Large Bowel


Preoperative Bowel Preparation: Is It Necessary?

Anna Chudnovets, MD,
Sandy Hwang Fang, MD

Patients who undergo colorectal surgery for cancer and inflammatory bowel disease often have associated risk factors that impair wound healing, such as chemoradiation therapy, steroid therapy, and malnutrition. Additional complicating factors include case complexity, perineal wounds, and combined multispecialty cases. Due to the nature of the colon and rectum yielding high bacterial inoculum and these contributing factors, the risk of postoperative infections after surgery is significant, with an incidence approaching 40%.

The overall objective for the use of bowel preparation in colorectal surgery is to reduce postoperative complications:

  • 1

    Decrease the rate of surgical site infections (SSIs)

  • 2

    Decrease anastomotic leak rates

  • 3

    Alter gut microbiome

A bowel preparation accomplishes these goals via two mechanisms: the evacuation of fecal material from the colon and the eradication of stool bacterial load. In addition, a bowel preparation should not alter the histologic appearance of the mucosa.

Historically, bowel preparations have created a harsh physiologic response to the human body. Newer bowel preparation formulations are smaller in volume, ingested in a short period of time with effective evacuation of stool, while also decreasing gastrointestinal discomfort and side effects, such as nausea and emesis. In addition, these newer drugs produce no fluid shifts, electrolyte imbalances, or dehydration, which optimizes the patient, leading into the operative stage of colorectal surgery with goal-directed fluid therapy (GDFT) as part of the enhanced recovery pathway or enhanced recovery after surgery (ERP or ERAS).

This chapter discusses the bowel preparation types, the efficacy of mechanical bowel preparations (MBP) versus oral antibiotics (OA) versus no bowel preparation, colorectal disease pathology and indications for bowel preparation, and its role in ERAS.

TYPES OF BOWEL PREPARATION

There are two types of bowel preparation: mechanical bowel preparation and oral antibiotics. MBPs are oral cathartics that clear feculent material from the colon. OA decrease intraluminal bacterial load.

Mechanical Bowel Preparation

Two types of MBPs exist: hyperosmotic versus isosmotic ( Table 1 ). Hyperosmotic MBPs (e.g., magnesium citrate, sodium phosphate) exert an osmotic effect, drawing fluid into the bowel lumen, as a mechanism of flushing out colonic contents. Although patients are able to tolerate ingestion of these lower volume formulations, they cause electrolyte and fluid imbalances and dehydration. Patients subsequently become orthostatic and may develop acute kidney injury. Contraindications to hyperosmotic solutions include renal failure, acute coronary syndrome, congestive heart failure, bowel obstruction, ileus, intestinal malabsorption, and ascites. The use of hyperosmotic MBP solutions is not recommended in current consensus guidelines for ERPs.

TABLE 1
Mechanical Bowel Preparations
Adjuncts to Mechanical Preparation Agent Volume/Dose Mechanism Use
PEG (electrolyte lavage) Colyte*GoLYTELY* 3785 mL4000 mL No solid food for at least 2 hours before ingestion of the solution; 240 mL (8 oz) every 10 minutes until rectal output is clear or 4 L are consumed Divided dose regimens (3 L the night before procedure, 1 L morning of procedure) may improve patient tolerance. PEG is considered safer than osmotic laxatives/NaP for patients with electrolyte/fluid imbalances, renal or liver insufficiency, CHF, or renal or liver failure.
Sulfate-free PEG (improved smell/taste, more palatable for patients) NuLYTELY*TriLyte* 4000 mL4000 mL No solid food for at least 2 hours before taking the solution; 240 mL (8 oz) every 10 minutes until rectal output is clear or 4 L are consumed Similar efficacy to PEG
Low-volume PEG and bisacodyl tablets (decrease volume-related discomfort [e.g., bloating, cramping]) HalfLytely and bisacodyl tablet bowel prepMiraLAX 2000 mL255 g in 2000 mL Only clear liquids on the day of the preparation. Dosage is four bisacodyl delayed-release tablets (5 mg) at noon. Wait for bowel movement or maximum of 6 hours; 240 mL (8 oz) low-volume PEG (i.e., HalfLytely) or 240 mL (8 oz) of clear liquid containing one capful of MiraLAX or other PEG-3350 regimen every 10 minutes until 2 L are consumed. Equally effective as 4 L solutions; additional studies needed regarding safety
Aqueous NaP solutions Fleet 90 mL with 48 oz additional liquid Only clear liquids can be consumed on the day of preparation. Two doses of 30–45 mL (2–3 tbsp.) of oral solution are given at least 10–12 hours apart. Each dose is taken with at least 8 oz of liquid followed by an additional minimum of at least 16 oz of liquid. The second dose must be taken at least 3 hours before the procedure. May cause significant fluid shifts. Not for use in pediatric or elderly patients or those with bowel obstruction, gut dysmotility, other structural intestinal disorders, renal or liver failure, or congestive heart failure. NaP may cause ulceration or mucosal abnormalities; do not use in patients with inflammatory bowel disease. Patients with compromised renal function or those taking ACE inhibitors or ARBs are at risk for phosphate nephropathy. In 2006, the FDA issued an alert regarding the risk for acute phosphate nephropathy, a type of acute renal failure, with use of oral sodium phosphate solution or tablets.
Oral sodium phosphate (tablet) Visicol (discontinued) 32–40 tablets with 48 oz clear liquid Dosage is 32–40 tablets: 20 tablets on the evening before the procedure and 12–20 tablets the day of the procedure (3–5 hours before). The 20 tablets are taken as 4 tablets every 15 minutes with 8 oz of clear liquid. Bisacodyl is prescribed by some physicians as an adjunct. Early tablet composition included higher concentration of microcrystalline cellulose per tablet, which left residue obscuring the mucosal surface. Later tablet composition decreased microcrystalline cellulose concentration. Overall, tablet NaP is not associated with significantly improved patient tolerance when compared with aqueous NaP.
Enemas Tap water 500–1000 mL Distention and lavage of rectum and distal colon Routine addition of enemas to oral preparation does not improve the quality of bowel cleansing but does increase patient discomfort. Use enemas in patients presenting for endoscopy with poor distal colon preparation and in patients with defunctionalized distal colon (e.g., Hartmann’s).
Soap suds 500–1000 mL
Fleet enema 135 mL
Fleet bisacodyl Enema 10 mg 1.25 oz 37.5 mL
Fleet mineral oil 480 mL
Bisacodyl Bisacodyl 5-mg tablet Poorly absorbed diphenylmethane that stimulates colonic peristalsis, used as adjunct for NaP or PEG preparations Has been found to decrease the volume of PEG preparation required
Saline laxatives Magnesium citrate (liquid)
Picolax (sodium picosulfate/magnesium citrate)
250–300 mL Hyperosmotic saline laxatives that increase motility by increased intraluminal volume Addition of magnesium citrate to PEG allows for lower volume preparation. Use with extreme caution in patients with renal insufficiency or renal failure because of exclusive renal excretion of magnesium.
Senna Senna
Senokot
X-Prep Syrup (8 mg/5 mL)
Anthraquinone derivatives (glycosides and sennosides) are activated by colonic bacteria and directly increase the rate of colonic motility, with a subsequent increase in colonic transit and reduced water and electrolyte secretion. Senna with PEG may improve the quality of preparation and reduce volume required.
Simethicone Gas-X
Mylicon
Mylanta
Generic formulations (80 mg)
Antiflatulent, often used to prevent foam formation after PEG preparation. Mechanism of action is unclear. May improve lumen visualization and patient toleration of bowel prep
Metoclopramide Reglan
Generic formulations also available
5 mg Dopamine antagonist gastro-prokinetic, increasing the amplitude of gastric contraction, with increased peristalsis in duodenum and jejunum but without change in colonic motility May reduce nausea, bloating; does not improve colonic cleansing
Carbohydrate-electrolyte solutions Gatorade
E-Lyte
Generic formulations
20 oz Used with PEG and/or NaP solution to improve flavor and prevent NaP-related fluid and electrolyte shifts Carbohydrate-based solutions more palatable for patients; however, associates with a theoretical risk of cautery-induced explosion if these carbohydrates are metabolized by colonic bacteria into explosive gases.
*Flavored options are available.
ACE, Angiotensin-converting enzyme; ARB, angiotensin receptor blocker; CHF, congestive heart failure; FDA, US Food and Drug Administration; NaP, sodium phosphate; PEG, polyethylene glycol.
Modified from Wexner SD, Beck DE, Baron TH, et al. A consensus document on bowel preparation before colonoscopy: prepared by a task force from the American Society of Colon and Rectal Surgeons (ASCRS), the American Society for Gastrointestinal Endoscopy (ASGE), and the Society of American Gastrointestinal and Endoscopic Surgeons (SAGES). Dis Colon Rectum. 2006;49:792–809.

ERAS consensus guidelines recommend the utilization of isosmotic MBPs, which consist of osmotically balanced, nonabsorbable solutions that do not produce significant fluid or electrolyte shifts. Isosmotic bowel preparations are deemed safe in patients with the comorbidities of hepatic disease, congestive heart failure, and renal failure.

Large volume polyethylene glycol (PEG) preparations include GoLYTELY, Colyte, NuLytely, and TriLyte. Patients have difficulty tolerating these large-volume solutions as they are not palatable and cause nausea and emesis in 4% to 17% of patients. New low-volume PEG (1–2 L) preparations combined with other agents have been developed and include MiraLAX, HalfLytely, MoviPrep, and BiPeglyte.

There are multiple studies/clinical trials and meta-analyses that show conflicting results for the effectiveness of MBPs in regard to surgical outcomes. In a Cochrane review in 2011, 18 randomized controlled trials were reviewed that included 5805 participants; 2906 patients were administered MBP while 2899 received no bowel preparation before elective colorectal surgery. There was no statistically significant difference between the MBP and non-MBP groups in regard to anastomotic leak for colonic resections and low anterior resection. These results are similar to a recent meta-analysis, published in 2018, evaluating eight studies with 1065 patients. However, both studies have reported that evidence quality is low due to the variation in bowel preparations. Because of this, MBP alone is generally not recommended for elective colorectal surgery.

Oral Antibiotics

First-line antibiotics include a combination of oral neomycin sulfate plus oral erythromycin base or oral neomycin sulfate plus oral metronidazole ( Table 2 ). The most common side effects are nausea and emesis. Multiple randomized controlled trials have reported a significant improvement in SSIs with the use of OA. Two large studies from American College of Surgeons National Surgical Quality Improvement Program (NSQIP) (>300,000 patients) and one large meta-analysis of prospective studies (69,000 patients) have confirmed that many medical centers are using OA alone for their bowel preparations before colorectal surgery. These studies suggest that OA alone is associated with similar SSI rates to OA plus MBP and lower SSI rates as compared with both MBP alone and no bowel preparation.

TABLE 2
Oral Antibiotic Regimens
Oral Antibiotic Prophylactic Regimen * Use in Prior Literature
Neomycin + erythromycin Coppa, 1988; Kaiser, 1983; Khubchandani, 1989; Lau, 1988; Nichols, 1973; Stellato, 1983
Metronidazole + neomycin Epsin-Basany, 005; Hanel, 1980; Lewis, 2002; Nohr, 1990 (included bacitracin); Reynolds, 1989
Metronidazole + kanamycin Lazorthes, 1982; Monrozies, 1983; Takesue, 2000
Tinidazole + neomycin Peruzzo, 1987
Kanamycin + erythromycin Ishida, 2001; Kobayashi, 2007

* Each of these oral antibiotics was combined with a range of intravenous antibiotics in the studies listed. Modified from Bellows CF, Mills KT, Kelly TN, Gagliardi G. Combination of oral non-absorbable and intravenous antibiotics versus intravenous antibiotics alone in the prevention of surgical site infections after colorectal surgery: a meta-analysis of randomized controlled trials. Tech Coloproctol. 2011;15:385–395.

Currently, there is a prospective randomized controlled trial using the Rethinking Clinical Trials (REaCT) platform and NSQIP to compare no bowel preparation versus preoperative OA alone to evaluate SSI rate in elective colorectal surgery. Results are pending at this time. A similar multicenter randomized controlled trial was performed in Spain (ORALEV) in which 536 patients were randomized to no OA versus OA. The incidence of SSI in the no OA group (30/269, 11%) was significantly higher than in the OA group (13/267, 5%) (χ 2 test, p = 0.013). The no OA group had more complications (76/269, 28%) compared with the OA group (51/267) ( P = 0.017).

MECHANICAL BOWEL PREP VS ORAL ANTIBIOTICS VS NO BOWEL PREPARATION

The combination of MBP and OA was described as early as 1973 when the Nichols-Condon bowel prep (a combination of MBP and OA) reported a decrease in the SSI rate from 43% to 9%. In 2012, a retrospective study utilizing the Veterans Affairs Surgical Quality Improvement Program preoperative risk and SSI outcome data evaluated no bowel preparation versus MBP alone versus OA alone versus MBP + OA. Those receiving no bowel preparation had similar SSI rates to those who had MBP only (18.1% vs 20%). Patients receiving OA alone had a SSI rate of 8.3%, and those receiving OA + MBP had an SSI rate of 9.2%. In adjusted analysis, the use of OA alone was associated with a 67% decrease in SSI rate (OR = 0.33, 95% CI 0.21–0.50). OA + MBP was associated with a 57% decrease in SSI rate (OR = 0.43, 95% CI 0.34–0.55). A retrospective analysis of the NSQIP colectomy cohort from 2011 to 2012 subsequently showed that the use of OA alone decreased SSI rates, consequently decreasing postoperative length of stay (LOS) and readmission rates.

The rate of antibiotic resistance or Clostridium difficile infection is similar in the OA alone, no bowel preparation, and OA + MBP groups.

OUTCOMES OF SURGERY DEPENDENT ON TYPE OF SURGERY

Studies have demonstrated that right colon bacterial concentrations range from 10 6 to 10 7 bacteria/g of stool content, whereas these numbers rise to 10 11 to 10 12 bacteria/g in the rectosigmoid. Several studies have demonstrated a lower risk of SSIs after right colectomies versus left colectomies or rectal resections. Studies evaluating patients undergoing right-sided versus left-sided colectomies/proctectomies show a preventative function of OAs in left-sided colectomies/proctectomies with an overall incidence SSIs as 7.01% vs 15.89% (p= 0.004) and superficial SSIs as 2.34% vs 7.01% (p= 0.03).

BOWEL PREPARATION IN ERAS

ERAS guidelines recommend the routine use of combined isosmotic MBP with OA before elective colorectal surgery ( Table 3 ). With the addition of oral carbohydrate loading to MBP, in addition to the emphasis on intake of clear liquids up to 2 hours before surgery as denoted by American Society of Anesthesiologists (ASA) Fasting Guidelines, patients undergoing elective colorectal surgery better tolerate adverse side effects of MBP while also requiring less fluid administration intraoperatively.

TABLE 3
Our Protocol at Johns Hopkins Hospital
Medication Comments
Mechanical bowel preparation MiraLAX (238 g)
Bisacodyl (four 5-mg tablets)
MiraLAX (238 g) mixed in a clear liquid and started at noon the day before surgery and completed after hourly administrations. Bisacodyl tablets taken as an adjunct to MiraLAX.
Oral antibiotics Neomycin (six 500-mg tablets)
Metronidazole (six 500-mg tablets)
Taken in three doses the day before surgery
Clear liquid diet initiated the day before surgery up until 2 hours before surgery.

CONCLUSION

The use of isosmotic MBP + OA as part of the ERAS pathway prevents postoperative complications, such as SSIs, and reduces hospital LOS, costs, and readmission rates. Although evidence shows that MBP alone has not shown benefit, evidence is lacking for the use of OA alone. Future studies, such as an ongoing prospective randomized controlled trial using the REaCT platform and NSQIP will help to inform the perioperative benefits of OA alone.

Suggested Readings

  • Holubar S.D., Hedrick T., Gupta R., et. al.: American Society for Enhanced Recovery (ASER) and Perioperative Quality Initiative (POQI) joint consensus statement on prevention of postoperative infection within an enhanced recovery pathway for elective colorectal surgery. Perioperative Medicine 2017; 6: pp. 1-18.
  • Khangura S.D., La Fleur P., Argáez C., Adcock L.: Bowel preparation for elective colorectal procedures: a review of clinical effectiveness, cost-effectiveness, and guidelines. Canadian Agency for Drugs and Technologies in Health 2018 Jul 13; Available from: https://www.ncbi.nlm.nih.gov/books/NBK537801/

Management of Diverticular Disease of the Colon

Katharina M. Scheurlen, MD,
Susan Galandiuk, MD

INTRODUCTION

Diverticular disease of the colon is common and a progressively increasing burden on both patients and the healthcare system. This term includes both asymptomatic diverticulosis as well as complications arising from diverticulosis that require further medical evaluation and treatment. Approximately 50% of Americans older than 60 years of age are diagnosed with diverticulosis; however, hospital admission rates resulting from complications of the disease, such as diverticulitis and diverticular bleeding, are specifically increasing in patients younger than 45 years of age. Underestimation of the prevalence of this disease in the young often leads to misdiagnosis of diseases causing similar symptoms, such as appendicitis. Inpatient treatment rates for diverticulitis have risen in young adults and show geographic variation caused by dietary habits and environmental factors.

Colonic diverticula are typically not true diverticula as they do not involve all layers of the bowel wall, but only consist of mucosa and submucosa protruding through the muscularis propria. Intraluminal pressure is thought to cause this herniation of mucosa through weak points of the muscle layer, where intramural branches of the vasa recta penetrate to deliver blood to the luminal layers of the colonic wall. Contributing factors that cause increased intraluminal pressure include a low-fiber diet with small-caliber stools and constipation as well as consumption of red meat. Other risk factors for developing diverticular disease are overweight and obesity, physical inactivity, smoking, use of nonsteroidal antiinflammatory drugs (NSAIDs), and a positive family history. In Western countries, diverticula usually occur in the sigmoid colon, whereas in Asia, right-sided location is common.

Diverticular disease usually remains asymptomatic, but 4% to 15% of affected individuals may develop diverticulitis during their lifetime, and another 5% to 15% develop diverticular bleeding. Diverticulitis refers to inflammation of the diverticulum resulting from a combination of poorly understood factors including bacterial stasis, the presence of a fecalith, an altered microbiome, impaired mucosal barrier function, and a subsequent inflammatory cascade. Although its pathogenesis is not completely understood, clinical and histopathologic findings in patients with diverticulitis show some similarities to those found in ulcerative colitis. Certain types of diverticulitis, such as segmental colitis associated with diverticulosis (SCAD), is considered to be an inflammatory bowel disease, suggesting an autoimmune etiology. Recent studies suggest that genetic predispositions involving immunomodulatory genes may also play a role.

Diverticular bleeding is typically painless and usually occurs in the absence of colonic inflammation. It originates from ruptured vasa recta at either the dome or neck of the diverticulum and is arterial. Repeated microtrauma involving mechanical and chemical stress within the diverticular lumen leads to thinning of the mucosal layer and subsequent rupture of arterial branches. Diverticular bleeding is more likely to occur in right-sided diverticular disease. This is because diverticula of the right colon typically have a wider lumen, exposing a larger mucosal surface area to luminal stress, along with a thinner colonic wall.

This chapter will focus on evaluation and management of patients with diverticulitis. A brief section about the evaluation and treatment of patients with diverticular bleeding is included as this topic will also be covered in the chapter devoted to gastrointestinal bleeding.

MANAGEMENT OF DIVERTICULITIS

Clinical Presentation and Diagnosis

Uncomplicated acute diverticulitis is typically located in the sigmoid colon and characterized by the symptom triad of left lower quadrant pain, fever, and leukocytosis. A mobile sigmoid colon or atypical manifestation in other segments of the colon can lead to abdominal pain in locations other than the left lower quadrant, such as the suprapubic region or even the right-side of the abdomen.

Severe complications occur in approximately 25% of cases and can complicate the diagnosis of underlying diverticulitis. Complications involve abscesses that occur in 30% of cases, fistula in 14%, and free perforation with peritonitis in 1% to 2% of patients. Colonic stricture with subsequent bowel obstruction is uncommon. For patients with complicated diverticulitis, medical treatment may be ineffective, and the majority require surgical intervention. Complications can be highly variable, and differential diagnoses must be excluded; therefore findings on physical examination and on blood and urine analyses should be further evaluated with diagnostic imaging. Computed tomography (CT) is the mainstay imaging modality in the assessment of acute diverticulitis and accompanying complications. A CT of the abdomen and pelvis with intravenous and oral contrast is used to evaluate the severity of inflammation and the presence of bowel strictures, obstruction, and local or distant complications such as abscesses and fistulas. In patients with contrast allergy, a noncontrast study or an alternative approach may be performed (e.g., magnetic resonance imaging or ultrasonography), particularly in the pregnant patient. Ultrasonography may identify a hypoechoic colonic wall with a fecalith obstructing the diverticulum, an adjacent bowel wall, and mesenteric edema, which may indicate abscess formation. The diagnostic accuracy of ultrasound is, however, dependent on the examiner’s skill and experience.

Classification of diverticulitis is essential to follow a standardized treatment algorithm that provides the highest success rates for disease management. The Hinchey classification, originally published in 1978, is used as a basis to provide evidence-based management strategies for patients with diverticular disease. The modification by Wasvary et al. is widely used and is based on CT findings concerning abscess formation and peritonitis ( Table 1 ). Both imaging and clinical evaluation are required for a complete patient assessment.

TABLE 1
“Staging” or Grading of Diverticulitis Severity *
Hinchey Classification Modified Hinchey Classification by Wasvary
0 Mild clinical diverticulitis
I Pericolic abscess/phlegmon Ia
Ib
Colonic wall thickening/confined pericolic inflammation
Confined pericolic or mesocolic abscess (<5 cm)
II Pelvic, distant intraabdominal or retroperitoneal abscess II Pelvic, distant intraabdominal or retroperitoneal abscess
III Generalized purulent peritonitis III Generalized purulent peritonitis
IV Generalized fecal peritonitis IV Generalized fecal peritonitis

* Based on the Hinchey classification and modified Hinchey classification by Wasvary to reflect computed tomographic findings.

Management of the Patient with Acute Diverticular Disease

Uncomplicated Diverticular Disease

The inflammatory processes in uncomplicated diverticulitis (Stage 0 or Ia) are restricted to the colon only, while complicated diverticulitis shows signs of inflammation beyond the colon, involving the peritoneum and pericolic structures. Although antibiotic therapy for acute uncomplicated diverticulitis has been the traditional treatment recommendation for many years, there have now been several large prospective randomized clinical trials showing that uncomplicated diverticulitis can be treated without antibiotics ( Table 2 ). It will take clinical practice a long time to follow this evidence. Much of this is the result of established practice patterns and patient expectations. In the event that symptoms limit oral intake or are severe enough to warrant hospital admission, fluid resuscitation and bowel rest will result in improvement while gradually advancing to a low-residue diet. If antibiotics are used for diverticulitis, they should cover both gram-negative and anaerobic bacteria, which is provided by either ciprofloxacin plus metronidazole or levofloxacin plus metronidazole. Because of an increasing prevalence of Escherichia coli resistance to fluoroquinolones worldwide, amoxicillin-clavulanate monotherapy or trimethoprim-sulfamethoxazole plus metronidazole can be used as an alternative.

TABLE 2
Overview of Selected Multicenter Randomized Controlled Trials
Study Acronym Investigation Main Findings
DI verticulitis: A nti B iotics O r c L ose O bservation?) DIAB-OLO Cost-effectiveness of conservative treatment (hospital admission plus antibiotics) versus liberal treatment (no antibiotics and no strict need for hospital admission); primary endpoint: time-to-full recovery In patients with uncomplicated diverticulitis, observational treatment without antibiotics shortens hospital stay and does not prolong recovery
A ntibiotika V id O komplicerad D ivertikulit
(Swedish for “antibiotics in uncomplicated diverticulitis”)
AVOD Treatment of uncomplicated diverticulitis with antibiotics versus without antibiotics; primary endpoint: complications and emergency
surgery during hospital stay
No differences in diverticulitis recurrence, diverticulitis complications, or emergency surgery rates (sigmoid resection) in the long-term; antibiotic treatment had no favorable outcome
Outpatient versus Hospitalization Management for Uncomplicated Diverticulitis DIVER Hospitalization versus outpatient treatment in patients with uncomplicated diverticulitis; primary endpoint: treatment failure rate of outpatient protocol, need for hospital admission Outpatient antibiotic treatment and diet is effective and safe, cost-saving, and associated with a similar quality of life compared with hospitalization with intravenous antibiotic treatment
Perforated DIV erticulitis: sigmoid resection with or without A nastomosis DIVA (LADIES trial) Sigmoidectomy with primary anastomosis versus Hartmann’s procedure in purulent perforated diverticulitis; primary endpoint: 12-month stoma-free survival rate Primary anastomosis leads to increased stoma-free survival and is more cost-effective compared with the Hartmann’s procedure
L apar O scopic LA vage trial LOLA (LADIES trial) Laparoscopic lavage and drainage versus Hartmann’s procedure versus sigmoidectomy with primary anastomosis; primary endpoint: major morbidity and mortality Premature end of the LOLA group: higher rate of in-hospital major morbidity, mortality, and reintervention rate in the laparoscopic lavage arm
SCAN dinavian DIV erticulitis trial SCANDIV Laparoscopic lavage versus primary resection in patients with perforated diverticulitis; primary endpoint: severe postoperative complications within 90 days Laparoscopic lavage does not reduce severe postoperative complications, increases the reoperation rate, and leads to a higher risk of missing associated colon carcinomas
DI verticulitis– LA paroscopic La vage trial DILALA Laparoscopic lavage versus Hartmann’s procedure in patients with purulent perforated diverticulitis; primary endpoint: number of reoperations within 12 months Patients undergoing lavage have fewer reoperations and equal hospital readmission and mortality rates.

Two randomized controlled trials showed no difference in patient outcome comparing treatment with antibiotics versus fluid resuscitation only. The most recent trial from the Dutch Diverticular Disease Collaborative Study Group (DIAB-OLO) demonstrated that no difference concerning mild or serious adverse events could be found between groups, but a significantly higher rate of adverse events were associated with antibiotic treatment. Long-term follow-up data, 11 years after The Swedish Antibiotic Therapy of Acute Uncomplicated Colonic Diverticulitis (AVOD) trial, showed no difference in recurrence rates, complications, surgical treatment rates for the disease, and quality of life between the patient groups. Based on these data, otherwise healthy patients with uncomplicated diverticulitis can be treated without antibiotics. Evidence from randomized controlled trials has also shown that a 4-day course of intravenous antibiotics was as effective as a 7-day course, while clinical outcomes of inpatient and outpatient antibiotic treatment in otherwise healthy patients tolerating oral intake did not differ (DIVER trial). If a patient’s clinical status does not improve after 5 days of treatment with persistent fever, leukocytosis, and elevated acute-phase proteins (C-reactive protein), further CT evaluation for an abscess should be considered.

Patients who present with uncomplicated diverticulitis have a low incidence (<2%) of occult colonic malignancy in contrast with those with complicated diverticulitis, in whom the incidence is 8% to 11 %. Colonoscopy is typically performed 6 weeks after symptom resolution, especially in those of screening age and family history. In case of recurrent episodes affecting quality of life, smoldering disease, or immunocompromise, elective colon resection may be indicated.

Diverticular Disease with Pericolic Abscess

Complicated diverticulitis that is accompanied by either a mesocolonic (stage Ib) or more distant abscess formation (stage II) occurs in 15% to 40% of acute diverticulitis patients. It can be treated nonoperatively with antibiotics in an outpatient setting in stable patients with abscesses ≤3 cm in size. Abscesses >3 cm or patients who do not respond to antibiotic treatment alone require percutaneous drainage, typically via a transabdominal approach ( Fig. 1 ). Alternative approaches are transgluteal for pelvic abscesses (even though they are associated with greater patient discomfort and a higher risk of drain dislocation) and transvaginal or transrectal approaches. Conservative treatment strategies are typically effective with small abscesses, with an overall success rate in 80% of patients presenting such abscesses. There is a failure rate of up to 34% with nonoperative treatment of abscesses >3 cm in size.

FIG. 1, Management of acute diverticulitis. This evidence-based algorithm should be individualized and modified based on the patient’s clinical course.

If the patient is in stable condition with nonoperative treatment, the diet is gradually advanced to a low-residue diet. If a CT-guided drain is placed, decreasing white blood cell and drain output to <30 mL/day indicate when the drain can be considered for removal. In many cases, a drain contrast study will show the size of the residual abscess cavity and whether there is a communication to the colon. The rate of recurrent abscesses after conservative treatment is high, although it is considerably lower after percutaneous drainage (25%–60% vs. 15%–25%). The overall failure rate of these nonoperative approaches is 15% to 32%. In case of failure, surgery is indicated, with urgent abscess drainage and colectomy, typically with primary anastomosis and proximal diversion.

Diverticular Disease with Perforation/Peritonitis

Purulent peritonitis (Hinchey III) and feculent peritonitis (Hinchey IV) cannot be distinguished from one another clinically. Emergency surgery with abdominal exploration is necessary for diagnosis. Hinchey III is defined by an occult colonic diverticular perforation with abscess formation and subsequent abscess rupture. Hinchey IV denotes a free perforation of a diverticulum with peritoneal fecal contamination.

Emergency surgery is required in 15% to 32% of patients who are hospitalized for acute diverticulitis. Acutely ill patients with diverticulitis and peritonitis should undergo expedited intravenous antibiotic therapy, fluid resuscitation, and emergency surgery. Abdominal exploration, resection of the acutely inflamed colon containing the area of perforation, and thorough peritoneal lavage should be performed. Overall, 30-day hospital mortality after emergency colectomy as reported by a retrospective National Surgical Quality Improvement Program (NSQIP) study in 2013 was 5.1%. Significantly higher mortality rates up to 53% were found in patients with two or more of the following risk factors: age >80 years, ASA class 4 or 5, elevated serum creatinine (>1.2 mg/dL), and hypoalbuminemia (<2.5 g/dL).

Two types of colectomy can be performed: (1) resection with primary anastomosis with or without proximal diversion (loop-ileostomy) or (2) resection with discontinuity and a Hartmann rectal stump and proximal end colostomy. Although discontinuity with a Hartmann stump was once the standard in an emergency setting, several randomized controlled trials and meta-analyses have demonstrated that primary anastomosis with or without proximal diversion yields favorable outcomes. In 2019, the DIVA arm of the LADIES trial showed a significantly higher 12-month stoma-free survival rate in patients receiving primary anastomosis with or without a defunctioning ileostomy compared with patients undergoing Hartmann procedures in perforated diverticulitis (95% vs. 72%). There were no significant differences in terms of morbidity and mortality between groups (see Table 2 ).

Table 1
Mayo Endoscopic Subscore
Score Disease Activity Endoscopic Feature
0 Normal or inactive None
1 Mild Decreased vascular pattern, erythema, mild friability
2 Moderate Absent vascular pattern, marked erythema, friability, erosions
3 Severe Spontaneous bleeding, ulceration

Bowel reconstruction with closure of an end colostomy after a Hartmann procedure is associated with higher morbidity compared with closure of a loop ileostomy after primary anastomosis with proximal diversion because of the extent of the actual disease and procedure. For this reason, high-risk patients who are at substantial risk of anastomotic leakage and major complications often never undergo the second operation of colostomy closure and Hartmann takedown. It is estimated that there are more than 5-fold the number of Hartmann procedures performed than the number of Hartmann reversals, and most of these colostomies can be considered permanent. The decision on whether bowel continuity should be restored after resection in patients with Hinchey III or IV diverticulitis should consider patient factors (presence of shock, hemodynamic stability, immunosuppression, age) and intraoperative findings (quality of tissue, presence of pan-abdominal inflammatory changes). The experience of the respective surgeon should determine the most appropriate procedure in these high-risk settings.

An alternative to bowel resection in case of perforated diverticulitis is laparoscopic lavage with drain placement. This procedure is not recommended with feculent perforation (Hinchey IV). It can be considered in select patients with purulent peritonitis (Hinchey III); however, colectomy is generally preferred as the need for secondary interventions is higher with laparoscopic lavage. Three randomized controlled trials investigated laparoscopic lavage in Hinchey III disease: the LOLA trial, SCANDIV trial, and DILALA trial (see Table 2 ). These studies were heterogeneously designed and focused on different techniques and endpoints. Several meta-analyses have tried to make sense of this heterogeneity. Overall, laparoscopic lavage was associated with a higher rate of surgical reintervention (20% vs. 7%, LALA trial) and a high rate of subsequent abscess formation requiring drainage (20% vs. 0%, LALA trial). An acute or elective surgical intervention was necessary in 48% of cases following laparoscopic lavage (LALA trial). Deep surgical site infection (32% vs. 13%) and unplanned reoperation rates (27% vs. 10%) were also higher in the lavage group (SCANDIV trial). The DILALA trial, however, showed that the lavage group had a 45% reduced risk of undergoing reoperations 2 years after the initial procedure.

Laparoscopic lavage is, however, an appealing approach because of its shorter operative time and lower risk for cardiac complications, wound infection, and stoma formation. This procedure may have a limited role in select young patients with a low ASA score and high BMI to reduce the risk of a stoma in the acute setting, allowing for a future elective resection. The lack of homogenous supporting evidence does not, however, allow for a general recommendation for this procedure.

Treatment of Sequelae of Complicated Diverticulitis

Diverticular Stricture

Strictures of the colon can result from chronic inflammation in patients with smoldering or recurrent diverticular disease. In the case of a diverticular stricture, underlying malignancy must always be suspected, and an oncologic resection of the diseased colonic segment with lymph node clearance should be performed if colonoscopy cannot be performed preoperatively to confirm the diagnosis.

Diverticular Fistula

Fistulas can develop in up to 2% of patients with diverticular disease, with colovesical fistulas representing half of all fistulas. Other types of fistulas include colocutaneous fistulae following interventional drain placement for abscess drainage, colovaginal fistulas, which almost always occur in women who have undergone prior hysterectomy, or rarely coloenteric or colouterine fistulas. Fistulas can also occur in the presence of Crohn’s disease or cancer, and colonoscopy should be performed to clarify their etiology. In patients with fistulas, sepsis and clinical instability are uncommon. Patients can often be evaluated and monitored in the outpatient setting until the time of elective surgery.

Surgical Treatment

Route of Access

Surgical approaches for colectomy in patients with diverticular disease include open surgery with laparotomy or minimally invasive surgery with either a laparoscopic or robotic approach. Regardless of route of access, surgical management of diverticular disease follows the same principles. A minimally invasive approach is preferred if expertise is available, as this has been associated with superior outcomes in terms of reduced postoperative morbidity and reduced hospital stay. The robotic approach has been associated with decreased conversion rates because of improved optics and instrument maneuverability and is especially useful when dealing with inflammatory disease adherent to the pelvic side wall. Compared with the laparoscopic approach, robotic access has been associated with reduced rates of ileus and postoperative complications and a shorter length of hospital stay, but significantly increased hospital charges and longer operative times.

Clinical factors, such as hemodynamic stability, patient comorbidities, the presence of coexisting bowel obstruction, and prior abdominal procedures causing adhesions determine whether minimally invasive surgery is feasible or open surgery should be performed. Several studies have shown that a minimally invasive approach can be performed in the emergency setting with improved morbidity. Clinical factors in the high-risk patient and longer operative times of laparoscopic procedures can, however, influence the decision regarding choice of approach.

In the acute situation, it is important to mark the patient for a possible stoma site in the sitting position and to avoid skin folds and creases. In obese individuals, the upper abdomen may provide a thinner abdominal wall, with easier stoma creation. Marking several possible stoma sites is never wrong. Remember, the patient may have the stoma a long time, and a well-placed stoma is essential.

Open access is performed via midline laparotomy. For laparoscopic access, four port incisions are required, followed by a subsequent suprapubic incision to remove the resected specimen. A large periumbilical camera port and two 5-mm ports in the right upper quadrant and left lower quadrant are placed. The port in the right lower quadrant should be of larger size because access to an endoscopic stapler should be provided. Robotic access incisions are dependent on the robotic system used. The Si robot incisions are similar to those of laparoscopic access, with modified placement of the large camera port to the right of the umbilicus. The Xi robot requires ports oriented along a straight diagonal line from the lower right abdomen medial to the anterior superior iliac spine upward toward the patient’s left, with an additional 5-mm assistant port in the right lower quadrant.

Common Surgical Themes Among All Routes of Access

  • 1

    Work from normal to abnormal. Beginning to work in a phlegmon can be challenging.

  • Open: Begin dissection proximally, freeing up retroperitoneal structures superiorly and slowly working toward the area of inflammation

  • Minimally invasive: Lateral-to-medial: same as for open

  • Minimally invasive: Medial-to-lateral: essentially begins in “normal” retroperitoneal plane proceeding laterally toward inflammation

  • 2

    Visualize the left ureter. If technical difficulties are anticipated, ureteral stents are useful. See https://youtu.be/pVaFId9UuC4 for use of indocyanine green in ureteral stents for easy intraoperative visualization.

  • 3

    If not performing an anastomosis, resect the area of perforation. There is no need to go down lower to the rectum itself because this will make the subsequent surgery more difficult.

  • 4

    If performing an anastomosis, the distal part of the colorectal anastomosis should lie within the upper rectum. This is most easily located as the area where the colonic tenia confluence. If an anastomosis is made in the sigmoid colon, there is an up to 25% recurrence rate.

  • 5

    If extra colonic length is needed, the splenic flexure may need to be mobilized and the inferior mesenteric vein and/or artery may need to be divided.

  • 6

    The proximal line of transection should be in soft pliable bowel. All of the diverticula-bearing colon does not need to be removed.

  • 7

    If there is a dense inflammatory scar around the rectum, the rectum may need to be mobilized to allow a circular stapler to pass.

In case of bowel reconstruction with primary anastomosis, an end-to-end circular stapler is used to create a double-stapled or triple-stapled anastomosis. If there is a lot of edema or an inability to pass a stapler transanally, a hand-sewn colorectal anastomosis is an option. In cases in which fecal diversion is appropriate, either a protective loop-ileostomy (primary anastomosis and proximal diversion) or an end colostomy (Hartmann procedure with rectal stump and discontinuity) is created. Placement of a drain is generally not required if the dissection is above the level of the peritoneal reflection. In the case of a colovesical fistula, a drain is typically placed near the bladder repair.

Most Common Surgical Errors

  • 1

    No preoperative stoma marking

  • 2

    Anastomosis performed in the distal sigmoid colon

  • 3

    Attempt to resect all colonic diverticula

  • 4

    Very low Hartmann procedure performed, making subsequent closure more difficult

MANAGEMENT OF DIVERTICULAR BLEEDING

Clinical Presentation and Diagnosis

Three to five percent of patients with diverticular disease develop diverticular bleeding, presenting as either bright red blood per rectum, hematochezia, or melena. Risk factors include the use of NSAIDs, thrombocyte aggregation inhibitors, and anticoagulants, which is common among the elderly. Although diverticular bleeding can cause severe blood loss, it will stop spontaneously in approximately 80% of cases. The overall mortality rate is 2% to 4%. Recurrent bleeding is common and occurs in up to 38% of patients.

Diverticular disease is the underlying cause of more than 40% of cases of lower gastrointestinal bleeding. Typically, patients present with painless hematochezia. Initial standard management includes establishing large-gauge peripheral intravenous access at two different sites, cardiopulmonary monitoring, and supplemental oxygen. Initial laboratory analyses should include complete blood count, electrolytes, liver parameters, lactate levels, and coagulation parameters if the patient is on anticoagulant therapy. Hemoglobin levels are serially reevaluated, and blood transfusion is considered according to the patient’s clinical status and the dynamics of changing hemoglobin levels. A hemoglobin level of 6 g/dL absolutely indicates the need for a blood transfusion in the acute setting. Patients with a history of cardiovascular disease should receive blood at an Hb level of 8 g/dL, and a level of 10 g/dL should be maintained.

Recommendations for withholding anticoagulants or antiplatelet drugs in patients with acute diverticular bleeding are based on moderate to low quality evidence. The type of drug, the history of the patient determining their individual thrombotic risk, and their clinical status should all be considered. Aspirin as a primary prophylaxis for cardiovascular events generally can be continued, even perioperatively in the case of emergent surgical treatment. In patients with a low thrombotic risk and unstable gastrointestinal hemorrhage under warfarin therapy, anticoagulation can be immediately reversed using prothrombin complex and vitamin K. Once the bleeding has stopped, warfarin therapy should be restarted after an interval of 7 days. In case of high thrombotic risk (mechanical heart valve, atrial fibrillation with a prosthetic heart valve or mitral stenosis, recent venous thromboembolic event [within prior 3 months]), however, the pausing of warfarin therapy has to be evaluated carefully. Anticoagulant therapy with heparin can be considered as a transitional solution in an emergency setting. With heparin therapy, monitoring the partial thromboplastin time (PTT) every 6 hours is essential. Dosing should be carefully adjusted according to PTT levels and body weight.

The two main diagnostic approaches to distinguish diverticular bleeding from other etiologies of lower gastrointestinal bleeding are colonoscopy and computed tomography angiography (CT-A).

Endoscopic Strategies for Diverticular Bleeding

Colonoscopy is performed to identify the endoluminal source of bleeding, while also providing an opportunity for hemostatic intervention. Ideally, the clinically stable patient with lower gastrointestinal bleeding receives colonoscopy within 24 hours of hospital admission, following a bowel preparation. Endoscopic treatment includes epinephrine injection (1:10,000, 1–2 mL aliquots), bipolar cautery, endoclipping, band ligation, or application of topical hemostatic agents (e.g., Hemospray).

Radiologic Strategies for Diverticular Bleeding

Following unsuccessful endoscopic evaluation, recurrent bleeding, or clinical instability of a patient that does not allow for colonoscopy, CT-A of the abdomen and pelvis is considered. CT-A sensitivity is relatively low (85%), but the technique is sensitive enough to detect bleeding rates of 0.3 to 0.5 mL/min. Only active bleeding sources can be detected, which can be critical because of the intermittent dynamic of diverticular bleeding. Minimally invasive interventional treatment using selective angioembolization can be performed via vasopressin infusion or coil embolization. Once the bleeding source is identified, success rates of embolization are up to 85%.

Surgical Strategies for Diverticular Bleeding

Surgery in patients with diverticular bleeding is indicated in emergency settings with persistent or recurrent bleeding that cannot be managed via either endoscopic or angiographic approaches. Indications are transfusion of 6 units of blood within 24 hours to maintain a hemoglobin level, persistent bleeding for 72 hours, or bleeding recurrence after initial treatment.

If the source of bleeding cannot be identified preoperatively, a subtotal colectomy with end ileostomy should be considered. Even in cases of successful preoperative localization, a targeted segmental colectomy is associated with a rebleeding rate in 14% of cases. The mortality rate for colectomy for diverticular bleeding in an emergency setting is 25%. Overall rebleeding rates 1 year after hospital discharge range from 4% to 42%. Therefore, a prophylactic elective resection after initial bleeding in patients at high-risk for rebleeding episodes can be considered.

Clinically stable patients who tolerate longer operative times should receive colectomy via a laparoscopic approach. Hemodynamic instability and cardiovascular comorbidities indicate an open approach.

In case of subtotal colectomy, after mobilization of the distal ileum beyond the ligament of Treves, the entire colonic mesentery is taken down. An end ileostomy with a rectal stump is created in the event of hemodynamic instability. In select stable patients undergoing elective surgery for diverticular bleeding, an ileorectal anastomosis can be considered.

CONCLUSION

In summary, management of diverticular disease has a significant role in everyday clinical practice, and the therapeutic algorithm is largely based on the patient’s clinical status and on diagnostic imaging and endoscopy to exclude the presence of complications such as abscesses/fistula or cancer. Antibiotic therapy is a therapeutic standard in complicated diverticulitis; however, several randomized prospective studies have shown that they are not necessary in the treatment of uncomplicated diverticulitis. Although patients with recurrent disease generally benefit from clinical improvement and elective surgery in a symptom-free interval, patients with perforated diverticulitis require emergency surgical treatment. Once a Hartmann procedure is performed, reconstruction of fecal continuity is challenging and, in the end, often not successful. Recent studies show that primary anastomosis with or without a protective ileostomy can prevent many patients from having a lifelong stoma without excess morbidity. Stronger evidence, however, is required before including these strategies in the treatment algorithm.

Bleeding of the lower gastrointestinal tract often originates from diverticula, and treatment can be challenging in the acute setting because of the use of anticoagulants or antiplatelet medication, particularly in the elderly. Endoscopic control of bleeding and interventional angioembolization show high success rates. A laparoscopic approach is an option in hemodynamically stable patients with recurrent bleeding undergoing resection. In case of hemodynamic instability, a laparotomy with resection of the respective bowel segment up to a subtotal colectomy in cases of an unknown bleeding source must be considered.

Careful clinical examination of the patient accompanied by established diagnostic algorithms lead the way in treating the complications associated with diverticular disease.

Suggested Readings

  • Chabok A., Pahlman L., Hjern F., Haapaniemi S., Smedh K., Group A.S.: Randomized clinical trial of antibiotics in acute uncomplicated diverticulitis. Br J Surg 2012; 99: pp. 532-539.
  • Hall J., Hardiman K., Lee S., et. al.: The American Society of Colon and Rectal Surgeons clinical practice guidelines for the treatment of left-sided colonic diverticulitis. Dis Colon Rectum 2020; 63: pp. 728-747.
  • Hawkins A.T., Wise P.E., Chan T., et. al.: Diverticulitis: An update from the age old paradigm. Curr Probl Surg 2020; 57:
  • Hinchey E.J., Schaal P.G., Richards G.K.: Treatment of perforated diverticular disease of the colon. Adv Surg 1978; 12: pp. 85-109.
  • Kohl A., Rosenberg J., Bock D., et. al.: Two-year results of the randomized clinical trial DILALA comparing laparoscopic lavage with resection as treatment for perforated diverticulitis. Br J Surg 2018; 105: pp. 1128-1134.
  • Lambrichts D.P.V., Vennix S., Musters G.D., et. al.: Hartmann’s procedure versus sigmoidectomy with primary anastomosis for perforated diverticulitis with purulent or faecal peritonitis (LADIES): a multicentre, parallel-group, randomised, open-label, superiority trial. Lancet Gastroenterol Hepatol 2019; 4: pp. 599-610.
  • Roberts P.L.: Chapter 149. Surgery for Diverticulitis.7th ed.2019.Wolters Kluwer/Lippincott Williams & WilkinsPhiladelphiapp. 1752-1762.
  • Schug-Pass C., Geers P., Hugel O., Lippert H., Kockerling F.: Prospective randomized trial comparing short-term antibiotic therapy versus standard therapy for acute uncomplicated sigmoid diverticulitis. Int J Colorectal Dis 2010; 25: pp. 751-759.
  • Schultz J.K., Yaqub S., Wallon C., et. al.: Laparoscopic lavage vs primary resection for acute perforated diverticulitis: The SCANDIV randomized clinical trial. JAMA 2015; 314: pp. 1364-1375.
  • Strate L.L., Morris A.M.: Epidemiology, pathophysiology, and treatment of diverticulitis. Gastroenterology 2019; 156: pp. 1282-1298.
  • Unlu C., de Korte N., Daniels L., et. al.: A multicenter randomized clinical trial investigating the cost-effectiveness of treatment strategies with or without antibiotics for uncomplicated acute diverticulitis (DIABOLO trial). BMC Surg 2010; 10: pp. 23.
  • Vennix S., Musters G.D., Mulder I.M.: Laparoscopic peritoneal lavage or sigmoidectomy for perforated diverticulitis with purulent peritonitis: a multicentre, parallel-group, randomised, open-label trial. Lancet 2015; 386: pp. 1269-1277.
  • Wasvary H., Turfah F., Kadro O., Beauregard W.: Same hospitalization resection for acute diverticulitis. Am J Surg 1999; 65: pp. 632-635.

Management of Chronic Ulcerative Colitis

Kirkpatrick Beekman Fergus, MD
Michael G. Kattah, MD, PhD,
Elizabeth C. Wick, MD

INTRODUCTION

Ulcerative colitis (UC) is a chronic inflammatory bowel disorder (IBD) characterized by inflammation in the rectum (proctitis) that may extend proximally to include the colon (proctocolitis). Unlike Crohn’s disease, which can affect anywhere in the gastrointestinal (GI) tract from the mouth to the anus, UC only affects the mucosa and submucosa of the colon and rectum. In general, UC is continuous, starting in the rectum (proctitis) and, in many patients, extending more proximally into the colon, first the sigmoid or descending colon (left-sided or distal colitis) and, in more severe cases, proximal to the splenic flexure (extensive or pancolitis). Patients exhibit a range of symptoms including frequent, small-volume, bloody diarrhea, urgency, hematochezia, proctalgia, and colicky abdominal pain. Associated hematochezia varies in frequency but is usually small volume and typically does not cause rapid drops in hemoglobin. In severe cases, patients can have significant weight loss, anorexia, and fatigue. With rectal inflammation, patients can develop rectal pain, spasm, and difficulty evacuating. In general, UC is a chronic disease, but patients may experience acute exacerbations or periods of remission, particularly those undergoing medical therapy.

EXTRACOLONIC MANIFESTATIONS OF UC

Extracolonic manifestations of UC include arthritis, eye disease (uveitis, episcleritis, iritis, conjunctivitis), oral aphthous ulcers, hepatobiliary disorders (asymptomatic transaminitis and primary sclerosing cholangitis [PSC]), erythema nodosum, and pyoderma gangrenosum (pretibial area and peristomal). Many extracolonic manifestations improve after proctocolectomy, but surgery does not improve outcomes in PSC, ankylosing spondylitis, or sacroiliitis.

In patients with UC and PSC, the liver symptoms usually occur in the setting of significant colonic symptoms and, in general, persist even after proctocolectomy. Therefore, it is important that management be collaborative with gastroenterology, hepatology and/or liver transplant surgery, and colorectal surgery. Patients with UC and PSC are at higher risk for dysplasia and colorectal cancer and should undergo annual colonoscopic surveillance beginning when PSC is diagnosed.

RISK FACTORS

The etiology of UC is poorly understood, but it is multifactorial with genetic, microbial, and environmental contributing factors. The microbiome, GI infections (bacterial, viral, and parasitic), food allergies, antibiotic exposure, dietary habits such as high sugar intake, or toxins have all been implicated in animal models and/or population-based studies. Cigarette smoking, of note, has been shown to be protective against UC, even though it is a risk factor for Crohn’s disease. The relationship and data regarding cigarette smoking are complex, and the overall health benefits of smoking cessation outweigh the theoretical disease control benefits in UC, therefore all patients should be encouraged to cease smoking.

DIAGNOSIS

Patients are usually evaluated by a gastroenterologist in the outpatient setting or, in the case of acute severe ulcerative colitis (ASUC), the inpatient setting. By the time of surgical evaluation, the diagnosis has generally been made. Endoscopy (sigmoidoscopy and/or colonoscopy) with biopsy is the gold standard for making the diagnosis of UC. Examination of the terminal ileum can help avoid misdiagnosis of Crohn’s disease, and a complete examination can help rule out malignancy. The endoscopist should grade the mucosa using the Mayo endoscopic subscore ( Table 1 ) and include photographs in the report. The mucosa in UC is erythematous, friable, edematous, and in severe cases can present with large ulcerations. Although pseudopolyps or even strictures can be seen in a UC postinflammatory state, the presence of strictures or fibrotic disease should raise suspicion for Crohn’s colitis or malignancy. Biopsies should be taken from inflamed areas to confirm both chronic and active colitis, and to rule out viropathic effects seen with cytomegalovirus (CMV).

Supplemental tests include laboratory testing, stool testing, and imaging. In addition to complete blood count and complete metabolic panel, the inflammatory markers erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) are often associated with disease activity. Stool testing should be obtained to rule out Clostridioides difficile (C. diff) and other common bacterial, viral, and parasitic pathogens. Fecal calprotectin is a noninvasive fecal marker commonly used to differentiate between inflammatory versus noninflammatory diarrhea in the outpatient setting and correlates well with disease activity in many patients. CT scan is frequently used in the acute setting to rule out toxic megacolon or perforation but has limited utility for evaluating the small intestine. CT enterography and MR enterography delineate the bowel better and, importantly, can identify any small bowel disease that might suggest the patient’s diagnosis is in fact Crohn’s disease and not UC.

MEDICAL THERAPY

Medical therapy for UC aims to both improve symptoms and heal the colon mucosa. Treatment is tailored for induction of remission followed by maintenance of remission. Agents for induction and maintenance for mild to moderate UC include aminosalicylates (mesalamine or diazo-bonded 5-ASA) given orally and/or topically by suppository or enema. Oral prednisone, rectal steroids, or oral or rectal budesonide multimatrix (MMX) can also be used to aid in achieving remission.

Medical therapy for moderate to severe disease requires biologics (monoclonal antibodies) with or without an immunomodulator (e.g., azathioprine) or small molecules. Except for steroids, medications used for induction of remission for moderate to severe UC are continued for maintenance. Nonresponse or loss of response among individuals on biologic agents is managed by dose-intensification or switching to a different biologic agent or small molecule.

For patients with ASUC requiring hospitalization, management includes bowel rest, intravenous hydration, venous thromboembolism prophylaxis, minimizing narcotics, and cessation of antidiarrheal agents to avoid toxic megacolon. C. diff should be ruled out, and flexible sigmoidoscopy aids in both risk-stratification and to rule out superimposed CMV colitis. Medical management involves intravenous steroids and biologic agents. Surgical consultation for patients with acute colitis can be helpful to discuss possible surgery earlier in the hospital course. As described later, in the setting of ASUC, surgery results in an end ileostomy. Allowing patients to be partners in this decision and, if time permits, affording them the opportunity to talk to an enterostomal nurse and to patients who have previously undergone surgery for UC can improve the experience.

SURGERY AND ULCERATIVE COLITIS

Overview

The 10-year cumulative risk of colectomy for patients with UC is 10% to 15%, and the rate appears to be decreasing in the era of biologics. Despite evolving medical therapies, patients come to surgery for three main reasons: (1) failure of medical management to control either the intestinal or extraintestinal manifestations of UC, (2) dysplasia and colorectal cancer, or (3) toxic megacolon.

The first is far and away the most common indication for surgery. A patient with UC is cured of colitis by removing the entire colon and rectum (from the terminal ileum to the anorectal junction). This stands in contrast to Crohn’s disease where surgery is used to address immediate symptoms, but there is always the risk of the disease returning in a new area.

Patients with UC are at higher risk of colorectal cancer and require routine endoscopic surveillance beginning 8 to 10 years after disease onset or sometimes sooner. Colonoscopy should occur every 1 to 3 years depending on risk factors such as PSC, a family history of colorectal cancer, and whether active inflammation is present. In the past, patients with dysplasia were encouraged to undergo surgery due a high rate of colorectal cancer even with immediate colectomy. Now with improved endoscopic techniques, patients with visible and resectable dysplasia can be safely followed with close colonoscopic surveillance by experienced gastroenterologists using dye spray or virtual chromoendoscopy and high-definition endoscopes. In the event of persistent invisible multifocal or high-grade dysplasia, unresectable dysplasia, or colorectal cancer on biopsy, patients should undergo proctocolectomy.

Patients with advanced disease despite maximal medical therapy may also progress to toxic megacolon, characterized by colonic dilation, fevers, tachycardia, leukocytosis, and abdominal distension. This requires urgent surgical intervention to prevent perforation.

While the surgical concepts are the same for all UC patients, the approach should be tailored to each patient’s indications, urgency, comorbidities, and severity (corticosteroids, biologics, nutritional status) as well as long-term goals of care with regards to life with an ostomy versus bowel frequency. Shared decision making and a team-based approach with the patient, family, gastroenterologist, and surgeon should be embraced to ensure that the patient is knowledgeable and prepared for the operation(s) and long-term impact on lifestyle.

Total Abdominal Colectomy with End Ileostomy

For urgent indications (toxic megacolon, severe medically refractory disease, refractory ASUC, perforation), total abdominal colectomy with end ileostomy is the best course of action. It is a relatively short operation (2–3 hours) that removes a good amount of diseased bowel (colon) and diverts the fecal stream away from the rectum, allowing the mucosa to partially heal. The rectum is left in place during this operation to reduce the risk of complications associated with pelvic dissection in the setting of severe inflammation and to allow for restoration of fecal continence via an ileal pouch later. Before surgery, if time allows, the patient should have a visit with an enterostomal therapist and the site for the end ileostomy should be selected. It is usually in the right lower quadrant, but it is important that it is tailored to the patient’s body habitus. A poorly placed ileostomy (in a skin fold or scar, belt line, etc.) can be very challenging to manage, time consuming, and lifestyle limiting for the patient and family.

The approach should be laparoscopic if the surgeon has experience and the patient is hemodynamically stable. A fully laparoscopic, hand-assisted laparoscopic, or robotic approach have all been described. In general, after the pneumoperitoneum is established, the colon is mobilized, either with a medial to lateral approach with ligation of the vessels first (ileocolic, middle colic, and inferior mesenteric) or lateral to medial approach with ligation of the vessels second. High ligation is not needed unless there is concern for dysplasia or malignancy, and it is generally safest to stay close to the colon when dividing the mesentery.

In a medial to lateral approach, the dissection begins on the left by the sigmoid colon, which is retracted anteriorly to identify the inferior mesenteric artery pedicle. A window is created to isolate the pedicle, and before ligation it is important to identify and preserve the left ureter and gonadal vessels. After ligation, the dissection then proceeds superiorly separating the colon from Gerota’s fascia up to the splenic flexure. The lateral attachments are taken next, following the white line of Toldt to free the descending colon from the retroperitoneum. Division of the rectosigmoid junction with a stapler is performed next, followed by ligation of mesenteric vessels. Attention is then turned to the right colon with identification and ligation of the ileocolic artery. The medial dissection proceeds superiorly toward the hepatic flexure, followed by a lateral dissection releasing the peritoneal attachments. After dividing the hepatic flexure, all remaining attachments along the transverse colon are divided. The colectomy is completed with division of the terminal ileum. Of note, based on surgeon preference, the dissection either proceeds from left to right (as described) or right to left.

There are a few key considerations when selecting the proximal and distal resection sites. The rectosigmoid junction should be divided either intracorporeally or extracorporeally (via Pfannenstiel or lower midline incision). If the rectosigmoid bowel is thick and edematous, it is possible that the staples will not hold. If there is concern, the selected division point should allow for the rectosigmoid area to be matured to the skin as a mucous fistula. Other strategies for managing a challenging rectal stump include oversewing the staple line and decompressing the rectum with a rectal tube. Delayed intraabdominal breakdown of the rectal stump results in sepsis and usually needs to be managed with reoperation. The proximal division should be in the very distal terminal ileum so that sufficient small bowel remains for an ileal pouch if the patient desires.

After the colectomy specimen is removed, the ileostomy should be brought out through the previously marked area and everted or “Brooked.” By having the ileostomy above the skin, it will allow for a well-fitting ileostomy appliance and reduce risk of skin breakdown, pain, and discomfort. When bringing the ileostomy through the abdominal wall, it is also important to ensure that the mesentery of the small bowel is not twisted as this can lead to obstruction and/or ischemia requiring reoperation.

For most patients, the total abdominal colectomy will restore appetite, weight, energy, and overall quality of life. There will be some bloody mucus drainage from the rectum, but if patients are warned to expect this it is usually quite manageable.

Completion Proctectomy/Total Proctocolectomy

For patients undergoing surgery for dysplasia, cancer, or chronic medically refractory disease (not hospitalized, not on high dose steroids, and without signs of malnutrition), the best approach is to remove the colon and rectum (total proctocolectomy) in one operation. In this case, the patient and surgeon should plan to either proceed with ileal pouch anal anastomosis (IPAA) with a temporary diverting loop ileostomy or simply remove the rectum and remain with a permanent end ileostomy. If the plan is to complete the proctocolectomy in one operation, the colectomy is completed as described before and the rectum is removed as described later.

Proctectomy can be completed laparoscopic, hand-assisted laparoscopic, robotic, or open. The rectum is identified, and the inferior mesenteric artery is noted coursing along the retroperitoneum. The peritoneum is incised, and the left ureter is identified. The inferior mesenteric artery and vein are ligated. The dissection is then carried into the pelvis posteriorly by developing the plane between the mesorectum and the fascia propria of the rectum. The superior hypogastric plexus should be identified and preserved. Once the pelvic floor is reached, the anterior dissection should be initiated by developing the plane between the rectum and prostate or vagina. Care should be taken to err on the side of the rectum and, in men, avoid entering Denonvilliers’ fascia as nerves controlling sexual function course around the pelvic brim and are at risk of injury. The lateral stalks of the rectum should be divided on the right and left, and the rectum should now be free circumferentially to the pelvic floor. This can be confirmed by placing a finger in the anus. If the patient is not to have an ileal pouch constructed, the rectum and anus will be completely removed by completing the dissection from the anus. This dissection is initiated with a self-retaining retractor effacing the anus. It is generally a simple dissection conducted from the perineum in the intersphincteric (between the internal and external anal sphincter) groove. This is usually bloodless and leaves the external sphincter in place to help with perineal wound healing and prevent herniation later.

The perineum is closed in multiple layers approximating the muscle layers using Vicryl or other absorbable suture, followed by skin closure. Surgical closed suction drains are typically left in the presacral space to protect the perineal wound from seroma formation and reduce the risk of infection and dehiscence.

Creation of Ileal Pouch Anal Anastomosis

Most patients will desire ileal pouch creation; IPAA is the most common operation done for UC in the United States. Although it is associated with more risks (short and long term) than end ileostomy, for many patients restoring intestinal continuity is important to their quality of life. For most patients with an IPAA, it is reasonable to expect 6 to 10 bowel movements per day. The stool will be looser, although consistency may improve with time as the small bowel adapts to absorb some water. A significant number of patients will experience urgency and even some fecal incontinence, but for the most part the pain associated with bowel movements is gone.

It is important to recognize scenarios where IPAA is not appropriate. These include patients with a history of fecal incontinence or sphincter injury, history of anorectal disease (anal fistula), uncertainty related to the diagnosis of UC versus Crohn’s disease (indeterminate colitis or IBD unclassified), and those who are frail and would not be able to tolerate the complications associated with IPAA (pelvic sepsis). IPAA should not be undertaken in patients with Crohn’s disease except by expert surgeons in specific situations as it is associated with a much higher rate of postoperative complications and pouch failure.

If the plan is to create an ileal pouch, when the rectal dissection is at the level of the pelvic floor, the rectum is transected with a stapler, approximately 1 cm above the anal sphincters. After the rectum is removed, the ileal pouch is created. If the colectomy was done in a prior operation, the terminal ileum is freed up from the ileostomy site. The distal terminal ileum is identified as is the superior mesenteric artery. The “tip” of the small bowel is located—the area of the small bowel that will reach the furthest into the pelvis. This is usually 10 to 15 cm proximal from the distal end. If this “tip” can touch the pubic bone, it will likely reach the anus. The two limbs of the ileal pouch are aligned, and a linear stapler is used to fashion the pouch ( Fig. 1 ). Usually, two loads of the linear stapler are needed as the goal is to have the pouch 10 to 12 cm in length. The pouch to anus anastomosis is created with a circular stapler ( Fig. 2 ). The anvil is placed in the pouch, secured with purse-string suture, and the handle is placed in the anus. The anastomosis is fashioned by firing the stapler. It is essential that there is excellent visibility in the pelvis when creating the anastomosis to ensure that the small bowel, bladder, vagina, etc. do not fall into the staple lines. There should be two complete “donuts” on the stapler, and the pouch should be inspected for bleeding by sigmoidoscopy and for air tightness by inflating air into the pouch and looking for air bubbles in the pelvis (saline irrigation is used to submerge the pouch from the abdominal side).

FIG. 1, Ileal pouch anal anastomosis in place with diverting loop ileostomy.

FIG. 2, Ileal pouch anal anastomosis creation using a circular stapler.

Most surgeons, when creating an IPAA, protect the pouch with a diverting loop ileostomy, created using more proximal small bowel brought up as a loop in the right lower quadrant. Although this does not prevent postoperative pelvic sepsis, it mitigates the severity as the fecal stream is diverted. The diverting loop ileostomy is closed in a separate operation about 3 months later. In most cases, this can simply be done with an incision around the ileostomy site, and the hospital stay is 1 to 2 nights. Rarely, more extensive dissection is needed, and a laparotomy is required. Before the ileostomy closure operation, the ileal pouch is interrogated with a Gastrografin enema to check for leaks at the staple lines and sometimes a flexible sigmoidoscopy to dilate the anus and inspect the mucosa.

PERIOPERATIVE AND POSTOPERATIVE CONSIDERATIONS

Perioperative Management

Enhanced recovery protocols beginning in the days leading up to surgery and continuing through postoperative management should be used. Pain medication regimens should limit opioids and maximize nonopioid analgesia including regional analgesia as appropriate. For patients on high-dose, long-term steroids, a short course of steroid taper should be used to avoid manifestations of adrenal insufficiency. Surgical site infection prevention bundles including presurgery bathing with chlorhexidine, mechanical bowel preparation with oral antibiotics (elective surgery), preincisional prophylactic antibiotics (cefazolin and Flagyl, or as determined by the hospital), alcohol-based skin preparation in the operating room, and appropriate attention to sterile technique using a closing set when suturing on the fascia and skin should be employed. Early ambulation with deep venous thrombosis prophylaxis is essential to reduce the risk of clot formation as this patient population is at significantly increased risk. Catheter-associated urinary tract infection bundles should also be employed comprising of careful insertion by a trained provider and prewash with antiseptic soap; early removal either at the end of the surgical case or the following morning should also be integrated into the care.

Ileal Pouch Complications

Complications can be divided into immediate perioperative complications associated with pelvic surgery and longer-term complications more specific to the ileal pouch. Surgical risks include injury to surrounding structures, especially nerves associated with sexual and urinary function, infection related to leaking at staple lines leading to pelvic abscess and sepsis, bleeding and ileus, or early postoperative small bowel obstruction. Overall, the short-term complication rates are in the range of 10% to 15%. Postoperative management should also include monitoring for persistent frank blood in the stool consistent with intraluminal bleeding (although some bleeding is expected in the first few days), which may require endoscopy with inspection of the staple line. Hemodynamically unstable patients should be taken to the operating room. Early anastomotic leak will present with fever, tachycardia, leukocytosis, and pelvic abscess on cross-sectional imaging and is usually treated with a percutaneous drain, but sometimes it is very low in the pelvis and needs to be drained transanally in the operating room.

Pouch-specific issues include acute, recurrent, or chronic inflammation in the ileal pouch, termed pouchitis. This can occur in about half of patients and is generally treated with antibiotics, mesalamine, topical steroids, budesonide, or biologic agents. A small subset of patients may develop de novo Crohn’s disease after IPAA. Medically refractory pouchitis, especially secondary to de novo Crohn’s disease, may lead to pouch failure and necessitate end ileostomy and/or pouchectomy. Patients with inactive UC and no prior pelvic surgery have similar infertility rates to the general population. However, women should be counseled that IPAA has been reported to increase the infertility rate compared with medically managed patients, although laparoscopic approaches may preserve fertility.

SUMMARY

Patients with UC are best cared for by a patient-centered transdisciplinary team that includes an experienced surgeon, gastroenterologist, enterostomal therapist, and, if needed, hepatologist and dermatologist. Surgical approaches should be tailored to the patient’s goals of care, and patients who have previously undergone surgery for UC can serve as an important resource for future patients.

Suggested Readings

  • Beyer-Berjot L., Maggiori L., Birnbaum D., Lefevre J.H., Berdah S., Panis Y.: A total laparoscopic approach reduces the infertility rate after ileal pouch-anal anastomosis: a 2-center study. Ann Surg 2013; 258: pp. 275-282.
  • Fazio V.W., Kiran R.P., Remzi F.H., et. al.: Ileal pouch anal anastomosis: analysis of outcome and quality of life in 3707 patients. Ann Surg 2013; 257: pp. 679-685.
  • Lamore R.F., Hechenbleikner E.M., Ha C., et. al.: Perioperative glucocorticoid prescribing habits in patients with inflammatory bowel disease: a call for standardization. JAMA Surg 2014; 149: pp. 459-466.
  • Turner D., Ricciuto A., Lewis A., et. al.: STRIDE-II: An Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology 2021; 160: pp. 1570-1583.
  • Wilson M.Z., Connelly T.M., Tinsley A., Hollenbeak C.S., Koltun W.A., Messaris E.: Ulcerative Colitis Is Associated With an Increased Risk of Venous Thromboembolism in the Postoperative Period: The Results of a Matched Cohort Analysis. Ann Surg 2015; 261: pp. 1160-1166.

Management of Toxic Megacolon

Jonathan E. Efron, MD

Toxic megacolon was first described in 1933 at Massachusetts General Hospital as a case report but became more widely known in the 1950s. It is an infrequent, potentially life-threatening condition that results from any inflammatory condition of the colon. It is most commonly seen as a complication of inflammatory bowel disease (IBD), more commonly ulcerative colitis (UC) than Crohn’s disease (CD) and some infectious colitides, most commonly with Clostridium difficile –associated (pseudomembranous) disease (CDAD). In cases of IBD, toxic megacolon results as a progression from fulminant colitis. Although fulminant colitis is not precisely defined, this term generally refers to severe inflammation of the colon with associated systemic toxicity with or without colonic dilatation. According to the diagnostic criteria of Truelove and Witts for the disease activity in UC, fulminant colitis is diagnosed by the presence of bloody diarrhea more than 10 times, heart rate higher than 90 beats/min, temperature above 37.5°C, requirement of blood transfusion, erythrocyte sedimentation rate (ESR) more than 30 mm/hr, with the presence of abdominal distension and tenderness on clinical examination, and dilated colon on x-ray. In the context of CDAD, according to the Dallal classification of CDAD severity, fulminant colitis is diagnosed by the presence of a heart rate above 120 beats/min, leukocytosis with more than 30% bands, severe oliguria, and requirement of mechanical ventilator and vasopressors. Toxic megacolon is defined as segmental or total colonic distension of 6 cm or more in the presence of acute colitis with systemic toxicity. Radiologically, it typically exhibits dilatation of the proximal colon with thickened inflamed distal colon and associated pneumatosis. Unlike colonic obstruction, in which cecal dilation with perforation is a concern, the transverse colon is the most common area of dilatation in toxic megacolon.

Given the other conditions that cause colonic distention, such as colonic pseudo-obstruction and Hirschsprung’s disease, toxic megacolon is distinguished from these conditions by its systemic manifestations of toxicity. Early diagnosis and aggressive medical management are pivotal to prevent progression to the associated high morbidity and mortality. Moreover, prompt recognition of disease progression and severity and timely surgical intervention may be lifesaving. The mortality rate for patients with toxic colitis significantly increases when the disease progresses to perforation—2% for those patients without perforation or 40% to 50% for those patients with perforation; therefore, close observation and surgical intervention before perforation is essential.

CAUSE, INCIDENCE, AND PATHOGENESIS

Any inflammatory condition of the colon can result in toxic megacolon. These include IBD, infectious causes including pseudomembranous colitis caused by C. difficile or other bacteria, such as Salmonella, Shigella, Campylobacter, or Entamoeba, and ischemic colitis ( Box 1 ).

BOX 1
Causes of Toxic Megacolon

Most Common

Ulcerative colitis

Clostridium difficile –associated colitis

Less Common

Crohn’s disease

Salmonella

Shigella

Campylobacter

Yersinia

Cytomegalovirus

Entamoeba histolytica

Cryptosporidium

Ischemia colitis

Chemotherapy

Colonoscopy

Barium enema

Drugs that slow colonic motility (narcotics, antidiarrheal drugs, anticholinergic drugs)

BOX 1
PSOGI 2016 Classification of Noncarcinoid Appendiceal Epithelial Neoplasms

  • Tubular, tubulovillous or villous adenoma, low- or high-grade dysplasia

  • Serrated polyp with or without dysplasia (low or high grade)

  • Low-grade appendiceal mucinous neoplasm

  • High-grade appendiceal mucinous neoplasm

  • Mucinous adenocarcinoma: well, moderately, or poorly differentiated

  • Poorly differentiated (mucinous) adenocarcinoma with signet ring cells

  • (Mucinous) signet ring cell carcinoma

  • Adenocarcinoma: well, moderately, or poorly differentiated

The incidence of toxic megacolon varies by the underlying cause. In patients with UC, it is estimated to be 5%, and the risk is higher early on in the disease. Historically, the reported incidence of CDAD ranged between 0.4% and 3%. However, with the changes in the epidemiology of C. difficile infections and emergence of new strains, there has been a 23% annual increase in the rate of hospitalizations resulting from CDAD in the United States.

The pathogenesis of the toxic dilatation of the colon is not fully understood. However, it is thought to be a result of severe inflammation of the colon associated with release of inflammatory mediators that induce colonic smooth muscle relaxation and inhibit colon motility. The acute severe mucosal inflammation becomes transmural and extends into the smooth muscle layer, resulting in loss of motor tone and paralysis. The severely inflamed smooth muscle produces nitric oxide, which is released into the colonic wall and further inhibits smooth muscle tone and causes dysmotility and atony. This generally causes dilatation of the colon proximal to the colonic segment that is severely inflamed. The toxic systemic response results from bacterial translocation and subsequent bacteremia.

Several other factors such as hypokalemia, hypomagnesemia, opiates, anticholinergic or antimotility agents, antidepressants, barium enemas, and colonoscopy may affect adversely colonic motility and exacerbate colon dilatation.

DIAGNOSIS

The diagnosis of toxic megacolon is based on both clinical and radiologic findings. Therefore, a thorough history and physical examination are crucial. The diagnosis must be suspected in patients who have diarrhea, abdominal distension, and signs of systemic toxicity.

The patient’s history typically reveals symptoms of severe colitis that preceded the acute onset of colonic dilatation. These include severe diarrhea (usually bloody), abdominal pain, fever, chills, and tachycardia. Obtaining a history about a previous diagnosis of IBD with the extent of colonic involvement and medical therapy and recent use of antibiotic or other medications such as steroid, antimotility, and chemotherapeutic agents will help in determining the underlying cause.

Physical examination reveals significant localized or generalized abdominal tenderness and reduced bowel sounds accompanied with signs of systemic toxicity, such as fever, tachycardia, and hypotension. Presence of signs of peritonitis may indicate colonic perforation. However, it is not uncommon that peritoneal signs maybe masked by high dose steroid treatment typically used in IBD patients with fulminant colitis.

The best acceptable clinical criteria for the diagnosis of toxic megacolon were described by Jalan et al. in 1969. The presence of three of the following criteria is required for the clinical diagnosis: fever higher than 101.5°F (38.6°C), heart rate higher than 120 beats/min, white blood cell count above 10.5 (× 10 9 /L), or anemia. In addition, patients should have one of the following criteria: dehydration, mental changes, electrolyte disturbances, or hypotension.

A plain abdominal x-ray is useful in confirming the diagnosis of toxic megacolon as it identifies the proximal colonic distention and may be used to follow the disease course and the rate of colon expansion. It typically shows dilatation of the ascending and transverse colon that varies from 6 cm up to 15 cm. Once the transverse colon is dilated past 8 cm, there should be great concern for pending perforation. Other radiologic features include presence of air fluid levels and the loss of normal haustral pattern in the colon with thickening and edema of colonic wall. Small bowel and gastric distension may be seen as well, and they have been shown to be significant predictors of toxic megacolon and progression to multisystem organ dysfunction in UC.

Computed tomography (CT) scan of the abdomen and pelvis is useful in confirming the diagnosis, excluding other causes of colonic dilatation, such as obstructing colonic cancer or diverticular stricture, and helping to exclude other abdominal complications, such as colonic perforation and ascending pyelophlebitis. Presence of colonic wall thickening, submucosal edema, pericolic stranding, and thickened haustra are indicative of severe colitis. As with plain abdominal x-rays, the presence of dilatation of the transverse colon (greater than 6–8 cm) confirms the diagnosis of toxic megacolon ( Fig. 1 ).

FIG. 1, Computed tomography scan of the abdomen showing dilated and thickened wall of the colon.

Laboratory tests are not specific and show the findings of systemic inflammatory response with leukocytosis, anemia, elevated ESR or serum C-reactive protein, and electrolyte abnormalities with hypokalemia, hypomagnesemia, and hypoalbuminemia. These findings, if not corrected, may exacerbate the condition.

Stool sample for culture, sensitivity, and C. difficile toxin assay should be sent as well as blood culture because bacteremia occurs in up to 25% of patients with toxic megacolon. Several tests to detect C. difficile are available. Sensitivity and specificity of each test vary; therefore, it is recommended to perform a two-stage test approach to improve the diagnosis accuracy. Stool culture is highly sensitive; however, it does not differentiate between the presence of Clostridium bacteria and active infection. It generally is used in conjunction with other diagnostic tests ( Table 1 ). The commonly used two-stage test approach includes initial screening with glutamate dehydrogenase assay followed by confirmation of a positive test with cell cytotoxicity assay. Some centers use toxin B gene PCR testing with nucleic acid amplification test as a single test to diagnose C. difficile.

TABLE 1
Laboratory Tests for the Diagnosis of Clostridium difficile Infection
Test Sensitivity Specificity
Cell cytotoxicity assays 60%–100% 96%–99%
Cell culture neutralization assay 67%–86% 97%–100%
Enzymatic detection of glutamate dehydrogenase 71%–91% 76%–98%
Enzyme immunoassay tests for toxins A and B 39%–76% 84%–100%
Nucleic acid amplification test for toxins A and B 84%–100% 94%–100%

Limited endoscopy, proctoscopy, or sigmoidoscopy may be considered to determine the cause of toxic megacolon in patients who are not known to have IBD. It can differentiate between the infectious causes of toxic megacolon because the finding of pseudomembranes is suggestive of CDAD, whereas the presence of inclusion bodies in the biopsies indicates cytomegalovirus (CMV) colitis as an underlying cause, which may occur in patients with IBD. It should be performed with extreme caution, without bowel preparation, and with minimal air insufflation; the endoscope should be advanced only as far as necessary to make a diagnosis. Complete colonoscopy should not be performed because of the high risk of perforation.

THERAPY

Management of toxic megacolon requires coordination between medical and surgical services with aggressive attempts of medical therapy and early surgical intervention in the absence of improvement, development of complications, or deterioration.

Medical Therapy

Regardless of the underlying cause of toxic megacolon, immediate aggressive supportive management in an intensive care unit must be initiated.

There is no evidence to support the use of nasogastric tube decompression because it does not decompress the colon. Frequent patient repositioning has been described as a method of colon decompression based on the observation that gas tends to accumulate in the transverse colon if the patient remains in the prone position and may be redistributed to the distal colon and rectum with frequent repositioning. There is no strong evidence to support use of this technique; however, it is simple and may be attempted.

Complete bowel rest with adequate intravenous fluid replacement is required. Electrolyte abnormalities, especially hypokalemia, dehydration, and anemia can exacerbate colonic dysmotility and must be corrected aggressively. Any medications that affect colonic motility such as opiates, antimotility agents, and anticholinergics should be discontinued immediately. Prophylaxis for deep venous thrombosis and gastric ulcer should be administered. Broad-spectrum antibiotics were found to reduce the mortality from septic complications that result from associated bacteremia or colonic perforation and should be initiated, whereas any agent that may have led to C. difficile overgrowth should be discontinued. Frequent clinical assessment and close monitoring with physical examination, serial complete blood counts, electrolyte monitoring, and abdominal x-rays must be performed ( Fig. 2 ).

FIG. 2, Management algorithm for toxic megacolon.

Management of Patients with Inflammatory Bowel Disease

High-dose intravenous steroid (hydrocortisone 100 mg every 6 hours) should be administered immediately to patients known to have IBD who have symptoms of fulminant colitis to prevent progression to toxic megacolon. There is no evidence that steroid therapy increases the risk of perforation; however, it may mask the signs of colonic perforation, so again close surveillance is required.

Aminosalicylic acid products are used in mild to moderate cases of UC; however, there are no data to support their benefit in treating toxic megacolon. Similarly, cyclosporine and antitumor necrosis factor-alpha (TNF-α) are immunosuppressant medications used in severe cases of UC. Both medications are initiated if there is no response to high-dose intravenous steroids within 3 days. Although generally not started for patients with acute toxic megacolon, as mentioned these drugs are often given to patients with fulminant colitis who are not responding to steroid therapy and so are often seen in patients who progress to surgery. Some data suggests that cyclosporin may have an initial effect in 80% of patients with severe fulminant colitis bordering on toxic megacolon and therefore may reduce the need for emergent surgery. The medical management is the same regardless of the defined IBD: UC, CD, or indeterminant colitis.

Management of Patients with Clostridium difficile–Associated Disease

If toxic megacolon is thought to be caused by CDAD, the antibiotics thought to have initiated the C. difficile infection should be withdrawn immediately, and treatment with oral vancomycin (125–500 mg four times a day) and/or oral metronidazole (200–500 mg four times a day, or 500 –750 mg three times a day) is initiated. Intravenous metronidazole is also acceptable. If a patient cannot tolerate oral vancomycin because of severe ileus, it may be administered via an enema or nasogastric tube. There have been reports of successful treatment of severe C. difficile colitis with colonoscopy and intracolonic vancomycin lavage. We would advise against this in patients suspected of having toxic megacolon due to the risk of perforation, which significantly increases the mortality rate of the patient.

Surgical Therapy

Timing of surgical intervention is paramount in decreasing the morbidity and mortality of toxic megacolon and improving the patient’s overall outcome. Medical management has been reported to be effective and successful in 50% to 70% of patients with toxic megacolon. However, delay in surgical intervention carries a risk of developing abdominal complications such as colonic perforation and abdominal compartment syndrome, which increases the mortality rate from 2% to 50%.

The possible need for surgical intervention and the nature of surgery must be discussed with patients and their families on admission. All patients must be evaluated and marked by an enterostomal therapist who should mark the best site for an end ileostomy.

Surgery is absolutely indicated in the presence of progressive colonic dilatation, uncontrolled hemorrhage, development of complications such as free perforation, and with general clinical deterioration. This includes progressive sepsis with continued tachycardia, hypotension, or the need for presser agents to maintain blood pressure. Lack of improvement within 48 hours is also a relative indication for surgical intervention. It is better to proceed to the operating room sooner as opposed to later, and any of the previously mentioned findings should push the surgeon to operate.

Mechanical bowel preparation is contraindicated, and the surgery typically is performed through an open approach for two reasons. The significant colon dilatation and friability of the colonic wall do not allow for a workable space or graspable tissue and therefore preclude a laparoscopic approach. The patient is often unstable and needs significant resuscitation from sepsis, requiring a quick efficient operation. Patients with fulminant colitis who have not progressed to toxic megacolon are often managed with a laparoscopic total colectomy and end ileostomy. If the surgeon feels the colonic distention will not interfere with laparoscopic visibility, the patient is hemodynamically stable and not on any pressor medications, and colonic wall does not appear significantly thinned or necrotic, an attempt at a laparoscopic total colectomy is feasible.

The current surgical standard of care for patients with toxic megacolon who require surgery is total colectomy with end ileostomy. This removes the diseased colon and allows restoration of intestinal continuity after the patient has recovered. The rectum should not be resected at the time of this emergent operation despite how inflamed it may appear. The bowel is often fragile with a high likelihood of intraoperative perforation with manipulation resulting from the significant dilatation and inflammatory process, and thus it should be handled with extra care. During hepatic and splenic flexures mobilization, the colonic mesentery is divided close to the bowel wall to avoid damage to retroperitoneal structures.

The rectal stump is often just as severely inflamed and, therefore, difficult to manage. If the rectosigmoid junction appears too inflamed to hold staples or sutures, then the surgeon should leave a short segment of sigmoid colon to form a mucous fistula to decompress the remaining colon and rectum. In obese patients, the rectal stump may be brought through the inferior aspect of the midline fascial incision and left buried in the subcutaneous space. This will be removed at time of stoma reversal, and if the stump blows out, it allows for decompression through the wound as opposed to in the peritoneal cavity. If a rectal stump is left in the peritoneal cavity, it should be decompressed in the operating room with a rectal tube that is left in place to allow for further postoperative decompression and possible vancomycin enemas. Drains should be left on top of the stump. A rectal stump leak typically occurs 5 to 10 days after surgery; therefore, if a patient manifests signs of peritonitis 5 to 7 days after surgery after initially recovering well, the surgeon should have a high suspicion for rectal leak. Emergent return to the operating room with washout and drainage is necessary.

Postoperative care requires transferring the patient to an intensive care unit where all supportive measures are continued as needed. Preoperative antibiotics are discontinued within 24 hours, and intravenous steroids are tapered to a maintenance dose (equivalent of 10–20 mg of prednisone per day). On restoration of gastrointestinal motility, enteral feeding is given. The rectal tube is removed on the fifth to seventh postoperative day.

An alternative, less invasive, colon-preserving surgical approach for the treatment of CDAD is the formation of a loop ileostomy with antegrade vancomycin irrigation of the colon. In a case-controlled study, Neal et al. treated 42 patients with creation of a loop ileostomy after visual assessment of colon viability. Intraoperatively, colonic lavage with 8 L of warmed polyethylene glycol 3350/electrolyte solution was performed via the ileostomy and drained via rectal tube. Postoperatively, patients received antegrade vancomycin enema (500 mg in 500 mL of lactated Ringer’s every 8 hours for 10 days) via the efferent limb of the ileostomy. All patients received intravenous metronidazole (500 mg every 8 hour for 10 days). These patients were compared with matching historical controls treated with total colectomy and end ileostomy. In the study cohort, 35 (83%) cases were performed laparoscopically. The colon was preserved in 39 (93%) patients, and subsequent colectomy was required for continued sepsis in one patient and for abdominal compartment syndrome in two patients. When compared with a historical population, mortality was reduced from 50% to 19%.

Although this novel surgical approach may represent a less-invasive surgical treatment with promising outcomes, the results of this study are limited by the retrospective nature and lack of randomization that introduce a selection bias. Furthermore, there are no clear criteria to suggest which patient may benefit from this approach. Finally, the results were never reproduced by other investigators. There has been little written recently, but the technique may still be considered for those patients who are less severe. The 2015 practice parameters of the American Society of Colon and Rectal Surgeons for the management of C. difficile infection considered that the evidence for loop ileostomy formation in the management of toxic megacolon from C. difficile to be weak and strongly recommend the standard surgical approach in managing these patients, which is subtotal colectomy with end ileostomy. Loop ileostomy with antegrade colonic vancomycin lavage should not be considered in the severely septic patient requiring significant (presser or ventilatory) support.

OUTCOMES

The mortality rates from IBD-associated toxic megacolon have changed dramatically over the years. In an early review (1976) of 604 patients, the overall mortality was 19%, and it was higher in medically managed patients when compared with patients who were treated with early surgery (27% vs. 19.5%). The presence of perforation increased the mortality to 41.5% compared with 8.8% in the absence of perforation. In more recent reports, mortality rate after colectomy is 2% to 5%. Older age and multiple comorbidities are associated with a higher mortality rate. The mortality rate after colectomy for CDAD remains high. In a systematic review (2009) of 1433 patients, it was found to be 41.3%. Preoperative intubation, acute renal failure, multiorgan failure, and requirement of vasopressors were found to be predictors of postoperative mortality.

SUMMARY

Toxic megacolon can complicate inflammatory or infectious colitis. It is a life-threatening emergency characterized by severe colonic distension and systemic toxicity. Early recognition, a multidisciplinary management approach, and the time of surgical intervention are crucial to improving outcomes.

Suggested Readings

  • Ausch C., Madoff R.D., Gnant M., et. al.: Aetiology and surgical management of toxic megacolon. Colorectal Dis 2006; 8: pp. 195-201.
  • Autenrieth D.M., Baumgart D.C.: Toxic megacolon. Inflamm Bowel Dis 2012; 18: pp. 584-591.
  • Carchman E.H., Peitzman A.B., Simmons R.L., et. al.: The role of acute care surgery in the treatment of severe, complicated Clostridium difficile -associated disease. J Trauma Acute Care Surg 2012; 73: pp. 789-800.
  • Desai J., Elnaggar M., Hanfy A.A., Dshi R.: Toxic Megacolon: Background, Pathophysiology, Management Challenges and Solutions. Clinical and Experimental Gastroenterology 2020; 13: pp. 203-210.
  • Klobuka A.J., Markelov A.: Current status of surgical treatment for fulminant Clostridium difficile colitis. World J Gastrointest Surg 2013; 5: pp. 167-172.
  • Neal M.D., Alverdy J.C., Hall D.E., et. al.: Diverting loop ileostomy and colonic lavage: an alternative to total abdominal colectomy for the treatment of severe, complicated Clostridium difficile associated disease. Ann Surg 2011; 254: pp. 423-427. discussion 427–429

Surgical Management of Crohn’s Colitis

Hanjoo Lee, MD
Alessandro Fichera, MD,
James W. Fleshman Jr., MD

INTRODUCTION

Crohn’s disease (CD) is a chronic inflammatory disorder of the gastrointestinal tract first described by Burrill Crohn, Leon Ginzburg, and Gordon Oppenheimer in 1932. The disease was thought to be limited to the small intestine and was referred to as “regional enteritis.” Subsequent observations confirmed that CD may not only affect any segment of the gastrointestinal tract but also involve a slew of extraintestinal organs including the bronchopulmonary, integumentary, ocular, and joint systems. Currently, 3 million patients are affected by the disease in the United States with an incidence of 3 to 20 cases per 100,000 people. Interestingly, the past five decades have seen a gradual increase in incidence. Populations with Northern European and Jewish heritage exhibit the highest incidence of CD. However, an increasing trend amongst Asian and Hispanic populations has also been noted in recent years. The disease onset shows bimodal peaks around ages 20 and 50, although diagnosis in children is not uncommon. Few pathologies involving the gastrointestinal tract have puzzled the medical community as much as CD. The pathophysiology of the disease is exceedingly difficult to characterize due to the complex interplay between multiple environmental and genetic factors involved. The environmental factors include gastrointestinal infection, chronic use of nonsteroidal antiinflammatory medications, and exposure to antibiotics to name a few. Generally, a disruption in the integrity of the intestinal mucosa and natural gut flora are thought to be involved. Perhaps the most remarkable known risk factor is smoking. Active and passive smoking as well as previous history of smoking are all shown to increase the risk of development and worsening disease severity. Several genetic risk factors have been identified including mutations in MLH1, a DNA mismatch repair gene, and in CARD15 , a nuclear factor-kappa B transcription factor. Increased incidence in identical twins and first-degree members of Crohn’s patients also points to hereditary contribution of disease development. Clinical presentation of CD is multifaceted. It may range from minimal symptoms to fulminant colitis or debilitating perianal disease. As mentioned before, CD can affect the entire gastrointestinal tract. In 20% to 30% of cases, the disease presents as isolated Crohn’s colitis (CC). When CC is suspected, it is extremely important to differentiate this diagnosis from ulcerative colitis (UC). Although there is a symptom overlap between CC and UC, the goal of surgical treatment is different. A prudent practitioner must be aware of several key differences between these two entities. Skip lesions, rectal sparing, longitudinal ulcers, intestinal and perianal fistulizing disease, and mucosal cobblestoning are all notable characteristics pointing toward CC rather than UC ( Fig. 1 ). Abdominal pain, fever, and diarrhea are common initial symptoms of CC, and occasionally a palpable abdominal mass can be present. Unlike UC, rectal bleeding is uncommon in CC. Development of carcinoma of the colon is well described in patients with CC based on the extent of involvement and duration of the disease, and it is not dissimilar to the risk in UC.

FIG. 1, Mucosal appearance of CC. (A) Entire colon is involved on a TAC specimen. (B) Extensive mucosal cobblestoning is present. (C) Normal-appearing terminal ileum.

The presentation of CC as a solely perianal disease is rare (<6%), but almost 20% of patients with CC will develop perianal disease over their lifetime. History and physical exam alone can be useful in establishing the diagnosis of CC. Laboratory workup should include C-reactive protein, erythrocyte sedimentation rate, fecal calprotectin, and albumin levels in addition to the standard complete blood count and basic metabolic panel. These values can also be used to assess disease progression. Plain x-rays of the abdomen are helpful initial radiographic tools to identify free perforation and evaluate severity of colonic dilation. Barium enema can show several characteristic findings of CC including longitudinal or transverse ulcers, deep fissuring of the bowel wall, coarse mucosa cobblestoning, or longitudinal intramural fistulas. Single or double contrast barium enemas are rarely used due to the excellent tomographic techniques available that can provide enterography with no heavy contrast or risk of perforation. Computed tomography (CT) with enterography is useful in evaluating bowel wall thickness, stricturing, intraabdominal abscess, internal hernias, and/or extraintestinal involvement. If previous surgical history is unclear, CT can help clarify the anatomy. Endoscopic examination of the colon with biopsy is crucial for diagnosis and grading of disease intensity. Proctosigmoidoscopy may be sufficient in diagnosing CC, but most practitioners prefer full colonoscopy, especially in the preoperative setting. This is because 40% of CD spares the rectum regardless of perianal involvement. If UC is suspected, biopsy of the rectum, even if normal appearing on visual inspection, is helpful to rule out pathologic inflammatory changes. In 10% to 15% of cases, differentiation between UC and CC may truly not be possible even under pathologic examination. These cases are categorized as inflammatory bowel disease unclassified (IBDU) and called indeterminate colitis. In the absence of definitive microscopic findings (i.e., noncaseating granulomas), pathologists often refer to clinical information to arrive at their definitive diagnosis.

MEDICAL MANAGEMENT

Similar to UC, initial management of CC should be supportive care with bowel rest, intravenous hydration, and antibiotics. Placement of a nasogastric tube may alleviate symptoms if the stomach and small bowel are dilated. Intravenous glucocorticoids should be initiated early in the course of management. Serial abdominal exam, plain radiographic films, and serologic markers described previously can be used to monitor disease progression. If symptoms do not improve in 72 hours, use of an anti-TNF antibody (infliximab) should be considered. Generally, response to anti-TNF should occur within 5 to 7 days.

INDICATION FOR SURGICAL INTERVENTION

Despite the remarkable advances in medical therapies in the past two decades, surgery still plays an important role in the management of CD. In fact, lifetime risk of needing a resection ranges between 50% to 80%, and this rate remained relatively stable in the postbiologics era. Therefore, discussion with patients about indication for surgery should occur early in the disease process so that a fully informed decision can be made when the need for surgery arises. The indications for surgical intervention can be categorized into emergent, urgent, and elective ( Table 1 ). Acute abdomen with diffuse peritonitis, severe bleeding, and hemodynamic instability undoubtedly warrant emergent exploration. Toxic megacolon, acute large bowel obstruction unresponsive to medical management, and intraabdominal abscess without successful control of sepsis by percutaneous drainage often require urgent exploration within the same hospitalization. Elective surgery is appropriate in cases where disease continues to progress despite extensive medical therapy, partial obstruction with fecalization of the small bowel, persistent intraabdominal abscess despite percutaneous drainage and antibiotics, presence of high-grade dysplasia or malignancy, and failure to thrive in children. Every effort should be made to transition urgent surgery to an elective surgery setting to gain invaluable time needed to optimize the patient and reduce the risk of postoperative complications.

TABLE 1
Indications for Surgical Intervention for Colonic Crohn’s Disease
Emergent
  • Acute abdomen with diffuse peritonitis

  • Severe bleeding

  • Hemodynamic instability

Urgent
  • Toxic megacolon

  • Acute large bowel obstruction unresponsive to medical management

  • Intraabdominal abscess without successful control of sepsis by percutaneous drainage

Elective
  • Continued disease progression despite extensive medical therapy

  • Partial large bowel obstruction with fecalization of the small bowel

  • Persistent intraabdominal abscess despite percutaneous drainage and antibiotics

  • Presence of high-grade dysplasia or malignancy

  • Failure to thrive in children

PREOPERATIVE OPTIMIZATION

Colonic resection for CC, regardless of the specific indication, is associated with significant morbidity. Postoperative outcomes are intimately associated with preoperative optimization. Multidisciplinary management including gastroenterology, infectious disease, enterostomal nursing, nutrition, surgery, and interventional radiology is critical. Since inflammatory bowel disease management has numerous nuances, it is best managed in a facility that cares for tertiary and quaternary level patients and provides optimal resources for the high-risk complex gastrointestinal procedures. Patients referred for surgery are typically immunocompromised and malnourished. Dose-dependent operative morbidity with steroid therapy is well documented. In contrast, no clear association has been established between use of immunomodulators such as azathioprine, 6-mercaptutopurine, and methotrexate and postoperative morbidity. Safety of preoperative exposure to biologic agents is a highly controversial subject due to a number of large-scale studies that showed conflicting results. Given the lack of conclusive data, urgent surgery generally should not be delayed based on recent exposure to biologic agents alone, and fecal diversion should be individualized based on the overall clinical picture.

Nutritional status is a significant preoperative factor to consider not only in CD but in any complex surgical case. Malabsorption of nutrients and change in dietary habits are inherently associated with CD before resection. Therefore, nutritional optimization is of critical importance. Notably, weight loss >10% is associated with postoperative intraabdominal septic complication. Nutritional supplementation can be administered via either elemental enteral alimentation (EEA) or total parenteral nutrition (TPN). EEA is preferred over TPN due to maintenance of the physiologic route of nutrient absorption and avoidance of complications associated with TPN and central line placement. Preoperative EEA supplementation, for 3 months when feasible, has been shown to reduce the rate of postoperative septic complications. If intraabdominal sepsis is present, medical management should be initiated immediately, and it should include intravenous hydration and initiation of antibiotics to cover enteric flora. The antibiotic regiment can then be tailored according to culture results. Any intraabdominal collection >3 cm should be managed with percutaneous drainage with interventional radiology whenever possible. These measures may lead to a definitive resolution with medical therapy or serve as a bridge to surgery. If an enterocutaneous fistula is present, early involvement of a wound and ostomy nurse is critical. The reduction of the surrounding secondary inflammatory response to an area of severe disease or local perforation should be a primary goal of preoperative optimization.

The tissue handling characteristics of a “phlegmon” will often result in secondary injury of secondarily involved normal intestine and a wider resection of intestine than actually required to manage the disease. The risk of short bowel syndrome is thus increased. Patience on the part of the surgeon and patient will soften the tissue to allow sharp, atraumatic dissection in normal tissue planes. Patients with CD are at increased risk of developing venous thromboembolism (VTE) due to several associated factors including hypercoagulability in a proinflammatory state, malnutrition, anemia, thrombocytosis, prolonged hospital stay with limited mobility, and use of steroids. Venous thromboprophylaxis should be initiated during the optimization period and carried through the operation unless significant bleeding risk is present. The risk of VTE continues to be elevated at least 30 days after an operation, and VTE prophylaxis should be continued postoperatively. Judicious use of postdischarge VTE prophylaxis should be considered, especially if other risk factors are involved such as smoking, obesity, prolonged pelvic surgery, immobilization, and malignancy.

SURGICAL MANAGEMENT

Surgical options for CC are segmental colectomy, total abdominal colectomy (TAC) with or without ileorectal anastomosis, total proctocolectomy (TPC) with a permanent end ileostomy, and restorative proctocolectomy with ilea-anal pouch anastomosis in a selected patient population ( Table 2 ). While minimally invasive surgery for CD disease has been found to be associated with better short-term outcomes, the approach should be tailored to the clinical presentation and surgeon’s skills and expertise. The critical principles of small bowel preservation apply to CC. This is especially important if the patient has already lost a significant amount of small bowel. The absorptive capacity of the colon that is not diseased can reduce diarrhea and possible “short bowel syndrome” secondary to operative treatment of ileocolonic CD. If <20 cm colon is affected, especially in the proximal colon, segmental resection with primary anastomosis should be considered. Leaving behind a segment of colon does put the patient at increased risk of recurrence, up to 62% at 5.5 years. This risk should be clearly discussed with the patient before surgery. Typical surgical considerations and principles apply to segmental colectomies for CC.

TABLE 2
Surgical Options for Colonic Crohn’s Disease
Segmental colectomy
  • Previous multiple small bowel resections

  • <20 cm involved segment in the proximal colon, segmental resection with or without primary anastomosis should be considered

Total abdominal colectomy (TAC)
  • More than two segments of colon are involved with rectal sparing and anastomosis is deemed unsafe

  • Patient is in extremis with pancolitis and a total proctocolectomy is not indicated

  • Definitive diagnosis is not established, and indeterminate colitis is considered

Total proctocolectomy (TPC)
  • Involvement of two or more segments of colon especially with perianal fistulizing disease

  • Visible dysplasia not amenable for complete endoscopic removal

  • Multifocal dysplasia, dysplasia in the surrounding flat mucosa

  • Carcinoma in the setting of pancolitis

Ileal pouch-anal anastomosis (IPAA)
  • Absence of small bowel or perianal involvement with a definitive preoperative diagnosis of CC

Total Abdominal Colectomy

In a patient without free perforation, the preferred methods for TAC are laparoscopic mobilization of the abdominal colon and transection of the terminal ileum and rectosigmoid junction through a Pfannenstiel incision or intracorporeal transection at the rectosigmoid junction and extraction through the ileostomy site incision. There are several indications for TAC: (1) when more than two segments of colon are involved, with rectal sparing, and anastomosis is deemed unsafe; (2) when a patient presents in extremis with pancolitis, but a TPC is not indicated; and (3) when definitive diagnosis is not established, and indeterminate colitis is considered. The dissection for laparoscopic TAC begins with transection of the ileocolic pedicle and mobilizing the cecum away from the right iliac vessels at the right pelvic brim in a medial to lateral fashion and continues in a clockwise rotation to the hepatic flexure taking care to avoid injuring the duodenum. The gastrocolic ligament and any ligamentous attachments to the liver, spleen, and pancreas are divided to mobilize the transverse colon. The lateral attachments of the right colon to the lateral abdominal sidewall are divided last to finally release the right colon to the midline. The authors prefer to divide the omentum distal to the gastroepiploic arcade and remove it with the specimen. Many surgeons would try to preserve the omentum, but the difficulty in releasing the omentum from an inflamed transverse colon is likely to result in a colotomy and fecal spillage.

The splenic flexure is mobilized in an antegrade fashion from the right to the left, when feasible. When severely inflamed or in obese patients, it may be necessary to return to the splenic flexure in the retrograde fashion after partial mobilization of the left colon. Care must be taken to divide the base of the mesentery of the splenic flexure with sealing energy in case the arc of Riolan is a significant vessel and the branches of the inferior mesenteric vein (IMV) persist. All ligamentous attachments of the splenic flexure should then be dissected close to the colon to avoid injury to the splenic capsule. The left colon is mobilized by dividing the lateral attachments and freeing it off of the retroperitoneum starting at the left pelvic brim and dissecting medially, taking care to avoid injury to the left ureter. Ureteral stents placed at the induction of anesthesia can facilitate identification and protection of the ureters in a patient with severe inflammation in the descending colon and sigmoid regions. A medial approach to releasing the left colon and rectosigmoid junction allows avoidance of inflammatory attachments of the sigmoid and left colon to the critical structures at the pelvic brim. Working from soft normal dissection planes toward the diseased areas gives a better chance to complete the dissection and avoid injury to diseased and normal structures. Unlike TAC for malignancy, ligation of the vascular supply to the colon does not need to occur close to its origin in CC, and the mesentery can be ligated closer to the colon.

To date, the benefit of extended mesenteric resection for CD in reducing recurrence rate is not clearly established. However, the mesentery near the diseased bowel is often inflamed and thickened, so the soft, normal embryologic plane at the base of the mesentery may be the preferred dissection plane. Care must be taken to avoid significant bleeding from the thickened, friable mesentery during the transection of the vessels and at the point of transection of the colon. The rectum is identified at the distal sigmoid colon where the tenia coli “splay” out onto the anterior surface of the rectum. The peritoneal reflection is incised around the pelvic sidewalls and the proximal rectum is mobilized using the areolar tissue plane posterior to the rectum at the sacral promontory. The terminal ileum and the rectosigmoid junction are transected with a linear cutter stapler to control spillage after dividing the mesentery up to the bowel wall, either intracorporeally or through a small Pfannenstiel incision. The rectosigmoid junction is the preferred level of distal transection. This provides adequate rectal length for ileorectal anastomosis or creation of a mucus fistula and removes all colonic mucosa. Depending on the clinical scenario either an end ileostomy or ileorectal anastomosis is performed using either hand-sewn or stapled technique.

Patients who are restored to intestinal continuity with an ileorectal anastomosis often experience a period of rapid return of bowel function followed by a sudden decrease in bowel function, marked abdominal distension, and vomiting. This phenomenon, sometimes described as “ileorectal syndrome”, is caused by exposure of the terminal ileum to a high back pressure from the rectal vault. Drainage of rectal contents and relief of build-up of pressure in the ileum using an in-dwelling large catheter (24–34F) in the rectum can alleviate this ileal response to perceived obstruction caused by normal rectal pressures withholding stool from evacuations until a steady bowel function returns.

There are certain factors that need to be considered when deciding between ileorectal anastomosis and end ileostomy. The rectum and the perianal region must be examined carefully to make sure they are free of stricturing or fistulizing disease before committing to ileorectal anastomosis. Adequate sphincter resting tone and maximal squeeze pressures are required to prevent incontinence of liquid stool. The presence of fistulotomy or episiotomy scars should raise concern for postoperative inadequate control of liquid stool. The patient’s general condition such as nutritional status, weight loss, usage and dosage of steroidal agents, and smoking status should be reviewed to determine the feasibility of anastomosis. Opting for an end ileostomy confers a lower rate of recurrence of CD, especially in active smokers and patients with penetrating disease. The ileostomy and Hartmann stump of the rectum allows the surgeon to appreciate the natural history of the patient’s disease to determine whether an ileorectal anastomosis is possible in the future. A Brook ileostomy should be performed in the acute setting, in cases of indeterminate colitis, and when the rectum is involved. Management of the rectal stump remains controversial. It is the authors’ preferences to drain the stump with a rectal tube and leave a drain by the staple line for the duration of the hospital stay. The transected Hartmann rectal stump of an inflamed rectum is at risk for dehiscence of the upper staple line due to active disease and tissue friability. Other surgeons bring the stump to the incision and either suture it to the fascia or open it as a formal mucus fistula. These two options require a longer stump that may not be feasible with sigmoid involvement or in the obese patient.

Total Proctocolectomy

If two or more segments of colon are involved, and if associated perianal fistulizing disease is present, TPC with end ileostomy should be performed. Any presence of visible dysplasia not amenable for complete endoscopic removal, multifocal dysplasia, dysplasia in the surrounding flat mucosa, or carcinoma in the setting of pancolitis should also warrant TPC and end ileostomy. This is based on the observation that more than a third of patients who undergo colectomy for dysplasia are found to have multifocal dysplasia. In addition, 14% to 40% of patients who undergo colectomy for carcinoma in CC eventually develop metachronous cancer. The proctectomy portion of TPC proceeds after the abdominal portion as described earlier. Typically, the pelvic dissection follows the total mesorectal excision (TME) plane performed for rectal cancer surgery. This plane allows for decreased blood loss especially in the presence of inflamed mesorectum. Using electrocautery, the loose avascular areolar tissue of the retrorectal space is dissected to mobilize the rectum with the mesorectum intact. Once the posterior dissection is complete to the levator ani, the lateral attachments and the anterior Denonvilliers’ fascia/rectovaginal septum are dissected to the levator ani to completely mobilize the mesorectum. Extra care must be taken not to mistake the pelvic parietal fascia for the fascia propria of the rectum because the loose avascular areolar tissue between presacral fascia and pelvic parietal fascia can look identical to the one present in the retrorectal space. Surgeons often realize this mistake mid-dissection and, in an attempt to return to the correct retrorectal space, transect the hypogastric nerve embedded within the pelvic parietal fascia. A helpful hint is that if median sacral vessels running on the sacral periosteum are visualized, the dissection plane is already in a posterior plane deep to the desired plane anterior to the pelvic parietal fascia. One technique to reduce potential pelvic nerve injuries is to perform close rectal dissection (CRD). In this technique, the dissection is performed close to the rectal muscle wall within the mesorectum. This minimizes the risk of injury to the hypogastric nerves, the pelvic splanchnic nerves, and the inferior hypogastric plexus.

When ileal pouch-anal anastomosis (IPAA) for benign disease was randomized to CRD and TME, the CRD group suffered significantly fewer severe complications and improved quality of life. However, dissection through the mesorectal tissue often results in bleeding and is understandably associated with longer operating time (approximately 30 minutes). Once the rectum is completely mobilized intraabdominally as close to the anal canal as possible, the transperineal portion of the procedure is begun. Intersphincteric dissection for the completion proctectomy is preferred, whenever possible. The dissection is begun at the anal verge in the palpable intersphincteric groove with a circumferential incision into the space between the internal and external sphincters. The autonomic internal sphincter circular fibers do not twitch when touched with electrocautery. The somatic external sphincter fibers that encircle the internal sphincter fibers do twitch. The longitudinal rectal muscle fibers run at right angles to the circular fibers in the intersphincteric space and guide the dissection cephalad to the level of the puborectalis muscle. Preserving the well-vascularized external sphincter reduces the size of the perineal wound, allows better pelvic closure and healing, and reduces the risk of injury to the vagina/prostate and neurovascular bundle. Notwithstanding, extra care must be taken during anterior dissection to avoid injury to the vagina or the prostatic capsule. Frequent palpation of the posterior vaginal wall or the bladder catheter in the perineal and prostatic urethra helps to maintain orientation in the correct dissection plane. The dissection progresses circumferentially in a cephalad direction, and the levator ani muscles are finally separated from the rectum at the puborectalis sling to join with the dissection plane from the intraabdominal portion of the procedure. Once the rectum is removed, a drain is left in place in the pelvis through the abdominal wall or perineum. The levator ani, the external sphincter, and the ischiorectal fat are approximated in layers across the midline, but the subcutaneous tissue and skin are left open. This serves the purpose of draining the wound as well as leaving an opening that cosmetically resembles an anal orifice. The drain is typically removed in two weeks if drainage is minimal.

Restorative Proctocolectomy with Ileal Pouch Anal Anastomosis

There is a rare, select patient group with isolated CC without small bowel or perianal involvement who may be candidates for IPAA. If the diagnosis of CD is made before IPAA, pouch failure rate is 15%. If the patient is able to keep the pouch, functional outcomes and quality of life are similar between IPAA for CD and UC.

Minimally Invasive Surgery

Adaptation of laparoscopic surgery for CD has been rather unhurried in the surgical community compared with other arenas of abdominopelvic surgery. This is because the disease involves chronically inflamed bowel with friable and thickened mesentery that makes laparoscopic surgery particularly challenging. The initial randomized and observational studies comparing the outcomes of laparoscopic colectomy to open surgery have been promising. Similar to comparative studies for other colorectal disorders, laparoscopic colectomy showed decreased morbidity, decreased blood loss, faster return of bowel function, shorter hospital stays, and, in some cases, decreased costs. Furthermore, laparoscopic surgery allows specimen extraction through a Pfannenstiel incision, which affords decreased incisional hernia rate and pain ( Fig. 2 ). Fistulizing disease warrants a special mention as it adds another layer of complexity. So far, comparative observational studies have shown that although operating time, conversion rate to open surgery, and rate of stoma creation are all increased, overall morbidity rate remains the same between laparoscopic surgeries for fistulizing and nonfistulizing disease. Individual surgeon’s skill and comfort level should dictate if laparoscopic surgery is feasible in these cases. The use of hand-assisted laparoscopic surgery is an intermediate step between open operation and totally laparoscopic operations. The experienced hand, inserted through an airtight seal in the Pfannenstiel incision, can facilitate dissection by finding planes, identifying structures, encircling vessels bluntly, and guiding the incision by feel to speed the operation, thus reducing stress and risk and achieving the same outcomes as a pure laparoscopic procedure.

FIG. 2, Hand-assisted laparoscopic TAC. (A) 5-mm umbilical laparoscope port for the camera, 12-mm right working port at the site of the future stoma (this is also the entry site), 5-mm left lateral assistant port, Pfannenstiel incision for hand assist and specimen extraction. (B) The entire colon is externalized through the Pfannenstiel incision.

Robotic Surgery

Advantages of robotic surgery including intracorporeal anastomosis, smaller incision, and minimal manipulation of the bowel generally apply to CD. Robotic surgery is associated with a median of 2-day shorter length of stay and lower 30-day complication rate compared with open surgery. When robotic TAC for CC was compared with laparoscopy, no differences were noted including complications. Evidence in favor of robotic surgery in the management of CD is still in development.

SURGICAL PEARLS

  • Utilize transversus abdominis plane (TAP) block and enhanced recovery after surgery (ERAS) pathway to improve outcomes.

  • If proceeding with laparoscopy, have predetermined criteria for conversion to open operation. This can be based on operative findings, inability to make progress, or hemodynamic instability.

  • Position with memory foams and chest straps in obese patients to avoid brachial plexus injury caused by shoulder brace and sliding of the patient on the table.

  • Tilt operating table at appropriate angles to improve visualization with gravity movement of abdominal contents.

  • Set up stirrups to keep legs no more than 10 degrees flexed at hips to allow adequate motion of instruments.

  • Foley catheter should be in place before trocars are inserted.

  • Keep insufflation pressure below 15 mm Hg if possible.

  • Insert trocars under direct vision with camera.

  • Insert instruments with camera guidance if at all possible.

  • Lift tissue without grasping whenever possible.

  • Start dissection at the base of the mesentery at the vascular bundle to make bloodless dissection possible.

  • Ensure no twist in the mesentery of the small bowel for ileostomy. This may require reinsufflation of abdomen for laparoscopic view of cut edge of mesentery without twist.

  • Know the protocol for air embolism: Trendelenburg and left lateral decubitus positioning to move the air into the right atrium to be aspirated.

POSTOPERATIVE MANAGEMENT

To date, there is no validated scoring system to stratify risk factors for postoperative recurrences. However, some of the known risk factors include penetrating disease, two or more previous CD surgeries, history of extensive small bowel resection of >10 cm, age <30 at diagnosis, an interval <10 years between diagnosis and surgery, and presence of perianal disease. Finally, smoking is an important modifiable risk factor that portends higher risk of recurrence. The number of risk factors involved can help categorize patients into low, moderate, and high risk of recurrence. Patients with no risk factor are considered low risk. These patients may stay off of medical therapy after surgery unless recurrence is noted at 6-month surveillance colonoscopy. Presence of several risk factors put the patient at moderate risk of recurrence. These patients should be aggressively managed with medical therapies including anti-TNF therapy in combination with immunomodulator. Alternatively fecal calprotectin monitoring 3 months after surgery can serve as an early indication for endoscopic examination.

CONCLUSION

Despite a century of advances in medical and surgical therapy, CD remains a clinical challenge for the treatment team and a chronic malady for patients. The complexity of the disease process and multifaceted phenotypic presentations demand multidisciplinary management. To date, the majority of patients suffering from CD ultimately will require surgery, and therefore surgeons are an integral part of this multidisciplinary team. A thorough preoperative workup is imperative to establish a clear diagnosis of CC and to adequately optimize nutritional status. A number of surgical techniques are available as minimally invasive options enter the fray. Each technique should be tailored to the patient’s condition and the surgeon’s skill and clinical judgment.

Suggested Readings

  • Bartels S.A.L., Gardenbroek T.J., Aarts M., et. al.: Short-term morbidity and quality of life from a randomized clinical trial of close rectal dissection and total mesorectal excision in ileal pouch-anal anastomosis. Br J Surg 2015; 102: pp. 281-287.
  • Fazio V.W., et. al.: Crohn’s disease and indeterminate colitis.Corman M.L. et. al.Corman’s colon and rectal surgery.2013.Springer
  • Feuerstein J.D., Cheifetz A.S.: Crohn disease: epidemiology, diagnosis, and management. Mayo Clin Proc 2017; 92: pp. 1088-1103.
  • Lightner A.L., Vogel J.D., Carmichael J.C., et. al.: The American Society of Colon and Rectal Surgeons clinical practice guidelines for the surgical management of Crohn’s disease. Dis Colon Rectum 2020; 63: pp. 1028-1052.
  • Lin A.Y.: Completion proctectomy for Crohn’s disease.Fleshman J.W. et. al.Atlas of surgical techniques for the colon, rectum, and anus.2013.ElsevierPhiladelphia:

Ischemic Colitis

Ranim Alsaad, MD,
Pamela A. Lipsett, MD, MHPE

DEFINITION AND EPIDEMIOLOGY

Ischemic colitis (IC) is a condition that occurs when the blood supply to colonocytes does not meet metabolic demands. It is the most common cause of gastrointestinal (GI) ischemia. Injury can occur in one of two ways: at the initial incident of decrease in blood flow phase and/or after reperfusion. If the injury occurred only at the mucosa, it can be reversible. However, a transmural injury can present as a life-threatening condition that can lead to stricture formation, perforation, sepsis, and death. The incidence of IC ranges from 4.5 to 44 cases per 100,000 persons per year. Patients generally present after the age of 60, and there is a higher prevalence in women. The incidence of IC is expected to rise given the growing proportion of elderly patients. It most commonly occurs after aortic or cardiac surgeries. The severity of the disease dictates specific management strategies. About 20% of patients diagnosed with IC will require operative intervention, typically in an emergent setting.

ETIOLOGY

To understand the pathophysiology behind IC, we must consider the arterial anatomy of the colon. As shown in Figure 1 , the colon is supplied by the superior mesenteric artery (SMA), inferior mesenteric artery (IMA), and branches of the superior rectal artery. In addition to these primary arterial branches, there is a rich collateral circulation. Griffith’s point, at the splenic flexure, and Sudek’s point, at the rectosigmoid junction, are particularly prone to ischemia, cumulatively compromising 80% of IC cases. These high-risk, or “watershed,” areas identify the regions in the colon between two major arteries; the splenic flexure is the area between the SMA and the IMA arterial supply, and the rectosigmoid junction is the region between the IMA and the superior rectal artery supply. The former area is mostly supplied by the marginal artery; however, in 50% of the population, this artery is poorly developed. During aortic surgery, when intraperitoneal, efforts should be made to ensure that the left colon has adequate blood supply if the IMA is acutely sacrificed.

FIG. 1, Colon blood supply.

IC is classified according to the mechanism of decreased blood flow to the colon. This is most commonly nonocclusive, as in cases of shock, drugs, and colon obstruction. Less commonly, IC occurs after a vascular insult, which could be due to an arterial thrombosis, embolism, or even a venous occlusion. A special entity is the postoperative IC that can occur after cardiac and vascular surgeries, in which there is an intraoperative temporary cessation of blood flow to the colon. For example, following abdominal aortic aneurysm repair or bypass, if the IMA was sacrificed or if there was prolonged cross clamp time, IC can occur.

During periods of hypotension, blood flow is redirected to the brain at the expense of the splanchnic circulation. At a histologic level, the initial ischemic changes are always in the mucosa on the antimesenteric side. These changes will eventually spread through the colon wall to the serosa if the insult continues. Mucosal injury will develop in 20 minutes to 1 hour of decreased blood flow, whereas transmural infarction occurs within 8 to 16 hours. Additional insult occurs when blood flow is reestablished, causing reperfusion injury. Reperfusion injury is associated with the release of reactive oxygen species, which causes lipid peroxidation within cell membranes and leads to cell necrosis.

Risk factors for IC include advanced age, female gender, peripheral artery occlusive disease, coronary artery disease, heart failure, chronic obstructive pulmonary disease, and inflammatory bowel disease (IBD). Postoperative IC occurs most commonly after aortic surgery or after cardiac surgery with temporary GI vascular exclusion. In addition, the odds of developing IC are higher in cigarette smokers, both current and former (relative to never smokers). The use of cardiovascular medications such as diuretics or digoxin and psychotropic medications within the past month of presentation is also significantly associated with IC.

CLASSIFICATION

One can classify IC into mucosal vs. transmural types with further classification by severity as mild, moderate, and severe IC. Severe IC often involves transmural infarcts of the colon wall, which leads to peritonitis, sepsis, perforation, and death. Another way to look at different types of IC is by anatomic location or distribution: segmental colitis or sidedness. A special entity is isolated right-sided colon ischemia (IRCI) as it is associated with poor outcomes. IRCI has 30-day mortality rate of 20.3% compared with 9.0% in those with non-IRCI, higher frequency of severe cases requiring surgical intervention (40.9% of patients compared with 10.3% in those with non-IRCI), and is associated with acute mesenteric ischemia. Pancolitis and IRCI may be seen frequently in patients with sepsis, and IRCI is associated more frequently in patients with coronary artery disease and chronic kidney disease on hemodialysis. 61% of episodes of IC that required surgical intervention had either IRCI or bilateral (pancolonic) patterns of ischemia.

PRESENTATION

The presentation of IC is often vague, and the diagnosis is often delayed while other differential diagnoses are ruled out. The most common symptoms of IC are acute onset abdominal pain, hematochezia, and an urgent desire to defecate. Gangrenous colitis is characterized by increasing abdominal tenderness, guarding, rebound tenderness, rising temperature, and paralytic ileus. The sudden onset of a toxic colitis with signs of peritonitis and a rapidly progressive course are typical of universal fulminant colitis, a rare variant of IC. Rectal bleeding is found more frequently in non-IRCI (69.9%) compared with IRCI (39.4%). One must have a high index of suspicion when patients present with severe acute abdominal pain without bleeding, especially if they have the common risk factors associated with IRCI. A timely diagnosis of IRCI is important for patient survival.

DIAGNOSIS

After a thorough history and physical examination looking for specific risk factors for IC, laboratory studies, imaging, and endoscopy might be used to reach the correct diagnosis.

Routine laboratory studies are unhelpful in reaching a diagnosis of IC but are important for prognostication and assessing severity. Leukocytosis is a frequent finding. Elevated lactate, urea, and creatinine are sometimes found. Some patients may present with metabolic acidosis and a base deficit in cases of severe ischemia, gangrene, and sepsis. Decreased hemoglobin levels, low serum albumin, and the presence of metabolic acidosis can be used to predict severity of IC.

Imaging usually involves a CT scan, but sometimes in entertaining other differential diagnoses, an abdominal x-ray is ordered. Classic findings on an x-ray include thumbprinting, which indicates mucosal edema. In cases of bowel perforation, an x-ray is a quick way to see free air under the diaphragm. The use of contrast enemas had previously been the diagnostic modality of choice, but has been replaced, for the most part, by CT imaging and colonoscopy.

CT is the most helpful modality in the initial assessment of the patient with abdominal pain. It can exclude other causes of abdominal pain, suggest a location and source of ischemia, and identify complications associated with more advanced disease ( Figs. 2 and 3 ). CT will frequently show bowel wall thickening, thumbprinting, and pericolonic stranding with or without ascites. After reperfusion, there may be evidence of submucosal edema or hemorrhage. Emboli or thrombi causing complete arterial occlusion are occasionally seen with corresponding thin, unenhancing colonic wall due to complete lack of reperfusion. CT findings of colonic pneumatosis and portomesenteric venous gas can be used to predict the presence of transmural colonic infarction but can also be present with other conditions like chronic obstructive pulmonary disease and infectious colitis. CT can also be diagnostic in severe cases, showing bowel perforation and free air in the peritoneal cavity. Of course this should be suspected on physical examination. In a patient in whom the presentation of IC may be a heralding sign of acute mesenteric ischemia (e.g., IRCI, severe pain without bleeding, and atrial fibrillation) and the multiphasic CT is negative for vascular occlusive disease, traditional splanchnic angiography should be considered for further assessment. Angiography is otherwise rarely helpful in diagnosing IC as most cases are caused by transient hypoperfusion.

FIG. 2, Contrast enhanced axial CT image demonstrating foci of gas in the portal veins (arrow). Peripherally branching air on CT helps differentiating portal venous gas from pneumobilia, which typically is more central. Significant bowel distention is also noted.

FIG. 3, A and B , Contrast enhanced axial CT image showing extensive dilatation and pneumatosis (arrow) of the small bowel. Pneumatosis was also seen in the cecum, corresponding to the superior mesenteric artery vascular tract. Mesenteric venous gas is noted as well (arrow with *).

The gold standard for confirming diagnosis of IC is endoscopy, where ischemic mucosa can be identified and biopsied for confirmation. Thus, early colonoscopy (within 48 h of presentation) should be performed in suspected IC cases to confirm the diagnosis. Typical findings of IC include segmental erythema, edema, mucosal ulceration, submucosal hemorrhagic nodules, and involvement of watershed areas. Occasionally, pseudomembranes related to mucosal sloughing are observed. The colonic single stripe sign, a single linear ulcer running longitudinally along the antimesenteric colonic wall, is associated with IC ( Fig. 4 ). After 48 hours, sloughing occurs, the purple submucosal hemorrhages dissipate, and ulcerations develop. In more severe ischemia with transmural infarction, the mucosa may appear gray-green or even black ( Fig. 5 ).

FIG. 4, Colonoscopic image of linear ulcerations placed along the longitudinal axis of the colon.

FIG. 5, Colonoscopic image showing cyanotic/black mucosal nodules with deep ulcerations until mucosal necrosis.

Endoscopy will also allow biopsies to be taken to further confirm diagnosis. On histologic examination, signs of IC include mucosa of normal architecture with necrosis and sloughing of the surface epithelium, loss of epithelium in the superficial aspects of the crypts (with or without ghosts of crypts), mucin depletion and reactive changes in the residual crypt epithelium with nuclear hyperchromasia and increased mitoses, paucity or complete absence of acute inflammatory cells, and the presence of hyalinosis in the lamina propria ( Fig. 6 ).

FIG. 6, Ischemic colitis, H&E, 100×. This image shows characteristic features of ischemic colitis, including superficial epithelial injury, crypts with atypia including nuclear hyperchromasia, and hyalinized lamina propria. The crypts appear closer together due to lamina propria collapse.

SEVERITY

IC is classified into three categories: mild, moderate, and severe. Most cases of IC are mild or moderate, and about 20% are severe at initial presentation.

The classification is based on clinical, laboratory, imaging, and endoscopic criteria. Management is driven accordingly. For mild and moderate disease, treatment is supportive care, which includes bowel rest, intravenous fluids, optimizing cardiac output, avoiding medications that could worsen colonic ischemia like vasopressors, correction of electrolytes and nasogastric tube decompression for ileus. For confirmed IC, intravenous antibiotics are used to treat bacterial translocation that occurs when the mucosal barrier is damaged. The antibiotic(s) selected should include both aerobic and anaerobic pathogens. You should not expect to cover highly resistant pathogens. The duration of antibiotic therapy needed has yet to be studied and determined, although some authors have recommended 2 weeks. However, a period covering the time of mucosal injury when translocation is theoretically occurring seems reasonable.

A heart rate of >100 beats per min and systolic blood pressure of <90 mm Hg at the time of diagnosis are associated with the need for surgical intervention and/or mortality as are Hgb <12 mg/dL, hyponatremia (Na <136 mEq/L), LDH >450 U/L, and blood urea nitrogen (>28 mg/dL). A pancolonic distribution of disease and IRCI also portend a poor outcome.

Patients who meet criteria for severe disease need a surgical evaluation and most often will require operative intervention. This was the initial management in 53.6% of those with IRCI versus 14.5% in those with non-IRCI.

Surgical intervention should be considered in the presence of IC accompanied by hypotension, tachycardia, and abdominal pain without rectal bleeding; for IRCI and pancolonic IC; and in the presence of gangrene.

SURGICAL APPROACH

Operative management of patients with IC is either in the acute setting or for treatment of the sequelae of colon ischemia. In the acute setting, operative indications include peritonitis on examination, massive bleeding, fulminant colitis, portal venous gas or pneumatosis on imaging with clinical picture of IC, or worsening clinical condition on medical management alone.

The general principle of surgery is to remove all segments of the colon that are grossly ischemic ( Fig. 7 ). Surgery is usually approached with a midline laparotomy; however, in certain circumstances, a diagnostic laparoscopy may be warranted. However, it can be difficult to determine the extent of resection as the serosa can be misleading as it will appear normal until transmural necrosis occurs. Therefore, a preoperative CT scan or endoscopic evaluation of the colon is important to establish the extent of bowel involvement. Intraoperatively, one may use Doppler ultrasonography, endoscopy, or photoplethysmography to assess mucosal viability or colonic blood flow. After adequate resection has occurred, in patients where there is concern for ongoing ischemia, it is not uncommon to leave the fascia open and bowel in discontinuity and take the patient to the intensive care unit for continued resuscitation for a planned second-look laparotomy and closure in 12 to 48 hours to make sure all necrotic tissue has been removed.

FIG. 7, Portion of intestine with ischemia (cecum, top right of image ). Compare with normal pink-tan mucosa (bottom right of image).

The decision to construct an anastomosis versus an ostomy depends on many factors. The main concern is anastomotic leak or nonhealing anastomosis. A primary anastomosis can be done safely in uncomplicated isolated right colon ischemia when the rest of the bowel appears very well perfused. However, for left-sided IC, published opinion advocates for an end colostomy and rectal stump (Hartmann procedure), which can be reversed later (i.e., no sooner than 6–8 weeks postsurgery). In selected cases, it may be reasonable even with left-sided disease to perform a primary anastomosis with or without a protective diverting loop ileostomy. With additional experience with preoperative localization of the extent of ischemia, less invasive surgical approaches may become more attractive.

POSTOPERATIVE COURSE

Overall, the postoperative course of patients undergoing surgery for acute IC is associated with high rates of morbidity and mortality. Two-thirds of patients with severe IC requiring surgery develop medical complications such as pneumonia, urinary tract infections, atrial fibrillation, postoperative myocardial infarction, acute renal failure, or the need for hemodialysis. There is a 37% in-hospital mortality rate for severe IC.

Independent risk factors of mortality after emergent colectomy for IC include elderly age, poor functional status, multiple comorbidities, low output heart failure (e.g., cardiac ejection fraction <20% on echocardiogram), preoperative septic shock, preoperative blood transfusions, preoperative acute renal failure, and delay from hospital admission to surgery. Postoperative death is associated with the peak preoperative lactate level (if above 2.5 mmol/L), amount of intraoperative blood loss, pre- and intraoperative catecholamine administration, subtotal or total colectomy, need for dialysis postoperatively, and an American Society of Anesthesiologists (ASA) class 4.

Operative intervention is sometimes required for sequelae of IC such as symptomatic colonic strictures or for ostomy reversal. Studies showed high rates of complications after ostomy reversal (i.e., longer length of stay, anastomotic leaks, and mortality). Other indications for a second operation include facial dehiscence and wound complications. Patients with nongangrenous IC sometimes present with abdominal pain, bloating, and obstructive symptoms weeks after their initial episode. If a stricture is suspected, either a colonoscopy or a contrast enema can identify it; the latter is helpful to describe the length and location of the stricture. In 10% of the cases, strictures develop as the colon heals. If clinically significant, causing obstruction or severe abdominal pain, strictures need operative intervention by elective resection and primary anastomosis. Alternatively, there are nonoperative methods to temporize symptoms when surgery is not feasible, such as balloon dilation, and in some instances, stenting.

CONCLUSION

The incidence of IC has increased during the last few decades. Acute IC continues to represent a very deadly disease associated with high rates of morbidity and mortality. IC in the community typically presents in older patients with multiple comorbidities. Patients who survive their initial episode may require additional intervention, and around 7% of patients will have another episode within 5 years. IC is challenging to diagnose due to its nonspecific presentation, but efforts should be directed to early consideration of this diagnosis when risk factors and clinical conditions warrant.

Suggested Readings

  • Brandt L.J., Feuerstadt P., Longstreth G.F., Boley S.J.: American College of Gastroenterology. ACG clinical guideline: epidemiology, risk factors, patterns of presentation, diagnosis, and management of colon ischemia (CI). Am J Gastroenterol 2015; 110: pp. 18-44. quiz 45
  • Castleberry A.W., Turley R.S., Hanna J.M., Hopkins T.J., Barbas A.S., Worni M., Mantyh C.R., Migaly J.: A 10-year longitudinal analysis of surgical management for acute ischemic colitis. J Gastrointest Surg 2013; 17: pp. 784-792.
  • Yadav S., Dave M., Edakkanambeth Varayil J., Harmsen W.S., Tremaine W.J., Zinsmeister A.R., Sweetser S.R., Melton L.J., Sandborn W.J., Loftus E.V.: A population-based study of incidence, risk factors, clinical spectrum, and outcomes of ischemic colitis. Clin Gastroenterol Hepatol 2015; 13: pp. 731-738. e1-6; quiz e41

Management of Clostridioides Difficile Colitis

Marylise Boutros, MD
Maria Abou Khalil, MD,
Steven D. Wexner, MD, PhD (Hon)

Clostridioides (formerly Clostridium ) difficile infection (CDI) is an important cause of community and nosocomial acquired diarrhea. It is associated with significant morbidity and mortality, especially amongst the immunosuppressed and elderly. This chapter will review the clinical presentation and treatment options for patients with CDI.

BACKGROUND

While C. difficile was first described as an important cause of antibiotic-associated colitis in 1978, it was not until the early 2000s that CDI was experienced as a threatening nosocomial problem due to an increase in disease frequency, severity, and mortality. This change was attributed in part to the emergence of hypervirulent strains, including NAP1/BI/027. Antibiotic stewardship programs and strict infection control measures allowed for early case identification and treatment, leading to a dampening of CDI incidence. Nonetheless, CDI remains an important cause of nosocomial acquired infection and an increasingly recognized cause of community acquired disease and transmission.

C. difficile is an anaerobic toxin-producing gram-positive bacillus that can exist in spore form (outside the colon) or vegetative forms. The bacteria produce two exotoxins (toxin A and toxin B) responsible for the colitis and diarrhea. Hypervirulent strains have been associated with significantly increased toxin production and subsequently a more severe clinical course. Ingestion of spores of C. difficile via a fecal-oral route is a common cause of transmissibility, and transmission is higher in patients with active disease compared with asymptomatic carriers.

Clinical Risk Factors for CDI

Several risk factors for contracting CDI have been identified. The most widely recognized and modifiable risk factor for CDI is antibiotic use; CDI can occur up to 3 months after antibiotic discontinuation. Antibiotics result in a disruption of the balanced normal colonic microbiota allowing pathogenic C. difficile bacteria to multiply and cause disease. While clindamycin was the first antibiotic to be associated with CDI, many other antibiotics have since been implicated. Antibiotic resistance patterns of C. difficile have also been found to be correlated to virulence. Another important risk factor for CDI is advanced age. Furthermore, immunosuppression, inflammatory bowel disease (IBD), gastrointestinal surgery, and length of hospitalization have all been associated with increased risk of CDI. Special attention needs to be paid to patients with IBD, as disease exacerbation can mimic C. difficile colitis. Although some epidemiologic studies have implicated gastric acid suppression as a risk factor, this association was not consistently found after adjusting for confounding factors and is most likely to be the result of patient illness and hospital length of stay.

CLINICAL PRESENTATION

Although some patients can be asymptomatic carriers of C. difficile , CDI usually has clinical manifestations that can range from mild colitis to fulminant severe disease with toxic megacolon. Patient factors and pathogen characteristics have been implicated in the severity of the clinical course. Increasing age, immunosuppression, and comorbidities are amongst the most reported predictors of severe disease presentation.

Different CDI severity classification systems exist; however, there is no universally accepted system. This limitation is likely because of the inability of one scoring system to accurately predict the clinical course, be simple enough to allow widespread point-of-care use, and allow for time-dependent variables to be adjusted as the patient’s clinical course evolves. In general, the existing CDI severity classifications include a combination of factors associated with increased morbidity and mortality. These factors include physical examination findings, signs of shock, and laboratory data. A useful disease classification system was published in 2017 by the Infectious Disease Society of America and Society for Healthcare Epidemiology of America (IDSA/SHEA) and is summarized in Table 1 . Thus, the evaluation of patients with CDI should include a history, physical examination including vitals, urine output, fluid requirements, as well as laboratory tests including complete blood count (with significant leukocytosis being a hallmark, but not a pathognomonic feature, of CDI), renal function tests, albumin, and lactate as a marker of end-organ perfusion.

TABLE 1
Disease Severity and Recommended Treatment
Disease Severity Clinical Characteristics Recommended Treatment
Nonsevere disease Leukocytosis, WBC ≤15,000 or serum creatinine <1.5 mg/dL Vancomycin 125 mg PO QID × 10d OR fidaxomicin 200 mg PO BID × 10d
Use metronidazole 500 mg PO TID if none of the above available
Severe disease Leukocytosis WBC ≥15,000 or serum creatinine >1.5 mg/dL Vancomycin 125 mg PO QID × 10d OR fidaxomicin 200 mg PO BID × 10d
Fulminant disease Hypotension/shock, ileus, or megacolon Vancomycin 500 mg QID PO/PT (consider adding rectal vancomycin if ileus) + IV metronidazole (500 mg q8h)
Recurrent disease Pulse-tapered vancomycin regimen or fidaxomicin regimen is recommended. Fidaxomicin, vancomycin with or without rifaximin, and fecal microbiota transplant (FMT) are used for second or subsequent recurrences.
With recurrences presenting with fulminant disease, in addition to the antibiotic regimen administered, some advocate for the addition of FMT.

DIAGNOSIS

Testing is recommended for patients with unexplained new onset diarrhea (3 stools/day) especially in the presence of risk factors for CDI. Patients should be placed on preemptive isolation while awaiting results. Hand hygiene with soap and water is superior to alcohol-based hand-hygiene products for elimination of C. difficile spores and is recommended in addition to contact precautions including gloves and gowns.

A variety of laboratory tests for C. difficile exist. They can detect either the organism itself or the toxins in the stools. To avoid false positive results, laboratory protocols recommend that only liquid or loose stool samples be tested. Stool toxin tests are often used as a multistep algorithm either with glutamate dehydrogenase (GDH) assay or nucleic acid amplification tests (NAAT) assay followed by toxin-recognition to help differentiate between asymptomatic carriers and patients with the disease. A summary of the commonly used tests is presented in Table 2 .

TABLE 2
Diagnostic Tests for C. difficile
Test Details Sensitivity Specificity Substance Detected
Selective anaerobic culture Rarely used
Takes a long time to finalize
Can be useful in patient with ileus (rectal swab)
High Low C. difficile vegetative cells or spores
Cell culture cytotoxicity assay Resource intensive and time consuming, not routinely done
Has been used as a gold standard test
High High Free toxins
Nucleic acid amplification tests (NAAT) Detects one or more genes specific to toxigenic strains
Capable of detecting asymptomatic carriers
High Low/Moderate C. difficile nucleic acid (toxic genes)
Enzyme immunoassay C. difficile glutamate dehydrogenase (GDH) GDH antigen is an essential enzyme produced by all C. difficile isolates, but cannot differentiate between toxigenic and nontoxigenic strains High Low C. difficile common antigen
Enzyme immunoassay for C. difficile toxins A and B Most strains produce toxins A and B although some strains produce toxin B only, but CDI due to strains producing toxin A alone has not been reported Low Moderate Free toxins

Endoscopy

Pseudomembranes are a characteristic endoscopic feature of CDI that appear as raised white and yellow plaques and consist of toxin-induced ulcers with inflammatory cells and mucous ( Fig. 1 ). Confirmatory endoscopy with flexible or rigid sigmoidoscopy is not necessary; however, it is often performed to exclude other causes of colitis such as cytomegalovirus infection, graft-versus-host disease, IBD exacerbation, or ischemic colitis. Endoscopic confirmation has also been used in cases where a decision on surgical management needs to be expediently made and the situation does not allow waiting for confirmatory laboratory testing.

FIG. 1, Endoscopic appearance of pseudomembranes.

Imaging

Abdominal imaging can aid in the diagnosis of colitis and identify complications requiring operative intervention. Rarely, complications such as free air may also be seen. Classically, plain films were used to assess for “thumb printing” secondary to submucosal edema or toxic megacolon with colonic distention. Nowadays, an infused computed tomography (CT) scan of the abdomen and pelvis is the preferred imaging modality to assess the degree, extent, and complications of the colitis. Typical findings in severe or fulminant disease include pancolitis with significant colonic thickening and ascites ( Fig. 2A ). If gastrointestinal contrast is given, it can be seen trapped between the edematous haustral folds (accordion sign) ( Fig. 2B ). Evidence of complications such as bowel perforation, toxic megacolon, and ischemia can also be seen on CT.

FIG. 2, Computed tomography scan showing fulminant Clostridioides difficile infection, with ( A ) and without ( B ) contrast.

MEDICAL TREATMENT

Given the role of antibiotics in the development of CDI and its potential recurrence, it is recommended to stop the inciting antibiotics as soon as possible. Early treatment for CDI while awaiting confirmatory tests should be initiated, especially in patients with severe or fulminant disease. Although probiotics can decrease the occurrence of CDI, they have no role in the treatment of the disease. Metronidazole was commonly used for treatment; however, vancomycin or fidaxomicin have been found to be superior to metronidazole and have largely replaced it. The recommended treatment regimen with these agents is based on disease severity and is outlined in Table 2 . For the first recurrence, a pulse-tapered vancomycin regimen or fidaxomicin regimen is recommended. Fidaxomicin, vancomycin with or without rifaximin, and fecal microbiota transplant (FMT) are used for second or subsequent recurrences.

With recurrences presenting with fulminant disease, in addition to the antibiotic regimen administered as outlined in Table 2 , some advocate for the addition of FMT.

Patients with severe colitis require special attention. They should have large bore intravenous access and accurate measurements of intake and output to guide aggressive fluid resuscitation with crystalloids. Acute kidney injury is a common complication of severe colitis and resuscitation is generally the initial approach. Patients should be kept fasting with complete bowel rest until their symptoms improve. We recommend serial clinical examinations of patients with severe disease including vital signs, abdominal examination, and laboratory investigations as the clinical course can evolve rapidly. Prompt recognition of complications associated with the colitis will allow early management and improve outcomes. A multidisciplinary approach including surgeons, intensive care and infectious disease specialists, may aid in the care of these patients.

SURGICAL MANAGEMENT

Although rare (0.3%–6.2% depending on the epidemic status), surgical management is reserved for patients with fulminant disease who fail to respond to medical management, worsen on treatment, or in whom complications related to the colitis arise including hemodynamic compromise, abdominal compartment syndrome, or colonic perforation ( Box 1 ).

BOX 1
Indications for Surgical Management

  • Colonic perforation

  • Full-thickness ischemia

  • Peritonitis, worsening abdominal exam despite adequate medical treatment

  • Abdominal compartment syndrome

  • Hemodynamic instability with ongoing or increasing need for vasopressor support

  • Need for intubation and mechanical ventilation

  • Worsening end-organ failure (especially renal failure)

In the absence of absolute indications for surgery, no clear guidelines exist to define failure of medical management or the optimal timing to intervene with surgery. Despite the morbidity associated with emergency surgery in critically ill patients, evidence from retrospective studies suggests that earlier time to surgical intervention with fulminant disease improves survival compared with continued medical management alone.

Total Abdominal Colectomy with End Ileostomy

An open total abdominal colectomy (TAC) with an end ileostomy remains the gold standard operation for patients with fulminant C. difficile colitis (FCDC) requiring surgery and is the operation to which all others are compared. Outcomes after segmental colectomies for the treatment of FCDC have been shown to be inferior to TAC, and thus segmental colectomy is not recommended even if the colitis appears to be limited to a part of the colon on CT imaging. Upon entry into the abdominal cavity, usually profuse ascites is encountered. The colon usually appears dilated and edematous but otherwise has a normal serosal surface, as this is a mucosal disease. The distal extent of the resection is at the rectosigmoid junction, where the rectum is transected and stapled; however, if the rectosigmoid appears too friable and there is heightened concern for rectal stump blowout, the surgeon can consider a more proximal transection to allow delivery of a distal stapled end of more proximal sigmoid as a mucous fistula. It is our preference to place a Malecot rectal catheter in the rectal stump for the first few postoperative days to allow decompression while healing. Postoperatively, patients should remain in a monitored setting until their vital functions are stable without the supportive measures offered in the intensive care unit and until their systemic inflammatory response syndrome (SIRS) starts improving. Postoperative length of stay is often prolonged by the patient’s comorbidities, and rapid recovery is limited by the severity of the multiorgan system involvement. After recovery, ileorectal anastomosis can be performed if the patient has returned to a functional baseline. This usually occurs starting at 3 to 6 months postoperatively. Given the significant postoperative morbidity associated with a colectomy for severe CDI, many patients do not reach a level of fitness suitable to endure another extensive operation for restoration of gastrointestinal continuity with an ileorectal anastomosis. In the literature, most patients who undergo a TAC do not eventually have restoration of gastrointestinal continuity.

Colonic Lavage and Diverting Loop Ileostomy

The creation of a diverting loop ileostomy (DLI) with colonic lavage has been advocated as a less-invasive alternative to TAC in this critically ill-patient population. In 2011, Neil et al. described this approach, which consists of the creation of a DLI with colonic lavage with warm polyethylene glycol solution and postoperative antegrade installation of vancomycin flashes by the ileostomy ( Fig. 3 ). In their single-institution, single-surgeon series, the authors compared 42 patients who underwent surgery for FCDC with 42 historical patients who had undergone TAC. The authors observed a decreased 30-day postoperative mortality for patients who underwent a DLI compared with TAC. In addition to the survival benefit, the authors demonstrated increased ileostomy closure rates at six months. A minority of patients in the DLI group required conversion to a TAC either due to the development of abdominal compartment syndrome or for failure of improvement. Since the first description of this novel procedure, few studies have compared it to TAC. There remains a lack of high-quality prospective data comparing these two operations given the difficulty in conducting such studies in the setting of a disease associated with high mortality in the emergency setting. A recent meta-analysis of 733 patients with DLI and 2950 patients with TAC found no difference in postoperative morbidity or mortality but found higher gastrointestinal restoration rates for patients with DLI.

FIG. 3, Example of set-up for diverting loop ileostomy and colonic lavage . (A) Foley catheter inserted into distal limb of the ileostomy, sutured to the rod. (B) Feeding access tubing used to administer vancomycin flushes.

Thus, while no recommendations can be made on which patients would best benefit from DLI vs. TAC, we believe patients with severe disease early in their disease course, who do not have any complications associated with their colitis necessitating a TAC (perforation, ischemia, or toxic megacolon), can be offered a DLI with colonic lavage. However, one must be cautious that patients need close follow-up as the diseased colon is still in situ, and thus there is a longer time to resolution of SIRS. Typically, patients may take up to a few days to a week in this SIRS state before improving. Tables 3 and 4 summarize the current evidence and the advantages and disadvantages of each operation.

TABLE 3
Comparison Between Total Abdominal Colectomy and Diverting Loop Ileostomy and Colonic Lavage
Pros Cons
Diverting loop ileostomy Less invasive operation and minimally invasive option
Higher gastrointestinal restoration rates
Improved postoperative morbidity compared with total abdominal colectomy (TAC)
Limited data supporting its use
Optimal patient population not defined
Patients may require reoperation in the event of failure
Slower resolve of systemic inflammatory response
Total abdominal colectomy Definitive management
Faster resolution of the systemic inflammatory response
High morbidity and mortality
Low gastrointestinal restoration rates

TABLE 4
Summary of Published Papers Mortality Following Total Abdominal Colectomy and Diverting Loop Ileostomy
Study Design and Location Study Interval Patient Population (n) 30-Day Postoperative Mortality
Neal et al. (2011) Prospective cohort, single center 2009–2011 DLI: 42
TAC: 42
DLI 19% vs. TAC 50% ( P = 0.006)
Fashandi et al. (2017) Retrospective, single center 2011–2015 DLI: 10
TAC: 13
DLI 30% vs. 23% 50%, ( P = 1)
Ferrada et al. (2017) Retrospective, multicentric 2010–2014 DLI: 21
TAC: 77
DLI 17.2% vs. TAC 39.7% ( P = 0.002)
Hall et al. (2018) Retrospective review of prospectively maintained database (ACS-NSQIP) 2011–2016 DLI: 47
TAC: 410
DLI 36% vs. TAC 31% ( P = 0.451)
Juo et al. (2019) Retrospective review of administrative database (NIS database) 2011–2015 DLI: 613
TAC: 2408
DLI 26.0% vs. TAC 31.1% ( P = 0.28)
*In-hospital mortality
ACS-NSQIP , American College of Surgeons National Surgical Quality Improvement Program; DLI , diverting loop ileostomy; NIS , Nationwide Inpatient Sample; TAC , Total abdominal colectomy.

Theoretically, the creation of a DLI is relatively simple; however, there are a few technical challenges associated with fulminant disease state and with performing the colonic lavage. We summarized the steps of the operation in Box 2 and at the following link https://www.youtube.com/watch?v=1VMQrEI6jro&t=26s . Furthermore, comorbidities such as elevated BMI may be challenging and may be overcome with the creation of an end loop ileostomy.

BOX 2
Technical Details and Tips

  • Exploratory laparotomy or diagnostic laparoscopy confirming diagnosis and ruling out contraindications to DLI (laparoscopy is preferable if patient is a candidate and the surgeon is comfortable with this approach).

  • Creation of a diverting loop ileostomy 20 cm from the ileocecal valve in a typical fashion (Brooke proximal limb, flush distal limb; Fig. 3A ).

  • Inserting a catheter, typically 18F Foley urinary catheter into the distal limb of the ileostomy, ideally positioned in the cecum ( Fig. 3B ).

  • Lavage of the colon is performed with 8L of polyethylene glycol (PEG) solution warmed to 37°C.

  • Foley catheter connected to the bag containing the PEG solution using urologic connection tubing (used in cystoscopy), allowing flow control of the PEG solution to the diseased colon.

  • PEG solution administered in increments, ensuring that effluent drainage is collected in the rectal tube.

  • If the procedure is performed laparoscopically, pneumoperitoneum can be maintained at 7–10 mm Hg during lavage. Laparoscopic bowel graspers may be used to aid in pushing the fluid along the colon.

  • If performed by a laparotomy, the abdomen is kept open, and the surgeon can manually aid the movement of the fluid through the colon.

  • If trouble is encountered getting fluid through the colon, the patient may be moved into the Trendelenburg/reverse Trendelenburg positions as well as left side up/down and right side up/down to move the fluid along the colon.

  • Rarely, flexure mobilization may be necessary.

  • Due to fluid sequestration in the diseased and atonic colon, an abdominal compartment syndrome (ACS) may occur during or after the operation and the surgeon should be aware of this possibility.

  • Abdominal drain can be left to remove excess ascites and potentially reduce the risk of ACS

  • Postoperatively, vancomycin flushes (500 mg in 500 mL of lactated Ringer’s) are delivered to the diseased colon through the Foley catheter that was left in the efferent limb of the ileostomy. The first vancomycin flush is given after completion of the PEG flushes, and administration should be continued every 8 h for 10 days or until the patient has clinically recovered ( Fig. 3A and B ).

BOX 2
Rome IV Criteria for Functional Constipation
From Lacy BE, Mearin F, Chang L, et al. Bowel disorders. Gastroenterology. 2016;150(6):1393–1407.

Requires two or more of the following:

  • Straining with more than 25% of defecations

  • Lumpy or hard stools (Bristol stool form scale 1 or 2) more than 25% of defecations

  • Sensation of incomplete evacuation more than 25% of defecations

  • Sensation of anorectal obstruction/blockage more than 25% of defecations

  • Manual maneuvers to facilitate more than 25% of defecations (such as digital evacuation or support of the pelvic floor)

  • Fewer than three spontaneous bowel movements per week

Plus:

  • Loose stools are rarely present without the use of laxatives

  • Insufficient criteria for irritable bowel syndrome

  • Criteria fulfilled for the last 3 months with symptom onset at least 6 months before diagnosis

SPECIAL POPULATION: PATIENTS WITH INFLAMMATORY BOWEL DISEASE

IBD, particularly ulcerative colitis, is a risk factor for the development of CDI. Patients with IBD are also more likely to need surgical intervention and have longer length of stay and increased mortality with CDI.

Given that CDI can mimic IBD flares, it should be considered in patients with worsening, smoldering, or relapsing IBD. While the treatment for CDI in patients with IBD is identical to patients without IBD, given the increased morbidity and mortality in this population, they should be monitored closely and have a lower threshold for early surgical intervention. It remains important to note that while CDI enteritis is a very rare event, it can occur after TAC with end ileostomy or following ileal pouch anal anastomosis, as a rare cause of pouchitis, and should be excluded in the event of a high-output ileostomy with SIRS.

CONCLUSION

CDI is a common cause of colitis especially in the surgical population. Most patients respond well to treatment with medical management alone. A small proportion of patients need intensive care unit admission to support the multisystem organ failure that can arise. Surgery may be required for the treatment of fulminant disease or to treat complications of the colitis needing urgent surgical attention. Although TAC is the standard operation, DLI and lavage has recently emerged as an alternative in select patients early in their disease.

Acknowledgments

The authors would like to acknowledge Dr. Rachel L. Choron and Dr. Pamela A. Lipsett for the previous version of this chapter.

Suggested Readings

  • Abou-Khalil M., Garfinkle R., Alqahtani M., Morin N., Vasilevsky C.A., Boutros M.: Diverting loop ileostomy versus total abdominal colectomy for Clostridioides difficile colitis: outcomes beyond the index admission. Surgical Endoscopy 2021; 35: pp. 3147-3153.
  • McDonald L.C., Gerding D.N., Johnson S., Bakken J.S., Carroll K.C., Coffin S.E., Wilcox M.H.: Clinical practice guidelines for Clostridioides difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clinical Infectious Diseases 2018; 66: pp. e1-e48.
  • Neal M.D., Alverdy J.C., Hall D.E., Simmons R.L., Zuckerbraun B.S.: Diverting loop ileostomy and colonic lavage: an alternative to total abdominal colectomy for the treatment of severe, complicated Clostridioides difficile associated disease. Annals of Surgery 2011; 254: pp. 423-429.
  • Poylin V., Hawkins A.T., Bhama A.R., Boutros M., Lightner A.L., Khanna S., Feingold D.L.: The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Management of Clostridioides difficile Infection. Diseases of the Colon & Rectum 2021; 64: pp. 650-668.

Management of Large Bowel Obstruction

Anthony J. Senagore, MD, MS, MBA,
Yesenia Rojas-Khalil, MD

Large bowel obstruction has traditionally been managed with urgent if not emergent surgery resulting inevitably in a laparotomy and stoma. However, in the past two decades with increasing use of endoscopy and experience with minimally invasive techniques, management can be more specifically tailored to the severity of obstruction and etiology. Nonetheless the management of large bowel obstruction requires prompt evaluation and diagnosis.

ETIOLOGY

The cause of large bowel obstruction can be categorized broadly as either mechanical or functional. Mechanical obstruction is more common than functional and often results from a neoplastic process. Mechanical obstructions can result from intrinsic or extrinsic processes, such as colorectal and ovarian cancer, respectively. Colorectal carcinoma is the most common etiology and accounts for nearly half of mechanical large bowel obstructions. Up to one-third of patients with colorectal cancer have a near or complete mechanical obstruction because of tumor burden, often representing an advanced stage.

Diverticular disease is the second most common cause of mechanical large bowel obstruction. Chronic inflammation in the sigmoid can cause a stricture in up to 17% of patients. This often results in a more chronic presentation of a large bowel obstruction. However, complicated diverticulitis with significant inflammation can also present as a large bowel obstruction due to the inflammatory stricture, phlegmon, or abscess. The third most common cause of large bowel obstruction is volvulus, and this occurs in 5% to 10% of all large bowel obstructions. Volvulus is a result of a long redundant segment of colon that has developed an axial rotation about its colonic mesentery. Because the blockage is not only of the colon lumen but also involves the mesentery, the risk of strangulation and ischemia is significantly higher that other causes of large bowel obstruction. The most common location for volvulus is the sigmoid colon (60%–70%), followed by the cecum (25%–40%), and very rarely the transverse colon (1%–4%).

Functional causes of large bowel obstruction are less common than mechanical obstructions. These include colonic pseudo-obstruction (Ogilvie’s syndrome), narcotic-induced adynamic ileus, and adynamic ileus due to systemic illness such as toxic megacolon from Clostridium difficile infection. Treating the underlying cause in functional large bowel obstruction often improves the symptoms.

CLINICAL PRESENTATION

Large bowel obstruction symptoms vary based on the acuity of the obstruction, resulting in a wide range of symptoms. Typically, patients will present with crampy abdominal pain, distension, and obstipation. Patients with an incompetent ileocecal valve will eventually present with nausea and emesis. Often, acute large bowel obstruction is associated with signs of hypovolemia and electrolyte imbalances secondary to fluid sequestration in the intestines as well as oral intolerance. Symptoms from chronic obstruction are typically mild and often associated with change in bowel habits, bloating, narrow caliber stools, and unintentional weight loss. This constellation of findings should raise suspicion for malignancy.

DIAGNOSIS

History and Physical Examination

Prompt evaluation and diagnosis beginning with a focused history and physical examination should occur in patients presenting with signs and symptoms of a large bowel obstruction. Obtaining a careful history of the development of symptoms can often cue surgeons to the most likely diagnosis. Patients may report a rapid or slow progression of their symptoms depending on the etiology. Recent orthopedic or gynecologic procedures are commonly associated with the development of colonic pseudo-obstruction. A family or personal history of colonic neoplasms, diverticular disease, or inflammatory bowel disease, as well as findings on previous colonoscopy (if performed) should be ascertained as this helps narrow the diagnosis. A review of current medications may indicate prescription medication-induced constipation but equally importantly can provide an overview of the patient’s overall underlying health status and comorbid conditions.

The physical examination classically reveals a tympanitic and distended abdomen. On occasion, a mass can be palpated through the abdominal wall in thin patients. Focal tenderness on examination may suggest ischemia or localized perforation, whereas diffuse tenderness is concerning for peritonitis. A digital rectal examination is an important part of the exam and should be performed on all patients to rule out fecal impaction, foreign body, or anorectal malignancy as the cause. In elderly patients and/or those with severe comorbid medical problems, it is important to ascertain goals of care, especially when advanced malignancy is suspected.

Imaging

Because of ease in accessibility, upright and supine abdominal radiographs are often the first images obtained. For the diagnosis of large bowel obstruction, plain films have a sensitivity and specificity of 84% and 72%, respectively. Importantly, they can help rule out an abdominal disaster such as pneumatosis coli or pneumoperitoneum, which in the right clinical context may obviate additional imaging. Cecal distension of 9 to 12 cm on plain abdominal film is concerning for impending perforation. In the setting of an incompetent ileocecal valve, reflux of contents back into the small intestine may reflect as air-fluid levels of the small bowel with distension. When colonic volvulus is the cause of large bowel obstruction, plain films can confirm the diagnosis. The classic findings of a sigmoid volvulus are seen in about two-thirds of patients and are commonly described as the “bent inner tube” sign or an “omega loop.” In the case of cecal volvulus, this classically presents as the “coffee bean” sign; however, it is only seen radiographically in less than 20% of the patients.

Contrast enemas (CEs) were once the gold standard in the diagnosis of large bowel obstruction. However, because these tests are labor intensive and require additional time, CEs are not readily accessible and are underutilized. CEs have a 96% sensitivity and a 98% specificity for diagnosing large bowel obstruction, compared to the 84% and 72%, respectively, of plain abdominal films. Classic finding is a “bird’s beak” deformity at the site of a volvulus or an “apple core lesion” at the site of an obstructing cancer. CE can also provide additional details such as the length, width, and tortuosity of the narrowing that may be helpful when intraluminal stents are being considered. In cases of colonic pseudo-obstruction, contrast will freely flow without evidence of transition point. Additionally, in the setting of fecal impaction, contrast may be therapeutic.

Computed tomography (CT) has become the preferred imaging modality in the evaluation of a patient with signs and symptoms of a large bowel obstruction. CT is much more readily accessible than CE and has largely supplanted CE as the diagnostic modality of choice. CT confirms the diagnosis of large bowel obstruction with almost 100% sensitivity and 90% specificity; it also allows multiplanar reconstruction that further facilitates the definitive etiology of large bowel obstruction. Finally, CT may have a substantial impact on the clinical management of a large bowel obstruction by demonstrating signs of intestinal ischemia, early pneumatosis, or pneumoperitoneum leading to earlier interventions.

TREATMENT

Initial Management

In concert with the initial history and physical, laboratory assessment should include serum chemistry and electrolytes, complete blood count, and lactate level. Hypovolemia and electrolyte imbalances are common and should prompt aggressive resuscitation with accurate monitoring of intake and output with a urinary catheter. Decompression with a nasogastric tube should be performed early. If surgery is a consideration, preoperative antibiotics and stoma marking should occur as an ostomy is frequently required.

Surgical Management

The optimal surgical management and approach, either laparoscopic or open, will vary according to the degree of obstruction, the underlying etiology of large bowel obstruction, as well as the clinical condition of the patient. Broadly, the surgical management of large bowel obstructions can be guided by the urgency of decompression, emergent or nonemergent.

Emergent Surgical Management

Patients who present with impending perforation on imaging, pneumoperitoneum (evidence of existing perforation), peritonitis, and/or with clinical signs of sepsis require emergent intervention. The best approach in this setting is a midline laparotomy that allows for decompression of the bowel, prompt identification of the lesion, and adequate exposure for a variety of procedures. Laparoscopic intervention may be difficult secondary to colonic distension and/or clinical instability. The primary goal of surgery in this setting is to decompress the large bowel as expeditiously as possible to prevent ischemia or perforation, if it has not already occurred. Ideally, this would entail treating the underlying etiology of the obstruction. When the obstructing lesion is resectable, the next decision is whether to restore intestinal continuity following resection. In general, under emergent setting where the patient is unstable or with gross intraabdominal contamination, primary anastomosis is contraindicated. For these patients, segmental resection with an end stoma is the safest choice.

For lesions on the right side, proximal to the splenic flexure, a right hemicolectomy with end ileostomy is recommended. For left-sided lesions, a Hartmann procedure should be performed. Introduced in 1923 by Henri Hartmann specifically for the management of large bowel obstruction, the Hartmann procedure involves resection of the distal obstruction and the formation of an end colostomy. To perform a Hartmann procedure, the patient should be placed in the lithotomy position, which greatly enhances pelvic dissection and allows for access to the rectum if necessary. After resection of the lesion, decompression of the colon is best achieved with an end colostomy. In a review of 35 studies by van de Wall and colleagues, of the 6249 patients who underwent a Hartmann procedure, only 44% underwent reversal with a mean time to reversal of 7.5 months. Morbidity rate was 3% to 50% (mean, 16.3%) and mortality rate from 0% to 7.1% (mean, 1%).

It is important to inspect the entire colon proximally to rule out any injury due to distension or synchronous pathology and, when feasible, to exteriorize the distal colon conduit as a mucus fistula or in the subcutaneous tissue at the inferior midline incision or the stoma site to facilitate reversal in the future. Given the emergent nature of the procedure, distinguishing between malignancy and inflammation can be difficult; however, attempts should be made to provide an oncologic resection when feasible.

Conversion from segmental resection to subtotal colectomy is rarely needed and determined by the integrity of the colon proximal to the obstruction. A subtotal colectomy is indicated when perforation, large serosal injuries, or synchronous lesions are found at the time of exploration. When performing a subtotal colectomy in the emergent setting, creation of an end ileostomy is often necessary.

Less commonly, there are circumstances in which addressing the underlying cause is not feasible. In instances of unresectable malignancy, diffuse carcinomatosis, severe inflammation, or in patients who are extremely unstable, a diverting loop colostomy proximal to the obstruction is indicated. A loop colostomy is preferred over an end colostomy as the blind end that is left in the abdomen may perforate. A colostomy relieves patients of their symptoms and limits subsequent concerns for an anastomotic leak. However, colostomies can be associated with significant morbidity, including high rates of parastomal hernia (50%), decreased quality of life, and low rates of stoma closure.

Nonemergent Surgical Management

In the absence of generalized peritonitis, perforation, or sepsis, the nonemergent management of a stable patient allows for more surgical treatment options. The surgical technique will depend on the skill of the surgeon as well as available hospital resources.

In patients with a right-sided obstruction and the point of obstruction is proximal to the splenic flexure, urgent right hemicolectomy with primary anastomosis should be considered and can often be performed laparoscopically in a stable patient with limited bowel distension. This procedure is associated with low anastomotic leak rates (<5%) in a hemodynamically stable patient who has not undergone a bowel preparation. An alternative is performing a right hemicolectomy with primary anastomosis and proximal diverting loop ileostomy . This can be considered in patients who may have associated small bowel dilation or who are immunosuppressed, on steroids, or malnourished from chronic obstruction where concern for anastomotic leak is higher or where a leak would preclude or delay further treatment such as chemotherapy in the setting of cancer. Additionally, reversal of a loop ileostomy is a less morbid surgery than reversal of an end ileostomy.

There is more debate regarding the preferred surgical procedure for an obstructing lesion arising in the descending sigmoid colon, rectum, or anus. Traditionally, primary anastomosis at the initial procedure was avoided because of the higher rates of anastomotic leak (20%). Currently, segmental resection with primary anastomosis is a good option for carefully selected patients if the proximal colon is not dilated significantly. Retrospective reviews have shown similar rates of operative mortality and anastomotic leak in carefully selected patients undergoing left-sided colectomy compared with right-sided colectomy. Furthermore, quality of life is improved. A good option for a high-risk patient is segmental colectomy, anastomosis, and a diverting loop ileostomy . This still allows for diversion of the fecal stream, and in the event of an anastomotic leak, intraabdominal sepsis is contained and can usually be managed nonoperatively with percutaneous drainage and antibiotics. Loop ileostomy reversal is a less invasive operation and patients will more likely undergo this procedure to have their intestinal continuity restored as compared to reversing a Hartmann’s procedure. The disadvantage is that ileostomy management in some patients (particularly the elderly) may be challenging with fluid and electrolyte shifts.

Colonic Lavage

In select patients, the use of an on-table colonic lavage may permit a single-stage surgery in the setting of a left-sided large bowel obstruction. Following resection of the lesion, the colon should be fully mobilized at both hepatic and splenic flexures to facilitate irrigation and allow descending colon to extend beyond the abdominal cavity. Next, an appendectomy is performed, and a catheter is passed into the cecum and secured with a purse-string suture. This catheter is attached to a large bag of warmed saline and elevated on a pole. If the cecum is thinned, an alternative is to insert the catheter into the terminal ileum and secure with a purse-string suture. The staple line at the distal segment of colon is opened and a generous length of sterile corrugated tubing is placed into the lumen of the descending colon. The tubing is secured in place with umbilical tape or a purse string and then draped off the table into a basin. Three to six liters of warmed saline is then flushed through the colon. This is continued until the effluent clears ( Fig. 1 ). The catheter and tubing are removed, and the site of the appendectomy is closed. Before the anastomosis, the colon is inspected for injuries that may have occurred during the lavage. The anastomosis is then performed in the standard fashion, either end-to-end or side-to-side if there is a large bowel size discrepancy. A modified version of this method uses a Y-shaped connector ( Fig. 2 ). One limb allows for retrograde infusion of saline through the descending colon and the other is attached to drainage tubing. This modification is appealing as it obviates the need for the appendectomy or cecostomy. In a study by Jung and colleagues, 171 patients with an obstructing left-sided lesion received intraoperative colonic lavage with primary anastomosis. Compared with elective, nonobstructed patients during the same interval, anastomotic leakage and wound infection were not significantly different.

FIG. 1, On-table colonic lavage.

FIG. 2, Alternative colonic lavage using a Y-shaped connector.

Endoscopic Stenting

In the past decade, endoscopic management of obstructing large bowel lesions has gained significant attention. Much of the literature is on stenting of obstructing left-side lesions, and although reports of right-sided lesions exist, the indication is less clear. In the appropriately selected patient, endoscopic stents can be tremendously beneficial. Indications for stent placement include palliation of an inoperable obstructing lesion (e.g., stage IV colorectal cancer) and as a temporizing “bridge” to definitive therapy in a patient with a curable or potentially curable lesion. The ability to stent a curable or potentially curable lesion affords time for colonic decompression, medical optimization, endoscopic evaluation for synchronous lesions, and increases the likelihood of a single-stage operation. Contraindications to stent placement include any of the previously discussed indications for emergent surgery, an abscess/infection closely associated with the lesion, short and tethered colon, and a lesion less than 5 cm from the anal verge.

Perioperative antibiotic coverage is unnecessary for stent placement. Gentle tap water enemas should be used to evacuate stool distal to the lesion. If an oral bowel preparation is desired, it may be attempted only if the obstruction is partial and the patient is closely monitored for the development of new or worsening symptoms. Colonoscopic evaluation of the lesion must be done with the utmost care; forceful attempts to traverse beyond the lesion are strictly avoided. Perforation is the most feared complication of stent placement. To mitigate the procedural risk of perforation, insufflation with carbon dioxide is preferred over room air because of the dramatically faster clearance time. The most common stents used in the United States are uncovered, self-expanding metal stents (SEM). Through-the-scope (TTS) and over-the-wire, also referred to as non-TTS, are the two conventional approaches to stent placement. TTS is usually the first-line approach ( Fig. 3 ). The TTS system requires a colonoscope equipped with a working channel to pass the guidewire and stent. The scope is advanced and parked in an area immediately distal to the lesion. Contrast is administered and the lesion assessed with fluoroscopy. The guidewire is then advanced safely beyond the lesion followed by the stent (Seldinger’s technique). Endoscopy and fluoroscopy confirm the correct location of the wire and the stent. Most TTS systems encourage a proximal and distal stent overlap of 2 cm. For longer lesions, additional stents can be placed in series. If assessment of the lesion shows anatomy unfavorable for TTS (e.g., available size of the stents), the procedure is converted to over-the-wire. With the guidewire fixed, the colonoscope is withdrawn entirely. The appropriate stent is selected and passed directly over the wire into position using fluoroscopy. Fluoroscopic or postprocedural abdominal films are obtained to confirm the typical stent appearance of a narrowed or waisted midportion with proximal and distal flaring. The flaring represents adequate overlap into normal, low-resistance lumen. To reduce development of rapid restenosis, the intraluminal diameter at the midportion of the stent should be at least 24 mm. Balloon dilation should not be attempted because of significant risk of perforation. If imaging does not demonstrate flaring at one or both ends of the stent, additional stenting is likely necessary and typically done in an end-to-end fashion. Patients should expect gradual improvement of symptoms over 3 to 5 days as the stents reach maximal expansion and the colon decompresses. For descending colon and beyond, patients are advised to consume a low-residue, low-fiber diet along with daily laxatives (e.g., polyethylene glycol) to promote soft stools that are unlikely to become impacted at the stent. It is important that if definitive therapy is planned, it should be done within 7 to 14 days following placement of the stent. Patients who receive stenting for palliation and the anticipated duration greater than 2 weeks have a significantly increased risk of stent complications, primarily perforation. Multidisciplinary discussion should weigh the possibility of surgical palliation in this high-risk population. Current literature reports successful stent placement for acute large bowel obstruction between 70% and 90%. In a pooled analysis by Sebastian et al., 25% of the 1198 patients who underwent stent placement for obstruction developed a complication. Most common complications include stent migration, reobstruction, and perforation. The most serious complication is perforation and occurs in 4% to 5% of patients. Intrinsic factors that may increase the risk of stent-related perforation include longer segment of obstruction (median length of 64 mm), benign etiology, and extraluminal origin.

FIG. 3, Endoscopic stent placement for obstructing rectosigmoid cancer. (A) Arrow indicates obstructing pelvic lesion. (B) Fluoroscopic-guided passage of the wire. Note the obstructing lesion. (C) Poststent abdominal film with improvement of large bowel distension.

SPECIFIC SITUATIONS

Sigmoid Volvulus

Volvulus, from the Latin, volvere , means to roll. The classic patient with a sigmoid volvulus presents with colicky lower abdominal pain and obstipation. The demographic profile most affected by sigmoid volvulus is a black male older than 70 years with significant comorbidities. The two primary goals for management of sigmoid volvulus are to relieve the obstruction and prevent recurrence. The gold standard in the management of uncomplicated sigmoid volvulus is endoscopic detorsion, gross inspection to ensure viability of the colon, and placement of a drainage catheter (successful in up to 70%–90% of patients). Endoscopic reduction converts an emergency situation into an elective one and allows preparation of bowel and patient prior to definitive surgery. Failure of endoscopic reduction and/or presence of bowel ischemia is an indication for emergent resection. Primary anastomosis in this setting is an ongoing controversy and dependent of clinical picture of the patient. When endoscopic reduction is successful, definitive elective operation is recommended based on the high rate of recurrence; most studies cite a rate in the 50% to 80% range. Atamanalp and colleagues published data from a single-center review of 686 patients. They found morbidity and mortality of 35% and 16% for emergent surgery for recurrent sigmoid volvulus and 12.5% and 0% for elective surgery, respectively. Some suggest performing the definitive surgery as early as the index admission. Regardless of timing, the approach can be either open or minimally invasive. The redundant colon and narrowed mesentery leading to sigmoid volvulus results in an anatomical configuration that facilitates sigmoid resection with minimal difficulty. The gold standard is resection with primary anastomosis. For isolated sigmoid volvulus and the elective sigmoid colectomy, the recurrence rates are very low.

Cecal Volvulus or Bascule

The second most common site of colonic volvulus is the cecum ( Fig. 4 ). In patients with cecal volvulus, there is a female predominance, and typically patients are in the second and third decade of life, younger than those presenting with sigmoid volvulus. Presentation is similar to small bowel obstruction with abdominal distension, nausea, and vomiting; however, this can vary. There are three types of cecal volvulus:

  • 1

    Axial cecal volvulus: A twist of the intestines in which the affected cecum remains in the right lower quadrant.

  • 2

    Loop cecal volvulus: The cecum and terminal ileum are twisted in the axial plane and the affected cecum typically lies in the left upper quadrant.

  • 3

    Cecal bascule: A redundant cecum folds in the sagittal plane onto itself and the proximal ascending colon leaving the affected cecum within the right upper quadrant.

FIG. 4, Types of cecal volvulus. (A) Type 1, axial cecal volvulus. (B) Type 2, loop cecal volvulus. (C) Type 3, cecal bascule.

All three types necessitate either an acquired or congenital hypermobile cecum and ascending colon. Types 1 and 2 are the most common, accounting for roughly 80% of cecal volvuli. Unlike types 1 and 2, type 3 does not exhibit torsion. Patients who present with type 3 typically have a much more insidious presentation. Endoscopic management is not recommended for cecal volvulus or bascule because of a low rate of successful detorsion, and colonic ischemia can be missed in up to 25% of patients. For the patient presenting with a high probability of cecal volvulus, the operation of choice is an exploratory laparotomy. Severe ischemia, necrosis, or perforation occurs in up to 44% of patients with cecal volvulus; if present, mortality rates are equally as high. Management of the patient with nonviable cecum is resection and creation of end ileostomy, with consideration for mucous fistula. If the cecum is viable or with patchy ischemia, there is less consensus on the appropriate management. Nonresectional options include detorsion with cecopexy and cecostomy. Widely variable rates of recurrence, morbidity, and mortality have been demonstrated, although there is a paucity of recent data that compares these options and with respect to viable versus nonviable bowel. Distillation of the current data favors ileocecectomy with primary anastomosis for patients with acute cecal volvulus, even when bowel appears viable. Laparoscopic approach is an acceptable option in stable patients with limited bowel distension if performed by an experienced surgeon.

Acute Colonic Pseudo-Obstruction

Acute colonic pseudo-obstruction (ACPO) is also referred to as Ogilvie’s syndrome in homage to British surgeon Sir Heneage Ogilvie who in 1948 identified two patients who presented with large bowel distension without an identifiable lesion. This syndrome is most encountered in the hospitalized, elderly male patient. Hospitalization is notably unrelated to the presenting symptoms rather for unrelated surgical procedure (most commonly involving pelvic dissection) or traumatic bony injury. Additional predisposing factors include severe electrolyte disturbances, certain medications, and acute deconditioning. Presentation is consistent with other etiologies of large bowel obstruction, including nausea, vomiting, and persistent abdominal pain. A significant portion of these patients will continue to pass loose stool. Focal peritonitis confined to the right lower quadrant may be a warning sign of impending perforation because of the profound cecal distension. These findings should encourage expeditious intervention. Perforation at the time of presentation occurs in about 15% of patients and often is associated with abdominal tenderness, cecal diameter greater than 12 cm, fever, and leukocytosis. Abdominal plain films are the best initial imaging study, although findings are nonspecific. Second-line imaging has traditionally been with water-soluble CE. Unfamiliarity with the technique coupled with the risk of perforation makes this option less favorable. The gold standard imaging modality is CT; findings include diffuse colonic distension, possible transition point near splenic flexure, and, importantly, no evidence of a mechanical obstruction. Nonoperative management of ACPO is successful in more than two-thirds of patients. This consists of aggressive fluid resuscitation, repletion of serum electrolytes as necessary, cessation of possible offending medications (e.g., opioids, anticholinergics), and bowel rest. The use of nasogastric and rectal tubes for decompression may also encourage the passage of flatus. These principles of nonoperative management are continued for up to 72 hours. During this period, it is essential to monitor the patient with the use of serial imaging and physical examination.

Failure to improve over this period necessitates the next step in the algorithm, neostigmine. Neostigmine increases the availability of acetylcholine through its antiacetylcholinesterase mechanism. In the colon, additional acetylcholine results in improved contractility and generalized motility. Before administration, the patient should be transferred to a unit with continuous cardiopulmonary monitoring and the ability to administer atropine, glycopyrrolate, or both. Neostigmine is contraindicated in patients in whom there is suspicion for ischemia or perforation, severe acute bronchospasm, poorly controlled cardiac dysrhythmia, or pregnancy. With the patient connected to continuous monitoring, the first dose of neostigmine of 2 to 5 mg is given intravenously over 1 to 5 minutes. Success is achieved with passage of flatus, stool, or decreased abdominal distension. Patients are observed over the next 80 minutes. Atropine and glycopyrrolate should be readily available if there is development of bradycardia or bronchospasm, respectively. Neostigmine is successful at treating ACPO in more than 90% of cases. For patients who are partial responders or nonresponders after one dose, a second can be administered with high rates of success. For those who fail to respond to neostigmine, the next step in management is endoscopic decompression. Geller et al. reviewed 50 patients with ACPO treated with colonoscopic decompression and found clinical success in 95% following a single intervention, 18% needed at least one additional colonoscopy; for patients who did not receive a decompression tube at the time of the procedure, clinical success was only achieved in 25%. Regarding technical considerations, insufflation should be minimized and preferably with carbon dioxide. The scope should be advanced into the right colon. A decompression tube is strongly recommended and should originate from the right colon; however, randomized controlled trial data are lacking to support the use of decompression tubes. Evidence does, however, support the use of polyethylene glycol solution after endoscopy and neostigmine to lower recurrence rate. The endoscopist should remove as much gas as possible during withdrawal of the scope. If endoscopic decompression is not possible or unsuccessful, percutaneous cecostomy can be used with variable success. When all these interventions have been unsuccessful, operative management is indicated. The surgery is dictated by the intraoperative findings. A minilaparotomy may be enough to assess the viability of the colon. If there is no evidence of perforation or ischemia, a tube cecostomy or surgical cecostomy may be sufficient. When ischemia is identified, the affected region should be resected, including subtotal colectomy if indicated. Primary anastomosis is not recommended but rather the creation of end colostomy with or without mucous fistula.

SUMMARY

Large bowel obstruction is a serious disorder that often necessitates surgical intervention. Rapid evaluation is essential to providing appropriate management. The approach and type of surgery will be largely dependent on the clinical condition of the patient and surgeon comfort. Emergent laparotomy should be performed in unstable patients with peritonitis and/or signs of symptoms of ischemia. Segmental resection with primary anastomosis should be considered in patients who are stable and have minimal barriers to healing. Endoscopic stenting can be considered as a bridge to surgery or as palliation in select patients.

Suggested Readings

  • Atamanalp S.S.: Sigmoid volvulus: diagnosis in 938 patients over 45.5 years. Tech Coloproctol 2013; 17: pp. 419.
  • Fiori E., Lamazza A., Schillaci A., et. al.: Palliative management for patients with subacute obstruction and stage IV unresectable rectosigmoid cancer: colostomy versus endoscopic stenting: final results of a prospective randomized trial. Am J Surg 2012; 204: pp. 321.
  • Nag H.J., Yule M., Twoon M., Binnie N.B., Aly E.H.: Current outcomes of emergency large bowel surgery. Ann R Coll Surg Engl 2015; 97: pp. 151-156.
  • Sebastian S., Johnston S., Geoghegan T., et. al.: Pooled analysis of the efficacy and safety of self-expanding metal stenting in malignant colorectal obstruction. Am J Gastroenterol 2004; 99: pp. 2051.
  • SCOTIA Study Group: Single-stage treatment for malignant left-sided colonic obstruction: a prospective randomized clinical trial comparing subtotal colectomy with segmental resection following intraoperative irrigation. The Subtotal Colectomy versus On-table Irrigation and Anastomosis. Br J Surg 1995; 82: pp. 1622.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here