen and ventilating the Lung. Chinese Medicine Modern Distance Education of China.2013,11(8):20–21.
  39. 39. Zhang XP, Clinical Research of Constipation-Predominant Irritable Bowel Syndrome treated with Guipi Decoction and Xiaoyao Powder. China Journal of Chinese Medicine.2014,29(11): 1667–1668.
  40. 40. Wang FD. Clinical Study of the San Zang Tiao He Run Chang Decoction in Treating Irritable Bowel Syndrome with Constipation[D]. Zhejiang:Zhejiang University of Chinese Medical; 2015.
  41. 41. Zhang ZY. Clinical Observation of TCM Syndrome Differentiation Therapy for Constipation- Predominant Irritable Bowel Syndrome in 60 Cases. China Practical Medical. 2016, 11(21):37–38.
  42. 42. Fan L, Yu H. Traditional Chinese medicine decoction and Fried condition survey analysis. China Pharmacist. 2009;9: 1360–1362.
  43. 43. Stanghellini V, Tosetti C, Barbara G, De Giorgio R, Cogliandro L, Cogliandro R, Corinaldesi R. Dyspeptic symptoms and gastric emptying in the irritable bowel syndrome. The American journal of gastroenterology. 2002; 97:2738–2743. pmid:12425541
  44. 44. Tillisch K, Labus JS. Advances in imaging the brain-gut axis: functional gastrointestinal disorders. Gastroenterology. 2011;140:407–411. pmid:21167161
  45. 45. Tillisch K, Mayer EA, Labus JS. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology. 2011; 140:91–100. pmid:20696168
  46. 46. Barbara G, Zecchi L, Barbaro R, Cremon C, Bellacosa L, Marcellini M, et al. Mucosal permeability and immune activation as potential therapeutic targets of probiotics in irritable bowel syndrome. Journal of clinical gastroenterology. 2012;46 Suppl:S52–55.
  47. 47. Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EM, Rajilic-Stojanovic M, Schemann M, Schwille-Kiuntke J, Simren M, Zipfel S, Spiller RC. Irritable bowel syndrome. Nature reviews Disease primers. 2016;2:16014. pmid:27159638
  48. 48. Qiu B. Experimental Study on Ma Zhi Jiang Zhuo Improving 5-HT of Intestinal Tissue in the Rat Model of Constipation-predominant Irritable Bowel Syndrome[D].Hebei: HeBei Medical University; 2013.
  49. 49. Lin ZY, Zhang SS, Dai HM, et al.Effect of Maziren Pills on the content of 5-HT in intestinal mucosa in rats with constipation-type irritable bowel syndrome. Modern Journal of Integrated Traditional Chinese and Western Medicine.2014,23(4): 343–346.
  50. 50. Fu F.The Clinical Study of Maren Pills flavored in treating constipation predominant Irritable Bowel Syndrome of children[D]. Fujian: Fujian university of traditional Chinese medicine; 2015.
  51. 51. Xu YY,Ai ZF,Zhu LJ. Effect of Liver-Discharging, Qi-Regulating and Intestines-Moistening Therapy on gastrointestinal hormone in patients with Constipation-Predominant Irritable Bowel Syndrome. LiShiZhen Medicine and Materia Medical Research. 2014,(09):2192–2194.
  52. 52. Jiang XY, The Clinical Study and mechanism of the Kuanzhongjieyutang in treating constipation predominant Irritable Bowel Syndrome(Liver–Qi stagnation)[D].ShanDong:ShanDong University of Traditional Chinese Medical;2013.
  53. 53. Yang SW,Liu XJ,Zhen HL, et al. Efficacy of Shanzha Xiaozhi Capsule on irritable bowel syndrome and its’ influence on motilin and endothelin.Zhejiang Journal of traditional Chinese medicine. 2016, (11): 815.