Summary
Background
Chronic constipation is a common, heterogeneous disorder with multiple symptoms and pathophysiological mechanisms. Patients are often referred to a gastroenterology provider after laxatives fail. However, there is limited knowledge of the spectrum and management of constipation disorders.
Aim
To discuss the latest understanding of the spectrum of constipation disorders, tools for identifying a pathophysiologic‐based diagnosis in the specialist setting, treatment options and the patient's perspective of constipation.
Methods
Literature searches were conducted using PubMed for constipation diagnostic criteria, diagnostic tools and approved treatments. The authors provided insight from their own practices.
Results
Clinical assessment, stool diaries and Rome IV diagnostic criteria can facilitate diagnosis, evaluate severity and distinguish between IBS with constipation, chronic idiopathic constipation and dyssynergic defecation. Novel smartphone applications can help track constipation symptoms. Rectal examinations, anorectal manometry and balloon expulsion, assessments of neuromuscular function with colonic transit time and colonic manometry can provide mechanistic understanding of underlying pathophysiology. Treatments include lifestyle and diet changes, biofeedback therapy and pharmacological agents. Several classes of laxatives, as well as prokinetic and prosecretory agents, are available; here we describe their mechanisms of action, efficacy and side effects.
Conclusions
Constipation includes multiple overlapping subtypes identifiable using detailed history, current diagnostic tools and smartphone applications. Recognition of individual subtype(s) could pave the way for optimal, evidence‐based treatments by a gastroenterology provider.
Gastroenterology providers should be familiar with the constipation subtypes and their underlying pathophysiology, as well as key signalling pathways that may contribute to constipation. Familiarisation with the varied mechanisms of action, expected efficacy and side effects of available therapies helps to select the appropriate treatment and meet patient expectations.
1. INTRODUCTION
Constipation affects 15%‐20% of the global population and carries a major health care burden. 1 , 2 The pathophysiology of chronic constipation is complex, exemplified by varying classifications. Patients often present with similar, overlapping symptoms. 3 , 4 Therefore, differentiation and appropriate classification of constipation types guides treatment selection. 4 , 5 Constipation is classified as primary or secondary, depending on the underlying cause. 3 , 4 Secondary constipation is associated with organic disease (eg, colonic stricture, mass or malignancy), medication use (eg, opioids, anti‐cholinergic medications) or an underlying condition (eg, metabolic, thyroid or diabetic disorders), while primary constipation is a consequence of neuromuscular dysfunction of the colon or anorectal sensory‐motor function. 3 , 6
The complexity of primary constipation—and failure to respond to first‐line treatments (eg, lifestyle changes and laxatives)—is why patients are referred to gastrointestinal (GI) specialists. 5 Constipation disorders are sometimes difficult to manage, and patients are often dissatisfied with their treatment. 7 Symptoms of constipation can be secondary to GI pathology, such as colonic strictures, advanced colorectal polyps or neoplasms, necessitating an age‐appropriate colon cancer screening. Failure to address constipation disorders may result in symptom progression over time. 8
IBS with constipation (IBS‐C), functional constipation or chronic idiopathic constipation (CIC) and defecatory disorders—especially dyssynergic defecation—comprise the most common spectrum of primary constipation disorders. 9 IBS‐C, CIC and dyssynergic defecation have overlapping symptoms. 8 Another growing problem is opioid‐induced constipation (OIC) and, although a secondary cause of constipation, significant new knowledge in the pathophysiology and treatment of OIC is helpful for GI specialists to consider when differentiating from a primary constipation diagnosis. 10 Our purpose is to provide an up‐to‐date review on the spectrum of primary constipation disorders, focusing on understanding pathophysiology, diagnostic and clinical assessment tools, and pharmacologic and biofeedback therapies.
2. METHODS
For this review, during the period from February 2020 through December 2020, literature searches were conducted using PubMed, CINAHL and Embase to identify publications reporting on the pathophysiology, diagnostic criteria, diagnostic tools and approved treatments for constipation. Various combinations of search terms were used for IBS‐C, CIC or functional constipation and defecation disorder. Review of the reference lists from the above searches provided additional references, as did additional targeted searches. In summary, 171 references were selected for inclusion in this review due to their relevance to the scope of this manuscript and based on the authors' insight, research experience and clinical practices in managing constipation.
3. PATHOPHYSIOLOGY
The pathophysiology of constipation is multifactorial and varies between patients. Patients with primary constipation are classified as having one of the three overlapping subtypes: defecatory disorders, slow‐transit constipation or normal transit constipation. 3 , 4 , 11
Defecatory disorders are often associated with underlying dyssynergic defecation, characterised by impaired rectal evacuation from inadequate rectal propulsion forces, high anal resting pressure and/or paradoxical contraction of the anal sphincter and pelvic floor muscles during defecation or incomplete relaxation. 3 , 4 , 11 , 12 Dyssynergic defecation results from uncoordinated contraction of the abdominal and pelvic floor muscles. 13 , 14 Difficulty defecating may also be compounded by anatomical abnormalities (eg, rectoceles, intussusception) or altered rectal sensitivity. 3 , 11
Primary slow colonic transit constipation can be caused by neuromuscular dysfunction of the colon wall that is not associated with other disorders or underlying systemic diseases. 3 , 11 Colonic motor disturbances, or myopathies, are associated with low‐amplitude contraction resulting in reduced propulsion and colonic stasis leading to water reabsorption and stool hardening, accompanied by a reduced feeling of the need to defecate. 3 , 11 Colonic neuropathy, associated with disorganised or uncoordinated contraction, may include abnormal colonic sensation contributing to abdominal pain and bloating and altered neuromuscular signalling in and/or to the colonic wall. 3 , 11 The colons of patients with chronic constipation have shown a reduced number of intrinsic nerves and interstitial cells of Cajal and a decreased response to cholinergic stimulation. 3 , 11 Evidence suggests that methane gas accumulation in the GI tract, or intestinal methanogenic overgrowth, is associated with reduced colonic transit and slow transit constipation 15 ; however, a separate study presented conflicting data where breath methane excretion was not associated with slow colonic transit. 16 The prevalence of methane‐producing flora—and baseline methane levels—is significantly higher in patients with delayed colonic transit than with normal transit, and negatively correlate with colonic transit. 17
The pathophysiology of CIC and IBS‐C is multifactorial, involving visceral hypersensitivity, neuropathy along the afferent gut‐brain axis, altered bile acid metabolism, neurohormonal regulation, immune dysfunction, gut microbiota dysbiosis, alterations in the epithelial barrier, secretory properties and brain and gut dysfunction. 18 , 19 , 20 These diagnoses of primary constipation are not exclusive, as patients may have co‐existing defecation disorder, slow transit, CIC or IBS‐C. 3 , 11 Consequently, with an unrecognised or untreated dyssynergia (or an overlapping rectal evacuation disorder), there is a potential for secondary delays in colonic transit.
Approximately, 40% of opioid‐using patients report OIC regardless of opioid potency. 21 Without treatment, opioid‐related bowel disorders, mainly constipation, significantly decrease the quality of life (QoL) and daily activities. 22 The robust interaction of opioids with µ‐receptors in the enteric nervous system, rather than effects of opioids in the central nervous system, results in decreased GI transit, stimulation of non‐propulsive activity, increased anal tone, decreased intestinal secretion, increased fluid adsorption and decreased rectal sensitivity. The net effort of these mechanisms induced by opioids is to cause constipation and bowel‐based dysfunctions. 10
4. DIAGNOSTIC TOOLS
The term ‘constipation’ is non‐specific, referring to a constellation of patient‐reported symptoms describing a bowel function disturbance. 23 , 24 However, clinicians should be able to characterise constipation's varied features. 23 , 24 Visual images of stool form, specifically the Bristol Stool Form Scale (BSFS; Figure 1), and bowel diaries are reliable methods to characterise bowel habits. A self‐administered mobile phone app, Constipation Stool Diary, provides information about constipation symptoms that a patient can keep prospectively for 1‐2 weeks, reducing recall bias and improving treatment response monitoring (Figure 2). 25
FIGURE 1.
Bristol Stool Form Scale. Copyright 2000 © by Rome Foundation. All Rights Reserved
FIGURE 2.
Parameters in Constipation Stool Diary in constipated and healthy subjects. This figure has been reproduced from Yan et al Gastroenterology 2020 with permission from Elsevier. 25 BM, bowel movement; CSBM, complete spontaneous bowel movement
Symptoms of chronic constipation alone do not accurately predict the underlying pathophysiology or response to treatment. 26 Providers should be familiar with ‘alarm features’ that suggest a more serious underlying health issue (eg, occult colon cancer), so appropriate testing (eg, colonoscopy) can be arranged. Alarm features include bloody stools, anaemia or iron deficiency, unintentional weight loss and family history of colorectal cancer. 5 , 27
4.1. Diagnostic criteria
Chronic constipation can be diagnosed via Rome IV criteria, which incorporate patient‐reported symptoms, stool consistency, physical examination and motility study findings (Table 1). 28 In addition to stool frequency, stool shape and consistency are also key components of the diagnostic criteria, and the BSFS is recommended for characterising stool appearance (Figure 1). 3 , 28 , 29 The BSFS describes seven stool consistency categories correlating with colonic transit time. 29 Longer transit times are associated with lower BSFS scores. Sensations of incomplete emptying, straining, use of digital manoeuvres for evacuation of stool and abdominal bloating/distension can indicate underlying constipation. 4
TABLE 1.
Rome IV diagnostic criteria for IBS with constipation, 43 functional constipation 43 and functional defecation disorders 28
IBS with Constipation (IBS‐C) |
---|
Criteria fulfilled for the last 3 mo with symptom onset ≥6 mo prior to diagnosis Recurrent abdominal pain, on average, ≥1 d per week in the last 3 mo, associated with two or more of the following criteria:
Diagnostic criteria for IBS subtypes Predominant bowel habits are based on stool form on days with at least one abnormal bowel movement a IBS with predominant constipation: More than one fourth (25%) of bowel movements with BSFS types 1 or 2 and less than one fourth (25%) of bowel movements with BSFS types 6 or 7 Alternative for epidemiology or clinical practice: Patient reports that abnormal bowel movements are usually constipation (like type 1 or 2 in the picture of BSFS). |
Functional constipation (Chronic idiopathic constipation [CIC]) |
---|
Criteria fulfilled for the last 3 mo with symptom onset at least 6 mo prior to diagnosis
|
Functional defecation disorders |
---|
Criteria fulfilled for the last 3 mo with symptom onset at least 6 mo prior to diagnosis
F3a. Diagnostic criteria for inadequate defecatory propulsion Inadequate propulsive forces as measured with manometry with or without inappropriate contraction of the anal sphincter and/or pelvic floor muscles b F3b. Diagnostic criteria for dyssynergic defecation Inappropriate contraction of the pelvic floor as measured with anal surface electromyography or manometry with adequate propulsive forces during attempted defecation b |
Abbreviations: BSFS, Bristol Stool Form Scale; CIC, chronic idiopathic constipation; IBS‐C, IBS with constipation.
IBS subtypes related to bowel habit abnormalities can only be confidently established when the patient is evaluated off medications used to treat bowel habit abnormalities.
These criteria are defined by age‐ and sex‐appropriate normal values for the technique.
Before diagnosing primary constipation, gastroenterology providers should first rule out secondary causes of constipation. A thorough medication history can identify exacerbating medications, opioids and anti‐cholinergic medications. Secondary constipation may also be a result of diabetes mellitus, chronic renal disease, dehydration, Parkinson's disease, Ehlers‐Danlos syndrome or other connective tissue disorders. 3 , 30 , 31
Primary chronic constipation can be further investigated by multiple tests. Defecatory disorders are evaluated by digital rectal examination, rectal balloon expulsion, anorectal manometry and defecography. 5 , 6 , 27
4.2. Physical examination
Comprehensive abdominal and thorough digital rectal examinations are useful in evaluating chronic constipation. 11 , 32 Close inspection of the perianal region can reveal excoriations, haemorrhoids, fissures or masses. A lubricated finger in the anal canal can assess anal sphincter tone. Placing one hand on the lower abdomen while a finger is inserted into the anal canal assesses rectoanal incoordination or dyssynergic defecation. 3 , 11 , 29 , 33 , 34 If constipation is fibre‐ and laxative refractory, then physiological testing should be considered. 29 Often, patients will have already attempted self‐treatment with fibre and laxatives before seeing a health care provider. 26
4.3. Functional tests
High‐resolution anorectal manometry and balloon expulsion testing are simple, inexpensive and the first anorectal physiological tests recommended for patients with laxative‐refractory CIC or suspected dyssynergic defecation, with high sensitivity for detecting dyssynergia and rectal hypersensitivity or hyposensitivity. 4 , 11 , 28 , 32
4.3.1. Balloon expulsion test
The balloon expulsion test is an office‐based test assessing a patient's ability, based on time taken, to expel a water‐ or air‐filled balloon inserted into the rectum. 28 , 32 , 35 , 36 Standardisation is lacking and methodology differs between GI motility laboratories, 32 , 35 but an expulsion time longer than 1‐2 minutes is generally considered abnormal. 28 An uncontrolled study of patients with constipation and defecation disorders illustrated that the balloon expulsion test may identify patients with dyssynergic defecation. 37 Some GI motility laboratories use the balloon expulsion test to screen for dyssynergic defecation. However, a normal test does not always exclude defecatory dysfunction, and correlation with other anorectal physiological testing and defecography is lacking. 5 , 11 , 29 , 38 Patient demographics can also affect test results: males typically have a shorter expulsion time, and expulsion time lengthens with increasing age. 35 Therefore, corresponding tests of anorectal physiological function should be obtained. 28 , 29
4.3.2. Anorectal manometry
Anorectal manometry assesses anorectal pressure changes during rest and simulated defecation of an intrarectal balloon, sphincter tone and rectoanal reflexes (which evaluate intrinsic and extrinsic innervation, and rectal compliance and sensitivity). 5 , 28 , 29 , 32 Although more expensive, high‐resolution anorectal manometry using a six‐sensor, solid‐state probe permits easier calibration and shorter procedure time compared to conventional water‐perfused anorectal manometry. 39 Anorectal manometry is useful in diagnosing dyssynergic or disordered defecation and other neuromuscular and sensory problems, and identifying patients who may benefit from biofeedback therapy. 5 , 32 , 40 , 41
While effective and widely used, anorectal manometry has limitations. Substantial variations in clinical practice in methodologies used for the balloon expulsion test and anorectal manometry 32 , 36 demand prompt efforts to standardise testing protocols. 36 , 40 A standardised protocol of high‐resolution anorectal manometry can characterise dyssynergic defecation subtype of rectoanal incoordination and guide therapists providing corrective biofeedback therapy treatment. 36 Furthermore, patient cooperation is key, 11 the procedure may be embarrassing 11 and test performance may not accurately replicate the actual act of defecation. 28 If the balloon expulsion or anorectal manometry test fails to diagnose or exclude a strongly suspected defecatory disorder, defecography is recommended. 11 , 28
4.3.3. Defecography
Defecography examines both the function and structure of the anorectum and the pelvic floor during voluntary defecation. 28 , 29 Defecography is performed in specialist centres, using X‐ray (barium) or magnetic resonance imaging. 28 , 32 , 35 X‐ray defecography assesses rectal wall structure and pelvic floor motion while seated. Magnetic resonance imaging defecography evaluates all pelvic compartments in the semi‐recumbent position, which may not replicate the everyday practice of defecation. 35 These tests can determine if chronic constipation is associated with incomplete anal opening, impaired puborectalis relaxation or contraction, abnormal perineal descent and anatomical abnormalities (eg, rectocele, prolapse or intussusception). 5 , 29
4.4. Assessing colonic transit time
Slow colonic transit time can contribute to constipation and is assessed by measuring the time taken for content to move through the GI system. 32 Colonic transit assessment is recommended in the evaluation of laxative‐refractory constipation. 11 , 32 Specialist centres evaluate colonic transit time either by radiopaque markers, wireless motility capsules or by scintigraphy. 5 , 11 , 27 , 28 , 42 Colonic transit tests should be performed while the patient is not taking a laxative. 43
4.4.1. Radiopaque marker test
Radiopaque marker testing is widely accessible, non‐invasive, inexpensive and the most common option for assessing colonic transit time. 5 , 28 , 29 , 42 In a radiopaque marker test, the patient ingests a dissolvable capsule containing 20‐50 plastic markers. 42 The radiopaque markers are visible on X‐ray. Transit time is calculated from abdominal radiographic images captured at set time points in the days following capsule ingestion. 42 Variations in the radiopaque marker test are used in gastroenterology clinical practice. 42 The most commonly applied method involves administering a capsule containing 20‐24 markers on day 1, followed by abdominal radiograph imaging after 5 days 29 , 42 ; retention of >20% of ingested markers indicates slow colonic transit. 29
4.4.2. Wireless motility capsule test
Wireless motility capsules enable radiation‐free, continuous monitoring of the intraluminal pH, pressure and temperature while passing through the GI tract. 29 , 42 , 44 Signals from the capsule are transmitted to a receiver the patient wears. After capsule ingestion following an overnight fast, patients record events (eg, meals, bowel movements, symptoms) over 3‐5 days by pressing a receiver button and maintain a diary. 44 Results have demonstrated good correlation with radiographic tests. 28 , 44 , 45 Wireless motility capsules provide a comprehensive picture with other regional GI transit times (gastric emptying time, small‐bowel transit time) and whole‐gut transit time. 44 , 46 The capsule measures colonic transit time by detecting pH changes while moving through the gut. 29 , 44 Demographics and study protocols can influence test findings. 47 Transit times are shorter among males and can vary with the menstrual cycle. 47
4.4.3. Scintigraphy
Scintigraphy uses repeated imaging of ingested radioisotopes over consecutive days to calculate overall and regional colonic transit times. 29 , 42 A range of γ‐emitting isotopes, administered in varying vehicles, may be used. 42 Scintigraphy testing is conducted at a few specialised centres, due to the need for specialist equipment and use of short‐lived radioactive isotopes. 42
4.4.4. Colonic manometry
In cases of severe slow‐transit constipation and suspected colonic inertia, ambulatory 24‐hour colonic manometry following a standardised protocol can help distinguish between underlying colonic myopathy and neuropathy, facilitating appropriate management. 48 Like scintigraphy, colonic manometry is limited to a few, specialised centres.
5. DIAGNOSTIC FEATURES
5.1. Chronic idiopathic constipation
CIC is estimated to affect 14% of the global population. 1 Patients with CIC may report lumpy or hard stools (BSFS type 1‐2) and infrequent bowel movements (<3 per week). Straining and abdominal bloating may be present. Pain may also be present, but it would not predominant. 43 Evaluation of patient's history may find sensation of incomplete evacuation or anorectal obstruction, and patients may report using manual manoeuvres to facilitate defecations. Discussion with the patient may reveal limited physical activity, QoL complaints, attempted use of laxatives and infrequent loose stools without the use of laxatives. A rectal examination and routine blood tests would likely be normal and there may be no immediate indication of alarm features or physical abnormalities. More invasive diagnostic tests may be necessary if symptoms are not relieved with prescription therapy. In patients with laxative‐refractory constipation, colonic transit assessment may be informative.
5.2. IBS with constipation
Approximately 5.2% of the North American population is estimated to experience IBS‐C. 49 In order to diagnose IBS‐C based on the Rome IV criteria, a review of the patient's medical history would reveal recurrent abdominal pain associated with defecation, reductions in stool frequency and/or a change in stool consistency. 43 Assessment of a patient's stools via BSFS would reveal more than 25% of bowel movements with BSFS types 1 or 2 and less than 25% of bowel movements with BSFS types 6 or 7. 43 Predominant abdominal pain would likely distinguish IBS‐C from CIC. Laxatives may help improve bowel movement frequency, but they may not improve abdominal symptoms (eg, bloating and pain). Digital rectal examination, anorectal manometry and balloon expulsion would determine the coexistence of disordered or dyssynergic defecation.
5.3. Defecatory disorder
As with CIC, patients may have infrequent and hard bowel movements, bowel movements associated with excessive straining, or a feeling of incomplete evacuation, and may use digital evacuation manoeuvres. Laxatives may not improve feelings of incomplete evacuation, straining and hard stools. A detailed history may reveal natural birth experience in women. Digital rectal examination, rectal balloon expulsion, anorectal manometry and defecography would help diagnose defecatory disorders. 27 In a digital rectal examination, a patient may have a normal sphincter tone at rest, but when asked to push and bear down may exhibit paradoxical contraction of anal sphincter with no perineal descent, suggesting dyssynergia. 34 The patient may be unable to pass the balloon expulsion test, 37 and results from anorectal manometry would show consistency with dyssynergic defecation. Biofeedback therapy using visual manometry‐based feedback would likely be helpful.
6. TREATMENT
The initial management approach to IBS‐C and CIC tends to be similar, but therapy response varies between the two conditions. 9 For example, stimulant laxatives and polyethylene glycol (PEG) can be effective for CIC but not for IBS‐C. 9 The symptomatology of defecatory disorders, such as dyssynergic defecation, can overlap with IBS‐C and CIC; however, symptoms of excessive straining, feeling of incomplete evacuation and use of digital manoeuvres to defecate are more prevalent. Defecatory disorders are less likely to respond to laxative therapy and more likely to require biofeedback therapy. 2
6.1. Lifestyle and dietary modifications
Initial therapy for chronic constipation includes lifestyle and dietary modifications (eg, increasing fluid and fibre intake) and physical activity. 28 , 29 , 32 Little evidence suggests that increasing fluid intake alone improves stool consistency among adequately hydrated patients. 28 , 29 , 50 Instead, increased fluid intake is most beneficial when combined with additional fibre in patients with mild constipation. 28 , 32 , 50
Increasing fibre intake, which should be done gradually to prevent abdominal distension, 11 , 28 , 32 may improve constipation by stimulating the gut mucosa to secrete water and mucus and improve stool consistency by increasing its water‐holding capacity. 29 However, not all constipation patients benefit from additional dietary fibre. 50 Insoluble fibre (eg, fibre in wheat bran and whole grains) may worsen symptoms of abdominal pain, distension and flatulence in some patients. 28 , 29 , 32 Fruit fibre (eg, prunes 51 ) or mixed soluble fibre 52 has demonstrated a short‐term efficacy in the management of chronic constipation, somewhat better than psyllium. In a recent randomised controlled trial of natural treatments for CIC in 79 patients, kiwifruit, psyllium and prunes were found to be effective. 53
Physical activity has been associated with reduced GI transit times. 50 Although exercise alone does not appear to improve constipation, patients report improved QoL and a reduction in symptom severity. 50
6.2. Pharmacological therapy
6.2.1. Laxatives
Laxatives are an inexpensive, widely available and often over‐the‐counter (OTC) treatment option for chronic constipation refractory to lifestyle and dietary modifications. 11 , 32 Laxatives can improve stool consistency, increase stool frequency and reduce defecation straining. 54
Osmotic laxatives
Osmotic laxatives, such as PEG, lactulose, sorbitol, glycerol and magnesium salts, contain non‐absorbable ions or molecules. 28 , 32 These compounds create an osmotic gradient that promotes water and electrolyte secretion into the intestinal lumen, increasing faecal volume and improving peristalsis. 29 , 32 Most studies have focused on PEG, which has demonstrated superiority over placebo and lactulose in improving symptoms of chronic constipation. 29 , 50 PEG treatment shows greater resolution of constipation symptoms, 55 , 56 improved stool consistency and frequency, 55 , 56 , 57 shorter GI transit time, 57 less straining, 55 , 56 , 57 and less severe abdominal bloating and pain compared with placebo. 55 PEG is also better than lactulose in improving stool consistency and frequency, reducing abdominal pain, and need for additional constipation‐related treatment. 58 , 59 Improvements in bowel movements, stool consistency and straining with PEG treatment have also been observed in patients with IBS‐C, though abdominal pain is largely unaffected. 50 , 60 , 61 Osmotic laxatives are generally well tolerated. 32 , 55 , 60 The most common adverse events (AEs) are abdominal pain and distension, diarrhoea, nausea, flatulence and vomiting. 28 , 56 , 57 , 61 Magnesium compounds should be used with caution in patients with renal impairment.
Stimulant laxatives
Stimulant laxatives are recommended after patients have failed to respond to osmotic laxatives. 11 , 29 Diphenylmethane derivatives (eg, bisacodyl, sodium picosulfate) and anthraquinones (ie, sennosides, cascara) are inactive, non‐absorbable glycosides that stimulate fluid, electrolyte secretion and peristalsis upon activation by glycosidases in the colon. 28 , 29 , 32 , 50 Stimulant laxatives are commonly used for patients with CIC and IBS‐C, though large controlled studies are lacking. 29 , 50 Both bisacodyl and sodium picosulfate can improve stool consistency and frequency, straining and QoL compared with placebo in CIC patients in randomised controlled trials. 62 , 63 , 64 The most common AEs with stimulant laxatives include diarrhoea, abdominal pain, nausea, vomiting and headache. 28 , 32 , 62
6.2.2. Prokinetic and prosecretory agents
Table 2 and Figure 3 detail the mechanisms of action by which the following types of prokinetic and prosecretory agents improve constipation symptoms.
TABLE 2.
Mechanism of action of pharmacological treatments for chronic constipation
Drug | Recommended dose a | Mechanism of action | Efficacy | Adverse events | |
---|---|---|---|---|---|
Polyethylene glycol | Non‐prescription | Osmotic laxative |
|
||
Bisacodyl | Non‐prescription | Stimulant laxative |
|
||
Sodium picosulfate | Non‐prescription | ||||
Anthraquinones | Non‐prescription | ||||
Plecanatide | CIC or IBS‐C: 3 mg q.d. | GC‐C agonist |
|
||
Linaclotide |
CIC: 145 mcg q.d. and 72 mcg q.d. IBS‐C: 290 mcg q.d. |
||||
Prucalopride | CIC: 2 mg q.d. b | 5‐HT4 agonist |
|
||
Tegaserod | IBS‐C: 6 mg b.i.d. c | ||||
Tenapanor | IBS‐C: 50 mg b.i.d. | Sodium/hydrogen exchanger isoform 3 inhibitor |
|
|
|
Lubiprostone |
CIC: 24 mcg b.i.d. IBS‐C: 8 mcg b.i.d. d |
Type‐2 chloride channel activator |
|
Abbreviations: b.i.d., twice daily; CIC, chronic idiopathic constipation GC‐C, guanylate cyclase‐C; GI, gastrointestinal; IBS‐C, IBS with constipation; q.d., once daily; QoL, quality of life.
Prescription doses are based on US Food and Drug Administration approval. Not all prescription therapies are approved outside the United States; treatment options should take into account therapy availability.
Indicated for patients with CIC (2 mg q.d.) or for patients with severe renal impairment (1 mg q.d.)
Indicated for women aged <65 y.
Indicated for women aged ≥18 y.
FIGURE 3.
Mechanism of action of agents used for the treatment of constipation. This figure has been modified from Simrén et al Nature Reviews Gastroenterology & Hepatology 2018 with permission from Springer Nature Customer Service GmBH: Wiley. 171 ACh, acetylcholine; CFTR, cystic fibrosis transmembrane conductance regulator; ClC‐2, type‐2 chloride channel; GC‐C, guanylate cyclase‐C; IBAT, ileal bile acid transporter; NHE3, sodium/hydrogen exchanger 3; VIP, vasoactive intestinal polypeptide
Guanylate cyclase‐C receptor agonists
Guanylate cyclase‐C (GC‐C) receptors are transmembrane proteins expressed by intestinal epithelial cells and help maintain bowel function by regulating fluid and electrolyte balance in the gut. 65 Activated GC‐C receptors facilitate the production of an ion gradient between the intestinal membrane and intestinal lumen that promotes net water movement into the gut 27 , 66 by simultaneous activation of cystic fibrosis transmembrane conductance regulator channels and inhibition of sodium/hydrogen exchanger isoform 3 channels. 65 , 67 Furthermore, GC‐C receptor activation helps maintain the intestinal mucosal barrier, prevent inflammation and attenuate visceral pain sensations. 67
The GC‐C receptor is activated by the hormones uroguanylin and guanylin and by heat‐stable enterotoxins produced by diarrhoeagenic bacteria. 67 Uroguanylin and guanylin both have two disulphide bonds resulting in a flexible structure with active forms stabilised at a specific pH. 67 Uroguanylin binds more readily to GC‐C receptors in slightly acidic conditions of the duodenum and jejunum (pH 5‐6), while guanylin binds to GC‐C receptors in more neutral to slightly basic conditions of the ileum and the colon (pH 7‐8). 65 , 67 In contrast to uroguanylin and guanylin, heat‐stable enterotoxins produced by diarrhoeagenic bacteria are stabilised by a third disulphide bond, producing a more stable and higher affinity structure. 67 Heat‐stable enterotoxins lack pH‐sensitive amino acids, allowing for GC‐C binding throughout the GI tract without being affected by gut pH. 67
GC‐C also helps regulate pain experienced in patients with chronic constipation, especially those with IBS‐C. 26 , 68 Activation of GC‐C inhibits nociception in the gut, reducing pain. 68 GC‐C is a target for pharmacological therapy because activating this receptor can increase fluid secretion and accelerate colonic transit time, restoring normal bowel function. 65 , 68 Furthermore, a recent report has demonstrated reduced uroguanylin levels in IBS‐C and CIC patients when compared to healthy control subjects during fasting and after meals. 69
Linaclotide, a synthetic analogue of exogenous diarrhoeagenic bacterial heat‐stable enterotoxins, and plecanatide, a synthetic uroguanylin analogue, are GC‐C agonists approved by the US Food and Drug Administration for the treatment of CIC and IBS‐C. 66 In clinical studies, linaclotide and plecanatide significantly improved stool consistency and frequency, and reduced straining, in patients with CIC or IBS‐C. 54 , 81 Patients also report improvements in abdominal discomfort, constipation severity and QoL, 54 , 82 , 83 with sustained efficacy over 24 and 52 weeks of treatment. 82 Linaclotide and plecanatide also reduce abdominal pain, bloating and cramping in patients with IBS‐C. 71 , 72 , 78 , 80 , 81 , 82 , 83 Improvements in IBS‐C symptoms were maintained over 12 and 26 weeks of treatment. 83 Japanese patients with IBS‐C may require a higher dose of linaclotide, possibly due to differences in GC‐C polymorphisms, bacterial proteases that metabolise linaclotide and/or diet compared with Western patients. 82
Linaclotide and plecanatide are well tolerated. Diarrhoea is the most frequent AE, but is generally mild or moderate in severity and occurs during the first 4 weeks of treatment. 54 , 84 , 85 No head‐to‐head trials have been conducted, and so direct comparisons cannot be made between the products; however, rates of diarrhoea reported in clinical trials of linaclotide were higher relative to those in plecanatide trials. 71 , 73 , 76 , 77 , 78 , 79 While this may be related to the similarity of plecanatide to endogenous uroguanylin and linaclotide to heat‐stable enterotoxins, 67 there were also differences between the plecanatide and linaclotide study designs that should be considered. Linaclotide, but not plecanatide, trials permitted dose interruptions (ie, patients could halt treatment to resolve a diarrhoea AE), which may have contributed to artificially lower rates of discontinuation due to diarrhoea. Additionally, while in both trials any verbatim report of ‘diarrhoea’ was recorded as an AE, in plecanatide studies, an extra level of scrutiny was added. When a patient reported increased stool frequency or looser stools, an assessment of ‘bothersome’ was made to determine if this report should be recorded as diarrhoea AE or a desired effect of the drug. 86
Serotonin agonists
Serotonin (5‐hydroxytryptamine) is a gut neurotransmitter that promotes motility via several serotonin receptor subtypes in the GI tract. 28 , 87 Serotonin controls gut smooth‐muscle contractions and relaxations. 88 Patients with IBS‐C may have dysfunctional serotonin neurotransmission in the gut. 87 , 88 In a healthy individual, serotonin plasma levels increase after food consumption; however, in IBS‐C patients, there may be limited or no response. 88 , 89 Alternatives in serotonin transporter gene polymorphism in IBS and in constipation have been reported. 90 This dysfunctional serotonin signalling may be associated with altered colonic transit. 89 In patients with slow‐transit constipation, levels of serotonin‐immunoreactive cells are significantly lower, so cell secretory indexes are decreased in these patients' colons. 91
Several agents have been developed that target the serotonin receptor 5‐HT4 to promote peristalsis and secretion. 92 , 93 Prucalopride and velusetrag (currently under clinical investigation) are selective, high‐affinity 5‐HT4 agonists that increase GI motility and reduce colonic transit times. 92 , 93 , 94 Earlier 5‐HT4 agonists, including cisapride and tegaserod, had poor selectivity and low affinity for the 5‐HT4 receptor, were associated with serious cardiovascular AEs, and, consequently, market withdrawal. 92 , 93 After data re‐examination, tegaserod has been re‐approved for the treatment of IBS‐C in women <65 years old, with a contraindication in patients with a history of cardiovascular issues. 95 In randomised, placebo‐controlled phase 3 studies, prucalopride was superior to placebo in improving symptoms, stool consistency and frequency, and reduced straining on defecation in patients with CIC. 96 , 97 , 98 Improvements in disease severity and QoL, 96 , 97 , 98 with patient satisfaction of bowel pattern, were noted and treatment efficacy was maintained for ≥18 months. 99 Prucalopride's efficacy for improving symptoms of constipation was confirmed in several integrated analyses and meta‐analyses; however, no evidence has been published demonstrating efficacy for pain so prucalopride is not approved for the treatment of IBS‐C. 54 , 100 , 101 , 102
Prucalopride‐associated AEs include GI complaints (nausea, abdominal pain, diarrhoea), flatulence and headache. 54 , 96 , 97 , 98 , 100 , 101 , 102 However, prucalopride is well tolerated, with AEs generally being mild, transient and resolving after the first day of treatment. 96 , 99 , 102
Sodium/hydrogen exchanger isoform 3 inhibitors
Inhibition of GI sodium/hydrogen exchanger isoform 3 expressed on the apical surface of the small intestine and colon reduces absorption of sodium from the small intestines and colon. 103 This results in water secretion into the intestinal lumen, increases intestinal transit time and softens stool consistency. Tenapanor, a sodium/hydrogen exchanger isoform 3 inhibitor, is approved by the US Food and Drug Administration for IBS‐C treatment. 103
In a phase 3 study, patients treated with tenapanor demonstrated improvements in stool frequency and reductions in abdominal pain. 103 Diarrhoea was the most common adverse reaction, with treatment‐related diarrhoea reported in 13.3% of tenapanor‐treated patients. 103
Bile acid modulation
Dysfunction in the enterohepatic circulation of bile acids can cause constipation. 87 Bile acids that are not absorbed in the terminal ileum can stimulate water and electrolyte secretion in the colon and directly contribute to colonic motility independent of secretory effects. 104 A subset of patients with IBS‐C may exhibit lower bile acid levels in their stools than in those of healthy controls and of patients with diarrhoea‐predominant IBS, 87 which may be attributed to altered bile acid synthesis in these patients. 105 Modulation of the bile acid cycle may help treat chronic constipation and involves bile acid supplementation or inhibition of the ileal bile acid transporter. Sodium chenodeoxycholate is a bile salt that can accelerate colonic transit times and improve stool consistency and frequency in patients with chronic constipation, including IBS‐C. 106 , 107
Elobixibat (approved in Japan) is a minimally absorbed, highly selective ileal bile acid transporter inhibitor. By inhibiting the active reabsorption of bile acids in the ileum, elobixibat increases bile acid concentrations in the colon, promoting fluid secretion and motility. 108 Elobixibat increases stool frequency and improves constipation‐associated symptoms, including stool consistency, constipation severity, straining and abdominal bloating in CIC and IBS‐C patients. 108 , 109 , 110
Both sodium chenodeoxycholate and elobixibat are generally well tolerated. AEs are typically mild and GI‐related, most commonly abdominal pain and diarrhoea. 107 , 108 , 109 , 110 Given elobixibat demonstrates minimal systemic absorption, 110 increased bile acids may stimulate propagated contractions in the colon, resulting in abdominal pain and cramping. 111
Chloride channel activators
Chloride channels located on intestinal epithelial cells regulate intestinal motility and fluid secretion. 92 Activation of type‐2 chloride channels triggers the release of chloride ions into the intestinal lumen. 92 The resulting ion gradient promotes sodium and water release into the lumen, increasing stool volume and GI motility while reducing colonic transit time. 92
Lubiprostone, a bicyclic fatty acid and a prostaglandin E1 analogue, is a locally acting selective type‐2 chloride channel agonist. 28 , 32 , 92 , 112 Lubiprostone has demonstrated efficacy in improving stool consistency and frequency and in reducing straining, bloating and constipation severity in patients with CIC or IBS‐C. 7 , 121 Lubiprostone treatment also reduces abdominal pain/discomfort and improves QoL for patients with IBS‐C. 115 , 117 , 118 , 120 , 121 Improvements are maintained with long‐term treatment. 113 , 117 , 120
GI‐related AEs are most common with lubiprostone treatment (predominantly nausea and diarrhoea), though headaches were also reported. 54 , 113 , 114 , 115 , 116 , 117 , 118 , 121 , 122 AEs tend to be mild or moderate in intensity and short lived. 113 , 114 , 115 , 117 , 118 , 121 A recent study using pooled trial data demonstrated that lubiprostone did not affect electrolyte homeostasis in the short or long term. 123
Opioid receptor antagonists
The mechanism by which opioid medications induce constipation is complex, involving peripheral and central effects. Peripheral activation of µ‐opioid receptors in the stomach and intestines inhibits both excitatory and inhibitory neural pathways, diminishing peristalsis and colonic transit time and delaying gastric emptying. 124 Evidence for a central mechanism is supported by a study in rats where intra‐cerebroventricular morphine administration inhibited GI propulsion. 124 In patients with laxative‐refractory OIC, opioid receptor antagonists with peripheral or peripheral and central action may be used. Although OIC is a secondary cause of constipation, many of which may be relieved by treating the primary problem, it is the only secondary cause of constipation with specific therapies and is potentially reversible with treatment (unlike other secondary causes). Several therapies for OIC are approved by the US Food and Drug Administration. 125 The American Gastroenterological Association's guidelines on the management of OIC distinguishes between traditional laxatives and peripherally acting µ‐opioid receptor antagonists and other prescription therapies for this condition. These guidelines recommend traditional laxatives (including PEG) as first‐line treatment for OIC, while peripherally acting µ‐opioid receptor antagonists and other prescription therapies are recommended for patients who fail traditional laxatives. 126 Meta‐analysis of the safety and efficacy of current OIC treatments has recently been published. 127
6.3. Biofeedback therapy for dyssynergic defecation
Biofeedback therapy is a robust treatment for dyssynergic defecation diagnosed by symptoms and anorectal motility testing, receiving a Grade A recommendation by the American and European Societies of Neurogastroenterology and Motility. 128 Multiple randomised controlled studies have proven that four to six sessions of electromyography or manometry‐based biofeedback therapy carries a 70%‐80% efficacy rate for dyssynergic defecation compared to standard treatment, 129 diltiazem 130 or laxatives. 131 Furthermore, biofeedback therapy, incorporating rectoanal coordination, simulated defecation and sensory conditioning, is durable, and separate studies have shown a sustained response for 12 and 44 months. 132 , 133 Severe constipation, digital facilitation of defecation, delayed colonic transit, impaired rectal sensation and increased anorectal angle during squeeze are predictors of poor response to biofeedback therapy. 134 , 135 , 136 Standard office‐based biofeedback therapy suffers from limitations: the need for skilled staff, multiple visits and limited availability at expert centres. Home biofeedback therapy is appealing for dyssynergic defecation and has similar efficacy in improvement of bowel symptoms and balloon expulsion compared to office‐based therapy. 137 Furthermore, home biofeedback therapy is more cost‐effective and shows promise for the future. 138
6.4. Clinical approach to managing patients with constipation
Management algorithms are available to help guide treatment decisions (Figure 4) and a recent meta‐analysis evaluating the efficacy of constipation treatments has been published. 139 First‐line treatment for chronic constipation patients failing lifestyle modifications include fibre supplements or osmotic laxatives (such as PEG) on a regular basis, or stimulant laxatives intermittently, 50 which are available to patients as OTC products. 11 , 28 , 29 , 32 , 140 Indeed, many patients with chronic constipation will use some OTC laxative before consulting a health care provider. 141 Patients try an average of three OTC products before seeking help 141 ; however, only half feel satisfied. 7 , 141
FIGURE 4.
Management algorithm for the treatment of constipation. This figure has been modified from Sharma Handb Exp Pharmacol. 2017 with permission by Springer Nature Customer Service GmBH: Wiley. 4 Mg, magnesium; PEG, polyethylene glycol; WMC, wireless motility capsule
Primary care providers should refer patients who are laxative refractory or intolerant to treatment to a gastroenterology provider for further assessment. 28 , 140 Primary care providers may try a promotility agent or prosecretory agent while awaiting gastroenterology evaluation. Gastroenterology providers may further titrate promotility agents or prosecretory agents and/or consider further testing—such as anorectal manometry, barium or magnetic resonance defecography, or colon transit time assessment—to identify the underlying pathophysiology and guide treatment in treatment‐refractory patients. 11 , 28 , 50 While there is a lack of evidence in the literature to guide a symptom‐based therapeutics approach, there is some evidence that different therapies may improve certain underlying mechanisms. For example, biofeedback therapy—a behavioural training technique using visual feedback to correct pelvic floor contractions—is safe, effective and recommended for patients with defecation disorders on the basis of anorectal manometry and defecography findings, 11 , 29 , 32 , 50 , 140 linaclotide and prucalopride accelerate colonic transit time, and linaclotide and plecanatide improve abdominal pain (and the former has been shown recently to modify afferent gut and brain interactions). 19 Likewise, rectal sensory retraining may be helpful in addition to biofeedback therapy to address both rectal hyper‐ and hyposensitivity.
A gastroenterology provider may recommend alternative treatments if other treatment options have failed. 50 Acetylcholinesterase inhibitors, such as neostigmine, can dramatically increase GI motility and may be considered in hospitalised patients with colonic pseudo‐obstruction; however, its use requires close observation and cardiorespiratory monitoring in the intensive care setting. 50 , 142 Furthermore, pyridostigmine, an oral acetylcholinesterase inhibitor, may be considered in severe constipation, especially with co‐existing autonomic dysfunction in patients who have previously responded to neostigmine or diabetes. 143 , 144 Oral ingestion of a vibrating capsule alters colonic circadian rhythm and may improve constipation by inducing more complete spontaneous bowel movements. 145 Habit training (bowel or pelvic floor retraining) and psychological interventions (cognitive behavioural therapy, hypnotherapy, psychological therapy) are also recommended for patients failing to respond to standard care. 50 , 146 Total abdominal colectomy is reserved for rare, severe cases of colonic inertia after all non‐surgical options have failed and specialised testing such as colonic manometry suggests underlying colonic neuropathy. 11 , 32 , 50 , 147 Continuous direct nerve stimulation (sacral nerve stimulation) is the least‐invasive surgical option available, with fewer complications than other surgeries; however, data demonstrating efficacy are lacking. 50 , 147 Subcutaneous electrodes can stimulate the sacral nerve to induce propagating contractions and increase stool frequency. 32 , 147 Antegrade continence enema, an endosurgical procedure where appendicocecostomy or caecostomy is created to serve as a conduit to directly administer enemas into the caecum, has been described as an efficacious option to treat faecal incontinence and refractory constipation. 148 The majority of cases and much of the literature are focused on the treatment of children; however, single‐centre experiences have described their long‐term outcomes in adults. 149 , 150 , 151 , 152 , 153 , 154 , 155 , 156 , 157 , 158 , 159 , 160 , 161 A meta‐analysis of observational studies, only three of which were prospective, suggested that approximately two thirds of antegrade continence enema procedures for constipation were efficacious, defined by continued use on follow‐up or successful resolution of symptoms, and approximately 45% of antegrade continence enema procedures were associated with morbidity, most commonly wound infection and stomal stenosis, with a re‐operation rate of >25%. 162 Given the low quality of evidence without randomised controlled trials and associated morbidity, antegrade continence enema cannot be recommended for treatment of severe constipation.
6.5. Patient perspective
6.5.1. Tools for assessing symptom severity
Patient QoL decreases with severity and duration of constipation and associated symptoms. 7 Furthermore, patients with chronic constipation exhibit significant psychological stress and impaired health‐related QoL compared to control subjects. 163 Symptoms reported as most bothersome are often also the most severely perceived. 7 Therefore, treatments focused on these symptoms are important to improve QoL. 7 Improvement in constipation with both secretagogues 73 , 75 , 76 , 77 , 79 and biofeedback therapy 138 can improve QoL.
In the absence of objective biomarkers for assessing constipation severity, prospective stool diaries and validated severity scales help assess patient‐reported outcomes in clinical trials and manage patients clinically. 164 The most commonly used scales include the Constipation Assessment Scale, Constipation Scoring System, Symptom Severity Score and Patient Assessment Constipation‐Symptom (Table 3). These scales assess the presence of constipation, constipation severity and the most bothersome constipation‐associated symptoms. Thorough review of stool diaries and responses to severity scales may offer clinicians an opportunity to assess treatment efficacy from the patient's perspective. However, if physical stool diaries and scales are not completed in real time, they may be limited by their dependence on patient recall of bowel habits and symptoms.
TABLE 3.
Commonly used scales for assessing the severity of constipation
Scale | Description | Scoring system | Scoring interpretation |
---|---|---|---|
Constipation Assessment Scale 168 |
Evaluates eight items:
|
Each item rated on a 3‐point scale:
|
Total score range: 0‐16
Score ≥1 indicates constipation |
Constipation Scoring System 169 |
Evaluates eight items:
|
Each item rated on a five‐point scale:
One item is rated from 0 to 2 |
Total score range: 0‐30
Score ≥15 indicates constipation |
Patient Assessment of Constipation‐Symptoms 170 |
Evaluates 12 items with three subscales:
|
Each item rated on a 5‐point scale:
|
Total score range: 0‐4
|
New tools are becoming available to support the monitoring of patients with constipation, such as electronic health records, smartphone apps and electronic stool diaries. The National Institutes of Health Patient‐Reported Outcomes Measurement Information System is increasingly incorporated into electronic health records as a measure of recording patient‐reported outcomes. 164 Within the GI domains of the National Institutes of Health Patient Reported Outcomes Measurement Information System, abdominal pain and constipation, as well as diarrhoea as a potentially treatment‐related adverse event, are relevant to patients with constipation. 164
With smartphone apps and diaries, gastroenterology providers can potentially review patients' symptoms as recorded in real time. The GI Patient Reported Outcomes Measurement Information System scales can be accessed via the MyGiHealth app online and on smartphones, which compare a patient's symptoms scores with those of the general population in the United States and generates a heat map of symptoms. 164 This app can also track changes across time, allowing gastroenterology providers to assess treatment efficacy and facilitate improved outcomes for both providers and patients. 164 The Constipation Stool Diary is an app‐based questionnaire used to track patient symptoms and medication use. 25 Apps have demonstrated a level of accuracy comparable to a physical diary and may be patient preferred. 165 For example, apps may assist in tracking diet and the symptoms of IBS‐C to see if there is a correlation. 166 Administering scales via an app either in the clinic or in the advance of an appointment may also optimise resource use.
6.6. Summary
CIC and IBS‐C are historically classified as idiopathic; however, pathophysiological dysfunctions can be commonly identified in these constipation disorders, which could aid effective management. In addition, pelvic floor disorders such as dyssynergic defecation are common and often may present with overlapping symptoms of infrequent defecation and difficulty with defecation. Gastroenterology providers should be familiar with the constipation subtypes and their underlying pathophysiology, as well as key signalling pathways that may contribute to constipation. Although lifestyle and diet modifications are useful, laxatives remain the first‐line treatment of constipation. For laxative‐refractory cases, several classes of prokinetic and prosecretory agents are available. Familiarisation with the varied mechanisms of action, expected efficacy and side effects helps to select the appropriate treatment and meet patient expectations. Treatment algorithms are available to guide decision‐making. Smartphone apps can help gastroenterology providers and patients track constipation symptoms and treatment response.
AUTHORSHIP
Guarantor of the article: Amol Sharma, MD.
Author contributions: Amol Sharma, Satish S. C. Rao, Kimberly Kearns, Kimberly D. Orleck and Scott A. Waldman made substantial contributions to conception and design of the manuscript, performed the research, were involved in the drafting of the manuscript and revising it critically for important intellectual content and provided final approval of the version to be published. All the authors approved the final version of this manuscript, including the authorship list.
ACKNOWLEDGEMENTS
The authors thank Blair Hesp, PhD, CMPP, Julie O'Grady, BA, and Nicole Coolbaugh, CMPP, of The Medicine Group, LLC (New Hope, PA, USA), who provided medical writing and editorial assistance, which was funded by Salix Pharmaceuticals, Inc, in accordance with Good Publication Practice guidelines.
Declaration of personal interests: Dr. A. Sharma served on advisory boards for Ironwood Pharmaceuticals, Phathom Pharmaceuticals and Salix Pharmaceuticals. Dr. S. S. C. Rao has served on the advisory board for Medtronic, Takeda Pharmaceuticals and Salix Pharmaceuticals. K. Kearns has served as a speaker for Medtronic and Takeda Pharmaceuticals, and a speaker and advisory board member for Salix Pharmaceuticals. K. D. Orleck has served as a speaker for AbbVie Pharmaceuticals, Allergan Pharmaceuticals and Salix Pharmaceuticals. Dr. S. A. Waldman has served on advisory boards for Salix Pharmaceuticals. He serves on the Board of Directors and is Chair of the Scientific Advisory Board for Targeted Diagnostics & Therapeutics, Inc.
Sharma A, Rao SSC, Kearns K, Orleck KD, Waldman SA. Review article: diagnosis, management and patient perspectives of the spectrum of constipation disorders. Aliment Pharmacol Ther. 2021;53:1250–1267. 10.1111/apt.16369
The Handling Editor for this article was Professor Jonathan Rhodes, and this uncommissioned review was accepted for publication after full peer‐review.
Funding information
The writing and preparation of this manuscript was funded by Salix Pharmaceuticals, Inc (Bridgewater, NJ, USA). SAW is supported by the National Institute of Health (1R01 CA204881, 1R01 CA206026, P30 CA56036), Department of Defense Congressionally Directed Medical Research Program W81XWH‐17‐PRCRP‐TTSA, The Courtney Ann Diacont Memorial Foundation and Targeted Diagnostic & Therapeutics, Inc.
DATA AVAILABILITY STATEMENT
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.
REFERENCES
- 1. Suares NC, Ford AC. Prevalence of, and risk factors for, chronic idiopathic constipation in the community: systematic review and meta‐analysis. Am J Gastroenterol. 2011;106:1582‐1591. [DOI] [PubMed] [Google Scholar]
- 2. Rao SS, Camilleri M. Approach to the patient with constipation. In: Podolsky DK, Camilleri M, Fitz JG, Kalloo AN, Shanahan F, Wang TC, eds. Yamada's Textbook of Gastroenterology. Hoboken, NJ: Wiley; 2015:757‐780. [Google Scholar]
- 3. Serra J, Mascort‐Roca J, Marzo‐Castillejo M, et al. Clinical practice guidelines for the management of constipation in adults. Part 1: definition, aetiology and clinical manifestations. Gastroenterol Hepatol. 2017;40:132‐141. [DOI] [PubMed] [Google Scholar]
- 4. Sharma A, Rao S. Constipation: pathophysiology and current therapeutic approaches. Handb Exp Pharmacol. 2017;239:59‐74. [DOI] [PubMed] [Google Scholar]
- 5. Basilisco G, Coletta M. Chronic constipation: a critical review. Dig Liver Dis. 2013;45:886‐893. [DOI] [PubMed] [Google Scholar]
- 6. Andrews CN, Storr M. The pathophysiology of chronic constipation. Can J Gastroenterol. 2011;25:16b‐21b. [PMC free article] [PubMed] [Google Scholar]
- 7. Johanson JF, Kralstein J. Chronic constipation: a survey of the patient perspective. Aliment Pharmacol Ther. 2007;25:599‐608. [DOI] [PubMed] [Google Scholar]
- 8. Bellini M, Usai‐Satta P, Bove A, et al. Chronic constipation diagnosis and treatment evaluation: the "CHRO.CO.DI.T.E." study. BMC Gastroenterol. 2017;17:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Chey WD. Symposium report: an evidence‐based approach to IBS and CIC: applying new advances to daily practice: a review of an adjunct clinical symposium of the American College of Gastroenterology Meeting October 16, 2016 * Las Vegas, Nevada. Gastroenterol Hepatol. 2017;13:1‐16. [PMC free article] [PubMed] [Google Scholar]
- 10. Allen‐Brady K, Tuteja AK. Chapter 33 ‐ Opioid‐induced bowel disorder and narcotic bowel syndrome. In: Rao SSC, Lee YY, Ghoshal UC, eds. Clinical and Basic Neurogastroenterology and Motility. Cambridge, MA: Academic Press; 2020:463‐475. [Google Scholar]
- 11. Bharucha AE, Dorn SD, Lembo A, Pressman A. American Gastroenterological Association medical position statement on constipation. Gastroenterology. 2013;144:211‐217. [DOI] [PubMed] [Google Scholar]
- 12. Sharma A, Rao S, Harrison JH. Anorectal function. Encyclopedia of Gastroenterology. 2nd edn. Vol. 1. Oxford: Academic Press; 2020:99‐104. [Google Scholar]
- 13. Patcharatrakul T, Rao SSC. Update on the pathophysiology and management of anorectal disorders. Gut Liv. 2018;12:375‐384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Rao SS. Dyssynergic defecation. Gastroenterol Clin North Am. 2001;30:97‐114. [DOI] [PubMed] [Google Scholar]
- 15. Kunkel D, Basseri RJ, Makhani MD, Chong K, Chang C, Pimentel M. Methane on breath testing is associated with constipation: a systematic review and meta‐analysis. Dig Dis Sci. 2011;56:1612‐1618. [DOI] [PubMed] [Google Scholar]
- 16. Wolf PG, Parthasarathy G, Chen J, et al. Assessing the colonic microbiome, hydrogenogenic and hydrogenotrophic genes, transit and breath methane in constipation. Neurogastroenterol Motil. 2017;29:1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Attaluri A, Jackson M, Valestin J, Rao SS. Methanogenic flora is associated with altered colonic transit but not stool characteristics in constipation without IBS. Am J Gastroenterol. 2010;105:1407‐1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Camilleri M, Ford AC. Irritable bowel syndrome: pathophysiology and current therapeutic approaches. Handb Exp Pharmacol. 2017;239:75‐113. [DOI] [PubMed] [Google Scholar]
- 19. Rao SSC, Xiang X, Yan Y, et al. Randomised clinical trial: linaclotide vs placebo—a study of bi‐directional gut and brain axis. Aliment Pharmacol Ther. 2020;51:1332‐1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ghoshal U, Shukla R, Srivastava D, Ghoshal UC. Irritable bowel syndrome, particularly the constipation‐predominant form, involves an increase in Methanobrevibacter smithii, which is associated with higher methane production. Gut Liv. 2016;10:932‐938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Andresen V, Banerji V, Hall G, Lass A, Emmanuel AV. The patient burden of opioid‐induced constipation: new insights from a large, multinational survey in five European countries. United European Gastroenterol J. 2018;6:1254‐1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Bell TJ, Panchal SJ, Miaskowski C, Bolge SC, Milanova T, Williamson R. The prevalence, severity, and impact of opioid‐induced bowel dysfunction: results of a US and European Patient Survey (PROBE 1). Pain Med. 2009;10:35‐42. [DOI] [PubMed] [Google Scholar]
- 23. Rao SS, Rattanakovit K, Patcharatrakul T. Diagnosis and management of chronic constipation in adults. Nat Rev Gastroenterol Hepatol. 2016;13:295‐305. [DOI] [PubMed] [Google Scholar]
- 24. Bharucha AE, Pemberton JH, Locke GR 3rd. American Gastroenterological Association technical review on constipation. Gastroenterology. 2013;144:218‐238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Yan Y, Jimenez E, Sharma A, et al. Sa1728 How useful is Constipation Stool APP compared to paper stool diary ‐ randomized study of constipation and healthy subjects. Gastroenterology. 2020;158:S‐400. [Google Scholar]
- 26. Lacy BE, Levenick JM, Crowell M. Chronic constipation: new diagnostic and treatment approaches. Therap Adv Gastroenterol. 2012;5:233‐247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Rao SSC. Plecanatide: a new guanylate cyclase agonist for the treatment of chronic idiopathic constipation. Therap Adv Gastroenterol. 2018;11:1756284818777945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Mearin F, Ciriza C, Mínguez M, et al. Clinical Practice Guideline: Irritable bowel syndrome with constipation and functional constipation in the adult. Rev Esp Enferm Dig. 2016;108:332‐363. [DOI] [PubMed] [Google Scholar]
- 29. Camilleri M, Ford AC, Mawe GM, et al. Chronic constipation. Nat Rev Dis Primers. 2017;3:17095. [DOI] [PubMed] [Google Scholar]
- 30. Sharma A, Kurek J, Morgan JC, Wakade C, Rao SSC. Constipation in Parkinson's disease: a nuisance or nuanced answer to the pathophysiological puzzle? Curr Gastroenterol Rep. 2018;20:1. [DOI] [PubMed] [Google Scholar]
- 31. Lee YY, Haque M, Lawenko R, Sharma A. Systemic disorders that affect gastrointestinal motility. Clinical and Basic Neurogastroenterology and Motility. Cambridge, MA: Academic Press; 2020:601‐618. [Google Scholar]
- 32. Serra J, Mascort‐Roca J, Marzo‐Castillejo M, et al. Clinical practice guidelines for the management of constipation in adults. Part 2: diagnosis and treatment. Gastroenterol Hepatol. 2017;40:303‐316. [DOI] [PubMed] [Google Scholar]
- 33. Tantiphlachiva K, Rao P, Attaluri A, Rao SS. Digital rectal examination is a useful tool for identifying patients with dyssynergia. Clin Gastroenterol Hepatol. 2010;8:955‐960. [DOI] [PubMed] [Google Scholar]
- 34. Rao SSC. Rectal exam: yes, it can and should be done in a busy practice! Am J Gastroenterol. 2018;113:635‐638. [DOI] [PubMed] [Google Scholar]
- 35. Carrington EV, Scott SM, Bharucha A, et al. Expert consensus document: advances in the evaluation of anorectal function. Nat Rev Gastroenterol Hepatol. 2018;15:309‐323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Carrington EV, Heinrich H, Knowles CH, et al. The international anorectal physiology working group (IAPWG) recommendations: Standardized testing protocol and the London classification for disorders of anorectal function. Neurogastroenterol Motil. 2020;32:e13679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Minguez M, Herreros B, Sanchiz V, et al. Predictive value of the balloon expulsion test for excluding the diagnosis of pelvic floor dyssynergia in constipation. Gastroenterology. 2004;126:57‐62. [DOI] [PubMed] [Google Scholar]
- 38. Bharucha AE, Wald A, Enck P, Rao S. Functional anorectal disorders. Gastroenterology. 2006;130:1510‐1518. [DOI] [PubMed] [Google Scholar]
- 39. Kang HR, Lee JE, Lee JS, et al. Comparison of high‐resolution anorectal manometry with water‐perfused anorectal manometry. J Neurogastroenterol Motil. 2015;21:126‐132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Rao SS, Azpiroz F, Diamant N, Enck P, Tougas G, Wald A. Minimum standards of anorectal manometry. Neurogastroenterol Motil. 2002;14:553‐559. [DOI] [PubMed] [Google Scholar]
- 41. Rao SS. Dyssynergic defecation and biofeedback therapy. Gastroenterol Clin North Am. 2008;37:569‐586, viii. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Sharif H, Devadason D, Abrehart N, Stevenson R, Marciani L. Imaging measurement of whole gut transit time in paediatric and adult functional gastrointestinal disorders: a systematic review and narrative synthesis. Diagnostics. 2019;9:221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lacy BE, Mearin F, Chang L, et al. Bowel disorders. Gastroenterology. 2016;150:1393‐1407.e5. [DOI] [PubMed] [Google Scholar]
- 44. Lee YY, Erdogan A, Rao SS. How to assess regional and whole gut transit time with wireless motility capsule. J Neurogastroenterol Motil. 2014;20:265‐270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Rao SSC, Kuo B, McCallum RW, et al. Investigation of colonic and whole‐gut transit with wireless motility capsule and radiopaque markers in constipation. Clin Gastroenterol Hepatol. 2009;7:537‐544. [DOI] [PubMed] [Google Scholar]
- 46. Rao SSC, Camilleri M, Hasler WL, et al. Evaluation of gastrointestinal transit in clinical practice: position paper of the American and European Neurogastroenterology and Motility Societies. Neurogastroenterol Motil. 2011;23:8‐23. [DOI] [PubMed] [Google Scholar]
- 47. Wang YT, Mohammed SD, Farmer AD, et al. Regional gastrointestinal transit and pH studied in 215 healthy volunteers using the wireless motility capsule: influence of age, gender, study country and testing protocol. Aliment Pharmacol Ther. 2015;42:761‐772. [DOI] [PubMed] [Google Scholar]
- 48. Singh S, Heady S, Coss‐Adame E, Rao SS. Clinical utility of colonic manometry in slow transit constipation. Neurogastroenterol Motil. 2013;25:487‐495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Saito YA, Schoenfeld P, Locke GR 3rd. The epidemiology of irritable bowel syndrome in North America: a systematic review. Am J Gastroenterol. 2002;97:1910‐1915. [DOI] [PubMed] [Google Scholar]
- 50. Serra J, Pohl D, Azpiroz F, et al. European Society of Neurogastroenterology and Motility guidelines on functional constipation in adults. Neurogastroenterol Motil. 2020;32:e13762. [DOI] [PubMed] [Google Scholar]
- 51. Attaluri A, Donahoe R, Valestin J, Brown K, Rao SS. Randomised clinical trial: dried plums (prunes) vs. psyllium for constipation. Aliment Pharmacol Ther. 2011;33:822‐828. [DOI] [PubMed] [Google Scholar]
- 52. Erdogan A, Rao SSC, Thiruvaiyaru D, et al. Randomised clinical trial: mixed soluble/insoluble fibre vs. psyllium for chronic constipation. Aliment Pharmacol Ther. 2016;44:35‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Chey SW, Chey WD, Jackson K, Eswaran S. S0454 Randomized, comparative effectiveness trial of green kiwifruit, psyllium, or prunes in U.S. patients with chronic constipation. Am J Gastroenterol. 2020;115:S229. [DOI] [PubMed] [Google Scholar]
- 54. Ford AC, Suares NC. Effect of laxatives and pharmacological therapies in chronic idiopathic constipation: systematic review and meta‐analysis. Gut. 2011;60:209‐218. [DOI] [PubMed] [Google Scholar]
- 55. Corazziari E, Badiali D, Bazzocchi G, et al. Long term efficacy, safety, and tolerabilitity of low daily doses of isosmotic polyethylene glycol electrolyte balanced solution (PMF‐100) in the treatment of functional chronic constipation. Gut. 2000;46:522‐526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Dipalma JA, Cleveland MV, McGowan J, Herrera JL. A randomized, multicenter, placebo‐controlled trial of polyethylene glycol laxative for chronic treatment of chronic constipation. Am J Gastroenterol. 2007;102:1436‐1441. [DOI] [PubMed] [Google Scholar]
- 57. Corazziari E, Badiali D, Habib FI, et al. Small volume isosmotic polyethylene glycol electrolyte balanced solution (PMF‐100) in treatment of chronic nonorganic constipation. Dig Dis Sci. 1996;41:1636‐1642. [DOI] [PubMed] [Google Scholar]
- 58. Lee‐Robichaud H, Thomas K, Morgan J, Nelson RL. Lactulose versus polyethylene glycol for chronic constipation. Cochrane Database Syst Rev. 2010;(7):Cd007570. [DOI] [PubMed] [Google Scholar]
- 59. Belsey JD, Geraint M, Dixon TA. Systematic review and meta analysis: polyethylene glycol in adults with non‐organic constipation. Int J Clin Pract. 2010;64:944‐955. [DOI] [PubMed] [Google Scholar]
- 60. Awad RA, Camacho S. A randomized, double‐blind, placebo‐controlled trial of polyethylene glycol effects on fasting and postprandial rectal sensitivity and symptoms in hypersensitive constipation‐predominant irritable bowel syndrome. Colorectal Dis. 2010;12:1131‐1138. [DOI] [PubMed] [Google Scholar]
- 61. Chapman RW, Stanghellini V, Geraint M, Halphen M. Randomized clinical trial: macrogol/PEG 3350 plus electrolytes for treatment of patients with constipation associated with irritable bowel syndrome. Am J Gastroenterol. 2013;108:1508‐1515. [DOI] [PubMed] [Google Scholar]
- 62. Kamm MA, Mueller‐Lissner S, Wald A, Richter E, Swallow R, Gessner U. Oral bisacodyl is effective and well‐tolerated in patients with chronic constipation. Clin Gastroenterol Hepatol. 2011;9:577‐583. [DOI] [PubMed] [Google Scholar]
- 63. Kienzle‐Horn S, Vix JM, Schuijt C, Peil H, Jordan CC, Kamm MA. Efficacy and safety of bisacodyl in the acute treatment of constipation: a double‐blind, randomized, placebo‐controlled study. Aliment Pharmacol Ther. 2006;23:1479‐1488. [DOI] [PubMed] [Google Scholar]
- 64. Mueller‐Lissner S, Kamm MA, Wald A, et al. Multicenter, 4‐week, double‐blind, randomized, placebo‐controlled trial of sodium picosulfate in patients with chronic constipation. Am J Gastroenterol. 2010;105:897‐903. [DOI] [PubMed] [Google Scholar]
- 65. Brancale A, Shailubhai K, Ferla S, Ricci A, Bassetto M, Jacob GS. Therapeutically targeting guanylate cyclase‐C: computational modeling of plecanatide, a uroguanylin analog. Pharmacol Res Perspect. 2017;5:e00295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Islam BN, Sharman SK, Browning DD. Clinical utility of plecanatide in the treatment of chronic idiopathic constipation. Int J Gen Med. 2018;11:323‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Waldman SA, Camilleri M. Guanylate cyclase‐C as a therapeutic target in gastrointestinal disorders. Gut. 2018;67:1543‐1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Hannig G, Tchernychev B, Kurtz CB, Bryant AP, Currie MG, Silos‐Santiago I. Guanylate cyclase‐C/cGMP: an emerging pathway in the regulation of visceral pain. Front Mol Neurosci. 2014;7:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Waldman SA, Tenenbaum R, Foehl HC, Winkle P, Griffin P. Blunted evoked prouroguanylin endocrine secretion in chronic constipation. Clin Transl Gastroenterol. 2019;10:e00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Atluri DK, Chandar AK, Bharucha AE, Falck‐Ytter Y. Effect of linaclotide in irritable bowel syndrome with constipation (IBS‐C): a systematic review and meta‐analysis. Neurogastroenterol Motil. 2014;26:499‐509. [DOI] [PubMed] [Google Scholar]
- 71. Brenner DM, Fogel R, Dorn SD, et al. Efficacy, safety, and tolerability of plecanatide in patients with irritable bowel syndrome with constipation: results of two phase 3 randomized clinical trials. Am J Gastroenterol. 2018;113:735‐745. [DOI] [PubMed] [Google Scholar]
- 72. Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: a 26‐week, randomized, double‐blind, placebo‐controlled trial to evaluate efficacy and safety. Am J Gastroenterol. 2012;107:1702‐1712. [DOI] [PubMed] [Google Scholar]
- 73. DeMicco M, Barrow L, Hickey B, Shailubhai K, Griffin P. Randomized clinical trial: efficacy and safety of plecanatide in the treatment of chronic idiopathic constipation. Therap Adv Gastroenterol. 2017;10:837‐851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lacy BE, Schey R, Shiff SJ, et al. Linaclotide in chronic idiopathic constipation patients with moderate to severe abdominal bloating: a randomized, controlled trial. PLoS One. 2015;10:e0134349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Lembo AJ, Kurtz CB, MacDougall JE, et al. Efficacy of linaclotide for patients with chronic constipation. Gastroenterology. 2010;138:886‐895.e1. [DOI] [PubMed] [Google Scholar]
- 76. Lembo AJ, Schneier HA, Shiff SJ, et al. Two randomized trials of linaclotide for chronic constipation. N Engl J Med. 2011;365:527‐536. [DOI] [PubMed] [Google Scholar]
- 77. Miner PB, Koltun WD, Wiener GJ, et al. A randomized phase III clinical trial of plecanatide, a uroguanylin analog, in patients with chronic idiopathic constipation. Am J Gastroenterol. 2017;112:613‐621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Rao S, Lembo AJ, Shiff SJ, et al. A 12‐week, randomized, controlled trial with a 4‐week randomized withdrawal period to evaluate the efficacy and safety of linaclotide in irritable bowel syndrome with constipation. Am J Gastroenterol. 2012;107:1714‐1724; quiz p.1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Schoenfeld P, Lacy BE, Chey WD, et al. Low‐dose linaclotide (72μg) for chronic idiopathic constipation: a 12‐week, randomized, double‐blind, placebo‐controlled trial. Am J Gastroenterol. 2018;113:105‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Videlock EJ, Cheng V, Cremonini F. Effects of linaclotide in patients with irritable bowel syndrome with constipation or chronic constipation: a meta‐analysis. Clin Gastroenterol Hepatol. 2013;11:1084‐1092.e3; quiz e68. [DOI] [PubMed] [Google Scholar]
- 81. Yang Y, Fang J, Guo X, et al. Linaclotide in irritable bowel syndrome with constipation: a Phase 3 randomized trial in China and other regions. J Gastroenterol Hepatol. 2018;33:980‐989. [DOI] [PubMed] [Google Scholar]
- 82. Fukudo S, Miwa H, Nakajima A, et al. A randomized controlled and long‐term linaclotide study of irritable bowel syndrome with constipation patients in Japan. Neurogastroenterol Motil. 2018;30:e13444. [DOI] [PubMed] [Google Scholar]
- 83. Quigley EM, Tack J, Chey WD, et al. Randomised clinical trials: linaclotide phase 3 studies in IBS‐C ‐ a prespecified further analysis based on European Medicines Agency‐specified endpoints. Aliment Pharmacol Ther. 2013;37:49‐61. [DOI] [PubMed] [Google Scholar]
- 84. Barish CF, Crozier RA, Griffin PH. Long‐term treatment with plecanatide was safe and tolerable in patients with irritable bowel syndrome with constipation. Curr Med Res Opin. 2019;35:81‐85. [DOI] [PubMed] [Google Scholar]
- 85. Barish CF, Griffin P. Safety and tolerability of plecanatide in patients with chronic idiopathic constipation: long‐term evidence from an open‐label study. Curr Med Res Opin. 2018;34:751‐755. [DOI] [PubMed] [Google Scholar]
- 86. Griffin PH. ,Comment on meta analysis by Shah et al. Am J Gastroenterol. 2018;113:1395. [DOI] [PubMed] [Google Scholar]
- 87. Niewinna K, Zielinska A, Fichna J. Recent advances in the pharmacological management of constipation predominant irritable bowel syndrome. Expert Opin Pharmacother. 2020;21:73‐84. [DOI] [PubMed] [Google Scholar]
- 88. Atkinson W, Lockhart S, Whorwell PJ, Keevil B, Houghton LA. Altered 5‐hydroxytryptamine signaling in patients with constipation‐ and diarrhea‐predominant irritable bowel syndrome. Gastroenterology. 2006;130:34‐43. [DOI] [PubMed] [Google Scholar]
- 89. Dunlop SP, Coleman NS, Blackshaw E, et al. Abnormalities of 5‐hydroxytryptamine metabolism in irritable bowel syndrome. Clin Gastroenterol Hepatol. 2005;3:349‐357. [DOI] [PubMed] [Google Scholar]
- 90. Bonfiglio F, Liu X, Smillie C, et al. GWAS of stool frequency reveals genes, pathways, and cell types relevant to human gastrointestinal motility and irritable bowel syndrome. medRxiv. 2020:2020.2006.2017.20132555. [Google Scholar]
- 91. El‐Salhy M, Norrgard O, Spinnell S. Abnormal colonic endocrine cells in patients with chronic idiopathic slow‐transit constipation. Scand J Gastroenterol. 1999;34:1007‐1011. [DOI] [PubMed] [Google Scholar]
- 92. Jiang C, Xu Q, Wen X, Sun H. Current developments in pharmacological therapeutics for chronic constipation. Acta pharmaceutica Sinica B. 2015;5:300‐309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Prichard DO, Bharucha AE. Recent advances in understanding and managing chronic constipation. F1000Res. 2018;7:1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Bouras EP, Camilleri M, Burton DD, Thomforde G, McKinzie S, Zinsmeister AR. Prucalopride accelerates gastrointestinal and colonic transit in patients with constipation without a rectal evacuation disorder. Gastroenterology. 2001;120:354‐360. [DOI] [PubMed] [Google Scholar]
- 95. In brief: Tegaserod (Zelnorm) returns. Med Lett Drugs Ther. 2019;61:72. [PubMed] [Google Scholar]
- 96. Camilleri M, Kerstens R, Rykx A, Vandeplassche L. A placebo‐controlled trial of prucalopride for severe chronic constipation. N Engl J Med. 2008;358:2344‐2354. [DOI] [PubMed] [Google Scholar]
- 97. Quigley EM, Vandeplassche L, Kerstens R, Ausma J. Clinical trial: the efficacy, impact on quality of life, and safety and tolerability of prucalopride in severe chronic constipation–a 12‐week, randomized, double‐blind, placebo‐controlled study. Aliment Pharmacol Ther. 2009;29:315‐328. [DOI] [PubMed] [Google Scholar]
- 98. Tack J, van Outryve M, Beyens G, Kerstens R, Vandeplassche L. Prucalopride (Resolor) in the treatment of severe chronic constipation in patients dissatisfied with laxatives. Gut. 2009;58:357‐365. [DOI] [PubMed] [Google Scholar]
- 99. Camilleri M, Van Outryve MJ, Beyens G, Kerstens R, Robinson P, Vandeplassche L. Clinical trial: the efficacy of open‐label prucalopride treatment in patients with chronic constipation ‐ follow‐up of patients from the pivotal studies. Aliment Pharmacol Ther. 2010;32:1113‐1123. [DOI] [PubMed] [Google Scholar]
- 100. Camilleri M, Piessevaux H, Yiannakou Y, et al. Efficacy and safety of prucalopride in chronic constipation: an integrated analysis of six randomized, controlled clinical trials. Dig Dis Sci. 2016;61:2357‐2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Sajid MS, Hebbar M, Baig MK, Li A, Philipose Z. Use of prucalopride for chronic constipation: a systematic review and meta‐analysis of published randomized, controlled trials. J Neurogastroenterol Motil. 2016;22:412‐422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Tack J, Quigley E, Camilleri M, Vandeplassche L, Kerstens R. Efficacy and safety of oral prucalopride in women with chronic constipation in whom laxatives have failed: an integrated analysis. United European Gastroenterol J. 2013;1:48‐59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Chey WD, Lembo AJ, Rosenbaum DP. Efficacy of tenapanor in treating patients with irritable bowel syndrome with constipation: a 12‐week, placebo‐controlled phase 3 trial (T3MPO‐1). Am J Gastroenterol. 2020;115:281‐293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Eswaran S, Guentner A, Chey WD. Emerging pharmacologic therapies for constipation‐predominant irritable bowel syndrome and chronic constipation. J Neurogastroenterol Motil. 2014;20:141‐151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Abrahamsson H, Ostlund‐Lindqvist AM, Nilsson R, Simren M, Gillberg PG. Altered bile acid metabolism in patients with constipation‐predominant irritable bowel syndrome and functional constipation. Scand J Gastroenterol. 2008;43:1483‐1488. [DOI] [PubMed] [Google Scholar]
- 106. Bazzoli F, Malavolti M, Petronelli A, Barbara L, Roda E. Treatment of constipation with chenodeoxycholic acid. J Int Med Res. 1983;11:120‐123. [DOI] [PubMed] [Google Scholar]
- 107. Rao AS, Wong BS, Camilleri M, et al. Chenodeoxycholate in females with irritable bowel syndrome‐constipation: a pharmacodynamic and pharmacogenetic analysis. Gastroenterology. 2010;139:1549‐1558, 1558.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Chey WD, Camilleri M, Chang L, Rikner L, Graffner H. A randomized placebo‐controlled phase IIb trial of a3309, a bile acid transporter inhibitor, for chronic idiopathic constipation. Am J Gastroenterol. 2011;106:1803‐1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Nakajima A, Seki M, Taniguchi S, et al. Safety and efficacy of elobixibat for chronic constipation: results from a randomised, double‐blind, placebo‐controlled, phase 3 trial and an open‐label, single‐arm, phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3:537‐547. [DOI] [PubMed] [Google Scholar]
- 110. Kumagai Y, Amano H, Sasaki Y, et al. Effect of single and multiple doses of elobixibat, an ileal bile acid transporter inhibitor, on chronic constipation: a randomized controlled trial. Br J Clin Pharmacol. 2018;84:2393‐2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Nakajima A, Seki M, Taniguchi S. Determining an optimal clinical dose of elobixibat, a novel inhibitor of the ileal bile acid transporter, in Japanese patients with chronic constipation: a phase II, multicenter, double‐blind, placebo‐controlled randomized clinical trial. J Gastroenterol. 2018;53:525‐534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Bassil AK, Borman RA, Jarvie EM, et al. Activation of prostaglandin EP receptors by lubiprostone in rat and human stomach and colon. Br J Pharmacol. 2008;154:126‐135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Lembo AJ, Johanson JF, Parkman HP, Rao SS, Miner PB Jr, Ueno R. Long‐term safety and effectiveness of lubiprostone, a chloride channel (ClC‐2) activator, in patients with chronic idiopathic constipation. Dig Dis Sci. 2011;56:2639‐2645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Johanson JF, Morton D, Geenen J, Ueno R. Multicenter, 4‐week, double‐blind, randomized, placebo‐controlled trial of lubiprostone, a locally‐acting type‐2 chloride channel activator, in patients with chronic constipation. Am J Gastroenterol. 2008;103:170‐177. [DOI] [PubMed] [Google Scholar]
- 115. Johanson JF, Drossman DA, Panas R, Wahle A, Ueno R. Clinical trial: phase 2 study of lubiprostone for irritable bowel syndrome with constipation. Aliment Pharmacol Ther. 2008;27:685‐696. [DOI] [PubMed] [Google Scholar]
- 116. Barish CF, Drossman D, Johanson JF, Ueno R. Efficacy and safety of lubiprostone in patients with chronic constipation. Dig Dis Sci. 2010;55:1090‐1097. [DOI] [PubMed] [Google Scholar]
- 117. Chey WD, Drossman DA, Johanson JF, Scott C, Panas RM, Ueno R. Safety and patient outcomes with lubiprostone for up to 52 weeks in patients with irritable bowel syndrome with constipation. Aliment Pharmacol Ther. 2012;35:587‐599. [DOI] [PubMed] [Google Scholar]
- 118. Li F, Fu T, Tong W‐D, et al. Lubiprostone is effective in the treatment of chronic idiopathic constipation and irritable bowel syndrome. Mayo Clin Proc. 2016;91:456‐468. [DOI] [PubMed] [Google Scholar]
- 119. Fukudo S, Hongo M, Kaneko H, Ueno R. Efficacy and safety of oral lubiprostone in constipated patients with or without irritable bowel syndrome: a randomized, placebo‐controlled and dose‐finding study. Neurogastroenterol Motil. 2011;23:544‐e205. [DOI] [PubMed] [Google Scholar]
- 120. Fukudo S, Hongo M, Kaneko H, Takano M, Ueno R. Lubiprostone increases spontaneous bowel movement frequency and quality of life in patients with chronic idiopathic constipation. Clin Gastroenterol Hepatol. 2015;13:294‐301.e5. [DOI] [PubMed] [Google Scholar]
- 121. Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation‐associated irritable bowel syndrome–results of two randomized, placebo‐controlled studies. Aliment Pharmacol Ther. 2009;29:329‐341. [DOI] [PubMed] [Google Scholar]
- 122. Johanson JF, Ueno R. Lubiprostone, a locally acting chloride channel activator, in adult patients with chronic constipation: a double‐blind, placebo‐controlled, dose‐ranging study to evaluate efficacy and safety. Aliment Pharmacol Ther. 2007;25:1351‐1361. [DOI] [PubMed] [Google Scholar]
- 123. Rao SSC, Lichtlen P, Habibi S. Effects of lubiprostone, an intestinal secretagogue, on electrolyte homeostasis in chronic idiopathic and opioid‐induced constipation. J Clin Gastroenterol. 2020;32(11). Publish Ahead of Print. 10.1097/MCG.0000000000001385 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Leppert W. The role of opioid receptor antagonists in the treatment of opioid‐induced constipation: a review. Adv Ther. 2010;27:714‐730. [DOI] [PubMed] [Google Scholar]
- 125. Viscusi ER. Clinical overview and considerations for the management of opioid‐induced constipation in patients with chronic noncancer pain. Clin J Pain. 2019;35:174‐188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Crockett SD, Greer KB, Heidelbaugh JJ, Falck‐Ytter Y, Hanson BJ, Sultan S. American Gastroenterological Association Institute Guideline on the medical management of opioid‐induced constipation. Gastroenterology. 2019;156:218‐226. [DOI] [PubMed] [Google Scholar]
- 127. Vijayvargiya P, Camilleri M, Vijayvargiya P, Erwin P, Murad MH. Systematic review with meta‐analysis: efficacy and safety of treatments for opioid‐induced constipation. Aliment Pharmacol Ther. 2020;52:37‐53. [DOI] [PubMed] [Google Scholar]
- 128. Rao SS, Benninga MA, Bharucha AE, Chiarioni G, Di Lorenzo C, Whitehead WE. ANMS‐ESNM position paper and consensus guidelines on biofeedback therapy for anorectal disorders. Neurogastroenterol Motil. 2015;27:594‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Rao SSC, Seaton K, Miller M, et al. Randomized controlled trial of biofeedback, sham feedback, and standard therapy for dyssynergic defecation. Clin Gastroenterol Hepatol. 2007;5:331‐338. [DOI] [PubMed] [Google Scholar]
- 130. Heymen S, Scarlett Y, Jones K, Ringel Y, Drossman D, Whitehead WE. Randomized, controlled trial shows biofeedback to be superior to alternative treatments for patients with pelvic floor dyssynergia‐type constipation. Dis Colon Rectum. 2007;50:428‐441. [DOI] [PubMed] [Google Scholar]
- 131. Chiarioni G, Whitehead WE, Pezza V, Morelli A, Bassotti G. Biofeedback is superior to laxatives for normal transit constipation due to pelvic floor dyssynergia. Gastroenterology. 2006;130:657‐664. [DOI] [PubMed] [Google Scholar]
- 132. Rao SSC, Valestin J, Brown CK, Zimmerman B, Schulze K. Long‐term efficacy of biofeedback therapy for dyssynergic defecation: randomized controlled trial. Am J Gastroenterol. 2010;105:890‐896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Lee HJ, Boo SJ, Jung KW, et al. Long‐term efficacy of biofeedback therapy in patients with dyssynergic defecation: results of a median 44 months follow‐up. Neurogastroenterol Motil. 2015;27:787‐795. [DOI] [PubMed] [Google Scholar]
- 134. Patcharatrakul T, Gonlachanvit S. Outcome of biofeedback therapy in dyssynergic defecation patients with and without irritable bowel syndrome. J Clin Gastroenterol. 2011;45:593‐598. [DOI] [PubMed] [Google Scholar]
- 135. Chiarioni G, Salandini L, Whitehead WE. Biofeedback benefits only patients with outlet dysfunction, not patients with isolated slow transit constipation. Gastroenterology. 2005;129:86‐97. [DOI] [PubMed] [Google Scholar]
- 136. Shin JK, Cheon JH, Kim ES, et al. Predictive capability of anorectal physiologic tests for unfavorable outcomes following biofeedback therapy in dyssynergic defecation. J Korean Med Sci. 2010;25:1060‐1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Rao SSC, Valestin JA, Xiang X, Hamdy S, Bradley CS, Zimmerman MB. Home‐based versus office‐based biofeedback therapy for constipation with dyssynergic defecation: a randomised controlled trial. Lancet Gastroenterol Hepatol. 2018;3:768‐777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Rao SSC, Go JT, Valestin J, Schneider J. Home biofeedback for the treatment of dyssynergic defecation: does it improve quality of life and is it cost‐effective? Am J Gastroenterol. 2019;114:938‐944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Luthra P, Camilleri M, Burr NE, Quigley EMM, Black CJ, Ford AC. Efficacy of drugs in chronic idiopathic constipation: a systematic review and network meta‐analysis. Lancet Gastroenterol Hepatol. 2019;4:831‐844. [DOI] [PubMed] [Google Scholar]
- 140. Tse Y, Armstrong D, Andrews CN, et al. Treatment algorithm for chronic idiopathic constipation and constipation‐predominant irritable bowel syndrome derived from a Canadian national survey and needs assessment on choices of therapeutic agents. Can J Gastroenterol Hepatol. 2017;2017:8612189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Harris LA, Horn J, Kissous‐Hunt M, Magnus L, Quigley EMM. The Better Understanding and Recognition of the Disconnects, Experiences, and Needs of patients with Chronic Idiopathic Constipation (BURDEN‐CIC) study: results of an online questionnaire. Adv Ther. 2017;34:2661‐2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Soufi‐Afshar I, Moghadamnia A, Bijani A, Kazemi S, Shokri‐Shirvani J. Comparison of pyridostigmine and bisacodyl in the treatment of refractory chronic constipation. Caspian J Intern Med. 2016;7:19‐24. [PMC free article] [PubMed] [Google Scholar]
- 143. Bharucha AE, Low PA, Camilleri M, Burton D, Gehrking TL, Zinsmeister AR. Pilot study of pyridostigmine in constipated patients with autonomic neuropathy. Clin Auton Res. 2008;18:194‐202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Bharucha AE, Low P, Camilleri M, et al. A randomised controlled study of the effect of cholinesterase inhibition on colon function in patients with diabetes mellitus and constipation. Gut. 2013;62:708‐715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Rao SSC, Lembo A, Chey WD, Friedenberg K, Quigley EMM. Effects of the vibrating capsule on colonic circadian rhythm and bowel symptoms in chronic idiopathic constipation. Neurogastroenterol Motil. 2020;32:e13890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. NICE Guidance: Irritable bowel syndrome in adults: diagnosis and management. 2008. https://www.nice.org.uk/guidance/cg61.Date Accessed: March 10, 2020.
- 147. Hussain ZH, Everhart K, Lacy BE. Treatment of chronic constipation: prescription medications and surgical therapies. Gastroenterol Hepatol. 2015;11:104‐114. [PMC free article] [PubMed] [Google Scholar]
- 148. Griffiths DM, Malone PS. The Malone antegrade continence enema. J Pediatr Surg. 1995;30:68‐71. [DOI] [PubMed] [Google Scholar]
- 149. Lees NP, Hodson P, Hill J, Pearson RC, MacLennan I. Long‐term results of the antegrade continent enema procedure for constipation in adults. Colorectal Dis. 2004;6:362‐368. [DOI] [PubMed] [Google Scholar]
- 150. Portier G, Ghouti L, Kirzin S, Chauffour M, Lazorthes F. Malone antegrade colonic irrigation: ileal neoappendicostomy is the preferred procedure in adults. Int J Colorectal Dis. 2006;21:458‐460. [DOI] [PubMed] [Google Scholar]
- 151. Teichman JM, Zabihi N, Kraus SR, Harris JM, Barber DB. Long‐term results for Malone antegrade continence enema for adults with neurogenic bowel disease. Urology. 2003;61:502‐506. [DOI] [PubMed] [Google Scholar]
- 152. Meurette G, Lehur PA, Coron E, Regenet N. Long‐term results of Malone's procedure with antegrade irrigation for severe chronic constipation. Gastroenterol Clin Biol. 2010;34:209‐212. [DOI] [PubMed] [Google Scholar]
- 153. Duchalais E, Meurette G, Mantoo SK, et al. Percutaneous endoscopic caecostomy for severe constipation in adults: feasibility, durability, functional and quality of life results at 1 year follow‐up. Surg Endosc. 2015;29:620‐626. [DOI] [PubMed] [Google Scholar]
- 154. Uno Y. Introducer method of percutaneous endoscopic cecostomy and antegrade continence enema by use of the Chait Trapdoor cecostomy catheter in patients with adult neurogenic bowel. Gastrointest Endosc. 2006;63:666‐673. [DOI] [PubMed] [Google Scholar]
- 155. Gerharz EW, Vik V, Webb G, Leaver R, Shah PJ, Woodhouse CR. The value of the MACE (Malone antegrade colonic enema) procedure in adult patients. J Am Coll Surg. 1997;185:544‐547. [DOI] [PubMed] [Google Scholar]
- 156. Biyani D, Barrow E, Hodson P, Watson AJ, Maclennan I. Endoscopically placed caecostomy buttons: a trial ACE procedure. Colorectal Dis. 2007;9:373‐376. [DOI] [PubMed] [Google Scholar]
- 157. Hirst GR, Arumugam PJ, Watkins AJ, et al. Antegrade continence enema in the treatment of obstructed defaecation with or without faecal incontinence. Tech Coloproctol. 2005;9:217‐221. [DOI] [PubMed] [Google Scholar]
- 158. Altomare DF, Rinaldi M, Rubini D, et al. Long‐term functional assessment of antegrade colonic enema for combined incontinence and constipation using a modified Marsh and Kiff technique. Dis Colon Rectum. 2007;50:1023‐1031. [DOI] [PubMed] [Google Scholar]
- 159. Poirier M, Abcarian H, Nelson R. Malone antegrade continent enema: an alternative to resection in severe defecation disorders. Dis Colon Rectum. 2007;50:22‐28. [DOI] [PubMed] [Google Scholar]
- 160. Worsoe J, Christensen P, Krogh K, Buntzen S, Laurberg S. Long‐term results of antegrade colonic enema in adult patients: assessment of functional results. Dis Colon Rectum. 2008;51:1523‐1528. [DOI] [PubMed] [Google Scholar]
- 161. Whiteley I, Stewart P. Antegrade continence enema (ACE) in adults: a review of long‐term outcomes and complications from a single institution. Aust N Z Continence J. 2020;26. [Google Scholar]
- 162. Chan DS, Delicata RJ. Meta‐analysis of antegrade continence enema in adults with faecal incontinence and constipation. Br J Surg. 2016;103:322‐327. [DOI] [PubMed] [Google Scholar]
- 163. Rao SSC, Seaton K, Miller MJ, et al. Psychological profiles and quality of life differ between patients with dyssynergia and those with slow transit constipation. J Psychosom Res. 2007;63:441‐449. [DOI] [PubMed] [Google Scholar]
- 164. Almario CV, Spiegel BMR. Employing irritable bowel syndrome patient‐reported outcomes in the clinical trenches. Clin Gastroenterol Hepatol. 2018;16:462‐466.e2. [DOI] [PubMed] [Google Scholar]
- 165. Yan Y, Karunaratne T, Jimenez E, et al. Electronic APP vs. paper form stool diary for fecal incontinence: 2877. Am J Gastroenterol. 2019;114:S1577–S1578. (2019 ACG Annual Meeting Abstracts). [Google Scholar]
- 166. Zia J, Schroeder J, Munson S, et al. Feasibility and usability pilot study of a novel irritable bowel syndrome food and gastrointestinal symptom journal smartphone app. Clin Transl Gastroenterol. 2016;7:e147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Johanson JF, Wald A, Tougas G, et al. Effect of tegaserod in chronic constipation: a randomized, double‐blind, controlled trial. Clin Gastroenterol Hepatol. 2004;2:796‐805. [DOI] [PubMed] [Google Scholar]
- 168. Sharma S, Agarwal B. Scoring systems in evaluation of constipation and obstructed defecation syndrome (ODS). J Int Med Sci Acad. 2012;25:57‐59. [Google Scholar]
- 169. McCrea HJ, Ment LR. The dianeonategnosis, management, and postnatal prevention of intraventricular hemorrhage in the preterm. Clin Perinatol. 2008;35:777‐792, vii. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Frank L, Kleinman L, Farup C, Taylor L, Miner P Jr. Psychometric validation of a constipation symptom assessment questionnaire. Scand J Gastroenterol. 1999;34:870‐877. [DOI] [PubMed] [Google Scholar]
- 171. Simrén M, Tack J. New treatments and therapeutic targets for IBS and other functional bowel disorders. Nat Rev Gastroenterol Hepatol. 2018;15:589‐605. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing not applicable to this article as no datasets were generated or analysed during the current study.