Archives for category: Section 5 Gastrointestinal Tract and Abdomen

 


David E. Beck, M.D., F.A.C.S., F.A.S.C.R.S.

Chairman, Department of Colon and Rectal Surgery
Ochsner Clinic Foundation, New Orleans

Figure 1. Anatomy of anal canal

In this chapter, I briefly review the evaluation and management of common benign conditions affecting the rectum, the anus, and the perineum: hemorrhoidal disease, anal fissures, anorectal abscesses and fistulas, pilonidal disease, hidradenitis suppurativa, pruritus ani, and solitary rectal ulcer syndrome (SRUS). For proper diagnosis and treatment of these conditions, an understanding of the relevant anatomy is essential [see Figure 1].1

The dentate line divides the rectal mucosa, which is generally insensate and is lined with columnar mucosa, from the anoderm below it, which is highly sensitive (because of the somatic innervation provided by the inferior hemorrhoidal nerve) and is lined with modified squamous mucosa. The anal canal is surrounded by two muscles. The internal anal sphincter, innervated by the autonomic nervous system, maintains resting anal tone and is under involuntary control. The external sphincter, innervated by somatic nerve fibers, generates the voluntary anal squeeze and plays the key role in maintaining anal continence. The area surrounding the anorectum is divided into four spaces—perianal, ischioanal, supralevator, and intersphincteric (intermuscular)—familiarity with which is particularly important in the evaluation of perirectal abscesses and fistulas.

Hemorrhoids


Hemorrhoids are fibromuscular cushions that line the anal canal. They are classically found in three locations: right anterior, right posterior, and left lateral.2,3 On occasion, smaller secondary cushions may be found lying between these main cushions. Contrary to popular belief, hemorrhoids are not related to the superior hemorrhoidal artery and vein, to the portal vein, or to portal hypertension.4 In fact, hemorrhoids are part of the normal anal anatomy. Their engorgement during straining or performance of the Valsalva maneuver is a component of the normal mechanism of fecal continence: it most likely completes the occlusion of the anal canal and prevents stool loss associated with nondefecatory straining. In the medical literature, however, the term hemorrhoid is used almost exclusively to refer to pathologic hemorrhoids, and I follow this usage throughout the remainder of the chapter.

Figure 2. External and internal hemorrhoids

Hemorrhoids are broadly classified as either internal or external. Internal hemorrhoids are found proximal to the dentate line, whereas external hemorrhoids occur distally [see Figure 2]. External hemorrhoids are redundant folds of perianal skin that generally derive from previous anal swelling; they remain asymptomatic unless they are thrombosed and are treated entirely differently from internal hemorrhoids.

Internal

Clinical Evaluation

Internal hemorrhoidal disease is manifested by two main symptoms: painless bleeding and protrusion.4 Pain is rare because internal hemorrhoids originate above the dentate line in the insensate rectal mucosa. The most popular etiologic theory states that hemorrhoids result from chronic straining at defecation (with upright posture and heavy lifting possibly playing contributing roles as well). This straining not only causes hemorrhoidal engorgement but also generates forces that reduce the attachment between the hemorrhoids and the anal muscular wall. Continued straining causes further engorgement and bleeding, as well as hemorrhoidal prolapse. Internal hemorrhoids are classified into four grades on the basis of clinical findings and symptoms. Grade 1 represents bleeding without prolapse; grade 2, prolapse that spontaneously reduces; grade 3, prolapse necessitating manual reduction; and grade 4, irreducible prolapse.

Questioning often reveals a long history of constipation and straining at defecation. Many patients with internal hemorrhoids are extensive bathroom readers, spending many hours in the bathroom each week. Symptoms start with painless bleeding and may progress to anal protrusion. The physical examination begins with visual inspection and may reveal prolapsing hemorrhoidal tissue appearing as a rosette of three distinct pink-purple hemorrhoidal groups. Hemorrhoidal prolapse must be distinguished from true full-thickness rectal prolapse. When hemorrhoidal prolapse is not present, anoscopy typically reveals redundant anorectal mucosa just proximal to the dentate line in the classic locations.

Surgical treatment of hemorrhoids in a patient whose main disease process is Crohn disease, a pelvic floor abnormality, or fissure disease invariably yields imperfect results. It is especially important to recognize the anal pain and spasm associated with anal fissure because in patients with this problem, excision of the hemorrhoids without concomitant management of the fissure leads to increased postoperative pain and poor wound healing. Not uncommonly, patients are treated for hemorrhoids when the true primary condition is fissure disease.

Management

Treatment of symptomatic internal hemorrhoids ranges from simple reassurance to operative hemorrhoid excision, depending on hemorrhoid grade [see Table 1]. Therapies may be classified into three categories: (1) diet and lifestyle modifications, (2) nonoperative and office procedures, and (3) operative hemorrhoidectomies.

Diet and lifestyle modifications For all patients with grade 1 or 2 hemorrhoids and most patients with grade 3 hemorrhoids, treatment should begin with efforts to correct constipation. Recommendations should include a high-fiber diet, liberal water intake (six to eight 8 oz glasses of water daily), and fiber supplements (e.g., psyllium, methylcellulose, calcium polycarbophyl, and gum). Sitz baths are recommended for their soothing effect and their ability to relax the anal sphincter muscles. Topical creams may reduce some of the associated symptoms, though they do not affect the hemorrhoids themselves. Suppositories are not helpful, because they deliver medication to the rectum, not the anus. Patients are instructed to avoid prolonged trips to the bathroom and to read in the bathroom only when sitting atop the toilet lid.

Nonoperative and office proceduresIf initial diet and lifestyle modifications are not effective, there are a number of nonoperative therapies that may be tried next. At present, the main recommended options are rubber band ligation, infrared photocoagulation, and sclerotherapy.

Rubber band ligation. Ligation of hemorrhoids with elastic bands is the method most commonly used in the outpatient setting [see 5:37 Anal Procedures for Benign Disease].5 It is successful in two thirds to three quarters of all patients with grade 1 or 2 hemorrhoids.2 Repeated banding may be necessary to resolve all symptoms. On rare occasions, patients do not respond to banding at all or cannot tolerate it. Some of these patients will respond to infrared coagulation; others may require formal hemorrhoidectomy.

Complications of rubber band ligation include bleeding, pain, thrombosis, and life-threatening perineal sepsis.5,6 The cardinal signs of perineal sepsis are significant pain, fever, and difficult urination. Patients in whom any of these symptoms develop require urgent evaluation and treatment with broad-spectrum antibiotics, coupled with selective debridement of any necrotic anal tissue.

Figure 3. Use of infrared coagulator

Infrared coagulation. The infrared coagulator generates infrared radiation, which coagulates tissue protein and evaporates water from cells.7 The extent of the tissue destruction depends on the intensity and duration of the application. An anoscopic examination is performed, and the infrared coagulator is applied to the apex of each hemorrhoid at the top of the anal canal [see Figure 3]. Each hemorrhoid bundle receives three to four applications, each lasting 1 to 1.5 seconds.

The infrared coagulator was designed to decrease blood flow to the region. It is not particularly effective for treating large amounts of prolapsing tissue; it is most useful for treating grade 1 and small grade 2 hemorrhoids. Overall, it is slightly less painful than rubber banding. Infrared coagulation is especially beneficial for patients in whom rubber band ligation fails because of pain or who have symptomatic internal hemorrhoids that are too small to band.

Sclerotherapy. Sclerotherapy was once commonly employed to treat internal hemorrhoids, but with the advent of rubber band ligation, it has become less popular. The technique involves injection of a sclerosant into the anorectal submucosa to decrease vascularity and increase fibrosis. The agents used include phenol in oil, sodium morrhuate, and quinine urea.2 Hemorrhoids are identified via anoscopy, and the sclerosant is infiltrated at the apex of the hemorrhoid (at the proximal anal rectal ring). After injection, patients occasionally experience a dull ache lasting 24 to 48 hours, but substantial bleeding and other significant complications are uncommon. Misplacement of the sclerosant, resulting in significant perianal infection and fibrosis, has been reported, albeit rarely.

Comparison of methods. A meta-analysis comparing a variety of modalities used to treat hemorrhoids found that rubber band ligation was superior to sclerotherapy for grade 1 through grade 3 hemorrhoids.8 Patients who were treated with sclerotherapy or infrared coagulation were more likely to require further treatment than those treated with rubber band ligation. The authors concluded that hemorrhoidectomy should be reserved for patients in whom rubber band ligation or infrared coagulation fails or who have associated external hemorrhoidal disease.

 

Hemorrhoidectomies Various hemorrhoidectomies have been developed throughout the years. Although they differ with respect to detail, they incorporate similar basic principles—namely, reduction of blood flow to the anorectal ring, removal of redundant hemorrhoidal tissue, and fixation of redundant mucosa and anoderm.9

Open and closed hemorrhoidectomy. In the United Kingdom, open (Milligan and Morgan) hemorrhoidectomy, in which hemorrhoids are ligated and excised with the wound left open, is the most commonly performed operative excision procedure. In the United States, closed (Ferguson) hemorrhoidectomy is most commonly performed10; this procedure is described more fully elsewhere [see 5:37 Anal Procedures for Benign Disease]. With either type of hemorrhoidectomy, one, two, or three hemorrhoidal bundles may be excised. In performing a closed hemorrhoidectomy, it is essential to excise as little of the anoderm as possible: if large amounts are excised, closing the anal wounds or secondary healing can result in significant postoperative pain and perhaps even long-term anal stenosis.

Stapled hemorrhoidectomy. Stapled hemorrhoidectomy or anoplasty (commonly known as the procedure for prolapsing hemorrhoids [PPH]) is increasingly being performed as an alternative to standard open or closed hemorrhoidectomy with the aim of reducing the pain associated with traditional surgical techniques.9 PPH involves transanal circular stapling of redundant anorectal mucosa with a modified circular stapling instrument (PPH-01, Ethicon Endo-Surgery Inc., Cincinnati, Ohio).11 Because no incisions are made in the somatically innervated, highly sensitive anoderm, there should, theoretically, be significantly less postoperative pain.

Randomized, prospective trials comparing PPH with various operative hemorrhoidectomies and other therapies found it to be associated with significantly less pain then conventional treatments and to have similar complication rates.9 PPH may, however, have a greater potential for disastrous complications (e.g., rectovaginal or rectourethral fistula from inclusion of too much tissue within the purse-string suture, perforation caused by placing the purse-string too high, and incontinence caused by placing the purse-string too low or too deep). Bleeding also remains a problem with PPH, and leaks have been reported. Finally, stapled hemorrhoidectomy has not yet been adequately compared with office procedures for grade 1 and 2 hemorrhoids and thus should not replace these techniques for treatment of minimally symptomatic hemorrhoidal disease.

 

External

Clinical Evaluation

External hemorrhoids are asymptomatic except when secondary thrombosis occurs. Thrombosis may result from defecatory straining or extreme physical activity, or it may be a random event.1 Patients present with constant anal pain of acute onset and often report feeling the sensation of sitting on a tender marble. Physical examination identifies the external thrombosis as a purple mass at the anal verge.

Management

Figure 4. Timing of excision of external hemorrhoids

Treatment depends on the patient’s symptoms [see Figure 4].4 In the first 24 to 72 hours after the onset of thrombosis, pain increases, and excision is warranted. After 72 hours, pain generally diminishes, and expectant treatment is all that is necessary. Patients should be advised that some drainage may occur. If operative treatment is chosen, the entire thrombosed hemorrhoid is excised with the patient under local anesthesia. Incision and drainage of the clot are avoided because they typically lead to rethrombosis and exacerbation of symptoms.

Anal Fissure


Anal fissures are tears or splits in the anoderm just distal to the dentate line.12 They are characterized as acute or chronic. Generally, acute fissures are caused by the mechanical force generated by the passage of a large, hard bowel movement through an anal canal that is too small to accommodate it safely and easily (though they can also be caused by diarrhea). These mechanical forces usually cause a split to occur in the posterior midline: 90% of the fissures in females and 99% of those in males are located posteriorly. Either decreased local blood flow or increased mechanical stress may account for the propensity of these fissures to occur in this location.13 Repeated injury (e.g., from hard or watery bowel movements) may result in the development of a chronic fissure.

Clinical Evaluation

Symptoms associated with anal fissures include anal pain and bright-red rectal bleeding after bowel movements. The pain is usually described as a knifelike or tearing sensation, and the associated anal sphincter spasm may persist for several hours after each bowel movement. The bleeding is usually minor and is seen mainly on the toilet paper.

Figure 5. Chronic anal fissures

Physical examination is difficult because the patient has an extremely tender anus and is fearful of further pain. Often, visual inspection with gentle eversion of the anoderm in the posterior midline is all that is required. Physical findings include a split in the anoderm approximately 1 cm long in the posterior midline just distal to the dentate line. In chronic fissures, the classic triad may be present: hypertrophy of the anal papilla, an anal fissure, and a sentinel skin tag [see Figure 5]. Once an anal fissure has been diagnosed, further examination is very painful, unrewarding, and unnecessary; more extensive investigations can be performed after the fissure has healed.

Figure 6. Association of fissures with other conditions

Multiple fissures are unusual, as are fissures that occur away from the anterior or posterior midline; either should raise suspicions that other problems may be present [see Figure 6].

Management

Acute fissures usually have been present for less than 4 to 6 weeks. As a rule, they are treated nonoperatively: fiber supplements, stool softeners, and generous intake of water, along with sitz baths and local anesthetic ointments, rapidly alleviate symptoms and usually bring about complete healing. Anal suppositories are avoided both because they are painful and because they rest in the rectum rather than the anal canal.

Chronic fissures have been present for periods longer than 4 to 6 weeks. As noted (see above), they are typically characterized by an associated hypertrophied papilla, a skin tag, rolled skin edges, and exposed internal anal sphincter muscle at the base of the fissure. Chronic fissures respond less well to nonoperative measures than acute fissures do. The surgical procedure most frequently performed for anal fissure is lateral internal anal sphincterotomy [see 5:37 Anal Procedures for Benign Disease],14 which results in cure in 95% to 98% of patients. Complications of this procedure include incontinence to flatus (0% to 18% of cases), soiling (0% to 7%), fecal incontinence (0% to 0.17%), and various other problems (0% to 7%).14

Several therapeutic alternatives for anal fissures have been proposed with the aim of avoiding the need for operation and the consequent risk of surgical complications.15 On the basis of the theory that a fissure is actually an ischemic ulcer of the anoderm, topical nitroglycerin (NTG) ointment has been used to treat fissures, with success rates ranging from 48% to 78%. When metabolized, NTG releases nitric oxide, which is believed to be an inhibitory neurotransmitter for smooth muscle. The resulting neurogenic relaxation of the internal sphincter brings about a reduction in anal canal pressure, which diminishes pain and spasm. Typically, NTG is given in a concentration of 0.2% three to five times daily, but concentrations as high as 0.5% have been recommended. In practice, dosing is limited by NTG’s side effects, which arise in as many as 88% of patients. Headaches are the predominant complaint, but dizziness, lightheadedness, and hypotension have also been reported. Caution must be exercised when NTG therapy is employed in patients receiving cardiac medications or those with sensitivities to nitrates. A meta-analysis comparing topical NTG therapy with sphincterotomy demonstrated that sphincterotomy results in better healing of chronic fissures.16

Another nonsurgical option is the use of nifedipine gel or ointment, which also has met with varying degrees of success. Nifedipine acts as a calcium antagonist, preventing calcium from flowing into the sarcoplasm of smooth muscle and thereby reducing local demand for oxygen and mechanical contraction of the muscle.12 Like NTG ointment, nifedipine ointment is usually applied topically in a 0.2% concentration, but it seems to have fewer side effects than NTG does. Nifedipine should be used with caution in cardiac patients and patients who have demonstrated previous sensitivities. One multicenter study reported a 95% complete healing rate after 21 days of treatment.12

Topical 2% diltiazem has also been employed to treat chronic anal fissures, yielding a 67% healing rate.12

Botulinum toxin has been reported to facilitate healing in 78% to 90% of anal fissure patients, with an 8% recurrence rate at 6 months.15,17 The toxin, produced by the bacterium Clostridium botulinum, acts by inhibiting the release of acetylcholine at the presynaptic membrane. The resulting blockage of neurotransmission diminishes or eliminates spasms and contractions of the sphincter mechanism. Typically, 2.5 to 10 units are injected at two to four sites in the internal sphincter at the level of the dentate line.18 Pain relief is generally noted within 24 hours, though it still takes days for the fissure to heal. Postinjection incontinence is rare.12 The major drawback is the cost of the toxin, which may be as high as $400 per 100 unit vial.

Anorectal Abscess


Anorectal abscesses, like abscesses elsewhere in the body, are the result of local, walled-off infections. Most perirectal abscesses are of cryptogenic origin—that is, they begin as infections in the anal glands that surround the anal canal and empty into the anal crypts at the dentate line.18,19 It is thought that the ducts leading to and from these glands become obstructed by feces or traumatized tissue, and a secondary infection then develops that follows the path of least resistance, resulting in an anorectal abscess.

Clinical Evaluation

Figure 7. Anorectal abscesses

Abscesses are categorized according to the space in which they occur: perianal, ischioanal, supralevator, or intersphincteric (intermuscular) [see Figure 7]. Perianal abscesses are the most common—together with ischioanal abscesses, they account for more than 90% of perianal infections. Perianal abscesses occur in the perianal space immediately adjacent to the anal verge. Ischioanal abscesses are larger and often more complex than their perianal counterparts, and they usually manifest themselves as a tender buttock mass. Supralevator abscesses occur above the levator ani muscles and are characterized by poorly localized pain; they are exceedingly rare. Intersphincteric abscesses occur in the plane between the internal and external sphincter muscles, high within the anal canal; these are also rare. The location of an abscess is important in that it dictates subsequent therapy.20

Regardless of location, all anorectal abscesses are associated with constant perianal pain. Accompanying symptoms may include fever, chills, and malaise. In rare cases, systemic toxicity may be evident. The history reveals rectal pain of gradual onset that progressively worsens until the time of presentation. Occasionally, spontaneous drainage decompresses the abscess, and the patient presents with a purulent discharge.

As with fissures, visual inspection of the perineum often clinches the diagnosis. A fluctuant, erythematous, tender area identifies the abscess. In the rare event of a supralevator or intersphincteric abscess, there may be no external manifestations. When this is the case, the presence of a tender mass on digital examination above the anal canal, either adjacent to the rectal ampulla (supralevator abscess) or within the anal canal (intersphincteric abscess), provides the clue to the diagnosis.

Management

Treatment of anorectal abscesses consists of adequate drainage performed in the office, in the emergency department, or in the operating room.18 Most abscesses can be drained in the office, but recurrent or complex abscesses, abscesses in immunosupressed hosts (including some diabetic patients), intersphincteric abscesses, and supralevator abscesses are more appropriately drained in the OR.

Adequate drainage is essential and may be established in several ways. One method is to place a catheter (e.g., a 10 to 16 French Pezzar catheter) through a small stab incision [see 5:37 Anal Procedures for Benign Disease].18 This measure allows the pus to drain through the catheter as the cavity closes down. A second option involves creating a larger elliptical incision.20 Unroofing the abscess cavity allows it to heal without any need for packing. A small incision should be avoided because it would require painful packing to keep the skin open until the abscess cavity heals.

The most difficult abscess to diagnose and manage is a deep postanal space abscess. Such abscesses are caused by fistulization from the posterior anal canal, usually in the bed of a chronic posterior fissure. The patient is highly uncomfortable and febrile, but there is no apparent sign of a problem perianally. A simple digital examination pushing posteriorly towards the coccyx and the deep postanal space will evoke severe pain, and this response should lead the examiner to suspect the diagnosis. The patient should be taken to the OR and anesthetized. On digital examination, the deep postanal space is often felt to be bulging. The diagnosis is confirmed by aspirating the space with an 18-gauge needle. Once the diagnosis is confirmed, an incision is made in the perianal skin posterior to the anal verge and deepened into the space, and the space is drained. In selected patients, a cutting seton is placed from the primary site in the posterior anal canal directly into the deep postanal space abscess. Horseshoe fistulas are dealt with as described elsewhere [see 5:37 Anal Procedures for Benign Disease].

Fistula in Ano


An anal fistula is a communication between the anal canal and the perianal skin. It usually begins in a crypt at the dentate line and follows a course either between the internal and external sphincters (the most common location), resulting in an ischioanal abscess, or above the sphincters, leading to a supralevator abscess.18,20 After drainage of an abscess, one of three things typically occurs if a fistula is present: (1) the fistula heals spontaneously, and the patient experiences no further symptoms; (2) the abscess heals, only to recur in the future; or (3) the abscess heals, but a chronic draining fistula remains. Only the third scenario is addressed here.

Clinical Evaluation

After drainage of one or more abscesses, a fistula is usually associated with chronic serosanguineous to seropurulent drainage. As long as the fistula remains open and draining, patients report little pain. If the fistula closes externally, however, an anorectal abscess may develop.

Figure 8. Relation of external to internal opening of anal fistuala

Figure 9. Classification of fistula in ano

Physical examination reveals a 2 to 3 mm opening in the perianal skin, with surrounding induration. Often, a fistula tract can be palpated as a firm cord running between the external opening and the anal canal. The relationship of the external opening to the internal opening is suggested by Goodsall’s rule [see Figure 8]. Fistulas are classified into four main categories according to their relation to the anal sphincters [see Figure 9]: intersphincteric, transsphincteric, suprasphincteric, and extrasphincteric.21

Management

Essentially, all chronic fistulas call for surgical treatment, which consists of unroofing the entire fistula tract (fistulotomy) and leaving the wound open to heal secondarily. Fistulas that course through significant amounts of sphincter muscle, anterior fistulas in women, and fistulas associated with inflammatory bowel disease or weakened sphincter muscles, however, cannot be opened entirely, because incontinence will result. These fistulas may be partially opened, with the anal musculature left intact and encircled with a seton [see 5:37 Anal Procedures for Benign Disease].20 Although some surgeons sequentially tighten the seton (making it a cutting seton), I prefer not to, because of the resultant patient discomfort. Another surgical option is to close the internal fistula opening with an advancement flap.22 Advancement flap repairs result in high success rates with minimal effects on continence.

Pilonidal Disease


The term pilonidal disease is derived from the Latin words pilus (‘hair’) and nidus (‘nest’).23 It denotes a chronic subcutaneous infection and foreign-body reaction to hairs embedded in the skin or to abnormalities of hair follicles in the natal cleft.23 Pilonidal disease is most commonly seen in men between the onset of puberty and 40 years of age and in obese persons.23

Clinical Evaluation

Pilonidal disease has three common presentations. First, nearly all patients experience an episode of acute abscess formation. Second, after the abscess resolves, either spontaneously or with medical assistance, a pilonidal sinus tract develops in many cases. Third, although most of these sinus tracts resolve, chronic disease or recurrent disease after treatment develops in a small minority of cases.

Figure 10. Pilonidal disease

Physical examination typically reveals one or more small (1 to 2 mm) dermal pits at the base of the intergluteal cleft [see Figure 10]. Tracking from the pits (usually proceeding in a cranial and lateral direction) appears as areas of induration. If there is an associated abscess, the diseased area may be tender and erythematous, and draining pus may be evident. The more extensive the disease, the more prominent the findings. Treatment varies according to the stage of the disease.

Management

Abscesses must be drained. In one study, incision and drainage in an office setting with local anesthesia led to healing in 60% of patients.24 As many as 40% of acute pilonidal abscesses treated with incision and drainage develop into chronic sinuses for which additional treatment is necessary.

Several different approaches have been employed in the surgical treatment of pilonidal sinus tracts. A review of articles published over a period of 30 years on the treatment of pilonidal disease divided the procedures into the following four broad categories and reported the following findings.25

  1. Closed techniques (coring out follicles and brushing the tracts). These techniques necessitated shaving of the area but could be performed on an outpatient basis. Mean healing time was about 40 days, and recurrence rates were slightly higher than those seen with other forms of treatment.
  2. Laying open (unroofing) the tracts with healing by granulation. This approach resulted in average healing times of 48 days and necessitated frequent outpatient dressing changes. The incidence of recurrent sinus formation was lower than 13%.
  3. Wide and deep excision of the sinus alone. This procedure resulted in an average healing time of 72 days. The recurrence rate was similar to that of simple unroofing of the sinus tract with healing by granulation.
  4. Excision and primary closure. This technique resulted in wound healing within 2 weeks in successful cases (19 days overall). However, primary wound healing failed in as many as 30% of patients, and the average recurrence rate was 15%.

 

A nonoperative or conservative approach—involving meticulous hair control (through natal cleft shaving), improved perineal hygiene, and limited lateral incision and drainage for treatment of abscess—has been suggested as an alternative to conventional excision.26 This approach has brought about a significant reduction in the number of excisional procedures and occupied-bed days.

Even with proper treatment of pilonidal abscesses and sinuses, a small number of patients are left with persistent, nonhealing wounds. A number of more aggressive approaches have been advocated for treatment of complex or recurrent disease, including wide excision with split-thickness skin grafting, cleft closure, and excision with flap closure.24,27 Flap techniques, as a group, have been found to lead to primary healing within 15 days in 90% of cases.24 These aggressive approaches nevertheless have certain disadvantages. For nearly all of them, hospitalization and general anesthesia are mandatory. In addition, as many as 50% of procedures involving the use of skin flaps for wound coverage or closure in this setting result in some loss of skin sensation or some degree of flap-tip necrosis.

My policy is to treat acute abscesses with drainage, followed by measures aimed at keeping the cleft free of hair. If a chronic sinus develops, it is managed with unroofing or, in selected patients, excision and closure. Flap procedures are reserved for cases of extensive, recurrent, and complex disease.23

Hidradenitis Suppurativa


Clinical Evaluation

Hidradenitis suppurativa is a chronic, recurrent inflammatory process involving the apocrine glands of the axilla, the groin, the perineum, and the perianal region.28 The disease can result in chronically draining wounds and sinus tracts and can become quite painful and debilitating.29 Occlusion of follicles and abnormalities of apocrine ducts are believed to be causative factors.

Management

Medical management may afford temporary relief of symptoms; however, most patients eventually require surgical therapy.30 In select patients, incision and drainage or unroofing of sinuses may provide relief, but these measures should be reserved for cases of early and acute disease. Local excision provides adequate control of symptoms; however, recurrence rates higher than 50% may be anticipated. Wide excision with secondary granulation of perineal wounds constitutes the most definitive therapy and generally can be accomplished safely.29 Perianal disease is associated with considerably lower recurrence rates than perineal disease is and thus can more often be managed with local excision alone.

Pruritus Ani


Pruritus ani is a dermatologic condition of the perianal skin characterized by uneasiness or itching in the area around the anus.12,31 Multiple factors may predispose this area to irritation, including poor perianal hygiene (related to incontinence, diarrhea, or excessive hair), excessive moisture, irregularities of the perianal skin (from hemorrhoids, fistulas, or previous surgery), skin hypersensitivity, diet, decreased resistance to infection, and injury to the perianal skin.31 The variety of possible causes is what often makes pruritus ani difficult to treat. Some patients improve only after the offending agents or conditions are identified and specific therapy instituted. Fortunately, many patients’ symptoms can be alleviated by the application of nonspecific treatments.

Clinical Evaluation

Figure 11. Idiopathic pruritus ani

A thorough history and physical examination are necessary to suggest possible causes of pruritus. Initial inspection of the perianal skin should be conducted with gentle retraction of the buttocks under bright lighting. A characteristic finding is erythematous or thickened perianal skin [see Figure 11]. This thickening results in a pale, whitish appearance, with accentuation of the radial anal skin creases. In addition, the skin may be excoriated or ulcerated; this process, when combined with thickening, is referred to as lichenification. Occasionally, the skin is so excoriated that a large, coalescing, weeping ulcer forms. Digital rectal examination should be performed to assess the competence of the anal sphincter both at rest and during maximal squeeze. Anoscopy and proctosigmoidoscopy may be performed after the administration of an enema.

Management

The basic principles of therapy for pruritus ani are uncomplicated. If an inciting cause can be identified, it should of course be eliminated or corrected. Frequently, the inciting cause is elusive, but even so, most patients can still be effectively managed by applying several simple measures.

Generally, patients should keep the perianal area dry and avoid further trauma to the area. The perianal area should be gently washed, never scrubbed. After showering, the area should be patted dry or dried with a hair dryer on a low heat setting. After bowel movements, the anus should be cleaned with moistened toilet paper. Excessive rubbing or wiping should be discouraged: scratching or rubbing the anal area damages the perianal skin, making it more susceptible to irritation. Patients should also be instructed to avoid irritating foods and drinks, such as tomatoes, peppers, citrus fruits and juices, coffee, colas, beer and other alcoholic beverages, milk, nuts, popcorn, and any other foodstuffs found to be associated with increased gas, indigestion, or diarrhea. After 2 weeks, food items eliminated from the diet can be reintroduced one at a time in an attempt to identify the offending agent more specifically.

A regular bowel habit should be maintained with the help of fiber supplementation or a high-fiber diet. Patients should be instructed to eschew all proprietary creams, lotions, and emollients. If prescribed and supervised by a physician, however, a hydrocortisone cream may be applied sparingly to the affected area for a period of 1 week or less to attain control of symptoms. Occasionally, when pruritus ani is refractory to treatment, a candidal yeast infection is found to be present, in which case a trial of antifungal lotion, solution, or powder is worthwhile.

The fundamental goal in the treatment of pruritus ani is to establish and maintain intact, healthy, clean, and dry perianal skin. When standard measures fail to elicit improvement, fungal and viral cultures and perhaps even biopsy may be necessary to exclude an infectious or neoplastic cause.

Solitary Rectal Ulcer Syndrome


Clinical Evaluation

SRUS is a clinical condition characterized by rectal bleeding, copious mucous discharge, anorectal pain, and difficult evacuation.32 The name of the condition notwithstanding, SRUS patients can have a single rectal ulcer, multiple ulcers, or even no ulcers at all. When present, the ulcers usually occur on the anterior rectal wall, just above the anorectal ring; less commonly, they occur from just above the dentate line to 15 cm above it. The ulcers usually appear as shallow lesions with a punched-out gray-white base that is surrounded by hyperemia.33

Colitis cystica profunda (CCF) is a benign condition that is related to SRUS and is characterized by mucin-filled cysts located deep to the muscularis mucosae. CCF is a pathologic diagnosis, of which the most important aspect is differentiation of CCF from colorectal adenocarcinoma.

Precisely what causes SRUS and CCF remains unclear, but both conditions are known to be associated with chronic inflammation or trauma (e.g., internal intussception or prolapse of the rectum, direct digital trauma, or the forces associated with evacuating a hard stool). Endoscopic evaluation of the distal colon and rectum reveals the lesions described. For both CCF and SRUS, the differential diagnosis includes polyps, endometriosis, inflammatory granuloma, infection, drug-induced colitis, and mucus-producing adenocarcinoma. These entities can be confirmed or excluded by means of an adequate biopsy.

Management

Treatment is directed at alleviating symptoms or interfering with some of the proposed etiologic mechanisms. Conservative therapy (e.g., a high-fiber diet, lifestyle changes, and biofeedback) reduces symptoms in most patients and should be tried first. Patients without rectal intussusception are offered biofeedback for retraining their bowel function. Pharmacologic therapy (e.g., anti-inflammatory enemas and suppositories) has had only limited success but nonetheless may be worth trying before the decision is made to embark on surgery.

If symptoms persist, localized resection may be considered in selected patients. Patients with prolapse are considered for perineal procedures (i.e., mucosal or perineal proctectomy) and abdominal procedures (i.e., fixation or resection and rectopexy). Patients without prolapse may be offered excision, for which the options range from a transanal excision to a major resection with coloanal pull-through. Understandably, surgeons have been hesitant to offer surgical therapy for this benign condition; the results are often unsatisfactory.34

 


Nancy N. Baxter, MD, PHD, FRCSC, FASCRS

Assistant Professor, Department of Surgery
University of Minnesota Medical School

Robert D. Madoff, MD, FACS, FASCRS

Professor, Department of Surgery
University of Minnesota Medical School

Surgeons commonly encounter patients with gastrointestinal motility disorders. The management of such patients is frequently challenging, in that the etiology of the disorder is often multifactorial. Furthermore, even when surgical therapy is appropriate, management of symptoms remains a key component of effective treatment.

In what follows, we discuss two of the most common motility disorders, constipation and fecal incontinence. Although constipation usually is not treated surgically, surgeons regularly see patients with this presenting symptom. It is therefore critical that surgeons have a practical method of diagnosing and managing the primary and secondary causes of constipation. Fecal incontinence is an understudied and undertreated condition that can have a dramatic impact on quality of life. Effective treatment of incontinence has a dramatic positive influence on patients’ lives; thus, it is important for surgeons to have both an effective approach to diagnosis and an informed awareness of the various therapeutic options available (including experimental treatments).

Constipation


Clinical Evaluation

History

Figure 1. Workup and management of constipation

Constipation is the most common digestive complaint, with as much as 20% of the population reporting this symptom.1 The meaning of the term constipation, however, is variable: when patients describe themselves as constipated, they may be referring to decreased stool frequency, reduced stool volume, altered stool consistency, or difficulty with defecation.2 Accordingly, when a patient presents with a complaint of constipation, a thorough history of the presenting illness is essential [see Figure 1].

The patient should be asked about the frequency of bowel movements, the volume of stool per movement, the caliber of the stool, and, in particular, any changes in bowel habits over time. Patients with idiopathic constipation tend to have long-standing problems, with no abrupt change in bowel habits. Thus, if the history reveals constipation of sudden onset, an underlying cause (e.g., cancer) is more likely and should be sought. Other important symptoms that should lead to a search for a secondary cause are weight loss, anorexia, nausea and vomiting, rectal bleeding, changes in stool caliber, and fever. The patient should always be asked about previous colon cancer screening or other GI investigations. Although chronic constipation is common, severe constipation that has been present since early childhood should alert the clinician to the possibility of undiagnosed short-segment Hirschsprung disease; this rare diagnosis is easily missed if it is not given appropriate consideration. Other symptoms may be indicative of an outlet problem (e.g., rectocele or nonrelaxing puborectalis syndrome); such symptoms include requiring a prolonged period to evacuate stool from the rectum, a feeling of incomplete rectal emptying, and the need to support the perineum (through digitation of the vagina or rectum) to achieve complete evacuation.

Diet can contribute significantly to constipation. Because high-fiber foods tend to increase stool bulk and frequency, detailed information on dietary fiber intake should be obtained. Because dehydration increases fluid resorption from stool and thereby results in the formation of hard stools, total daily fluid intake should be determined as well. A specific effort should be made to assess intake of fluids that contain caffeine, which exerts a diuretic effect. Most patients with long-standing constipation will already have tried some form of self-medication. Such attempts should be documented, both to help assess the severity of the symptom and to determine the likelihood of response to simple measures.

Various other diseases and certain common medications [see Table 1] also can cause or contribute to constipation. When such factors are present, treating the underlying condition or changing medications can result in substantial improvement. Therefore, a thorough past medical history and an accurate medication history are essential. A family history of colonic neoplasia or inflammatory bowel disease is potentially suggestive and may lead to a more intensive search for secondary causes. Victims of physical or sexual abuse may present with constipation; however, they are unlikely to mention the abuse if not directly questioned about the possibility.

Physical Examination

During physical examination, it is important to make a quick assessment of the patient’s nutritional status. In general, patients with idiopathic constipation should not appear malnourished; the appearance of malnutrition should prompt a more extensive search for a secondary cause. An abdominal examination should be conducted to look for any significant abdominal distention, tenderness, or masses. Distention is a common and expected finding with idiopathic constipation, but significant tenderness or masses should prompt a full investigation.

All patients presenting with constipation should undergo a rectal examination. The anus should be examined for evidence of scarring or stricture. A digital rectal examination should be done to assess anal tone; high anal tone and inability to increase pressure when asked to squeeze are common findings in patients with obstructed defecation resulting from a nonrelaxing puborectalis. An effort should be made to look for any anterior defect in the rectovaginal septum, which would indicate the presence of a rectocele; such a defect, if present, may be made more prominent by having the patient strain. The finding of a rectal mass warrants further investigation.

Investigative Studies

In general, diagnostic studies are conducted to rule out an underlying cause of constipation (e.g., partially obstructing colon cancer) and to diagnose specific disorders associated with severe constipation (e.g., a nonrelaxing puborectalis and slow-transit constipation). Therefore, the choice of investigative studies should be individualized according to the clinical situation. In patients with mild symptoms and poor dietary habits who have no indications of any secondary causes of constipation, no investigations need be done on a routine basis. In patients with severe constipation, however, serum calcium concentrations, thyroid function tests, hemoglobin concentrations, glucose levels, serum electrolyte levels, and creatinine concentrations may be helpful.

Constipation with Suspected Underlying Cause

Whenever any of the findings from the history or the physical examination indicate a possible secondary cause of constipation, further investigation is mandatory. In particular, if a patient presents with any sign, symptom, or laboratory test result consistent with colorectal cancer (e.g., a sudden change in bowel habits, blood in the stool, weight loss, anorexia, a suggestive family history, abdominal masses, or anemia), colonoscopy is necessary, irrespective of the patient’s age or history of previous colonic investigations.

Patients with other secondary causes of constipation (e.g., hypothyroidism and hypercalcemia) often respond to treatment of the underlying disease or manipulation of medications. If such measures are ineffective, the constipation should be treated symptomatically, in much the same fashion as idiopathic constipation is. Patients requiring long-term opioid administration for pain control generally experience constipation as a side effect, and this effect does not dissipate with time. Thus, many of these patients will require laxative therapy for the duration of their opioid use.3

Mild to Moderate Idiopathic Constipation

In patients who have mild to moderate symptoms and no findings from the history or the physical examination that would indicate a secondary cause, extensive investigations are not necessary. Routine colonoscopy is not mandatory for patients younger than 50 years. For patients older than 50 years, the baseline risk of colorectal cancer is sufficiently high that screening colonoscopy is recommended even in the absence of symptoms. These older patients should therefore undergo routine colonoscopy, and many authors recommend that patients younger than 50 years undergo routine flexible sigmoidoscopy. Random endoscopic biopsies are unnecessary, because idiopathic constipation is not associated with abnormalities on routine processing of mucosal biopsies.

Severe, Long-standing Constipation or Refractory Constipation

In patients who have very severe constipation or in whom medical management fails, further investigative tests are warranted. These tests are conducted to classify patients into three categories, each of which calls for a different treatment approach: (1) slow-transit constipation, (2) nonrelaxing puborectalis, and (3) normal-transit constipation.4 The initial investigations should include assessment of colonic transit time to determine if slow-transit constipation is present, as well as evaluation of pelvic floor function to determine if a nonrelaxing puborectalis is present.

Figure 2. Colonic transit study

There are two main methods for evaluating colonic transit: the radiopaque marker study and colonic scintigraphy. Both tests have advantages and disadvantages. In general, the choice between them depends on local expertise; the radiopaque marker study is more widely available. For the radiopaque marker study, 20 radiopaque markers (prepackaged in gelatin capsules) are ingested, and an abdominal x-ray (which includes the pelvis) is taken on day 5. The patient abstains from laxatives for the duration of the study. At 3 days, most patients with normal transit have excreted more than 80% of the markers; however, because there is substantial variation among asymptomatic persons, only patients who retain more than 20% of the markers for at least 5 days are considered to have abnormal transit. Abnormal transit may be demonstrated either throughout the colon or within a limited portion thereof (most commonly, the sigmoid and the rectum) [see Figure 2].

Colonic scintigraphy shares certain principles with the radiopaque marker study. Patients ingest a meal containing a radioactive isotope, and abdominal images are obtained with a gamma camera at 12, 24, and 48 hours. The results provide a quantitative assessment of colonic transit. In addition, unlimited numbers of images may be taken with the single isotope dose, and this feature of the test may be especially useful in children. For optimal accuracy, this technique requires standardization, and its availability is generally limited to centers with specific interest and expertise in it.

Pelvic floor studies are valuable for ruling out obstructed defecation as a cause of constipation. The balloon expulsion test can be performed in the office as an initial screening measure.5 A balloon filled with 50 ml of water is attached to tubing and placed in the rectum; patients with a nonrelaxing puborectalis generally cannot expel the balloon from the rectum in 1 minute while sitting on a commode. It should be kept in mind, however, that as many as 12% of patients with normal pelvic floor function will have difficulty with balloon expulsion in this setting.6

A thorough pelvic floor evaluation is best conducted in a pelvic floor laboratory with a specific interest in anorectal function. In addition to the balloon expulsion test, the evaluation generally involves manometry, including assessment of the reflexive relaxation of the internal sphincter after rectal distention. The presence of this reflexive relaxation rules out Hirschsprung disease as a cause of constipation. In patients with a nonrelaxing puborectalis, manometry during straining effort demonstrates abnormal function of the external sphincter—either failure to relax to enable expulsion or, on occasion, paradoxical contraction. Similar findings during straining can be documented by means of electromyography (EMG) with a sponge electrode in the anal canal.

Figure 3. Schematic representation of normal defecation

Defecography is commonly performed as well. Barium paste is formulated so as to simulate a fecal bolus and placed in the rectum. The patient is asked to defecate on a radiolucent commode, and the event is recorded with fluoroscopy. During normal defecation, the puborectalis and the anal sphincter muscles relax, and the rectum assumes a more vertical position with respect to the anal canal, facilitating evacuation of stool [see Figure 3]. In a patient with a nonrelaxing puborectalis, defecography typically demonstrates failure to open the anorectal angle and persistence of the puborectalis impression during defecation, as well as failure to empty completely.7 Other important findings that may be noted include rectocele, internal intussusception, and rectal prolapse. When appropriately selected, patients with obstructed defecation resulting from such abnormalities may benefit from surgical correction; however, even when these anatomic abnormalities are present, they may not be the underlying cause of constipation. Interpretation of defecography is subjective, and there is wide normal variation. Therefore, the diagnosis of a nonrelaxing puborectalis should be based not on a single test result but, rather, on the totality of the diagnostic findings.

The diagnosis of nonrelaxing puborectalis syndrome is made in persons with constipation in whom there is evidence of failure of the pelvic floor to relax appropriately (or paradoxical contraction of the pelvic floor) with defecation. The cause of this condition is not known; however, the syndrome is thought to be acquired over time. Patients with an underlying neurologic disorder (e.g., multiple sclerosis or Parkinson disease) are prone to spasticity of the puborectalis and may experience severe constipation as a result.

Management

Mild to Moderate Idiopathic Constipation

Many cases of constipation can be managed with dietary and lifestyle changes, such as modifying the diet to include foods high in fiber and drinking adequate amounts of water. Physical inactivity is associated with constipation, and encouraging moderate exercise may lead to significant symptomatic relief. Fiber supplementation is a key component of therapy for mild constipation.8 Fiber products (e.g., psyllium, methlycellulose, and polycarbophil) increase stool bulk and stimulate colonic motility. Such products must be taken with sufficient amounts of fluid, or they may lead to stool hardening. Often, patients have already tried fiber products but did not achieve satisfactory results because the quantities were insufficient; daily doses as high as 20 g may be necessary for a therapeutic effect. Patients taking fiber products may experience an increase in flatulence, particularly with fermentable fiber products. To improve tolerance, the amount of fiber should be increased gradually, and patients should be informed that the effect of fiber may not be seen immediately.

Nonlaxative therapy should be stressed; however, if dietary changes and fiber supplementation fail, judicious use of laxatives can bring about significant symptomatic relief. It should be kept in mind that tachyphylaxis to laxatives is common and may lead to chronic dependence. Stool softeners, or emollient laxatives (e.g., ducosate sodium and mineral oil), enhance penetration of water and fat into the stool, thereby making it less hard. These agents may be of use on a relatively short-term basis. Ducosate sodium is less effective than fiber supplementation9; stool softeners should not be used as a substitute for fiber.

Stimulant laxatives, including cascara, anthraquinones (senna and rhubarb), castor oil, and bisacodyl, are common components of popular over-the-counter medications. These agents have direct neuromuscular or mucosal effects, resulting in enhanced GI motility and altered mucosal transport (and thus increased intestinal secretion).8 Long-term use or abuse of anthraquinones can lead to melanosis (discoloration of the colonic mucosa caused by pigment deposition in colonic macrophages).

Osmotic laxatives contain compounds that either are not absorbed or are poorly absorbed. If the solutions are hypertonic, they cause water to move into the bowel lumen to maintain tonicity.10 Common preparations include magnesium and phosphate salts. Ingestion of large amounts of such preparations can lead to hypermagnesemia or hyperphosphatemia, mainly in patients with renal failure. The large fluid shifts that result when these compounds are used for bowel preparation may be dangerous in patients with underlying heart disease. Polyethylene glycol (PEG) is a high-molecular-weight compound that is not absorbed and thus functions as an osmotic laxative. PEG preparations are commonly administered as isotonic solutions and therefore cause only minimal fluid or electrolyte shifts when consumed rapidly (as in bowel preparation). PEG compounds are available as laxatives that can be taken either intermittently or regularly.

Tegaserod, a 5-HT4 partial agonist, has been shown to alleviate bloating and increase stool frequency by improving gut motility and decreasing visceral sensitivity.11 It may be prescribed for women with constipation-predominant irritable bowel syndrome (IBS) (see below) and for either male or female patients younger than 65 years who have idiopathic constipation. Tegaserod has been associated with the development of diarrhea; typically, the diarrhea resolves when the drug is discontinued, but occasionally, it is severe. In addition, several cases of ischemic colitis have been reported in patients receiving tegaserod. Although no causal relation has been established, patients should be warned to cease taking tegaserod and immediately contact their physician if abdominal pain worsens.

Enemas and suppositories act via a number of mechanisms, including softening of the stool, stimulation of rectal contraction by rectal distention, and direct alteration of mucosal secretion. They may be useful for occasional administration.

Constipation-Predominant Irritable Bowel Syndrome

In patients with constipation, significant abdominal pain, and no identifiable secondary cause of constipation, the diagnosis of constipation-predominant IBS may be appropriate [see Table 2].12,13 Often, patients with constipation-predominant IBS respond to reassurance and fiber supplementation. Tegaserod may be employed in female patients who do not respond to conservative measures.

Severe, Long-standing Constipation or Refractory Constipation

Nonrelaxing puborectalis Patients with constipation arising from a nonrelaxing puborectalis often benefit from biofeedback.14 In this modality, a device (e.g., an anorectal manometer) is used to monitor pelvic floor activity; electrodes may also be used for EMG biofeedback. Patients observe pressure changes (or EMG activity) during attempts to evacuate. Through trial and error, they are taught to modify their responses until appropriate relaxation is achieved, the aim being to retrain the pelvic floor to relax during defecation. Training may have to be reinforced at intervals. Accurate determination of the success rate of biofeedback is difficult, in that the published literature consists primarily of case series and most of the trials that have been conducted have not included a placebo arm. It has been estimated that the success rate may be as high as 70%; however, this estimate is probably overoptimistic.15

If biofeedback fails, injection of botulinum toxin into the puborectalis under ultrasonographic guidance may be attempted. To date, published reports have evaluated this approach only in relatively small study groups; the results, though not decisive, are promising, in that the use of botulinum toxin clearly brought about noticeable improvements in manometric and defecographic findings16 and symptomatic improvements in the majority of patients.17 Other experimental techniques available for treatment of nonrelaxing puborectalis syndrome are electrogalvanic stimulation18 and sacral nerve stimulation (SNS).19 Currently, surgical approaches do not play a role in the treatment of constipation secondary to a nonrelaxing puborectalis.

 

Slow-transit constipation Slow-transit constipation, also known as colonic inertia, is most common in young women and often starts at puberty. It is characterized by abnormally slow forward propulsion of colonic contents. The cause of slow-transit constipation is unknown, though abnormalities in a number of cellular and neuromuscular modulators of GI motility have been found in patients with this condition.20,21 Although patients with idiopathic slow-transit constipation are frequently resistant to laxative therapy, many respond to osmotic PEG laxatives. Surgery should be considered as an option only in the most severely affected patients, in whom aggressive laxative therapy has repeatedly failed over a prolonged period. Even in specialized centers, only about 5% of patients presenting with constipation are considered appropriate candidates for surgical treatment.22

The operation most commonly performed to treat slow-transit constipation is subtotal colectomy with ileorectal anastomosis, performed via either an open or a laparoscopic approach. The colon is removed to the level of the sacral promontory in a standard fashion; the ileorectal anastomosis may be either stapled or handsewn. Constipation is less likely to recur with this anastomosis than with an ileosigmoid anastomosis.23

Surgical therapy is generally successful in improving bowel function: in most patients, stool frequency rises to one to three bowel movements a day. Unfortunately, surgery may not satisfactorily alleviate other symptoms (e.g., abdominal discomfort or bloating),24 and patients should be made aware of this possibility before operation. The key to successful surgical treatment is patient selection. Overall, the majority of well-selected patients are satisfied with the results of surgical treatment25,26; however, long-term postoperative complications, particularly small bowel obstruction, are common. In addition, patients may manifest symptoms of a more global GI dysmotility disorder in the long term.

Other surgical approaches sometimes employed in this setting are ileostomy [see 5:30 Intestinal Stomas], and colectomy with cecorectal anastomosis; however, data on the long-term effectiveness of these approaches in large numbers of patients are lacking. Completion proctectomy with IPAA and ileostomy are options for patients who remain severely symptomatic after ileorectal anastomosis but who manifest no evidence of proximal dysmotility.

Not infrequently, patients have both slow-transit constipation and a nonrelaxing puborectalis. In such cases, it is essential that the obstructed defecation be addressed before any surgical treatment is carried out. Even after biofeedback, if surgical therapy is attempted in this setting, as many as 50% of patients will be dissatisfied with the results.27

 

Severe idiopathic constipation Patients who have severe constipation but show no signs of slow-transit constipation, pelvic floor dysfunction, or IBS should be treated with reassurance and symptomatic management. Osmotic laxatives—in particular, PEG products—may be very useful in this group. Operative treatment plays no role in management; however, experimental approaches (e.g., SNS) are being evaluated for possible use in this setting.

Fecal Incontinence


Fecal incontinence may be defined as the involuntary loss of rectal contents through the anal canal. It is a relatively common condition, occurring in an estimated 2.2% of persons in the United States.28 Its exact prevalence is unknown, however, and appears to vary with the population being studied. For example, nearly 50% of nursing home patients are incontinent to stool.28 Fecal incontinence is often treated inadequately, either because of underreporting of symptoms to the physician29 or because of ignorance or disinterest on the physician’s part.

Fecal incontinence makes a significant contribution to medical morbidity (e.g., urinary tract infections and decubitus ulcers), but its main impact is on quality of life. Affected patients experience embarrassment and shame, and many dramatically alter their lifestyle in an effort to avoid accidents.

Normal continence depends on a chain of interdependent processes, and disruption of any of the links in the chain can lead to incontinence. Frequently, a combination of factors is responsible for the incontinence.

To care about continence, persons must have adequate mental function, and to maintain normal continence, they must have an intact neurologic arc from the brain to the anal sphincter. A wide array of neurologic disorders can lead to incontinence, including dementia, strokes, spinal cord injury, multiple sclerosis, and diabetic autonomic neuropathy. So-called idiopathic fecal incontinence is caused by pelvic floor denervation resulting from traction injury to the pudendal nerves.30 The injury is usually caused by straining and consequent pelvic floor descent during obstetrical delivery or by chronic straining at stool.

Conditions characterized by abnormal GI function, especially diarrheal states, can cause or exacerbate incontinence. Common causative conditions include infectious diarrhea and inflammatory bowel disease. Diarrhea-predominant IBS can contribute to incontinence in patients with other associated disorders. Fecal impaction is an important cause of incontinence, particularly in older and institutionalized populations.31

Figure 4. Obstetric sphincter injury

Abnormalities of the pelvic floor are frequent causes of incontinence. Some such abnormalities are congenital malformations (e.g., imperforate anus, rectal agenesis, and cloacal defect). More often, abnormalities are attributable to acquired sphincter injuries. Common causes of sphincter injury include obstetric injury, pelvic fracture, and traumatic impalement [see Figure 4]. One of the most frequent causes is an anorectal procedure, such as fistulotomy,32 sphincterotomy,33 or anal dilatation.34 Sphincter-sparing rectal resections can also lead to incontinence as a consequence of both the loss of the normal rectal reservoir and the sphincter injury caused by transanal introduction of intraluminal staplers.

Clinical Evaluation

History

Figure 5. Workup and management of fecal incontinence

A careful patient history and a directed physical examination are the most important elements of clinical evaluation for a patient with fecal incontinence [see Figure 5]. The patient should be asked about the onset and nature of the incontinence (e.g., whether the stool is liquid or solid and whether flatus is present), any associated changes in stool consistency or bowel habits, and the frequency of incontinence. A pertinent but thorough medical, surgical, and obstetric history should be obtained, and any underlying contributory conditions (e.g., colitis) should be treated. The impact of the incontinence on the patient’s quality of life should be assessed, at least qualitatively.

Physical Examination

Physical examination should focus primarily on the perineum. Seepage and secondary perineal skin breakdown should be noted, as should scars from previous surgical treatment or trauma. Perineal body deformity is an important sign of obstetric injury, and gaping of the anus with traction on the buttocks is suggestive of rectal prolapse. When prolapse is suspected but not evident, the patient should be asked to strain while seated on a commode. Digital rectal examination is useful for detecting low rectal tumors and fecal impaction; it also provides a qualitative assessment of both resting sphincter tone and voluntary squeeze pressure.

Investigative Studies

Endoscopy should be performed on all incontinent patients to exclude a neoplastic or inflammatory condition. In most cases, flexible sigmoidoscopy is adequate, but if the patient has unexplained diarrhea, bleeding, or changed bowel habits, complete colonoscopy should be performed.

Figure 6a. Endoanal ultrasonogram: normal sphincter

Figure 6b. Endoanal ultrasonogram: sphincter defect

Anorectal testing is indicated for most patients with significant incontinence, particularly if operative treatment is being considered. The most important test is endoanal ultrasonography (EAUS), which yields a highly accurate assessment of sphincter integrity [see Figures 6a and 6b].35 At some centers, magnetic resonance imaging has become the test of choice for evaluating the pelvic floor. Anal manometry provides a quantitative assessment of resting and squeeze anal pressures, which serve as indicators of internal anal sphincter function and external anal sphincter function, respectively. EMG may be used to diagnose neuropathic injury of the pelvic floor. Although concentric-needle EMG is the most accurate technique, most centers employ a glove-mounted intra-anal electrode to measure pudendal nerve conduction time (i.e., pudendal nerve terminal motor latency [PNTML]). The practical utility of PNTML testing is debatable, however, and opinions vary regarding the test’s ability to predict successful outcomes after anal sphincter repair.36,37 When the cause of incontinence is uncertain, dynamic imaging of the pelvic floor with defecography or MRI may reveal an occult pathologic state (e.g., occult rectal prolapse).

Management

Conservative Management

Minor incontinence should be treated first with conservative measures. Dietary changes (e.g., avoidance of foods that cause diarrhea or urgency), fiber supplementation, and bowel habit training are helpful for most patients, as is regular use of loperamide. Perianal skin excoriation should be treated with a barrier cream, and seepage may be controlled either with placement of a small cotton wick at the anal orifice or, occasionally, with rectal washouts.

Biofeedback

Biofeedback appears to be an effective therapy for fecal incontinence in a high percentage of patients.38,39 It is an inherently attractive approach because it is simple, painless, and risk-free. However, the biofeedback literature consists mostly of small, uncontrolled, retrospective studies; a randomized, controlled trial from 2003 found that biofeedback had no advantages over standardized medical and nursing care (i.e., advice) or advice plus sphincter exercises.40

Sphincteroplasty

Figure 7. Sphincteroplasty

Anal sphincter repair is the most widely accepted operation for fecal incontinence [see Figure 7]. In acute situations (e.g., when an obstetric sphincter injury is recognized), immediate direct repair is generally recommended. Unfortunately, as many as 75% of women have persistent external anal sphincter defects after primary repair, and about 60% have some degree of incontinence.41 If immediate repair is not attempted, surgical treatment should be delayed at least 3 to 6 months to permit resolution of local tissue inflammation and edema.

For incontinent patients with established sphincter defects, overlapping sphincteroplasty is the procedure of choice. Complete bowel preparation is carried out before the procedure, and prophylactic antibiotics are administered.

Operative technique Step 1: initial dissection. The patient is placed in the prone jackknife position, with the buttocks taped apart and a large roll beneath the hips. A curvilinear incision is made over the perineal body, and the anoderm and the anal canal mucosa are raised as an endodermal flap [see Figure 7, part a]. The vaginal wall is mobilized anteriorly.

Step 2: mobilization of sphincter muscle. It is often easiest first to identify normal muscle laterally in the ischiorectal fossa and then to work medially toward the attenuated tissue in the midline. Lateral dissection is extended back on either side until enough healthy muscle is mobilized to allow overlapping without tension. Generally, however, lateral dissection should not extend beyond the midcoronal line, so as not to risk injury to the inferior rectal branches of the pudendal nerves, which cross the ischiorectal fossae posterolaterally. Dissection is then carried out cranially in the rectovaginal septum to the level of the puborectalis. The muscle is divided through its midline scar, but the scar is preserved to help prevent the sutures from tearing through.

Step 3: overlapping repair. The tapes on the buttocks are then re leased, and an overlapping sphincter repair is performed with absorb able mattress sutures [see Figure 7, parts b and c]. A snug plication is universally advocated, but unfortunately, there are no generally accepted objective criteria to define exactly what ‘snug’ means in this context. Many authorities advise plication of the puborectalis (so-called levatorplasty) at the cranial aspect of the repair to maximize the length of the anal canal.42 Others favor individual dissection and repair of the internal and external sphincter muscles, but at present, there is no compelling evidence for the superiority of this approach.

Step 4: restoration of perineal body. The skin incision is closed in a V-Y configuration [see 3:7 Surface Reconstruction Procedures] to restore the perineal body and maximize the distance between the anus and the vaginal introitus. The wound is left partially open or closed loosely over small Penrose drains to minimize the risk of surgical site infection [see Figure 7, part d]. A diverting stoma is not generally indicated but may be considered in special situations (e.g., multiple previous failed repairs, Crohn disease, or various chronic diarrheal states).

 

Outcome evaluation Overlapping sphincteroplasty yields substantial clinical improvement in approximately 65% to 80% of patients.43,44 Unfortunately, current data indicate that results deteriorate significantly over time.45–47 When sphincteroplasty fails, repeat EAUS evaluation should be done to confirm that the muscle wrap is intact, and another repair should be performed after 6 to 12 months if a significant defect persists.48 If the muscle wrap is intact, the functional outcome can often be improved by means of biofeedback.49

Various surgical options are available for patients in whom sphincteroplasty has failed or who are not candidates for the procedure (e.g., those with pudendal neuropathy and an anatomically intact sphincter). A number of these options are investigational, and further study is needed to determine their eventual role (if any) in incontinence therapy.

 

Postanal Repair

Sir Alan Parks devised the postanal repair in 1975 to treat patients with incontinence and intact sphincters. The initial results were encouraging but tended to deteriorate over time. Consequently, despite evidence of lasting improvement in some patients, this operation is rarely performed today.50,51

Injectable Biomaterials

A number of studies have explored the use of injectable biomaterials to provide bulk around the anal sphincter and thereby improve continence. The materials employed have included autologous fat, cross-linked collagen, silicone, and carbon-coated beads.52,53 Several small, uncontrolled studies have reported promising results, but larger series with longer follow-up times are needed.

Nonstimulated Muscle Transposition

Attempts to restore continence by creating a neosphincter from transposed skeletal muscle date back to the early 20th century. Most such attempts have made use of either the gluteus maximus54 or the gracilis.55 Good results have frequently been reported, but many authorities believe that the quality of the resulting continence is poor. One of the main limitations of nonstimulated muscle transposition is that patients are typically unable to maintain voluntary contraction of the transposed muscle over the long term.

Stimulated (Dynamic) Graciloplasty

Successful electrical stimulation of a transposed gracilis by means of an implantable pulse generator was first reported in 1988.56 Such stimulation has two main effects. First, it converts the fast-twitch, rapidly fatigable gracilis to a slow-twitch, fatigue-resistant muscle that is capable of tonic contraction for prolonged periods.57 Second, electrical stimulation maintains tonic muscle contraction without the need for continuous voluntary control on the part of the patient. A small number of centers with particular expertise in dynamic graciloplasty and high patient volumes have reported good results with acceptable morbidities58; however, three large multicenter trials have reported less encouraging results with prohibitive morbidities.59–61 In the United States, dynamic graciloplasty is not available, because it has not been approved by the Food and Drug Administration. Elsewhere in the world, the operation can be considered a salvage option at centers with the requisite expertise and experience.

Artificial Anal Sphincter

Figure 8a. Artificial anal sphincter
Figure 8b. Placement of artificial anal sphincter

The artificial anal sphincter is an implantable system consisting of three parts: an inflatable perianal cuff, a pressure-regulating balloon, and a control pump that is implanted in the scrotum or the labia majora [see Figures 8a and 8b]. Good results have been reported in individual case series,62 but device infection has been a problem.63,64 In a large multicenter trial, 46% of patients required surgical revision of the device, including 25% who required revision or explantation because of infection. Of the patients who underwent implantation, 53% had successful results; among those with a functioning device in place, the success rate was 85%.

Sacral Nerve Stimulation

In SNS, an electrode is inserted through a sacral foramen and used to stimulate the sacral nerves. To date, the procedure has been employed mainly in patients with intact anal sphincters (including those with intact repairs). It is available for treatment of fecal incontinence in Europe but has not yet received FDA approval for this indication in the United States.

SNS is generally carried out in two stages. The first stage, peripheral nerve evaluation (PNE), is performed to confirm a muscular response to stimulation of the sacral nerves, to identify the optimal site for stimulation (S2, S3, or S4) and to determine the clinical response to stimulation with an external pulse generator. In most cases, stimulation of the S3 nerves provides the optimal response.

Figure 9a. Sacral nerve stimulation: lead
Figure 9b. Identification of sacral foramina
Figure 9c. Quadripolar lead in position

PNE is performed with the patient prone and under local anesthesia, with or without sedation. The sacral foramina are located by means of bony landmarks; S3 is typically about 1.5 cm off the midline at the level of the sciatic notch.65 Initial testing is performed with an insulated spinal needle and an external pulse generator. Stimulation of each foramen leads to a typical response: S3 causes a bellows-type contraction of the pelvic floor and dorsiflexion of the ipsilateral great toe. Usually, several levels are tested until the optimal site is identified. A temporary pacing wire or a permanent quadripolar lead is then inserted and connected to an external stimulator [see Figures 9a, 9b and 9c].

Patients are asked to provide a baseline continence diary, and a second diary is recorded during the test stimulation period. If continence is significantly improved (e.g., by 50% or more), the second stage of SNS, implantation of a permanent lead (if not already in place) and a pulse generator, is carried out. This second stage is also performed with the patient prone, under local anesthesia, and sedated. The pulse generator is implanted in a subcutaneous pocket on the same side as the stimulating electrode.

Both stages of SNS are performed as outpatient procedures. The pulse generator is activated and its stimulation parameters set by means of a telemetric programmer. If problems (e.g., pain) develop or if the results of stimulation are inadequate, the system can be reprogrammed in a variety of ways: stimulation frequency can be altered, voltage can be increased or de creased, and the configuration of the stimulating electrodes can be modified.

SNS has been shown to be a highly effective treatment for fecal incontinence.66–68 Unlike dynamic graciloplasty and the artificial anal sphincter, SNS is associated with only minimal morbidity. In a multicenter prospective trial, the frequency of incontinent events dropped from 16.4/wk at baseline to 3.1/wk at 12 months after SNS and 2.0/wk at 24 months. Fecal incontinence-related quality of life was significantly improved.

Because of its high success rate and excellent safety profile, many authorities now consider SNS the salvage procedure of choice for patients with refractory incontinence. If SNS fails, more aggressive treatments may still be tried at a later time.

Colostomy

Although creation of a colostomy does not restore continence, it does provide a degree of bowel control in a manner that allows patients to resume their normal activities without fear of accidents. Surprisingly few data are available regarding colostomy for incontinence; however, one questionnaire study of patients who underwent colostomy for incontinence reported extremely high levels of patient satisfaction and marked improvements in subjective quality of life.69 In most cases, a simple end sigmoid colostomy with a Hartmann pouch is the appropriate procedure, and it can often be performed with relatively little operative trauma by using a laparoscopic or minilaparotomy technique. Patients should receive preoperative counseling from an enterostomal therapist, and the optimal stoma site should be marked before the procedure is initiated.

 


Bruce M. Brenner, MD, FACS

Associate Professor, Division of Surgical Oncology
Medical College of Wisconsin

David M. Ota, MD, FACS

Teske Professor and Chief, Division of Surgical Oncology
Medical College of Wisconsin

NCCN Guidelines for Colon Cancer NCCN Guidelines for Rectal Cancer Colorectal cancer (CRC) remains a major public health problem throughout the world. In the United States, CRC is the third most frequently diagnosed cancer in both men and women and the second most common fatal cancer (behind lung cancer).1 During 2004, there were an estimated 106,000 cases of colon cancer and 41,000 cases of rectal cancer in the United States, resulting in 57,000 total deaths.1 The cost of treating colorectal cancer in the United States is believed to be between 5.5 and 6.5 billion dollars a year.2 Worldwide, the risk of death from CRC is highest in developed countries and especially low in Asia and Africa.3

Figure 1. Frequencies of CRC

Data from the Surveillance, Epidemiology, and End Results (SEER) program indicate that the overall incidence of and mortality from CRC have been decreasing in the United States among both men and women,4 though they remain generally higher among men than among women. Overall, the incidence of and mortality from CRC are highest among African Americans, somewhat lower among European Americans, and lowest among Native, Asian, and Hispanic Americans [see Table 1].5 Most CRCs still occur in the distal colon (beyond the splenic flexure), but the incidence of proximal adenocarcinomas relative to that of distal adenocarcinomas has been increasing over the past 25 years [see Figure 1].6 The cause of this shift is not known.

Genetics


Figure 2. Genetic model of colorectal tumorigenesis

The development of CRC involves a progression from normal mucosa through adenoma to carcinoma.7 A genetic model of colorectal carcinogenesis has been proposed that describes a sequence of key mutations driving the process of colorectal carcinogenesis [see Figure 2].8 This process may involve the accumulation of mutations in both tumor suppressor genes and proto-oncogenes, as well as epigenetic phenomena such as DNA hypermethylation or hypomethylation.9 The onset of genomic instability increases the mutation rate and accelerates this progression. Inactivation of the adenomatous polyposis coli (APC) gene on chromosome 5q is thought to be one of the earliest mutations in sporadic cancers and is seen as a germline mutation in patients with familial polyposis [see 5:14 Hereditary Colorectal Cancer and Polyposis Syndromes]. Mutations in other tumor suppressor genes play an important role in this pathway as well, including mutations in DCC, SMAD2, and SMAD4 on chromosome 18q and p53 on chromosome 17p; these events are thought to occur at a later stage of tumor progression. Mutations in the K-ras oncogene occur at an intermediate stage. The accumulation of additional mutations (as yet poorly defined) allows metastases to develop.

Microsatellite instability (MSI) is an alternative pathway to genomic instability and subsequent colorectal carcinogenesis. This phenomenon arises from defects in mismatch repair genes, which cause significantly increased mutation rates in comparison with those in normal cells. MSI in hereditary nonpolyposis colorectal cancer (HNPCC) [see Risk Factors, below] is most commonly attributable to germline mutations in the hMLH1 and hMSH2 genes.10 MSI in sporadic CRC is most frequently associated with hypermethylation of the promoter region of the hMLH1 gene,11 which leads to inactivation of the gene and loss of expression of the hMLH1 protein.

Risk Factors


A number of risk factors for CRC have been described, including a family history of cancer or adenomatous polyps, familial CRC syndromes, inflammatory bowel disease (both ulcerative colitis and Crohn disease), and dietary and lifestyle factors.12,13 The vast majority of CRCs worldwide are sporadic—that is, they are not associated with known genetic syndromes. In the United States, no more than 5% of CRCs are associated with known genetic syndromes.

In a meta-analysis of studies addressing CRC risk and family history, the relative risk of CRC in those with an affected first-degree relative was 2.25; this figure rose to 4.25 if more than one relative was involved and to 3.87 if CRC was diagnosed before the age of 45.14 The National Polyp Study found that the relative risk of CRC was 1.78 in first-degree relatives of patients with adenomatous polyps.15 In another study, the relative risk of CRC was 1.74 in first-degree relatives of patients with adenomatous polyps and was especially high (4.36) in those diagnosed with polyps at or before the age of 50.16

The most common of the genetic syndromes known to be associated with CRC is HNPCC, which accounts for the majority of patients with familial CRC. MSI is the characteristic finding of HNPCC, though it is also present in approximately 15% of all sporadic CRCs. HNPCC can be diagnosed clinically on the basis of what are known as the Amsterdam Criteria.17 Polyposis syndromes (e.g., familial polyposis and juvenile polyposis) account for the remainder of patients with familial CRC syndromes. HNPCC and polyposis syndromes are discussed further elsewhere [see 5:14 Hereditary Colorectal Cancer and Polyposis Syndromes].

As determined by a 2001 meta-analysis, the lifetime risk of CRC for patients with ulcerative colitis is 3.7%, which increases to 5.4% for patients with pancolitis and rises further with greater duration of disease.18 Despite the common misconception, Crohn disease may be associated with a similarly increased risk of CRC.19

Numerous lifestyle and dietary factors have been put forward as potential causes of increased CRC risk. Lower levels of physical activity and increased body mass are associated with an increased risk of CRC in both men and women.20 The Western-style diet, which is high in calories and fat and low in fiber, is associated with high rates of CRC. There is evidence that increased dietary intake of calcium may confer some protection against the development of CRC and adenomatous polyps. The Calcium Polyp Prevention Study, a large randomized trial done in the United States, reported a small but statistically significant reduction in the incidence of recurrent colorectal adenomas with dietary calcium supplementation.21 To date, the evidence from randomized trials has not shown dietary fiber supplementation to have a similar effect. In Japan, where the incidence of CRC has traditionally been low, CRC has become considerably more common in the past few decades.22 This increased incidence is believed to be the result of post—World War II lifestyle changes (e.g., increased consumption of animal fat and decreased expenditure of energy) that mirror Western habits.

Screening


Early diagnosis of colorectal neoplasms at a presymptomatic stage is important for improving survival. Polypectomy has consistently been shown to decrease the subsequent development of CRC: the National Polyp Study found that the incidence of CRC in patients who underwent colonoscopic polypectomy was as much as 90% less than would otherwise have been expected.23 Identifying patients with early-stage disease that has not yet metastasized can prevent many CRC-related deaths. Early detection of and screening for CRC have become important components of routine care and public health programs both in the United States and abroad. The benefits of screening for CRC are especially substantial in patients who are at high risk for CRC (e.g., those with affected first-degree relatives), but even average-risk patients derive some benefit.

There is no ideal method of screening for CRC that is applicable to all patients. Physical examination is generally not helpful in making the diagnosis; various investigative tests are used instead. Modalities commonly employed for CRC screening and early detection include fecal occult blood testing (FOBT), double-contrast barium enema (DCBE), flexible sigmoidoscopy, and colonoscopy. Of these, only FOBT and sigmoidoscopy have been tested in randomized trials.24 It is clear, however, that these tests are less sensitive and specific than colonoscopy. There is evidence that colonoscopy detects many CRCs in asymptomatic patients that would not be detected by sigmoidoscopy.25,26 Colonoscopy has been shown to be a safe and effective method of CRC screening in asymptomatic, average-risk patients.27

Newer screening modalities, such as virtual colonoscopy and stool DNA assays, are currently being developed and tested. Virtual colonoscopy, which uses high-resolution computed tomographic scanning to image the colon, has been evaluated in at least two multicenter trials in the United States, with varying results.28,29 One of the studies reported a sensitivity and a specificity of 89% and 80%, respectively, for polyps larger than 6 mm and up to 94% and 96%, respectively, for polyps larger than 10 mm.28 The sensitivities were equivalent to those of optical colonoscopy in this group of asymptomatic average-risk patients. The second study, however, found that virtual colonoscopy had a sensitivity of only 39% for lesions larger than 6 mm and 55% for lesions larger than 10 mm.29 Given these divergent findings, it appears that there are issues related to equipment, software, and training that remain to be addressed before virtual colonoscopy can be recommended as a routine screening modality. Another consideration is that patients with lesions detected by means of virtual colonoscopy must still undergo optical colonoscopy for treatment or tissue diagnosis. Fecal DNA assays have been developed to test for mutations in multiple genes known to be involved in colorectal neoplasia and are currently being evaluated in clinical trials.30 These assays are not as sensitive as colonoscopy but may be useful in patients who are unable or unwilling to comply with endoscopic screening.31

Many groups have advocated CRC screening, and published guidelines are available from several organizations, including the American Cancer Society,32 the American Gastroenterologic Association,33 and the U.S. Preventive Services Task Force.34 All of these guidelines recommend that screening begin at age 50 for average-risk patients. The recommended screening options are consistent among the various organizations and include (1) FOBT yearly, (2) flexible sigmoidoscopy every 5 years, (3) yearly FOBT and flexible sigmoidoscopy every 5 years, (4) DCBE every 5 years, and (5) colonoscopy every 10 years. In high-risk patients (e.g., those with a family history of CRC), screening may begin at an earlier age—generally, 10 years younger than the age of the affected first-degree relative. There are also specific intensive screening and follow-up regimens for patients with known or suspected familial cancer syndromes.

Clinical Evaluation


As a consequence of the use of screening modalities, patients with CRC are often asymptomatic at diagnosis. Some CRC patients present with occult GI bleeding and anemia. Many patients do not exhibit symptoms until relatively late in the course of the disease. The duration of symptoms, however, is not necessarily associated with the stage of the tumor.35

The most common symptoms of CRC are bleeding per rectum, abdominal or back pain, and changes in bowel habits or stool caliber. Other symptoms are fatigue, anorexia, weight loss, nausea, and vomiting. Some patients present with acute bowel obstruction or perforation.

Staging and Prognosis


Figure 3. Classification of CRC

Accurate staging of CRC is extremely important for determining patient prognosis and assessing the need for adjuvant therapy. Traditionally, staging of CRC has been based on modifications of the Dukes classification, which was initially developed as a prognostic tool for rectal cancer in the 1930s.36 Since this classification was first implemented, it has undergone multiple modifications, of which the most widely used is the modified Astler-Coller system, initially introduced in the 1950s.37 Currently, the TNM classification, developed by the American Joint Committee on Cancer (AJCC) and the International Union against Cancer (UICC), is the preferred staging system [see Tables 2 and 3].38 This system takes into account the depth of penetration into the bowel wall (T) [see Figure 3], the presence and number of involved mesenteric nodes (N), and the presence of distant metastases (M).

Clinical Staging

Clinical staging is based on the history and the physical examination, endoscopic findings, and biopsy results. If colonoscopy cannot be completed, an air-contrast barium enema study should be performed to evaluate the remainder of the colon. Additional staging information may be obtained by means of imaging studies (e.g., roentgenography, CT, magnetic resonance imaging, and positron emission tomography [PET]). A chest x-ray is routinely obtained to rule out metastases and prepare for operation.

There is some debate regarding the utility of preoperative CT scans in the management of primary colon cancer. The rationale for obtaining these scans includes evaluation of potential metastatic disease and assessment of the local extent of disease. In a 2002 study of preoperative CT in patients with intraperitoneal colon cancer, however, the results of the imaging changed management in only 19% of patients, and CT had a sensitivity of only 78% for all metastatic disease.39 Nonetheless, many surgeons routinely perform staging CT in patients with primary colon cancer. PET is a sensitive study, but its routine use for staging primary CRC is not generally recommended. PET may be considered for high-risk patients in whom the detection of metastases would change initial management.40

In cases of rectal cancer, locoregional staging may significantly affect therapeutic decision making. Such staging includes determination of the depth of invasion of the rectal wall and the degree of regional node involvement. Modalities commonly used include CT, MRI, and endoscopic ultrasonography (EUS). In a 2004 meta-analysis that examined the relative utility of each of these studies in rectal cancer staging,41 EUS proved to be the most accurate technique for evaluating muscularis propria involvement and perirectal tissue invasion. The various techniques were equally accurate in assessing lymph node involvement, with none of them being highly sensitive.

Pathologic Staging

Figure 4. Five-year survival rates for colon cancer
Figure 5. Five-year survival rates for rectal cancer
Figure 6. Five-year survival rates: stage II colon cancer

Definitive pathologic staging is carried out after surgical exploration and examination of the resected specimen. The final stage of the cancer is then determined on the basis of the TNM system [see Tables 2 and 3] . Survival is correlated with the stage of the tumor [see Figures 4 and 5] . In the current (sixth) edition of the AJCC staging system,38 stage II is subdivided into stages IIA and IIB, and stage III is subdivided into stages IIIA, IIIB, and IIIC on the basis of both the extent of wall penetration and the number of nodes involved. These changes were implemented as a result of studies demonstrating differences in survival among these subgroups [see Figure 6].42

Numerous other criteria have been evaluated as additional prognostic factors in CRC. The degree of lymphatic invasion and the extent of vascular invasion are important adjuncts to the TNM staging system and are incorporated in the current schema.38 Certain histologic types, including signet-ring and mucinous carcinomas, are associated with poor outcomes. The preoperative serum carcinoembryonic antigen (CEA) level may be an independent prognostic factor that is predictive of resectability and the presence of distant metastases.43

Molecular Markers

Various molecular markers have been investigated with respect to prognosis and response to therapy in CRC patients. Unfortunately, there are conflicting data on the prognostic impact and clinical utility of most of these markers. As noted [see Risk Factors, above], MSI is seen in as many as 15% of patients with sporadic CRC. Patients with MSI typically have proximal, poorly differentiated tumors with mucinous or signet-ring components, but they usually exhibit improved overall survival.44 These patients may be less sensitive to 5-fluorouracil (5-FU)-based chemotherapy.45

The long arm of chromosome 18 (18q) harbors at least three candidate tumor suppressor genes, including DCC, SMAD2, and SMAD4. Deletions of chromosome 18q in CRC patients are associated with decreased survival. One study found that patients with stage II cancers and 18q allelic loss had a prognosis similar to that of patients with stage III disease.46 In addition, p53 mutations and overexpression are associated with poor outcomes in CRC.47 Thymidylate synthase is an enzyme active in DNA synthesis that is targeted by 5-FU and similar chemotherapeutic agents. Overexpression of this enzyme is associated with a poor prognosis but also with improved sensitivity to 5-FU—based chemotherapy.48

All of these molecular alterations, as well as others (e.g., K-ras mutations and 5q deletions), are commonly observed in CRC patients, but further study is required to establish their real prognostic significance.

Management of Colon Cancer


Surgical Therapy

Figure 7. Treatment of colon cancer

Surgery with curative intent remains the mainstay of therapy for colon cancer [see Figure 7]. Complete R0 resection (leaving no gross or microscopic disease) with wide margins along the bowel wall, coupled with regional lymphadenectomy, is the standard of care. The major arterial vessels supplying the segment of the colon containing the tumor should be excised at their origins. A minimum margin of 5 cm of normal bowel on each side of the tumor is considered adequate.

Extent of Resection

The standard extent of resection for various colon cancers has been defined. For tumors of the cecum and the ascending colon, a right hemicolectomy that includes the right branch of the middle colic artery at its origin should be performed. For tumors of the hepatic flexure, an extended right colectomy that includes the entire middle colic artery is indicated. For tumors of the transverse colon, an extended right or left colectomy or a transverse colectomy may be performed. For tumors of the splenic flexure region, a left hemicolectomy is performed, and for sigmoid tumors, a sigmoid colectomy is performed.

In patients who have small or flat tumors or who are undergoing resection after a polypectomy, intraoperative identification of the tumor may be difficult. This is especially true with laparoscopic procedures, in which the bowel often cannot be palpated. If the lesion is in the cecum, the ileocecal valve and the appendiceal orifice are visualized endoscopically, and localization of the tumor is simple. If the lesion is at another location, endoscopic measurements of the distance from the anus or estimates of the location of the tumor may be inaccurate. Endoscopic tattooing, a process in which an agent is injected into the bowel wall submucosally at or near the site of the lesion, has been employed to facilitate intraoperative identification of the tumor site. India ink is the agent most commonly used for this purpose and generally yields excellent results.49 As an alternative, many institutions use a commercially available sterile suspension of carbon particles, which is also very safe and effective.50 Intraoperative endoscopy is another option for locating these lesions.

Surgical Staging

The selection of patients for adjuvant therapy relies heavily on accurate staging. A significant percentage of patients with early-stage node-negative disease present with recurrences or metastases; such a presentation implies that the patients had occult metastatic disease at the time of operation. Surgical resection of CRC should include division of the appropriate mesenteric vessels at their origins, along with resection of the regional nodes. Optimal staging of CRC patients, especially with regard to nodal status, remains controversial. One area of debate is the number of nodes that must be examined to confirm node-negativity. This number depends both on the surgeon’s technique (i.e., how many nodes were resected) and on the pathologist’s efforts to harvest nodes from the specimen. Most groups recommend analysis of at least 12 nodes to confirm node-negativity.51

Because of the importance of nodal status, ultrastaging of harvested nodes with techniques such as serial sectioning, immunohistochemistry (IHC), and reverse transcriptase polymerase chain reaction (RT-PCR) has been proposed as a means of detecting micrometastases. All of these techniques may result in upstaging of patients who are node negative on standard pathologic analysis, which involves only bivalving the nodes and examining a limited number of sections. The prognostic impact of micrometastases that are detected only by IHC or RT-PCR and are not verified by hematoxylin-eosin staining remains unclear. It is impractical to perform these assays on all nodes harvested; accordingly, the use of lymphatic mapping to identify sentinel lymph nodes (SLNs) has been proposed as a means of selecting a small number of nodes for further analysis.

SLN biopsy in the setting of CRC remains investigational. Lymphatic mapping may be done with either in vivo or ex vivo injection of tracer dye. The dye rapidly diffuses through the lymphatic vessels, and SLNs can be identified and marked in the mesocolon within minutes. This procedure has been shown to be feasible in a number of studies52; however, its sensitivity and false negative rates have been variable. In a 2004 multicenter trial, SLN biopsy with serial sectioning had a false negative rate of 54% in patients with node-positive colon cancer.53 In a large single-institution trial, both SLNs and non-SLNs were studied with serial sectioning and IHC in patients who were node negative on routine pathologic analysis54; 19.5% of patients were upstaged by the combination of serial sectioning and IHC of SLNs. These results imply that the main role of this technique may be in upstaging patients who are node negative on routine pathologic analysis. Further study is required before the use of SLN techniques in the context of CRC becomes standard clinical practice.

Occult metastatic disease may also be present in the peritoneal cavity or systemically in the blood or bone marrow at the time of operation. The presence of tumor cells in the peritoneum may be detected by performing cytologic analysis of washings done at the time of operation. In one study, disseminated tumor cells were identified in peritoneal washings or blood in 25% of patients, and their presence was found to be an independent prognostic factor for survival.55 In another study, patients with positive peritoneal washings had significantly higher rates of local recurrence and peritoneal carcinomatosis but manifested no differences in survival.56 Again, further study is required before these assays can be routinely used for staging CRC.

Laparoscopic versus Open Colectomy

At present, open colectomy is the most widely accepted treatment of resectable colon cancer. Laparoscopic techniques have been developed and are being tested in prospective, randomized multicenter trials [see 5:34 Segmental Colon Resection]. Initially, there were concerns about port-site recurrences,57 but current data suggest that these concerns are unfounded.58 With respect to comparative cost, data from a subset of patients in the European COlon cancer Laparoscopic or Open Resection (COLOR) trial demonstrated that although the total cost to society from laparoscopic colectomy is similar to that from open colectomy, the costs to the health care system are significantly higher with the former.59 The Clinical Outcomes of Surgical Therapy (COST) study group, a large, randomized, multicenter trial conducted in the United States, found that laparoscopic-assisted colectomy conferred only minimal (though statistically significant) short-term quality-of-life benefits when compared with open colectomy.60,61 Cancer-specific outcomes (e.g., recurrence rates, wound recurrences, and overall survival rates) were similar with the two approaches.61 The COST investigators concluded that laparoscopic colectomy is an acceptable alternative to open colectomy. Recurrence and survival data from other large multicenter trials (e.g., the COLOR trial62) are not yet available.

Special Situations

Obstructing and perforated cancersObstructing and perforated colon cancers are associated with a poor prognosis and with increased surgical morbidity (as a consequence of the need for emergency surgery). Perforation can occur either via direct erosion of the tumor through the wall of the colon or secondary to obstruction with resultant bowel distention proximal to the tumor. Patients with perforated colon cancer are managed with emergency laparotomy, washout, and resection of the primary lesion to prevent further soilage. A diverting stoma is usually indicated, with either a Hartmann pouch or a mucous fistula constructed distally. Select patients may be managed by means of primary anastomosis, with or without a proximal diverting colostomy or ileostomy.

Obstructing right-side cancers (up to the splenic flexure) can usually be treated with resection and primary anastomosis. The traditional emergency treatment of obstructing left-side colon cancers is a diverting colostomy, with or without resection of the lesion. In many such cases, the stoma is never taken down. Some surgeons advocate emergency treatment of these lesions with total abdominal colectomy and ileorectal anastomosis as a means of improving outcomes.63 Another treatment option is primary resection and anastomosis, with or without on-table intestinal lavage. Yet another option for managing obstructing left-side colon and rectal cancers is the use of colorectal stents with the aim of avoiding emergency surgery. Stents can serve as a bridge to definitive resection by decompressing the colon and thereby allowing subsequent bowel preparation. In patients with advanced disease, stents may also be employed for palliation as an alternative to surgical resection or a diverting stoma.

 

Synchronous primary colorectal cancers The incidence of synchronous CRCs is reported to range from 3% to 5%64,65 but may be as high as 11%.66 Stage for stage, there appear to be no differences in survival between synchronous cancers and single primary cancers.67,68 Synchronous adenomatous polyps are present in as many as 35% of patients undergoing surgical treatment of CRC.65,68 In one study, the presence of synchronous lesions made the surgical procedure more extensive than was initially planned for resection of the primary tumor in 11% of patients.65

Most synchronous polyps are identified on preoperative colonoscopy, and the colon can often be cleared of these lesions before operation. Management of adenomas not amenable to endoscopic resection and management of synchronous cancers are more challenging. Each primary cancer must be managed surgically according to sound oncologic principles. One option is to perform multiple segmental resections with multiple anastomoses. Another is to perform an extended resection that encompasses all of the lesions or even total abdominal colectomy if needed. The presence of a rectal cancer and a second synchronous lesion makes surgical treatment even more challenging, especially if sphincter preservation and a low rectal or coloanal anastomosis are contemplated.

 

Adjuvant Therapy

Significant progress in systemic adjuvant therapy for patients undergoing resection of a colorectal adenocarcinoma has been made in the past 20 years, primarily through a series of phase III randomized trials and the development of new drugs. The evolution of adjuvant therapies is likely to continue for the foreseeable future, and surgeons will play a pivotal role as the primary entrance point for standard adjuvant therapy and new phase III randomized trials. Surgeons’ awareness of past accomplishments, current study findings, and future phase III trials is crucial for improving the survival of potentially cured patients.

The 5-year survival rate after resection of colon cancer is inversely correlated with the pathologic stage [see Figure 4]. The diminishing 5-year survival rates for stage II and III colon cancer became the basis of several phase III randomized trials designed to test the hypothesis that postoperative systemic adjuvant chemotherapy would significantly improve survival in patients with resected but high-risk cancers. Multi-institutional, cooperative cancer group trials were necessary to obtain populations large enough to test this hypothesis. The North Central Cancer Treatment Group (NCCTG) initiated a randomized trial of postoperative systemic adjuvant 5-FU plus levamisole for Dukes stage B and C (AJCC stage II and III) colon carcinomas.69 Patients were randomly assigned to receive either levamisole alone or 5-FU plus levamisole. Overall survival was significantly improved in stage C patients treated with 5-FU plus levamisole. This was the first randomized trial to demonstrate the efficacy of systemic adjuvant therapy.

The NCCTG trial led to second-generation trials of adjuvant therapy for patients with resected colon cancers. In one such study, patients with high-risk stage II or stage III colon cancer were randomly assigned to receive either 5-FU plus leucovorin and levamisole or 5-FU plus levamisole.70 Survival rates after 12 months of adjuvant chemotherapy were no better than those after 6 months of chemotherapy; however, 5-FU plus levamisole proved to be inferior to 5-FU plus leucovorin and levamisole with respect to survival.

National Surgical Adjuvant Breast and Bowel Project (NSABP) protocol C-04 randomly assigned Dukes stage B and C colon cancer patients to receive (1) postoperative 5-FU plus leucovorin, (2) 5-FU plus levamisole, or (3) 5-FU plus leucovorin and levamisole.71 A slight improvement in 5-year disease-free survival was noted with 5-FU plus leucovorin, but overall 5-year survival did not differ significantly among the three treatment arms. Accordingly, 5-FU plus leucovorin became the standard adjuvant regimen.

Intergroup Trial 0089 randomly assigned patients with high-risk stage II and III disease to receive either 5-FU plus high-dose leucovorin or 5-FU plus low-dose leucovorin. The investigators concluded that (1) the high-dose and low-dose regimens were equivalent, (2) a regimen consisting of four cycles of 5-FU with high-dose weekly leucovorin was equivalent to the low-dose leucovorin Mayo Clinic regimen, and (3) the addition of levamisole to the 5-FU plus leucovorin regimen did not improve survival.

These clinical trials have established 5-FU plus leucovorin as standard therapy for patients with high-risk stage II and stage III colon cancer. The next generation of clinical investigations should provide data on the potential benefits of augmenting this regimen with irinotecan or oxaliplatin in an adjuvant setting.72

Routine use of systemic adjuvant therapy for stage II colon cancer remains controversial. Patients with stage II colon cancers, including those at high risk (e.g., those who present with large bowel obstruction or perforation), are typically included in adjuvant chemotherapy trials. A meta-analysis of stage II patients included in NSABP colon cancer trials demonstrated that adjuvant chemotherapy did confer a survival benefit at this disease stage.73 This study was criticized, however, for having included patients from trials that lacked a surgery-only arm, as well as from trials that employed outmoded chemotherapeutic regimens.74 Another meta-analysis, which included only trials that compared 5-FU plus leucovorin with observation after curative resection in stage II patients, found no statistically significant survival benefit with chemotherapy.75 A 2004 meta-analysis formulated recommendations on this controversial topic and provided a Web-based tool for calculating risk.76 This report included data from seven randomized trials that compared surgery alone with surgery plus chemotherapy. Patients with node-negative disease derived a much lower reduction in risk and no statistically significant improvement in overall survival. The authors concluded that the use of postoperative adjuvant chemotherapy for stage II colon cancer patients should be individualized on the basis of the estimated prognosis and the potential treatment benefit.

In summary, postoperative systemic adjuvant therapy is the standard of care in patients with stage III disease. In stage II colon cancer patients who have undergone complete surgical resection, the relative risk of recurrence is small enough that adjuvant chemotherapy yields relatively little benefit in terms of survival. There is, however, a subgroup of patients who have recognized prognostic factors that significantly reduce survival and in whom adjuvant therapy is therefore more likely to be beneficial. These risk factors include (1) bowel obstruction, (2) colonic perforation, (3) high-grade or lymphovascular invasion, and (4) the presence of fewer than 12 lymph nodes in the resected specimen.

Management of Rectal Cancer


Rectal cancer presents special management issues with respect to local recurrence after surgical resection. With cancer of the intraperitoneal colon, local recurrence is rare. With rectal cancer, however, surgical treatment alone results in recurrence rates of 16.2% after low anterior resection (LAR) and 19.3% after abdominoperineal resection (APR).77 Higher stages are associated with higher recurrence rates: 8.5% for Dukes stage A, 16.3% for stage B, and 26% for stage C.77 Multimodality management, including adjuvant radiation therapy or chemotherapy (or both) in combination with appropriate operative therapy, can reduce local recurrence rates significantly.

Surgical Therapy

Extent of Resection

Sphincter preservation has become a major goal in the multimodality treatment of rectal cancer. Surgical procedures are chosen and performed with this goal firmly in mind.

Radical resection

Figure 8. Treatment of rectal cancer

Traditionally, tumors of the rectum have been treated with either LAR or APR [see Figure 8]; in numerous series, APR rates of 60% or higher have been reported. Surgical techniques such as stapled or handsewn coloanal anastomoses [see 5:29 Intestinal Anastomosis].

The morbidity associated with radical rectal resection can be substantial. Anastomotic leakage rates vary widely, ranging from less than 10% to more than 30% after resection with anastomosis. Leaks can lead to substantial morbidity and mortality and can necessitate reoperation. Such concerns have prompted the use of temporary diverting ileostomies or colostomies in patients with low rectal anastomoses. Defunctioning stomas may be overused, however, thereby increasing the cost of care in low-risk patients.79 Preoperative chemoradiation therapy has not been shown to increase anastomotic leakage rates. Urinary and sexual dysfunction are also fairly common after radical resection of rectal cancer. Autonomic nerve preservation in conjunction with TME may improve the functional results of these procedures.80 The use of local resection techniques (see below) is another means of reducing surgical morbidity and mortality in rectal cancer patients.

 

Local excision Local excision-including transanal, transsphincteric, and transcoccygeal techniques, as well as transanal endoscopic microsurgery (TEM) [see 5:35 Procedures for Rectal Cancer]—is another option for curative resection of low rectal cancers with preservation of sphincter function. These procedures were initially implemented for local control in patients who were medically unfit for or unwilling to undergo major resections. Transsphincteric and transcoccygeal resections have been associated with an increased incidence of complications, including fecal fistulas and incontinence, and have largely been abandoned now that other, better techniques are available.

Local excision with curative intent is generally reserved for the treatment of early-stage (T1-2N0) lesions. Selection of patients for these procedures is critical and is based on preoperative staging and on the probability of harboring nodal metastases, which increases with the T stage. EUS has become an important staging procedure in these patients, both for assessing the depth of tissue invasion and for detecting the presence of nodal disease. CT is generally performed to rule out distant metastases. Palliative procedures (e.g., fulguration and endocavitary irradiation) may also be considered in patients who are unfit for major surgery.

Several criteria have been established to identify patients who may be candidates for transanal excision (TAE).81 Generally, the lesion must be no more than 4 cm in diameter, must encompass no more than one third of the circumference of the rectum, and must be less than 8 cm from the anal verge. With the advent of TEM, these criteria have been expanded to include patients with higher lesions. Poorly differentiated tumors and the presence of lymphovascular invasion may also be associated with increased nodal involvement and higher recurrence rates. At least two prospective trials have reported their results with TAE.82,83 Local recurrence rates ranged from 5% to 7% for T1 lesions treated with surgery alone. Results were not as promising for T2 lesions: local recurrence rates ranged from 14% to 16%, even when adjuvant radiation or chemoradiation therapy was provided.

The use of local excision in patients with more locally advanced disease is even more controversial. Such patients are at considerably greater risk for nodal metastases and thus for local recurrence even after adequate resection of the primary lesion. Traditionally, local excision in patients with locally advanced disease has been associated with unacceptably high recurrence rates. Some authors advocate combining chemoradiation therapy with local excision to manage these patients. In a 2004 retrospective series, the results of local excision were comparable to those of radical resection in T3 patients who had a good response to preoperative chemoradiation therapy and who refused or were medically unfit for major surgery.84 The role of local excision in these patients remains poorly defined.

Patients undergoing local resection must receive careful follow-up, including digital examination, measurement of CEA levels, proctoscopy, and, possibly, transanal ultrasonography. A subset of these patients with local-only recurrences who are medically fit for surgery may be candidates for resection. At present, few good data are available on the results of salvage surgery for local recurrence after local excision of rectal cancer, but it is unlikely that outcomes are equivalent to those of initial radical resection.85

 

Importance of Radial and Distal Resection Margins

There has been a great deal of debate about what constitutes an adequate margin of resection in surgical treatment of rectal cancer. With respect to distal margins, 2 to 5 cm has traditionally been considered to be the minimum necessary for curative resection. Growing interest in sphincter preservation has led investigators to consider smaller distal margins (i.e., < 2 cm). Studies have shown that clear margins smaller than 2 cm are not associated with higher local recurrence rates or reduced survival.86 Subsequent reports have suggested that even smaller histologically negative margins (i.e., < 1 cm) may be adequate in patients receiving adjuvant chemoradiation therapy.87,88

The importance of radial margin involvement after rectal cancer resection was not recognized until comparatively recently.89 Radial margins are assessed by means of serial slicing and evaluation of multiple coronal sections of the tumor and the mesorectum.89 Involvement of radial margins is a predictor of both local recurrence and survival after potentially curative rectal cancer surgery90 and may be associated with an increased risk of distant metastases.91 Radial margins smaller than 2 mm are associated with increased local recurrence rates.91 Adjuvant radiation therapy does not compensate for the adverse impact of positive margins on local recurrence rates.92

Adjuvant Therapy

Adjuvant therapy for rectal cancer has focused both on locoregional control of disease and on treatment of systemic disease. Several large studies have evaluated local recurrence of disease after surgical resection alone. Local failure rates of 30% to 40% for T2N0 disease and 50% to 70% for node-positive disease strongly suggested that postoperative adjuvant therapy was needed.93–95 In distinct contrast to these data, however, other series in which TME was performed reported extremely low local recurrence rates with surgery alone.96,97

A series of randomized trials were conducted to assess adjuvant therapy for rectal cancer. Initial studies reported a decrease in local recurrence rates with postoperative radiation therapy.98,99 In a multi-institutional trial conducted by the NCCTG, the combination of 5-FU with radiation therapy led to improvements in local control rates and in survival.100 These results were confirmed in large intergroup trials, the results of which indicated that continuous infusion of 5-FU during radiation therapy resulted in significantly better disease-free survival and overall survival than bolus infusion of 5-FU.

Simultaneously with the ongoing development of postoperative locoregional adjuvant therapy for rectal cancer, interest in preoperative therapy has been growing. Preoperative radiation therapy has been associated with excellent local control of disease, sphincter preservation, and acceptable postoperative recovery. There is evidence that rectal adenocarcinoma is sensitive to preoperative radiation therapy, with or without 5-FU. Pathologic complete response rates of 10% to 20% have been noted in resected rectal specimens101; pathologic complete response is associated with improved outcomes.102

Perhaps the strongest reason to consider preoperative therapy for rectal cancer is its potential for inducing significant tumor regression before surgical resection. Such regression makes clear radial and distal margins easier to obtain. Moreover, tumor regression with preoperative therapy may result in higher sphincter preservation rates. In many published series, the APR rate in rectal cancer patients is between 40% and 60%; more aggressive preoperative efforts to induce regression may give surgeons a better chance to achieve sphincter preservation without compromising local control of disease.101

There remains significant controversy regarding the choice between preoperative and postoperative radiation therapy for rectal cancer. An advantage of the postoperative approach is that the disease is more accurately staged before adjuvant therapy begins, and thus, patients with early-stage disease are less likely to be overtreated. Two trials attempted to compare preoperative and postoperative radiation therapy for rectal cancer in an effort to determine their relative effects on local control, overall survival, and sphincter preservation. Both studies were unsuccessful, however—probably because of bias on the part of the treating physicians in favor of either preoperative or postoperative radiation therapy—and were closed because of slow accrual. At present, there is greater enthusiasm for preoperative therapy and pretreatment staging with transrectal ultrasonography, which is 90% accurate for determining the T stage. Patients with T3 or T4 rectal cancer on ultrasonography would be eligible for preoperative treatment.

Two Swedish studies studied the role of preoperative radiation therapy in treating rectal cancer. The first demonstrated that a short course of preoperative radiation therapy (2.5 Gy in 5 fractions) was comparable to high-dose postoperative radiation therapy (60 Gy over a period of 8 weeks). The local recurrence rate was significantly lower with the short-course preoperative regimen (12% versus 21%), and there was no overall survival difference between the two regimens.103 In the second trial, patients received either a short course of preoperative radiation therapy or surgery alone.104 The local recurrence rate for preoperative therapy plus surgery was 11%, compared with 27% for surgery alone. The combined regimen also resulted in significantly better 5-year survival (58% versus 48%).

The question of the relative merits of preoperative and postoperative radiation therapy may be resolved by the findings from a 2004 German trial that randomly assigned patients to receive either preoperative or postoperative 5-FU plus radiation, followed by systemic 5-FU therapy.78 This study was limited to patients with locally advanced disease, including those who had T3 or T4 disease or were node positive on ultrasonography. TME was performed in all patients and was done 6 weeks after treatment in patients receiving preoperative chemoradiation therapy. The primary end point of this study was overall survival; secondary end points included disease-free survival, local and distant control of disease, sphincter preservation, toxicity of adjuvant therapy, surgical complications, and quality of life. There was no difference between the preoperative group and the postoperative group with respect to 5-year survival, but the local recurrence rate was significantly lower with the former (6% versus 13%), as were both the short-term and the long-term toxicity of adjuvant therapy. Although overall, the rates of complete (R0) resection and sphincter preservation were similar in the two groups, the APR rate was significantly lower in patients determined by the surgeon to require APR before randomization.

Special Considerations


Synchronous Metastatic (Stage IV) Disease

As many as 20% of CRC patients have metastatic disease at the time of initial presentation. The need for surgical intervention in this group of patients is not well defined. Clearly, surgical resection or diversion is indicated in patients who present with significant bleeding, perforation, or obstruction. In asymptomatic patients with unresectable metastatic disease, the role of surgical resection of the primary lesion remains controversial. In patients with resectable metastatic disease (e.g., isolated liver or lung metastases), curative resection may be undertaken.

In a retrospective review of patients presenting with unresectable stage IV CRC, there was no difference in survival between those who were initially managed surgically and those who were initially managed nonoperatively.105 In the surgical group, the morbidity rate was 30% and the mortality 5%. Only 9% of the nonoperative patients subsequently required surgical intervention for bowel obstruction. In another retrospective series, patients managed surgically had significantly better overall survival than those managed nonoperatively but had a lesser tumor burden106; 29% of the nonoperative patients eventually required surgery for bowel obstruction. When prognostic factors were evaluated in the surgical arm of this series, the only factor associated with improved outcomes was a less than 25% extent of liver involvement. On the basis of these and other studies, asymptomatic patients with unresectable metastatic CRC should be managed selectively: those with limited tumor burdens may benefit from surgical treatment, whereas those with more extensive disease (especially extensive liver involvement) may initially be managed nonoperatively.

Management of patients with synchronous resectable isolated liver metastases continues to evolve. Many studies have documented improved survival after liver resection in patients with metastatic disease that is confined to the liver. Patients presenting with synchronous lesions have a worse prognosis than those presenting with metachronous lesions.107 Many of these patients have been managed with staged resections of the primary cancers and the liver metastases. Several groups have reported that such combined procedures do not substantially increase surgical morbidity and mortality or compromise cancer survival.108,109 These combined procedures should be done only in carefully selected patients at specialized centers with significant experience in resection of both CRC and liver tumors.

Peritoneal Carcinomatosis

Peritoneal carcinomatosis develops in approximately 13% of all CRC patients.110 The survival rate of patients who present with peritoneal carcinomatosis from CRC is dismal. In patients with stage IV CRC, the presence of carcinomatosis is associated with a significant reduction in survival (from 18.1 months to 6.7 months).111 Treatment has traditionally included systemic chemotherapy, with surgery reserved for palliation of symptoms such as bowel obstruction. Newer chemotherapy regimens that include agents such as oxaliplatin may improve survival, but they certainly are not curative.

Peritoneal carcinomatosis is often associated with hematogenous metastases, but in some 25% of patients, the peritoneal cavity is the only site of disease. Several groups have advocated the use of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) as a means of improving survival in these patients.112 This treatment, however, is associated with significant morbidity and mortality.113 A randomized trial from the Netherlands that compared cytoreduction surgery plus HIPEC with systemic chemotherapy plus palliative surgery found that patients in the former group exhibited a statistically significant improvement in median survival (22.3 months versus 12.6 months).114 Cytoreductive surgery plus HIPEC seems to be a viable option for the treatment of peritoneal carcinomatosis. Patient selection for these aggressive procedures remains a major issue, given the substantial morbidity and mortality associated with them.

Follow-up and Management of Recurrent Colorectal Cancer


Guidelines for Colonoscopy Surveillance after Cancer Resection The goal of any CRC follow-up regimen should be to detect any recurrences or metachronous lesions that are potentially curable. In a large, multicenter trial, the incidence of second primary CRCs in patients with resected stage II and III lesions was found to be 1.5% at 5 years.115 Between 40% and 50% of patients experience relapses after potentially curative resection of CRC. Detection and treatment of recurrent disease before symptom development may improve survival. The time to recurrence is critical, in that as many as 80% of recurrences occur within the first 2 years and as many as 90% within the first 4 years. Patterns of recurrence should also be taken into account—for example, the markedly increased risk of local recurrence in rectal cancer patients compared with that in colon cancer patients. Even when recurrent CRC is detected, only a small percentage of patients are candidates for reoperation, and resection in these patients may not improve overall survival. Systemic therapy may improve survival in some patients who have unresectable recurrent lesions.

Various modalities are available for follow-up after surgical treatment of CRC. The history and the physical examination continue to be useful, in that a significant percentage of patients present with symptomatic recurrences. Measurement of serum CEA levels has proved effective in detecting asymptomatic recurrences. Other studies, such as liver function tests (LFTs), complete blood count (CBC), chest x-ray, and imaging studies (e.g., CT and ultrasonography), have not been consistently shown to detect asymptomatic resectable recurrences. One study that evaluated routine CEA measurement and CT scanning of the chest, the abdomen, and the pelvis for follow-up of stage II and III CRC demonstrated that both modalities were able to identify asymptomatic patients with resectable disease.116 Colonoscopy is valuable for detecting metachronous cancers and polyps.

Some authorities advocate so-called intensive follow-up. However, this term lacks a standard definition, and such follow-up has not been conclusively shown to be beneficial. In a meta-analysis that compared an intensive follow-up regimen (including history, physical examination, and CEA measurement) with no follow-up, the former detected more candidates for curative re-resection and led to improvements in both overall survival and survival of patients with recurrences.117 Two other meta-analyses have been published that assessed the value of intensive follow-up of CRC patients.118,119 Both of these meta-analyses included only randomized, controlled trials, and both documented a survival advantage with intensive follow-up. Some caution is required in interpreting these results, however, because the meta-analyses included trials with vastly different follow-up regimens in their baseline and intensive groups.

At present, the ideal follow-up regimen for CRC patients remains to be determined. Intensive follow-up regimens obviously are more costly. Patients with stage I disease are at very low risk for recurrence and therefore do not require intensive follow-up.120 Patients with stage II and III disease are at significantly higher risk for recurrence and therefore need more specific cancer-related follow-up, but how intensive such a follow-up regimen should be is still a matter of debate.

Several organizations, including the American Society of Clinical Oncology,121 the National Comprehensive Cancer Network (NCCN),122,123 and the American Society of Colon and Rectal Surgeons,124have developed algorithms for postoperative surveillance of CRC patients. Their recommendations generally apply to patients with stage II or III disease (and sometimes patients with T2 lesions) who are candidates for resection of recurrent disease. The recommendations vary somewhat among groups, but the following are generally agreed on:

  1. Measurement of CEA levels every 2 to 3 months for 2 years, then every 3 to 6 months for 3 years, then annually.
  2. Clinical examination every 3 to 6 months for 3 years, then annually.
  3. Colonoscopy perioperatively, then every 3 to 5 years if the patient remains free of polyps and cancer (the NCCN also recommends colonoscopy 1 year after primary therapy).

 

Imaging studies (e.g., CT and chest x-ray) are not routinely recommended, nor are other blood tests (e.g., CBC and LFTs).

A complete review of the treatment of recurrent CRC is beyond the scope of this chapter. The primary aim of postoperative surveillance is the detection of treatable recurrences or metastatic disease. The most common sites of metastasis in CRC patients are the liver and the peritoneal cavity. Surgery is the only potentially curative therapy for recurrent CRC. Only a select group of patients with isolated peritoneal, liver, or lung metastases are candidates for surgical resection. As noted [see Special Considerations, Peritoneal Carcinomatosis, above], cytoreductive surgery and HIPEC improve survival in patients with peritoneal carcinomatosis and may lead to long-term survival in a very select group of patients.

Numerous studies have addressed the treatment of patients with isolated liver metastases from CRC. Resection of isolated hepatic metastases has been reported to yield 5-year survival rates higher than 30%, with acceptable surgical morbidity and mortality. Investigators from the Memorial Sloan-Kettering Cancer Center developed a staging system known as the clinical score in an attempt to predict which patients are likely to benefit from aggressive surgical resection.125 This system used five factors that were found to be independent predictors of poor outcome: (1) node-positive primary disease, (2) a disease-free interval shorter than 12 months, (3) the presence of more than one hepatic tumor, (4) a maximum hepatic tumor size exceeding 5 cm, and (5) a CEA level higher than 200 ng/ml. Patients who met no more than two of these criteria generally had good outcomes, whereas those who met three or more were recommended for inclusion in adjuvant therapy trials.

PET scanning has also been used to detect occult metastatic disease and thus to aid in the selection of patients for surgical resection. In one series, a 5-year overall survival of 58% was reported after resection of CRC liver metastases in patients screened with PET.126 When combined with the clinical risk score, PET was found to be helpful only in patients with a score of 1 or higher.127

Modalities for treating unresectable disease confined to the liver include cryotherapy, radiofrequency (RF) ablation, hepatic artery infusion of chemotherapeutic agents, and hepatic perfusion. Of these, RF ablation is the one most commonly employed. It may be performed via an open approach, percutaneously, or laparoscopically; it may also be combined with resection and with local or systemic chemotherapy. The survival benefit (if any) associated with use of these modalities has not been well established.

Patients with isolated lung metastases from CRC may also benefit from surgical resection. Because there are relatively few of these patients, treatment of such metastases has not been studied as well as treatment of liver metastases. Some series have reported 5-year survival rates higher than 40% after complete resection. Patient selection remains a major issue. Several prognostic factors that may predict poor outcomes have been identified, including (1) a maximum tumor size greater than 3.75 cm, (2) a serum CEA level higher than 5 ng/ml, and (3) pulmonary or mediastinal lymph node involvement.128,129 Patients with both pulmonary and hepatic metastases may also be considered for surgical resection.

Pelvic recurrences of rectal cancer present another difficult management issue. These tumors may cause significant pain and disability, and if they are not treated, survival is measured in months. Radiation and chemotherapy provide symptomatic relief and yield a modest increase in survival. Surgery may provide excellent palliation and is potentially curative in patients who do not have distant metastases.

Multimodality therapy has been advocated as a means of improving the chances of cure. In one study, a 37% 5-year survival rate was reported in patients who underwent multimodality therapy, including resection with negative margins.130 A subgroup of patients in whom complete resection was impossible underwent intraoperative radiation therapy; the 5-year survival in this subgroup was 21%. Several predictors of poor outcomes were identified, including incomplete resection, multiple points of tumor fixation, and symptomatic pain. In another series, hydronephrosis was associated with the presence of unresectable disease.131 Selection of appropriate patients for curative surgery remains a major issue in the management of locally recurrent rectal cancer.

Chemotherapy is the mainstay of palliative treatment for patients with CRC and unresectable recurrent or metastatic disease. Combinations of 5-FU and leucovorin with newer agents such as irinotecan and oxaliplatin define the current standard. Patients in whom these regimens fail may be considered for treatment with other newer agents, including cetuximab, a monoclonal antibody against epidermal growth factor receptor, and bevacizumab, a monoclonal antibody against the vascular endothelial growth factor receptor.

 


Roger D. Hurst, MD, FACS, FRCS Ed

Associate Professor, Department of Surgery
University of Chicago Pritzker School of Medicine

Fabrizio Michelassi, MD, FACS

Lewis Atterbury Stimson Professor and Chairman, Department of Surgery
Joan & Sanford I. Weil Medical College of Cornell University

Fulminant ulcerative colitis is a potentially life-threatening disorder that must be expertly managed if optimal outcomes are to be achieved. This condition was once associated with a very high mortality,1 but medical and surgical treatments have improved dramatically, to the point where the mortality associated with fulminant ulcerative colitis is now lower than 3%.2,3 Optimal management depends on close coordination between medical and surgical therapy, and multidisciplinary strategies are essential.

Classification


The most commonly applied system of classifying the severity of ulcerative colitis has been the one devised by Truelove and Witts, who identified clinical parameters by which colitis could be categorized as mild, moderate, or severe.4 The Truelove-Witts classification does not, however, specify a unique category for fulminant disease. Accordingly, Hanauer modified this classification scheme to include a category for fulminant colitis [see Table 1].5 Unfortunately, there is no universally agreed upon distinction be tween severe ulcerative colitis and fulminant ulcerative colitis.6 Some authors use the terms severe and fulminant interchangeably, whereas others, concerned about the lack of a clear distinction between the two, recommend that the term fulminant ulcerative colitis be avoided altogether.7 This latter recommendation has not been widely followed: the term fulminant ulcerative colitis remains an established component of the medical vernacular, the absence of a clear definition notwithstanding.8–10

Fulminant ulcerative colitis is certainly a severe condition that is associated with systemic deterioration related to progressive ulcerative colitis. Most authorities would agree that a flare of ulcerative colitis can be considered fulminant if it is associated with one or more of the following: high fever, tachycardia, profound anemia necessitating blood transfusion, dehydration, low urine output, abdominal tenderness with distention, profound leukocytosis with a left shift, severe malaise, or prostration. Patients with these symptoms should be hospitalized for aggressive resuscitation while clinical assessment and treatment are being initiated.11

Clinical Evaluation


When a patient is admitted with severe or fulminant ulcerative colitis, a complete history and a thorough physical examination are required. The abdominal examination should focus on signs of peritoneal irritation that may suggest perforation or abscess formation. Any patient admitted with severe ulcerative colitis may have already received substantial doses of corticosteroids, which can mask the physical findings of peritonitis.

Investigative Studies


Laboratory Tests

Initial laboratory studies should include a complete blood count with differential, a coagulation profile, and a complete metabolic profile with assessment of nutritional parameters (e.g., serum albumin concentration). Multiple stool specimens should be sent to be tested for Clostridium difficile, cytomegalovirus, and Escherichia coli 0157:H7.12,13 It is important to rule out the presence of opportunistic infections, particularly with C. difficile, even in patients with an established diagnosis of ulcerative colitis; superinfection with C. difficile is common in such patients.

Imaging

Abdominal films and an upright chest x-ray should be obtained to look for colonic distention (indicative of toxic megacolon) and free intraperitoneal air (indicative of perforation).

Figure 1. Sigmoidoscopy reveals deep ulcerations

Endoscopic evaluation of the colon and rectum in the presence of fulminant ulcerative colitis is a controversial measure.14–16 Undoubtedly, colonoscopy with biopsy can provide useful diagnostic information in this setting, and numerous reports indicate that in experienced hands, colonoscopy poses little risk to patients with severe colitis.14,15 In general, however, it is recommended that endoscopic examination proceed no further than the minimum distance necessary to confirm severe colitis. If an endoscopic examination is to be performed, insufflation of air must be minimized; overdistention of the colon may lead to perforation or the development of megacolon. In addition to diagnostic information, endoscopy can provide useful prognostic information. In one study, the presence of deep, extensive colonic ulcerations indicated a low probability for successful medical treatment of fulminant ulcerative colitis: fewer than 10% of patients with such ulcerations responded to medical measures.14 Thus, an endoscopic finding of deep ulcers [see Figure 1] may facilitate the decision to proceed with early operative treatment if medical therapy does not lead to rapid and significant improvement.

Management


General Care

All patients with fulminant ulcerative colitis should be hospitalized. Blood products should be administered to treat significant anemia or coagulopathy. Metabolic derangements should be corrected.17 Patients with a perforation or massive lower GI hemorrhage are taken to the operating room for emergency surgical treatment; more stable patients are initially managed with medical therapy. Narcotics, antidiarrheal agents, and other anticholinergic medications should be avoided because they can precipitate toxic dilation of the colon.

Bowel rest typically reduces the volume of diarrhea, but whether it affects the clinical course of the fulminant colitis remains to be established.18,19 One study of patients with acute flares of ulcerative colitis reported no significant difference in outcome between those who were managed with total parenteral nutrition (TPN) and bowel rest and those who received enteral nutrition. This study, however, included patients with colitis of varying degrees of severity, and apparently, only a small number of them had fulminant ulcerative colitis.19 A subsequent study found that bowel rest and TPN did have a potential clinical advantage in patients with fulminant ulcerative colitis.18 The most common approach to nutritional management of these patients is first to place them on bowel rest with hyperalimentation, then to initiate oral feeding once the symptoms of the fulminant attack begin to be alleviated. Whether patients are being maintained on bowel rest or are receiving oral feedings, adequate nutritional support must always be ensured. Hence, TPN, if employed, should be maintained until the patient is receiving and tolerating full enteral feedings.

Medical Therapy

The standard medical approach to fulminant ulcerative colitis involves induction of remission by means of I.V. corticosteroid therapy, followed by long-term maintenance treatment (in the form of purine analogues) once remission has been achieved. If treatment with steroids fails to induce remission, I.V. cyclosporine therapy is considered.

Steroids

For decades, steroid treatment has been the frontline therapy for acute flares of ulcerative colitis. Response rates in cases of fulminant ulcerative colitis range from 50% to 60% when the steroids are given over a period of 5 to 10 days.20,21 Methylprednisolone, 40 to 60 mg/day in a continuous I.V. infusion, is a common regimen.5,22–24

The length of time that should be allowed for patients to respond to I.V. steroid therapy has been a subject of debate. In 1974, Truelove and Jewell recommended urgent operative treatment after 5 days if there is no response to I.V. steroid therapy.25 This 5-day rule has been widely adopted, but more recent experience suggests that steroids can be safely administered for as long as 7 to 10 days to allow patients more time to respond.12 Patients who respond to I.V. steroid therapy are switched to an oral steroid regimen (typically prednisone). It is important, however, to stress that corticosteroids should never be employed as long-term maintenance therapy.5,26 The toxic effects of corticosteroids are related to not only the dosage but also the duration of treatment. Severe complications are common with extended use of even modest doses of steroids. Accordingly, patients should be slowly but completely weaned from steroid therapy. Because symptomatic colitis recurs in 40% to 50% of patients who initially respond to I.V. therapy, maintenance therapy with either purine analogues or immuno salicylates should be instituted.5,20

Unfortunately, corticosteroid dependency is frequently encountered in patients with ulcerative colitis. Often, the steroid dosage cannot be tapered without an increase in disease activity and exacerbation of symptoms. In such cases, if the patient cannot be weaned from steroids and switched to purine analogues within 3 to 6 months, surgical consultation is indicated. In addition, if complications related to ulcerative colitis or to corticosteroid therapy develop, the colitis must be treated surgically.

Cyclosporine

At one time, patients who did not respond to I.V. steroid treatment were invariably referred for surgical treatment. Currently, such patients are most often treated with I.V. cyclosporine therapy. Cyclosporine is an immunosuppressant macrolide that suppresses the production of interleukin-2 by activated T cells through a calcineurin-dependent pathway.27 Originally employed to prevent tissue rejection after transplantation, cyclosporine has become the standard treatment of steroid-refractory severe ulcerative colitis.

The first report of the use of cyclosporine to treat ulcerative colitis was published in 1984.28 It was not until 10 years later, however, that a randomized, placebo-controlled trial of cyclosporine therapy for steroid-refractory ulcerative colitis convincingly demonstrated the effectiveness of cyclosporine in this setting.29 In this trial, patients with steroid-refractory ulcerative colitis who received cyclosporine (4 mg/kg) had an 82% response rate, compared with a 0% response rate in those treated with continued I.V. steroid therapy alone. Since this initial report, response rates ranging from 56% to 91% have been reported in the medical literature, confirming cyclosporine as a major advance in the treatment of severe and fulminant ulcerative colitis.30–32

The beneficial effects of cyclosporine therapy are not always durable: as many as 60% of patients experience recurrence of disease after initial cyclosporine-induced remission.33 Fortunately, recurrence rates can be substantially lowered by means of maintenance therapy with 6-mercaptopurine (6-MP) or azathioprine. With appropriate maintenance therapy, the rate of early recurrence of symptoms after successful I.V. cyclosporine treatment may be reduced to levels as low as 22%.31,34 Even if the disease does recur, the initial success of cyclosporine therapy in aborting the acute phase of the ulcerative colitis allows patients to recover from the acute illness, so that they are in better condition to undergo elective surgical treatment at a later date if such treatment ultimately proves necessary. This is a major benefit, in that operative management of ulcerative colitis carries a much higher risk of complications when carried out on an urgent basis than when carried out in an elective setting.18,35,36

The major side effects of cyclosporine treatment are renal insufficiency, opportunistic infections, and seizures. The risk of seizures appears to be highest in patients with hypocholesterolemia. Consequently, cyclosporine should not be given to patients with significant hypocholesterolemia (serum cholesterol concentration < 100 mg/dl). Hypomagnesemia is commonly seen in patients with fulminant ulcerative colitis who undergo cyclosporine treatment; accordingly, serum magnesium levels should be closely followed.

Dosing regimens for cyclosporine vary. The typical starting dosage is 4 mg/kg/day I.V., which is then adjusted to achieve a whole-blood level between 150 and 400 ng/ml, as measured by high-power liquid chromatography or radioimmunoassay.37,38 Whole-blood levels as high as 800 ng/ml are considered acceptable by some investigators.29 If the patient shows no improvement within 4 to 5 days or if complete remission is not achieved by 10 to 14 days, surgical treatment is advised.12 Most of the side effects of cyclosporine therapy are dose dependent. Several studies have shown that an initial dosage of 2 mg/kg/day I.V. can also be effective in achieving remission.39–41 Some physicians prefer to begin at this lower dosage and then increase it as necessary on the basis of the measured cyclosporine levels.

Concerns have been raised about the possibility that prolonging medical therapy in patients with severe colitis who have already received large amounts of corticosteroids may increase the risk of perioperative morbidity and mortality in those who respond to neither steroids nor cyclosporine therapy and thus require operative management. At present, however, there is no evidence that patients who do not respond to cyclosporine therapy are at increased risk for perioperative complications; such therapy does not appear to compromise surgical results.42

Surgical Therapy

Indications

Indications for surgical treatment of fulminant ulcerative colitis have been established [see Table 2]. One such indication, of course, is the exhaustion of options for appropriate medical treatment. Because most patients with fulminant ulcerative colitis respond to aggressive medical therapy, such treatment is warranted in almost all cases. Care must be exercised, however, not to overtreat patients with fulminant ulcerative colitis who are otherwise stable. The immunosuppressive effects of high-dose cortico steroids and I.V. cyclosporine, along with the debilitation induced by prolonged severe disease, can place patients at high risk for perioperative complications. Patients who do not show significant improvement in response to I.V. steroid therapy within 5 to 7 days should be started on I.V. cyclosporine therapy or referred for operative treatment.12 Those who do not respond to cyclosporine therapy within 4 days or in whom remission of major symptoms is not achieved within 2 weeks should be treated surgically. Patients whose symptoms progress during the course of I.V. therapy or who show no sign of improvement at all should be considered for early surgery. Patients known to have deep longitudinal ulcerations are less likely to respond to I.V. medical therapy and thus may also be referred for early surgery. The decision regarding when to abandon medical therapy for fulminant ulcerative colitis in favor of surgical therapy is difficult and requires considerable experience and special expertise. Accordingly, patients with fulminant ulcerative colitis are best managed in a center specializing in inflammatory bowel disease.

Patients with perforation or severe GI bleeding require urgent surgical treatment.43 The debilitation resulting from the disease, coupled with the immunosuppression resulting from intensive medical therapy, can mask the signs and symptoms of sepsis and peritonitis associated with perforation. Perioperative mortality in cases of fulminant colitis is as much as 10 times higher when perforation occurs than when it does not.44 For this reason, patients with high fever, marked leukocytosis, and persistent tachycardia should be referred for early surgery, regardless of whether other indications of perforation or peritonitis are noted.

Toxic megacolon, though an uncommon complication of severe ulcerative colitis, is important in that it is associated with impending colonic perforation and therefore must be watched for and aggressively managed if present. Two specific conditions must be satisfied to establish the diagnosis of toxic megacolon.45 First, there must be colonic dilatation; second, the patient must be in a toxic state. Patients with mild symptoms of ulcerative colitis may experience a degree of colonic dilatation, perhaps in conjunction with colonicileus. This condition is distinctly different from and considerably less worrisome than toxic megacolon. Patients admitted to the hospital with fulminant ulcerative colitis will, by definition, exhibit some degree of toxicity. Colonic dilatation in these patients is a very worrisome phenomenon, in that it completes the picture of toxic megacolon. Accordingly, in all patients with fulminant ulcerative colitis, an abdominal x-ray should be obtained to look for colonic dilatation. Those in whom abdominal distention develops or who experience a sudden decrease in the number of bowel movements without signs of significant clinical improvement should also be assessed for colonic dilatation and toxic megacolon.

In patients with toxic megacolon who are otherwise stable, conservative management, consisting of elimination of narcotics and anticholinergic agents, may be briefly tried. Changes in patient position may be tried as well: moving the patient from side to side, from supine to prone, and into the knee-elbow prone position is thought to facilitate expulsion of colonic gas.46 Patients with toxic megacolon should be kept on a nihil per os (NPO) regimen, and broad-spectrum I.V. antibiotics should be given. Endoscopic decompression is to be avoided. Blind placement of rectal tubes is ineffective and may be harmful. Patients who do not rapidly respond to conservative management and those who show signs of peritonitis or are otherwise unstable require urgent surgical treatment.47

Preparation for Operation

With patients who are stable but are not responding to medical therapy, there may be time for preoperative preparation. Patients who are not on NPO status should be maintained on clear liquids, then kept on an NPO regimen for 6 to 8 hours before operation. On occasion, a patient may be able to tolerate mild bowel preparation with either polyethylene glycol or Fleet Phospho-soda (Fleet Pharmaceuticals, Lynchburg, Virginia). Any bowel preparations that are used need be employed only until bowel movements are free of residue. If time allows, the patient should be counseled by an experienced enterostomal therapist, and an optimal site for the ostomy should be marked on the abdomen. Prophylactic antibiotics should be given before the the surgical incision is made, and appropriate stress-dose steroids should be administered.

Surgical Strategies

The operative strategies for treating fulminant ulcerative colitis are controversial. Ultimately, almost all patients end up undergoing a restorative proctocolectomy with ileoanal anastomosis [see 5:33 Procedures for Ulcerative Colitis]. In most cases, however, the final surgical goal is achieved in multiple steps. Performing an extensive resection in conjunction with a prolonged and delicate reconstruction in an acutely ill patient is a procedure of questionable safety. Accordingly, many surgeons elect first to perform a total abdominal colectomy with an ileostomy, leaving the rectal stump as either a Hartmann pouch or a mucous fistula,43,44 then to perform a restorative proctectomy with ileoanal anastomosis at a later date. This staged approach allows the patient to recover from the acute illness, to be weaned from immunosuppressive agents, and to achieve improved nutritional status. Although the remaining rectal stump continues to be affected by ulcerative colitis, the fecal diversion greatly diminishes disease activity, so that almost all patients can be completely weaned from steroids and other immunosuppressive medications. It is then possible to perform the proctectomy with ileoanal anastomosis in more controlled conditions.

The exact circumstances in which it is best to follow a staged approach have not been clearly defined. It is universally accepted that a staged procedure is mandatory in patients with perforation, peritonitis, or sepsis, but beyond this point, there is no clear consensus. The studies published to date have been inconclusive on this issue: either they included only a small number of patients, they did not clearly define what constituted fulminant colitis, or they did not directly compare the results of the two alternative strategies (i.e., staged colectomy and immediate ileoanal anastomosis). A 1995 study reported excellent long-term results and acceptable short-term morbidity in 12 patients undergoing immediate restorative proctocolectomy with ileal pouch-anal anastomosis (IPAA) for fulminant colitis.48 These 12 patients, however, represented an extraordinarily small percentage of the total number of ileoanal procedures performed by the authors. In addition, this study used a somewhat liberal definition of fulminant colitis and thus might have included a number of cases that would not have qualified as fulminant colitis—or, possibly, even as severe colitis—according to the criteria cited earlier [see Table 1]. Finally, the study provided no data on the experience of patients undergoing a staged procedure for the management of severe or fulminant colitis.

A 1994 study also reported excellent long-term results and exceptionally low perioperative morbidity in 20 patients undergoing restorative proctocolectomy with IPAA for urgent treatment of ulcerative colitis.49 Another study from the same year, however, reported a 41% anastomotic leakage rate in 12 patients also undergoing an urgent ileoanal procedure for ulcerative colitis, compared with an 11% leakage rate in patients undergoing ileoanal anastomosis under more controlled conditions.50 On the basis of these results, the authors counseled against ileoanal anastomosis in the urgent setting. A later study also noted a higher incidence of anastomotic leakage (36%) in patients undergoing urgent ileoanal anastomosis.51 These authors likewise advised against ileoanal anastomosis in the urgent setting.

The fact of the matter is that the distinction between severe and fulminant ulcerative colitis may be little more than an academic exercise. At one end of the disease spectrum, there is a small subset of patients who have symptoms severe enough to necessitate hospitalization yet are healthy enough to undergo a primary ileoanal anastomosis without undue risk. At the other end of the spectrum, there is a subset of severely ill patients with fulminant colitis for whom a staged procedure is mandatory. The middle of the spectrum remains something of a gray area. Because specific criteria for quantifying the risk have not been defined, the decision whether to follow a staged operative approach ultimately is made on the basis of the experienced surgeon’s clinical judgment. It has been our experience, however, that the majority of patients who fit the criteria of fulminant colitis [see Table 1] and who do not respond to maximal medical therapy are best managed with a staged approach.

Technical Considerations

Figure 2. Subtle changes on serosal aspect

Surgical exploration is performed via either a midline or a transverse incision. The abdomen is carefully examined, with particular attention paid to the small intestine in an effort to detect any signs of Crohn disease. The colon often shows the changes typical of colitis: serosal hyperemia, corkscrew vessels, and edema [see Figure 2].

Figure 3. Hartmann pouch

Colectomy may be performed in the standard fashion, with mesenteric division occurring at a convenient distance from the bowel; wide mesenteric resection is not necessary. If a staged colectomy is to be performed, the colon is removed, and the rectum is left either as a Hartmann pouch or as a mucous fistula. In most cases, a Hartmann pouch can safely be created. In the construction of a Hartmann pouch, it is important that the stump be of the appropriate length. If the stump is too short, the proctectomy to be performed in the second stage may prove very difficult; if it is too long, there is an increased risk of complications related to persistent disease in the rectum (e.g., bleeding, discharge, and tenesmus). Ideally, the Hartmann pouch should be made at the level of the sacral promontory [see Figure 3]. During the colectomy, the sigmoid branches of the inferior mesenteric artery should be divided and the terminal branches of the inferior mesenteric artery preserved. This measure ensures a good blood supply to the remaining rectal stump and helps the Hartmann closure to heal. Preservation of the terminal branches of the inferior mesenteric artery and the superior rectal artery also simplifies the subsequent proctectomy by keeping the pelvic sympathetic nerves free of surrounding scar tissue and by providing a key anatomic landmark that will assist the surgeon in locating the appropriate presacral dissection plane for any future planned proctectomy.

To create the Hartmann pouch, the mesenteric and pericolonic fat are removed from the bowel wall for a distance of approximately 2 cm. A transverse anastomosis (TA) stapler loaded with 4.8 mm staples is placed on the prepared bowel and fired to close the pouch. The bowel is then divided proximal to the staple line. The staple line should be closely examined to confirm that the staples are closed properly into two rows of well-formed Bs and that individual staples are not cutting into the muscularis propria of the bowel. To provide additional protection against dehiscence, the staple line may be oversewn with interrupted Lembert sutures [see Figure 3]. If sutures are employed, they should be carefully placed so that the anterior and posterior serosal surfaces are approximated without undue tension. With a well-constructed Hartmann pouch, pelvic drains are unnecessary and may even be harmful, in that they can promote dehiscence if situated close to the suture line.

Figure 4. Surgical specimen showing severe ulceration

In some cases, the colon at the level of the sacral promontory is affected by deep ulcerations and severe inflammation, to the point where closure of the Hartmann pouch at this level poses an unacceptably high risk of dehiscence [see Figure 4]. If the severity of disease precludes safe closure of the Hartmann pouch, creation of a mucous fistula should be considered. A mucous fistula requires a longer segment of bowel than a Hartmann pouch does and thus is associated with a higher risk of bleeding from the retained segment. In addition, a mucous fistula is unsightly and often generates a very foul odor. As a compromise approach, some surgeons advocate creating a Hartmann pouch of moderate length and placing the proximal end of the stump through the fascia at the lower edge of the midline incision; the end of the stump is then left buried in the subcutaneous tissue. The benefit of this approach is that if dehiscence of the staple line occurs, any ensuing infection is limited to the subcutaneous space and does not result in an intra-abdominal or pelvic abscess.

If attempts to fashion a secure Hartmann closure fail and the remaining rectal stump is too short to be brought out as a mucous fistula, the proximal rectum should be resected, and closure of the Hartmann pouch should be performed just below the peritoneal reflection. In this situation, closed suction drains should be placed deep in the pelvis, and the peritoneum should be closed over the rectal stump. Such a short Hartmann pouch, however, will be more difficult to locate during the subsequent restorative proctectomy and ileoanal anastomosis.

With a staged colectomy, an end ileostomy is created in the standard fashion [see 5:30 Intestinal Stomas], and the abdomen is closed. Placing a rectal tube to drain rectal secretions may be beneficial in reducing the risk of dehiscence of the Hartmann pouch.

Laparoscopic Approaches

Figure 5. Mobilization of splenic flexure

Experience has demonstrated that laparoscopic-assisted approaches to abdominal colectomy can be safely employed in patients with ulcerative colitis.52 Mobilization of the colon and division of the mesentery can be accomplished laparoscopically [see Figure 5], with the specimen being removed through a small Pfannenstiel incision. An end ileostomy can also be fashioned with the aid of inspection through the Pfannenstiel incision. Alternatively, the Pfannenstiel incision can be made early in the procedure and used for placement of a hand port, and the colon can be removed by means of a hand-assisted laparoscopic approach.

Whether a laparoscopic-assisted approach to the management of fulminate ulcerative colitis possesses any significant clinical advantages remains to be determined. However, a growing body of experience with this approach indicates that in experienced hands, laparoscopic-assisted colectomy is a safe and reasonable alternative that may well result in shorter hospital stays and decreased postoperative pain. The laparoscopic-assisted approach may therefore be considered as an option for patients with fulminant ulcerative colitis. Patients with toxic megacolon, however, should be managed by means of an open surgical approach; the instruments used to grasp the bowel in a laparoscopic-assisted colectomy are likely to cause perforation of the severely thinned walls of the dilated megacolon.


John P. Welch, M.D., F.A.C.S.

Clinical Professor of Surgery
University of Connecticut School of Medicine
Adjunct Professor of Surgery
Dartmouth Medical School

Jeffrey L. Cohen, M.D., F.A.C.S., F.A.S.C.R.S.

Associate Clinical Professor
University of Connecticut School of Medicine
Adjunct Assistant Clinical Professor of Surgery
Dartmouth Medical School

Figure 1. Colon segment containing diverticula

Diverticula are small (0.5 to 1.0 cm in diameter) outpouchings of the colon that occur in rows at sites of vascular penetration between the single mesenteric taenia and one of the antimesenteric taeniae. At the sites of most diverticula, the muscular layer is absent [see Figure 1]. Technically, such lesions are really pseudodiverticula; true diverticula (which are much less common than pseudodiverticula) involve all layers of the bowel wall. Nevertheless, both pseudodiverticula and true diverticula are generally referred to as diverticula.

The sigmoid colon is the most common site of diverticula: in 90% of patients with diverticulosis, the sigmoid colon is involved.1 If a diverticulum becomes inflamed as a result of obstruction by feces or hardened mucus or of mucosal erosion, a localized perforation (microperforation) may occur—a process known as diverticulitis. The incidence of diverticulitis is about 10% to 25% in patients with colonic diverticula.1 Both diverticulosis and variants of diverticulitis may be subsumed under the more encompassing term diverticular disease.

The incidence of diverticular disease increases with age. Diverticula are quite common in elderly patients, being present in more than 80% of patients older than 85 years.2 Consequently, as the population of the United States continues to age, the overall risk of diverticular complications continues to increase.3 Before the 20th century, diverticular disease was rare in the United States. By 1996, however, 131,000 patients were being admitted to hospitals with diverticulitis each year.4

Figure 2. Schematic representation: segmentation in the colon

A diet containing refined carbohydrates and low-fiber substances, such as is currently widespread in many developed countries (especially in the West), has been associated with the emergence of this disease entity.5,6 A low-residue diet facilitates the development of constipation, which can lead to increased intraluminal pressure in the large bowel. In addition, elevated elastin levels are commonly noted at colon wall sites containing diverticula,7 and this change causes shortening of the taeniae.1 High-pressure zones or areas of segmentation may develop [see Figure 2], usually in the sigmoid colon, and diverticula begin to protrude at these locations. If microperforation of a thin-walled diverticulum takes place, local or, sometimes, widespread contamination with fecal organisms may ensue. The pericolonic tissue (typically, the mesentery and the pericolic fat) thus becomes inflamed while the mucosa tends to remain otherwise normal.

Several factors appear to promote the development of diverticular disease and its complications, including decreased physical activity,8 intake of nonsteroidal anti-inflammatory drugs (NSAIDs),9,10 smoking,11 and constipation from any cause (e.g., diet or medications). The well-known Western afflictions cholelithiasis, diverticulosis, and hiatal hernia frequently occur together (Saint’s triad). Obesity has been associated with the intake of low-fiber diets,12 and growing numbers of young, obese patients with diverticulitis are being seen by physicians.

Clinical Evaluation


History

Uncomplicated (Simple) Diverticulitis

The classic symptoms of uncomplicated acute diverticulitis are left lower quadrant abdominal pain, a low-grade fever, irregular bowel habits, and, possibly, urinary symptoms if the affected colon is adjacent to the bladder. If the sigmoid colon is highly redundant, pain may be greatest in the right lower quadrant. Diarrhea or constipation may occur, together with rectal urgency.

Figure 3. Napkin-ring carcinoma

The differential diagnosis includes gynecologic and urinary disorders, perforated colon carcinoma, Crohn disease, ischemic colitis, and, sometimes, appendicitis. Chronic diarrhea, multiple areas of colon involvement, perianal disease, perineal or cutaneous fistulas, or extraintestinal signs are suggestive of Crohn disease. Rectal bleeding should raise the possibility of inflammatory bowel disease, ischemia, or carcinoma; such bleeding is uncommon with diverticulitis alone. Given the prevalence of diverticula, it is not surprising that colon carcinoma may coexist with diverticular disease [see Figure 3].

Complicated Diverticulitis

Figure 4. Major complications of diverticular disease

Some cases of diverticulitis are classified as complicated, meaning that the disease process has progressed to obstruction, abscess or fistula formation, or free perforation [see Figure 4]. Complicated diverticulitis may be particularly challenging to manage,13,14 especially because patients may have no known history of diverticular disease.15 Lower gastrointestinal bleeding is also a complication of diverticular disease in 30% to 50% of cases16; in fact, diverticula are the most common colonic cause of lower GI bleeding.16 When diverticular hemorrhage occurs [see 5:6 Lower Gastrointestinal Bleeding ], it is usually associated with diverticulosis rather than with diverticulitis. Approximately 50% of diverticular bleeding originates in the right colon, despite the low incidence of diverticula in this segment of the colon. Patients tend to be elderly13 and to have cardiovascular disease and hypertension. Regular intake of NSAIDs may increase the risk of this complication. Although patients may lose 1 to 2 units of blood, the bleeding usually ceases spontaneously,17 and expeditious operative treatment generally is not necessary.

Figure 5. Hinchey classification

The most common form of complicated diverticulitis involves the development of a pericolic abscess, typically signaled by high fever, chills, and lassitude. Such abscesses may be small and localized or may extend to more distant sites (e.g., the pelvis). They may be categorized according to the Hinchey classification of diverticular perforations,18 in which stage I refers to a localized pericolic abscess and stage II to a larger mesenteric abscess spreading toward the pelvis [see Figure 5]. On rare occasions, an abscess forms in the retroperitoneal tissues, subsequently extending to distant sites such as the thigh or the flank. The location of the abscess can be defined precisely by means of computed tomography with contrast.

Some abscesses rupture into adjacent tissues or viscera, resulting in the formation of fistulas. The fistulas most commonly seen in this setting (50% to 65% of cases) are colovesical fistulas. This complication is less common in women because of the protection afforded by the uterus. Symptoms of colovesical fistulas tend to involve the urinary tract (e.g., pneumaturia, hematuria, and urinary frequency). Fecaluria is diagnostic of colovesical or enterovesical fistulas. Colovaginal fistulas (which account for 25% of all diverticular fistulas) are usually seen in women who have undergone hysterectomies.19 The diseased colon is adherent to the vaginal cuff. Most commonly, patients complain of a foul vaginal discharge; however, some patients present with stool emanating from the vagina.

About 10% of colon obstructions are attributable to diverticulitis. Acute diverticulitis can cause colonic edema and a functional obstruction that usually resolves with antibiotic infusion and bowel rest. Stricture formation is more common, usually occurring as a consequence of recurrent attacks of diverticulitis. Circumferential pericolic fibrosis is noted, and marked angulation of the pelvic colon with adherence to the pelvic sidewall may be seen. Patients complain of constipation and narrowed stools. Colonoscopy can be difficult and potentially dangerous in this setting. Differentiating a diverticular stricture from carcinoma may be impossible by any means short of resection.

The term malignant diverticulitis has been employed to describe an extreme form of sigmoid diverticulitis that is characterized by an extensive phlegmon and inflammatory reaction extending below the peritoneal reflection, with a tendency toward obstruction and fistula formation.20 Malignant diverticulitis is seen in fewer than 5% of patients older than 50 years who are operated on for diverticulitis.20 The process is reminiscent of Crohn disease, and CT scans demonstrate extensive inflammation. In this setting, a staged resection might be preferable to attempting a primary resection through the pelvic phlegmon. The degree of pelvic inflammation may subside significantly after diversion.20

A dangerous but rare complication of acute diverticulitis (occurring in 1% to 2% of cases) is free perforation,21 a term that includes both perforation of a diverticular abscess throughout the abdomen leading to generalized peritonitis (purulent peritonitis; Hinchey stage III) and free spillage of stool thorough an open diverticulum into the peritoneal cavity (fecal peritonitis; Hinchey stage IV). The incidence of free perforations may be increasing, at least in the southwestern United States.22 The overall mortality in this group is between 20% and 30%; that for purulent peritonitis is approximately 13%, and that for fecal peritonitis is about 43%.21

Physical Examination

Uncomplicated Diverticulitis

Physical examination reveals localized left lower quadrant abdominal tenderness with variable degrees of guarding and rebound tenderness. A mass is occasionally felt. The stool may contain traces of blood, but gross bleeding is unusual. Localized inflammation of the perforated diverticulum and the adjacent mesentery is present, and a phlegmon may be seen as well. Depending on the severity of the physical findings, patients may be managed either as inpatients or outpatients.

Complicated Diverticulitis

In a patient with a pericolic abscess, a mass may be detectable on abdominal, rectal, or pelvic examination. In a patient with a colovaginal fistula, a site of granulation tissue and drainage is seen at the apex of the vaginal cuff. In a patient with obstruction, there may be marked abdominal distention, usually of slow onset; abdominal tenderness may or may not be present, but if tears develop in the cecal taeniae, right lower quadrant tenderness is typically seen. In a patient with a free perforation, there is marked abdominal tenderness, usually commencing suddenly in the left lower quadrant and spreading within hours to the remainder of the abdomen. Hypotension and oliguria may develop later. Patients with rectal bleeding usually have no complaints of abdominal pain or tenderness, and they may be hypovolemic and hypotensive, depending on the rapidity of the bleeding.

Investigative Studies


Imaging

Figure 6. CT: acute diverticulitis
Figure 7. CT: thickened colonic wall and diverticulum

The most useful diagnostic imaging study in the setting of suspected diverticulitis is a CT scan with oral and rectal contrast.23 Localized thickening of the bowel wall or inflammation of the adjacent pericolic fat is suggestive of diverticulitis; extraluminal air or fluid collections are sometimes seen together with diverticula [see Figure 6]. The most frequent findings (seen in 70% to 100% of cases) are bowel wall thickening, fat stranding, and diverticula.24 In some cases, small abscesses in the mesocolon or bowel wall are not detected. The diagnosis of carcinoma cannot be excluded definitively when there is thickening of the bowel wall [see Figure 7].2

Figure 8. Local extravasion from sigmoid colon
Figure 9. Extravasation into abscess cavity

Although CT scanning has tended to replace contrast studies in the evaluation of diverticulitis, the latter may be more useful in differentiating carcinoma from diverticulitis. A contrast study can also be complementary when the CT scan raises the suspicion of carcinoma.23 When diverticulitis is suspected, water-soluble contrast material should be used instead of barium because of the complications that follow extravasation of barium [see Figures 8 and 9]. Furthermore, in the acute setting, only the left colon should be evaluated. Carcinoma is suggested by an abrupt transition to an abnormal mucosa over a relatively short segment; diverticulitis is usually characterized by a gradual transition into diseased colon over a longer segment, with the mucosa remaining intact. If the contrast study reveals extravasation of contrast outlining an abscess cavity [see Figure 9], an intramural sinus tract, or a fistula, diverticulitis is likely.1

Figure 10. Colonoscopic view of sigmoid diverticula

Colonoscopy is avoided when acute diverticulitis is suspected, because of the risk of perforation. It may, however, be done 6 to 8 weeks after the process subsides to rule out other disorders (e.g., colon cancer) [see Figure 10]. If diverticular disease is advanced, the endoscopic procedure may be difficult; the diverticular segment must be fully traversed for the examiner to be able to exclude a neoplasm with confidence. When major lower GI bleeding occurs, colonoscopy is done to search for polyps, carcinoma, or a site of diverticular bleeding. In the case of massive bleeding, selective arteriography is useful for localizing the source, and superselective embolization frequently quells the hemorrhage. The actual risk of bowel ischemia is low when superselective techniques are employed. Bleeding at the time of arteriography may be facilitated by the infusion of heparin or urokinase; however, this is a risky approach that should be taken only when other attempts at localization have failed and recurrent bouts of bleeding have occurred.

Figure 11. CT: colovesical fistula

When a colovesical fistula occurs, contrast CT with narrow cuts in the pelvis can be very helpful. The classic findings are sigmoid diverticula, thickening of the bladder and the colon, air in the bladder, opacification of the fistula tract and the bladder, and, possibly, an abscess [see Figure 11]. Cystoscopy is less specific, showing possible edema or erythema at the site of the fistula. A contrast enema helps rule out malignant disease. The diagnostic tests that are most useful for detecting colovaginal fistulas are contrast CT and vaginography via a Foley catheter. Charcoal ingestion helps confirm the presence of colovesical or colovaginal fistulas. On rare occasions, colocutaneous fistulas may develop, causing erythema and breakdown of the skin. Colouterine fistulas may occur as well; these are also quite rare.25

Management


Medical

Uncomplicated diverticulitis is usually managed on an outpatient basis by instituting a liquid or low-residue diet and administering an oral antibiotic combination that covers anaerobes and gram-negative organisms (e.g., ciprofloxacin with metronidazole or clindamycin) over a period of 7 to 10 days. Provided that symptoms and signs have subsided, the colon may be evaluated more fully several weeks later with a contrast study or colonoscopy if the diagnosis of diverticular disease has not already been established. If symptoms worsen, hospitalization should be considered. Over the long term, patients should be maintained on a high-fiber diet, though it may take months for the diet to have an effect on symptoms.26

Figure 12. Treatment options for complicated diverticulitis

If more significant physical findings and symptoms of toxicity develop, hospitalization is warranted [see Figure 12]. Patients are placed on a nihil per os (NPO) regimen, and intravenous fluids and antibiotics are administered (e.g., a third-generation cephalosporin with metronidazole) until abdominal pain and tenderness have resolved and bowel function has returned. As a rule, resolution occurs within several days. If there is clinical evidence of intestinal obstruction or ileus, a nasogastric tube is placed. In most cases, ileus-related symptoms resolve with antibiotic treatment. CT scans are useful for establishing the correct diagnosis in the emergency department27; furthermore, the severity of diverticulitis on CT scans predicts the risk of subsequent medical failure.28 Following the sedimentation rate may be helpful in assessing the effectiveness of treatment. It has been estimated that 15% to 30% of patients admitted with acute diverticulitis will require surgical treatment during the same admission.1

Figure 13. CT scan: pericolonic abscess

If fever and leukocytosis persist despite antibiotic therapy, the presence of an abscess should be suspected. Small (< 5 cm) abscesses may respond to antibiotics and bowel rest. Larger abscesses that are localized and isolated may be accessible to percutaneous drainage [see Figure 13].28 Generally, this technique is reserved for abscesses greater than 5 cm in diameter in low-risk patients who are not immunocompromised. It often leads to resolution of sepsis and the resulting symptoms and signs (e.g., abdominal pain and tenderness and leukocytosis), usually within 72 hours, thereby facilitating subsequent elective surgical resection of the colon. In addition, percutaneous drainage offers cost advantages, in that it reduces the number of operative procedures required and shortens hospital stay.

Access to a pelvic collection may be difficult to obtain, and the drainage procedure typically must be done with the patient in a prone or lateral position. If the catheter drainage amounts to more than 500 ml/day after the first 24 hours, a fistula should be suspected. Before the catheter is removed, a CT scan is done with injection of contrast material through the tube to determine whether the cavity has collapsed. If this approach fails (as it usually does in patients with multiple or multiloculated abscesses), an expeditious operation may be necessary.22 An initial surgical procedure is required in about 20% of cases.29

Surgical

Overall, approximately 20% of patients with diverticulitis require surgical treatment.2,30 Most surgical procedures are reserved for patients who experience recurrent episodes of acute diverticulitis that necessitate treatment (inpatient or outpatient) or who have complicated diverticulitis. The most common indication for elective resection is recurrent attacks—that is, several episodes of acute diverticulitis documented by studies such as CT. Estimates of the risk of such attacks range from 30% to 45%. A task force of the American Society of Colon and Rectal Surgeons recommended sigmoid resection after two attacks of diverticulitis.31 A cost analysis using a Markov model suggested that cost savings can be achieved if resection is done after three attacks.32 Efforts are made to time surgical treatment so that it takes place during a quiescent period 8 to 10 weeks after the last attack. Barium enema or colonoscopy may be employed to evaluate the diverticular disease and rule out carcinoma. The bowel can then be prepared mechanically and with antibiotics (e.g., oral neomycin and metronidazole on the day before operation).

Elective resection is a common sequel to successful percutaneous drainage of a pericolic abscess in an otherwise healthy, well-nourished patient. The timing of surgery may be guided by the extent of the inflammatory changes (as documented by CT scanning) and the patient’s clinical course. Most patients can be operated upon within 6 weeks. Elective resection is the preferred approach to diverticular fistulas as well. Colovesical fistulas are usually resected because of the risk of urinary sepsis and the concern that a malignancy might be overlooked. Preferably, the operation is done when the acute inflammation has subsided.

Elective resection is done via either the open route or, increasingly, the laparoscopic route33; a few telerobotic-assisted laparoscopic colectomies have also been attempted.34 The learning curve for laparoscopic colectomy is 20 to 50 cases.35 Obese patients with severe colonic inflammation are poorer candidates for laparoscopic resection.33 In our institution, the development of hand-assisted procedures has widened the opportunities for utilizing minimally invasive surgery [see 5:32 Procedures for Diverticular Disease].40,41

Some patients with complicated diverticulitis require emergency resection because of free perforation and widespread peritonitis. In such patients, the American Society of Anesthesiologists (ASA) physical status score and the degree of preoperative organ failure may be significant predictors of outcome.42,43 Unfavorable systemic factors (e.g., hypotension, renal failure, diabetes, malnutrition, immune compromise, and ascites) play a vital role in determining patient outcome,43 as does the severity of the peritonitis (i.e., extent, contents, and speed of development).44,45 One of the unfortunate limitations of the Hinchey classification is that it does not take comorbidities into account.44 Because the bowel is not prepared before operation, the surgeon may feel uncomfortable doing an anastomosis. On-table lavage may be considered if contamination is minimal, but it adds to the time spent under anesthesia during an emergency procedure.

As a general rule, resection and immediate anastomosis are suitable for Hinchey stage I and perhaps stage II diverticular perforations, whereas resection with diversion (the Hartmann procedure) is the gold standard for stage III and especially stage IV.46–48 This recommendation is based on the finding that an anastomosis involving the left colon is risky when performed under emergency conditions.49 The once-popular three-stage procedures are now of historic interest only. There are some reports of successful outcomes for type III and type IV cases after extensive abdominal lavage and two-layer anastomoses50 or after on-table lavage of the colonic contents to allow primary anastomosis.51 Grading of comorbidities with classification systems such as APACHE II or the Mannheim peritonitis index can facilitate decision-making with respect to the question of anastomosis versus diversion.52 The surgeon’s decision must be individualized on the basis of each patient’s condition and needs. The literature on this topic is confusing, in that most of the published reports are small and retrospective, with only limited classification of disease severity.

Currently, surgeons encountering acute diverticulitis are more likely to do one-stage resections, as opposed to Hartmann procedures, than they once were.43,53 The advantage of the one-stage approach is that the colostomy takedown and the attendant 4% mortality are avoided.54 Furthermore, at least 30% of patients who undergo a Hartmann procedure never return for colostomy closure. A primary anastomosis can be protected with a proximal ileostomy as well.46,55,56 Transverse colostomy and loop ileostomy appear to be equally safe, though skin changes may be more problematic after a colostomy57 and an ileostomy closure tends to be less complex than a colostomy closure. On-table lavage may also be used as an adjunct to anastomosis.58

The risk of complications inherent in operations on the colon should always be kept in mind, especially in the relatively few patients undergoing emergency procedures. In this setting, the bowel is unprepared and systemic sepsis may be present. Potential complications include ureteral injuries; anastomotic leakage, anastomotic stricture, and postoperative intra-abdominal abscesses; perioperative bleeding involving the mesentery, adhesions, the splenic capsule, or the presacral venous plexus; postoperative small bowel obstruction; stomal complications; wound infection, wound dehiscence, and abdominal compartment syndrome; the acute respiratory distress syndrome (ARDS); and the multiple organ dysfunction syndrome (MODS).

Figure 14. CT scan: marked thickening of sigmoid wall
Figure 15. High-grade retrograde obstruction

Large bowel obstruction secondary to diverticulitis can lead to considerable morbidity and may necessitate surgical intervention.55 The obstruction is usually partial [see Figures 14 and 15], allowing preparation of the bowel in many cases. High-grade obstruction represents a complex problem. If the cecum is dilated to a diameter of 10 cm or greater and there is tenderness in the right lower quadrant, expeditious surgery is necessary because of the risk of cecal necrosis and perforation. High-grade obstruction with fecal loading of the colon is usually managed by performing a Hartmann procedure, though on-table lavage may be considered.22 A survey of GI surgeons in the United States indicated that 50% would opt for a one-stage procedure in low-risk patients with obstruction, whereas 94% would opt for a staged procedure in high-risk patients.59

Small bowel obstruction may also complicate the clinical picture. Mechanical small bowel obstruction may occur as a consequence of adherence of the small bowel to a focus of diverticulitis, especially in the presence of a large pericolic abscess. Whereas small bowel obstruction tends to cause periumbilical crampy abdominal pain and vomiting, these characteristic manifestations may be obscured in part by pain attributed to diverticulitis. The concern in this situation is that ischemic small bowel may be ignored, with potentially disastrous consequences. Diarrhea should trigger the suspicion of colonic disease, and formation of a fistula into the small bowel should raise the possibility of Crohn disease. CT scanning often helps the surgeon differentiate between primary and secondary small bowel obstruction, but ultimately, exploratory surgery may be required both for diagnosis and for treatment.

Lower GI bleeding caused by diverticular disease rarely calls for emergency resection, because the bleeding is self-limited in most patients (80% to 90%). Furthermore, active diverticulitis is rare when active bleeding is the presenting symptom. Attempts are made to establish the active bleeding site by means of colonoscopy, tagged red blood cell nuclear scans, or angiography; barium contrast studies have no role to play in this situation. Emergency resection is indicated if the bleeding is life-threatening and if colonic angiography and attempted superselective embolization prove unsuccessful. In an unstable patient, total abdominal colectomy is necessary if the site of bleeding is unknown, though identification of the bleeding site with intraoperative colonoscopy has been reported. In a stable patient with ongoing bleeding, repeat angiography at a later time is appropriate, or so-called pharmacoangiography (infusion of heparin) can be employed in an attempt to induce bleeding.

Special Types of Diverticulitis


Cecal Diverticulitis

Figure 16. Classification of pathologic types of cecal diverticulitis

In the United States, diverticulitis rarely involves the cecum or the right colon. Right-side diverticula occur in only 15% of patients in Western countries, compared with 75% in Singapore.1 The incidence of cecal diverticulitis appears to be related to the number of diverticula present.60 A classification system has been proposed that divides cecal diverticulitis into four grades [see Figure 16] to facilitate comparisons between different clinical series and to help surgeons formulate treatment plans in the OR.60 Some cecal diverticula are true diverticula, containing all layers of the bowel wall, but the majority are pseudodiverticula. Diverticulitis of the hepatic flexure and the transverse colon is even less common and can present with symptoms suggesting appendicitis.61,62

Figure 17. CT scan: inflammation in the pericecal area

Patients with right-side disease tend to be younger and to have less generalized peritonitis than patients with left-side diverticulitis.60,61 Because they typically present with right lower quadrant pain, fever, and leukocytosis, acute appendicitis is usually suspected. CT scans are helpful for differentiating cecal diverticulitis from appendicitis or colon cancer [see Figure 17].63,64 If cecal diverticulitis is suspected (as in a patient who has previously undergone appendectomy or in a patient with known right-side diverticulosis who has experienced similar attacks in the past), medical management with observation and antibiotics is generally the favored strategy, just as with simple sigmoid diverticulitis. In Japan, where right-side diverticulitis is more common, medical treatment has been successfully used for recurrent attacks of uncomplicated right-side diverticulitis.65 After a few weeks, colonoscopy should be performed to rule out a colonic neoplasm.

If the patient has significant peritonitis or the diagnosis is unclear, laparoscopy or laparotomy is indicated. It is important that one or the other be done because the mortality associated with delayed treatment of perforated cecal diverticulitis is high. In our institution, laparoscopy is usually employed; if the diagnosis is unclear, laparotomy is recommended. When inflammation is localized and minimal, colectomy is unnecessary, and incidental appendectomy should be considered if the cecum is uninvolved at the base of the appendix.66 If desired, the diverticulum may be removed as well.

Diverticulectomy should be done only if (1) carcinoma can be ruled out, (2) the resection margins are free of inflammation, (3) the ileocecal valve and the blood supply of the bowel are not compromised, and (4) perforation, gangrene, and abscess are absent.60 Localized diverticulectomy, in general, should be reserved for grade I and grade II disease.60 Sometimes, the ostium of the inflamed diverticulum is palpable if the cecum is mobilized surgically.67 On-table cecoscopy thorough the appendiceal stump has also been helpful in establishing the diagnosis in the OR.66 Grade III and IV cecal diverticulitis may be difficult to differentiate from carcinoma; resection is favored for these lesions.67 An anastomosis may be created if contamination is limited, but generally, primary resection, ileostomy, and a mucous fistula are favored for treatment of grade IV disease.

Diverticulitis in Young Patients

Diverticulitis in patients younger than 40 years has been a focus of considerable attention in the literature, though this group only represents about 2% to 5% of the patients in large series.31 The incidence of diverticulitis in young patients may be increasing, and obese Latino men appear to be at particular risk.68 This predominance in males reflects a tendency to underdiagnose acute diverticulitis in young women.69 Some authors have asserted that diverticulitis is particularly virulent in young patients; however, current data tend not to support this concept, suggesting that patients with mild diverticulitis are misdiagnosed when hospitalized or are treated as outpatients. The high rate of early operation in young patients probably reflects misdiagnosis of diverticulitis as acute appendicitis rather than the development of particularly severe forms of diverticulitis.68 Patients found to have uncomplicated acute diverticulitis may, if desired, undergo incidental appendectomy in conjunction with medical treatment of diverticulitis.

Unlike elderly patients, hospitalized young patients with diverticulitis tend to have few comorbidities other than obesity. Furthermore, young patients hospitalized for diverticulitis tend to have relatively advanced disease, perhaps as a consequence of delayed diagnosis,2 whereas elderly patients hospitalized with an admitting diagnosis of diverticulitis tend to exhibit a wider spectrum of disease severity. Young patients appear not to have a higher rate of recurrent diverticulitis than older patients do, and thus, aggressive resection is not necessary at the time of the first attack.42,68 However, a finding of advanced diverticulitis on CT scans is a predictor of subsequent disease complications in this population.70,71

In general, diverticulitis should be approached in the same fashion in younger patients as in older patients.71 The pathophysiology of the disease is probably identical. As in the elderly, elective resection is recommended after recurrent attacks, not after a single attack; with follow-up, the majority of patients hospitalized with acute diverticulitis do not require operation.71,72

Diverticulitis in Immunocompromised Patients

In view of their known predisposition to infection, immunocompromised patients (e.g., chronic alcoholics, transplant patients, and persons with metastatic tumors who are receiving chemotherapy) with diverticulitis are at particular risk. There is no evidence that the incidence of diverticulitis is higher in this population than in the general population, but it is clear that immunocompromised patients have higher rates of operation once diverticulitis develops and that their postoperative mortality is higher.73,74 Corticosteroid intake causes a number of significant problems, such as thinning of the colonic wall, lessening of the physical findings with diverticulitis, and an attenuated inflammatory response.

Any immunocompromised patient with abdominal pain should be evaluated aggressively. Contrast-enhanced CT is the imaging study of choice. The risk of perforation is increased in this setting, as is the risk of postoperative complications such as wound dehiscence. For an immunocompromised patient who has recovered from an episode of symptomatic diverticulitis, elective surgical treatment is recommended. A renal transplant patient with asymptomatic diverticulosis, however, need not undergo prophylactic colectomy. Pretransplantation colonic screening of patients older than 50 years does not reliably predict postransplantation colonic complications.75

Atypical Presentations

Diverticulitis may give rise to various unusual manifestations involving multiple organ systems [see Table 1]. Not surprisingly, immunocompromised patients are at particular risk.

Retroperitoneal abscesses can track into anatomic planes (e.g., along the psoas muscle) or through the obturator foramen to areas such as the neck, the thigh,76 the knee, the groin,77 and the genitalia.78,79 CT scanning is essential to outline the extent of such abscesses. Contrast enemas show the diverticula along with a sinus tract into the abscess cavity. Cultures of the abscess demonstrate the presence of colonic organisms such as Bacteroides fragilis. Definitive treatment consists of wide abscess drainage and colon resection. Without aggressive surgical management, mortality is high.

The protean manifestations of diverticulitis also include pylephlebitis (which causes liver abscesses), arthritis, and skin changes. Diverticulitis has in fact replaced appendicitis as the most common source of liver abscesses of portal origin. Simple abscesses may be drained percutaneously if they are not too large, and multiple loculated abscesses may be managed with open drainage. The main risk factors for mortality from liver abscesses are immunosuppression, underlying malignancy, the presence of multiple organisms, and liver dysfunction. If the decision is made to perform a colectomy, the procedure may be done after drainage of the liver abscess or simultaneously with drainage during an open procedure.

Giant Diverticula

An anatomic curiosity sometimes encountered in patients with diverticular disease is a giant diverticulum, also termed a giant gas cyst or a pneumocyst of the colon.80 These lesions, which may reach diameters of 40 cm, are believed to develop as a consequence of a ball-valve mechanism created by intermittent occlusion of the neck by fecal material that traps air in the diverticulum. Most giant diverticula are minimally symptomatic, causing only mild abdominal pain, and perforation is rare. A mobile mass may be palpable, and the gas-filled cyst can be seen on plain abdominal films. As many as two thirds of giant diverticula are opacified during a barium enema and can thereby be differentiated from other abnormalities (e.g., a mesenteric cyst, emphysematous cholecystitis, or a colon duplication). The cyst tends to adhere densely to adjacent structures (e.g., the bladder and the small bowel). The treatment of choice is resection of the colon and the cyst; performing diverticulectomy alone can lead to the development of a colocutaneous fistula.

Recurrent Diverticulitis after Resection

Recurrent diverticulitis is rare after a colectomy for diverticulitis, occurring in 1% to 10% of patients.81 As many as 3% of patients who have undergone resection for diverticulitis will require repeat resection.3 The differential diagnosis includes Crohn disease, irritable bowel syndrome, carcinoma, and ischemic colitis. CT imaging and colonoscopy should be carried out. Particular care should be taken to review pathologic specimens for evidence of Crohn disease.

The only significant determinant of recurrent diverticulitis is the level of the anastomosis; the high pressure in the sigmoid colon distal to the anastomosis appears to be responsible. In one study, the risk of recurrence was four times greater in patients with a colosigmoid anastomosis than in those with a colorectal anastomosis.82 Reoperation requires a dissection that commences in noninflamed tissue. Dissection may be particularly difficult near the pelvic sidewall because of fibrosis; ureteral stenting may facilitate identification of the ureters.

Subacute and Atypical Diverticulitis

A small number of patients experience recurrent episodes of left lower quadrant abdominal pain that are not accompanied by the classic findings of acute diverticulitis (e.g., fever and leukocytosis). The inflammatory changes associated with diverticula in this subgroup have been referred to as atypical, subacute, or smoldering diverticulitis.83,84 In this setting, there is not always a direct association between endoscopic and clinical findings; endoscopic evidence of diverticular inflammation has been seen in asymptomatic patients.85 It has been suggested that there is a relation between diverticular disease and colitis.86 Patients with chronic lower abdominal pain should undergo imaging studies and endoscopic evaluation, and other disorders (e.g., irritable bowel syndrome, inflammatory bowel disease, drug-induced symptoms, and bowel ischemia) should be excluded. In most cases of atypical diverticulitis, endoscopic findings are normal.84 In carefully selected patients, colectomy often eliminates the abdominal pain, and many of these patients are eventually found to have histologic signs of acute and chronic mucosal inflammation.84

 

Susan Galandiuk, MD, FACS, FASCRS

Professor, Department of Surgery
University of Louisville School of Medicine

The role of surgery in the management of Crohn disease has undergone a dramatic evolution over the past 50 years. Currently, surgical treatment of Crohn disease is seldom performed in the emergency setting; it is nearly always performed after failed medical therapy. The decision to proceed with operative management is based on careful patient evaluation, with full awareness of the potential complications and ramifications of treatment. In particular, attention must be paid to the risk of recurrent disease, the possible surgical sequelae, and the side effects of medical therapy.

Classification


There are many systems for classifying Crohn disease. One of the simplest is the classification developed by Farmer and associates,1 which categorizes the disease on the basis of disease location alone (ileocolic, purely colonic, small bowel, and perianal). A more elaborate system is the Vienna classification, which categorizes the disease on the basis not only of location but also of age of onset and disease behavior.2 In this system, there are four categories for disease location: terminal ileum (L1), colon (L2), terminal ileum and colon (L3), and any location proximal to the terminal ileum (L4). There are two categories for age of onset: less than 40 years of age (A1) and 40 years of age or older (A2). Finally, there are three categories for disease behavior: nonstricturing and nonpenetrating (B1), stricturing (B2), and penetrating (B3).

Given that there are as many types and combinations of Crohn disease as there are patients with this condition, the most sensible approach is probably to use some combination of these two classification schemes. Careful evaluation of the specifics of each case will yield the best treatment results; however, general classification of the disease can help guide therapy. Broadly speaking, Crohn disease of the small bowel has the highest recurrence rate. Because of the important function of the small bowel in digestion, surgeons tend to emphasize conserving small bowel length during operative treatment of Crohn disease. Currently, however, there is an increasing focus on colon conservation with the aims of maintaining water absorption in patients and delaying (or perhaps eliminating) the need for a stoma.

Roles of Medical Therapy and Surgical Therapy


In planning treatment of Crohn disease, it is important not to make the use of medical therapy or surgical therapy an either-or issue. Just as one tool cannot be expected to fill every household need, operative management cannot be expected to solve every problem related to Crohn disease. Overall, careful use of medical therapy, appropriately combined with surgical therapy, provides the best treatment of Crohn disease. Single-minded reliance on either therapy to the exclusion of the other often leads to inadequate patient care.

Generally speaking, except in the case of a free perforation, cancer, or dysplasia, one should not operate on a patient with Crohn disease without first attempting medical therapy. With the dramatically improved medical treatment options currently available, surgery can be avoided in many cases. This is often a desirable result, given the known risk of disease recurrence after surgical treatment of Crohn disease and the significant associated operative morbidity. In one single-center study, the reoperation rate for Crohn disease was 34% at 10 years.3 The agents used to treat Crohn disease can be divided into several broad groups: probiotics, antibiotics, anti-inflammatory drugs, immunosuppressive drugs, and biologic agents. These can be used alone or in combination to treat disease, as well as to maintain remission [see Table 1].

Few good studies have been done on the cost-effectiveness of medical or surgical therapy4,5 versus that of timely surgery followed by maintenance medical therapy. There is clearly a need for such studies. The use of potent and expensive immunomodulator therapy (e.g., maintenance infliximab) for simple ileocolic disease is questionable, especially in the light of studies indicating that such treatment is not at all innocuous.6,7

Changing Concepts in Surgery for Crohn Disease

Although first described in the beginning of the 19th century, Crohn disease was not recognized as a discrete clinical entity until the first part of the 20th century.8 At one point, it was treated surgically in much the same way as cancer, with frozen-section margins obtained at the time of resection. This approach did not yield any substantial reduction in the recurrence rate.9 In fact, overzealous resections often resulted in Crohn patients’ requiring lifelong parenteral nutritional support.10 Accordingly, conservative surgery is now the rule: only gross macroscopic disease is resected into palpably normal margins (in particular, a palpably normal mesenteric border of the bowel).

General Indications for Surgical Treatment


Side Effects of Medical Therapy

Significant side effects of medical therapy include those associated with failure to wean from prednisone (e.g., cataract formation, aseptic necrosis of the femoral head, and weight gain). Side effects of antimetabolite therapy include pancreatitis, neutropenia, and opportunistic infections.

Complications of Disease

Lack of Response to Medical Therapy

Many patients with so-called toxic colitis do not respond satisfactorily to medical treatment. In severe cases of refractory disease, if surgery is not performed, colonic perforation, peritonitis, and multiple organ failure may ensue. Such cases are much less frequent now than they once were.

Obstruction

Figure 1. Stenotic ileocolic Chron disease

In many patients with Crohn disease, the behavior of the disease changes over time, from a more inflammatory and edematous process to one characterized more by fibrosis and scarring. Where as anti-inflammatory drugs are ideal for treating the former, surgery is frequently necessary for the latter. Failure to refer for surgical treatment of obstruction is, unfortunately, a common error among gastroenterologists. Severe abdominal pain is always a warning sign of obstruction and should be taken seriously [see 5:1 Acute Abdominal Pain and see 5:4 Intestinal Obstruction]. The importance of this point is illustrated by a case from my experience, involving a patient who had obstructing ileocolic Crohn disease with gross proximal distention of the terminal ileum [see Figure 1]. This patient lost 20 lb, was experiencing severe abdominal pain, and was treated for more than a year with 6-mercaptopurine before being referred for operative management. Ileocolic resection led to rapid resolution of the symptoms.

Symptomatic Fistulas

Figure 2. Enterocutaneous fistula

Enteroenteric fistulas, by themselves, are no longer considered an absolute indication for operation in the absence of other complicating factors. Symptomatic fistulas, such as those associated with obstruction or those associated with disabling symptoms (e.g., rectovaginal fistulas or enterocutaneous fistulas [see Figure 2]), may have to be treated surgically. Ileosigmoid fistulas, which effectively bypass the entire colon, may be associated with profound and refractory diarrhea (i.e., ³ 20 bowel movements/day) and may also have to be treated operatively.

Abscess Formation

Abscesses are particularly common with ileocolic Crohn disease. If they cannot be controlled by means of computed tomography-guided drainage, surgical therapy may be indicated.

Cancer or Dysplasia

The risk of colorectal cancer is approximately three times higher in patients with Crohn disease than in the general population.11–13

Failure to Grow

In children, failure to grow and develop normally is one of the main indications that medical therapy for Crohn disease has been unsuccessful. Timely surgical therapy will permit normal development. On occasion, when bone age lags significantly behind chronological age, treatment with recombinant human growth hormone is required.

Special Considerations


Pregnancy

Persons who have Crohn disease may be less fertile than healthy age-matched persons. One possible explanation for this difference is that feeling ill may result in reduced sexual desire or decreased sexual activity. Another is that pelvic inflammation caused by Crohn disease or by scarring and adhesion formation resulting from surgery may impair fertility. To reduce the chances of the latter, hyaluronic acid sheets may be placed around the tubes and ovaries; alternatively, the ovaries may be tacked to the undersurface of the anterior abdominal wall with absorbable sutures and thereby prevented from entering the pelvis.

There is no evidence that pregnancy exacerbates Crohn disease; however, there are some specific concerns that apply to pregnant patients with this condition. Because patients with Crohn disease often have more-liquid bowel movements, they have a particular need for a well-functioning anal sphincter. If there is any chance of an obstetrics-related injury (e.g., from a large baby in a primagravida or from a breech presentation), a cesarean section is advisable to minimize the risk of sphincter trauma. The same is true in the presence of severe perianal Crohn disease. During pregnancy, prednisone and 5-aminosalicylic acid (5-ASA) medications are safe, whereas drugs such as metronidazole are not. If imaging studies are needed, magnetic resonance imaging and ultrasonography are the modalities of choice.

Marking of Stoma Sites and Choice of Incision

When a patient with Crohn disease is expected to need an ileostomy [see 5:30 Intestinal Stomas], it is extremely important to mark the site preoperatively. What looks flat when the patient is on the operating table may not be flat when he or she is upright. The patient must be asked to sit and lean over to confirm that the marked stoma site is in an area without folds, creases, or previous incisions. Stoma appliances do not adhere well to areas of previous scarring, and these should be avoided whenever possible.

Patients with Crohn disease do not react to intra-abdominal infection in a typical fashion. It is not unusual to find unsuspected abscesses that were not revealed by preoperative CT scans and other imaging studies. If there is even a remote chance of an unsuspected abscess (particularly in cases of obstructing ileocolic Crohn disease), the possibility of a temporary stoma should be raised with the patient and the proposed stoma site marked preoperatively.

A key point is the necessity of planning for the future. Many patients with Crohn disease will eventually require a stoma. Operating through a midline abdominal incision preserves all four quadrants for possible future stoma sites (if needed).

Laparoscopy

Laparoscopic surgical techniques have gained acceptance in the treatment of Crohn disease. In performing a laparoscopic operation for Crohn disease, it is essential to adhere to the same technical standards that apply to corresponding open procedures. Careful intraoperative exploration of the abdomen is important, in that many patients have multifocal disease. Without such exploration, patients may experience persistent postoperative symptoms as a consequence of persistent proximal pathologic states that were not addressed. As with other treatment modalities, there are some circumstances in which laparoscopy is particularly useful and others in which it should not be used. For example, a laparoscopic approach is ideal for fecal diversion in patients with perianal Crohn disease.

Ileocolic resection for Crohn disease also lends itself well to a laparoscopic-assisted approach; compared with open resection, laparoscopic resection has been reported to result in shorter hospital stays and reduced costs.14,15 The ileocolic vessels originate centrally, and they only lie over the retroperitoneum. Once the lateral peritoneal attachments are divided, the colon and the small bowel mesentery can be exteriorized, and the mesentery can be divided and the anastomosis performed extracorporeally.

Many studies have shown that even fistulizing Crohn disease can be safely addressed laparoscopically, depending on the skill of the surgeon. A hand-assisted approach is often useful with cases of dense fixation, in which fistulas are common and finger dissection may facilitate definition of the anatomy. If in doubt, one should not hesitate to convert to an open procedure. Typically, most areas that feel fibrotic or contain fibrotic adhesions are actually areas of fistulizing disease and should be treated as such until proved otherwise. In one study, patients with recurrent disease, those older than 40 years, and those with an abdominal mass were more likely to require conversion to an open procedure.16

Surgical Management of Crohn Disease at Specific Sites


Esophageal, Gastric, and Duodenal Disease

Crohn disease of the upper alimentary tract can be difficult to diagnose, largely because it is relatively uncommon. Obstructing strictures due to Crohn disease in this area are unusual; the unsuspected finding of noncaseating granulomas in biopsies of erythematous areas in a patient with Crohn disease in other locations is diagnostic.

Occasionally, a patient with Crohn disease of the distal esophagus requires dilatations, but this is uncommon. Surgical treatment for Crohn disease of the upper alimentary tract is almost exclusively reserved for disease affecting the duodenum. Diagnosis of duodenal Crohn disease can be difficult and requires a certain amount of suspicion. Frequently, the diagnosis is not made until relatively late, because diagnostic imaging tends to focus on endoscopy and because the degree of duodenal obstruction is often not evident except on barium studies. The rigidity and luminal narrowing of the second portion of the duodenum is typically much more readily apparent on contrast studies than on endoscopy. Duodenal Crohn disease can lead to gastric outlet obstruction. In children, it can be mistaken for annular pancreas.

When duodenal Crohn disease does not respond to medical therapy, gastrojejunostomy with vagotomy is the preferred surgical treatment.17,18 Failure to perform a vagotomy may result in marginal ulcer formation and obstruction. Some surgeons have performed duodenal strictureplasty to treat duodenal Crohn disease. The results have been conflicting19,20; the feasibility of this operative approach is limited by the pliability of the duodenum. Many patients experience prompt and full recovery of normal gastric emptying after operation, but some patients with long-standing gastric outlet obstruction continue to experience impaired emptying. The latter may benefit from administration of a prokinetic agent (e.g., metoclopramide or erythromycin).

Jejunoileal Disease

Short Bowel Syndrome

Although Crohn disease of the small bowel is not common and accounts for a relatively small proportion of all cases, disease in this area is associated with one of the highest overall recurrence rates. Resection of large portions of the small bowel can result in short bowel syndrome. For this reason, before proceeding with any type of small bowel or ileocolic resection, one should measure the length of the existing small bowel to determine the patient’s ‘bowel resource.’ One naturally would more readily perform a resection in a patient who has 400 cm of normal small bowel than in one who has only 200 cm.

Resection versus Strictureplasty

Figure 3. Heineke-Mikulicz strictureplasty
Figure 4. Finney strictureplasty
Figure 5. Short fibrotic stricture

The major advance in the surgical treatment of Crohn disease over the past quarter-century has been the technique of small bowel strictureplasty, first proposed by Lee and subsequently popularized by Williams, Fazio, and others.17,18 Currently, the two most prevalent strictureplasty techniques are Heineke-Mikulicz strictureplasty [see Figure 3] and Finney strictureplasty [see Figure 4]. The former is best suited for strictures up to 5 to 7 cm long [see Figure 5], the latter for strictures up to 10 to 15 cm long. The side-to-side strictureplasty described by Michelassi21 is suitable for longer areas of stricture; however, this technique involves longer suture lines and is mainly considered for patients who already have, or are at high risk for, short bowel syndrome.

The short, isolated strictures characteristic of diffuse jejunoileal Crohn disease are more frequently described in patients with long-standing Crohn disease. It has been postulated that over time, Crohn disease progresses from an edematous condition to a more fibrotic, stricturing condition.22 It is the fibrotic strictures characteristic of the later stage of the disease that are amenable to treatment with strictureplasty. Patients with these short fibrotic strictures typically have obstructive symptoms and often are unable to tolerate solid food, experiencing dramatic weight loss as a result. Although strictureplasty leaves active disease in situ, it usually leads to prompt resolution of obstructive symptoms, regaining of lost body weight, and restoration of normal nutritional status.

Figure 6. Large ulcer on mesenteric luminal border

A significant concern with strictureplasty is the possibility that small bowel adenocarcinoma may develop; several cases have been reported.23,24 I have treated a patient in whom a poorly differentiated jejunal adenocarcinoma developed at the site of a strictureplasty that had been performed 10 years earlier. Accordingly, many surgeons advocate routine biopsy of the active ulcer on the mesenteric side of the bowel at the time of strictureplasty [see Figure 6]. Another concern has to do with the number of strictureplasties that can safely be performed in a single patient in the course of a single operation. As many as 19 strictureplasties have been performed during one procedure without increased morbidity.25

Strictureplasty can be performed with either a single-layer or a double-layer anastomosis. It should not be performed in the presence of an abscess, a phlegmon, or a fistula; and like any other anastomosis, it should not be performed proximal to an existing obstruction that is not treated at the time of operation.

Areas of small bowel Crohn disease that are too long to be treated with strictureplasty can be treated with segmental resection. The area to be resected should be as short as possible. There is no need to obtain frozen-section margins to determine the extent of resection; doing so leads to unnecessary loss of small bowel length.26 The resection should extend into palpably normal areas of small bowel. The easiest way of determining the area to be resected is to feel the mesenteric margin of the bowel until palpably normal tissue is reached. Because Crohn disease is generally more severe on the mesenteric side of the bowel, palpation in this area gives the most accurate impression of the intraluminal character of the bowel. Because it is not uncommon for patients to have multifocal Crohn disease, the entire small bowel should always be inspected at the time of operation. Operating on one area of disease while failing to treat a more proximal lesion is clearly not in the patient’s interest.

Because of the high rate of recurrence in patients with isolated small bowel disease, postoperative chemoprophylaxis should be strongly considered. In these patients, I prefer to use a more potent agent, such as an antimetabolite, rather than a 5-ASA agent.

Ileocolic Disease

Approximately half of those diagnosed with Crohn disease have ileocolic disease. Ileocolic resection is, in fact, the operation most frequently performed to treat Crohn disease. Currently, there is a trend toward more aggressive medical management of Crohn disease; at the same time, surgeons are seeing more complicated disease at the time of operation. These developments have implications for management. An easy ileocolic resection is an experience that a patient generally tolerates well and recovers from very quickly; however, delaying operative management with years of aggressive medical therapy can lead to more complicated disease associated with enteroenteric fistulas, which can be difficult to treat. Ileosigmoid fistulas are among the most common fistulas associated with ileocolic Crohn disease, along with fistulas between the terminal ileum and the ascending colon and fistulas between the terminal ileum and adjacent loops of small bowel.

Disease recurrence is common after ileocolic resection. Colon os copy is the most accurate modality for postoperative surveillance and the easiest to use; it is more sensitive than either small bowel follow-through or air-contrast barium enema. For this reason, I favor an end-to-end anastomosis after ileocolic resection. In the event of recurrent disease, an end-to-side, side-to-end, or side-to-side anastomosis may be difficult to intubate. There is some evidence in the literature to suggest that the postoperative recurrence rate may be lower with a wider anastomosis.27 The anastomosis can be performed in either one or two layers. If the bowel is thicker, a handsewn anastomosis is preferred to a stapled one.

The incidence of reoperation for recurrent disease after ileocolic resection is high and increases with the number of resections.28 Postoperative chemoprophylaxis with mesalamine can significantly reduce the recurrence rate.29 Patients who smoke should be strongly encouraged to stop: the rate and severity of recurrence are increased in smokers.20

Special Circumstances

Ileocolic Crohn disease is often associated with intra-abdominal abscesses or fistulas. If an associated abscess is known to be present, CT-guided drainage should be done preoperatively so that a single-stage procedure can then be performed. If an unsuspected abscess is identified at the time of operation, the safest approach is to proceed with bowel resection, perform the posterior wall of the anastomosis, and exteriorize the anastomosis as a loop ileostomy. This loop ileostomy can then be safely closed, often without a formal laparotomy, 8 weeks after operation if there are no signs of ongoing sepsis. If the abscess or the terminal ileal loop is adherent to the sigmoid colon, an ileosigmoid fistula may be present. The decision whether to resect the sigmoid colon is dictated by the appearance and feel of the sigmoid in the involved areas. If only a portion of the anterior colon wall is involved, that portion can be excised in a wedgelike fashion and the excision site closed primarily. If the entire circumference of the sigmoid colon at that point is indurated and woody feeling, a short segmental resection with anastomosis is the best option.

Colonic Disease

Figure 7a. Pyoderma gangrenosum
Figure 7b. Peristomal gangrenosum of the breast

Colonic involvement is present in 29% to 44% of patients with Crohn disease.30 One of the challenges in treating colonic Crohn disease is obtaining the correct diagnosis. Whereas Crohn disease of the small bowel is fairly easy to diagnose, colonic disease often is not. Because granulomas are not present in most cases of colonic Crohn disease and because this condition can look very similar to ulcerative colitis both endoscopically and macroscopically, differentiation between Crohn colitis and ulcerative colitis can be difficult in the absence of small bowel or anal disease. Colonic Crohn disease appears to be more frequently associated with cutaneous manifestations (e.g., pyoderma gangrenosum) [see Figures 7a and 7b].

Indications for Surgical Treatment

Figure 8. Sigmoid colon stricture

The main indications for operative management of colonic Crohn disease are stricture [see Figure 8], malignancy, side effects of medical therapy, and failure of medical therapy. In children, failure to recognize and treat this condition promptly may result in growth retardation. It is important to monitor both bone age and insulinlike growth factor-1 levels. If these are abnormal, timely institution of human growth hormone therapy, operative management of inflammatory bowel disease, or both may still permit normal growth and development.

Side effects of medical therapy can be substantial. They may include such varied complications as aseptic necrosis of the femoral head and cataract formation (both related to steroid use), as well as an increased incidence of opportunistic infections (from immuno suppression secondary to antimetabolite therapy).

Figure 9a. Toxic megacolon
Figure 9b. Denuded muscle wall
Figure 9c. Incorporation of distal rectosigmoid and closure

Failure of medical therapy can refer to continuing severe disease activity or, at worst, to so-called toxic megacolon. The term toxic megacolon is actually a misnomer, in that not all patients with this condition actually have a true megacolon [see Figure 9a]. In common usage, the term toxic megacolon refers to any condition associated with colitis that is severe enough to result in sloughing of the colonic mucosa; such sloughing permits endotoxins to enter the circulatory system and evoke a septic response. The signs and symptoms of toxic megacolon include those characteristic of sepsis—leukocytosis, fever, tachycardia, and hypoalbuminemia. These patients are very ill and often manifest ileus, which is an ominous development that frequently signals impending perforation. Emergency surgical intervention is required. At operation, the colon is often distended, and when the specimen is opened, the colon may appear almost autolytic [see Figure 9b]. In this state, the bowel frequently does not hold staples well; accordingly, it is often helpful to sew the distal Hartmann stump between the left and right halves of the anterior inferior rectus fascia at the lower abdominal incision and then to close the skin over it.31 Thus, if the staple line is disrupted, the result is essentially a surgical site infection that can be opened and drained, rather than the pelvic abscess [see Figure 9c] that could develop if the rectal stump were located deep within the pelvis.

Types of Disease

Segmental disease In a 2003 review of 92 consecutive cases of patients with Crohn colitis, the number of patients with segmental colonic Crohn disease and the number of those with pancolonic disease were nearly equal.30 Approximately 63% of those with segmental colitis had other disease involvement as well (e.g., jejunoileal, ileocolic, or perianal), compared with only 12% of those with pancolitis. The recurrence rate, however, was higher in patients with segmental colitis than in those with pancolitis. In addition, the risk of recurrence was higher in patients who had granulomatous disease than in those who did not.

Pancolonic disease In cases of pancolonic Crohn disease with associated perianal, jejunoileal, or ileocolic involvement, diagnosis is not difficult. However, most patients with Crohn pancolitis do not have other sites of disease involvement, nor do they have granulomas.30 Consequently, differentiation of Crohn pancolitis from ulcerative colitis can be very difficult. Many patients with Crohn disease have been inappropriately subjected to colectomy with ileal pouch-anal anastomosis (IPAA) because they were initially presumed to have ulcerative colitis.

Operative Procedures

Total proctocolectomy with end ileostomy

Figure 10. Watering-can perineum

The traditional procedure for colonic Crohn disease is total proctocolectomy with end ileostomy, which is associated with an 8% to 15% rate of recurrence in the bowel proximal to the stoma.32–34 This operation remains the best choice in patients with severe rectal and anal Crohn disease (e.g., those with so-called watering-can perineum [see Figure 10]) and carries the lowest risk of disease recurrence. In contrast to the approach taken in patients with rectal cancer, which involves excising the external anal sphincter and a large portion of the levator muscles, the approach taken in those with colonic Crohn disease is intersphincteric, with dissection performed in the plane between the internal and external anal sphincters to reduce the size of the perineal wound and facilitate healing. Even with the intersphincteric approach, delayed healing of the perineal wound is common, occurring in as many as 30% of patients.

Subtotal colectomy with ileorectal or ileosigmoid anastomosis Because many patients with Crohn disease are young, surgeons have long been interested in operations that do not involve an ileostomy. In the absence of significant rectal and anal disease, subtotal colectomy with ileorectal or ileosigmoid anastomosis is an option. Unfortunately, this operation is associated with high recurrence rates (up to 70%)35; however, with the advent of more effective immunosuppressive and biologic therapy, it is hoped that these rates can be reduced. As much palpably normal distal rectum and colon as possible should be spared. The anastomosis can be stapled, though if the bowel wall is thickened, many surgeons would feel more secure with a handsewn anastomosis in either one or two layers.

Segmental resection Currently, more surgeons are advocating colon-sparing procedures [see 5:34 Segmental Colon Resection] for Crohn disease. Although this is a relatively new approach, there have already been some reports documenting the safety of segmental resection in cases of limited disease.36 In patients with colonic strictures resulting in obstruction, segmental resection into palpably normal areas of the bowel yields prompt resolution of symptoms. Because the colon performs an important water-ab sorbing function, many patients with a limited amount of small bowel can still live without intravenous supplementation if a significant segment of the colon is left in situ. However, patients with segmental Crohn disease appear to have a higher recurrence rate than those with pancolitis, as do patients with granulomas.30 Surgical treatment of Crohn disease continues to undergo reevaluation and reassessment of results on the basis of the availability of newer medical therapies.

Colectomy with IPAA Although colectomy with IPAA [see 5:33 Procedures for Ulcerative Colitis] is not an operation that one would knowingly perform in a patient with Crohn disease, every year there are many such patients who undergo this procedure as treatment of colonic inflammatory bowel disease that initially is incorrectly presumed to be ulcerative colitis but later is diagnosed as Crohn disease (on the basis of either final pathologic analysis of the resected specimen or the disease’s clinical behavior). Generally speaking, in the absence of fistulizing disease, most of these patients are able to maintain their pouch, but they require medical therapy for disease control.30,37–40

Anal Disease

Types of Disease

With stenosis For patients with anal strictures that are not regularly dilated, the outlook is poor. Such strictures pose functional obstructions and typically lead to continuing problems with fistulas and suppurative disease. They frequently become more and more fibrotic over time and often extend proximally. Most of these patients eventually require fecal diversion. Management generally involves self-dilation, which can often be done with Hegar dilators. If the stenosis is not dealt with, all other treatment of the Crohn disease is doomed to failure; obstruction at the level of the anal canal inevitably results in the persistence of anorectal disease.

Without stenosis

Figure 11. Drainage of abscesses

Anal Crohn disease without stenosis is much easier to treat medically. Long-term oral metronidazole therapy is often helpful; other medications (e.g., anti-tumor necrosis factor antibody) may be useful as well. Broad fissures are usually asymptomatic. Surgical treatment should be avoided unless the lesions are causing symptoms. Because they tend to have more liquid bowel movements, patients with Crohn disease need an optimally functioning anal sphincter; hence, fistulotomies, which divide portions of the sphincter, should be avoided if at all possible. Placement of setons through fistula tracts can often prevent abscess formation, provide drainage, and thereby prevent perianal pain while minimizing sphincter trauma. Silk sutures, vessel loops, or Penrose drains also can be used as setons [see Figure 11]. Rectovaginal fistulas pose a particular challenge. In the presence of active Crohn disease, advancement flap repair of such fistulas has a low success rate.41 Laparoscopic-assisted loop ileostomy improves the success rate, but unfortunately, the fistulas may recur when intestinal continuity is reestablished.

Postoperative Management


Chemoprophylaxis

In 1995, a prospective, randomized study showed that patients who underwent ileocolic resection and were given mesalamine postoperatively had a significant reduction in both the symptomatic and the endoscopic rate of recurrence.29 Not all of the work done since then has confirmed these results, but several studies and a meta-analysis have indicated that mesalamine does reduce the postoperative recurrence rate of Crohn disease.42 Many patients undergoing surgical treatment of Crohn disease are advised to take some type of postoperative preventive medical therapy—either a 5-ASA derivative (e.g., mesalamine) or a stronger immunosuppressive agent (e.g., 6-mercaptopurine or azathioprine). Better studies are required to document the efficacy of the latter agents in preventing recurrence. It is hoped that chemoprophylaxis will reduce the anticipated recurrence rates by 30% to 40%.

Surveillance

At present, there are no clear guidelines for surveillance after operative treatment of Crohn disease. In my opinion, however, given the increased risk of colorectal cancer in this setting, patients with Crohn disease who retain some colon should undergo colonoscopy every 2 years, not only to detect any development of colonic neoplasia but also to identify any recurrence of disease in a timely manner. If recurrent Crohn disease is detected, appropriate medical therapy should be promptly instituted, with the aim of avoiding subsequent operation if possible.

Behavioral Modification

Exposure to cigarette smoke is known to exacerbate the symptoms of Crohn disease. Smoking has been reported to affect the overall severity of the disease, with smokers having a 34% higher recurrence rate and a higher rate of reoperation than nonsmokers.43–45 A 1999 study of 141 Crohn disease patients who had undergone ileocolic resection, of whom 79 were nonsmokers and the remainder were smokers, found that the respective 5- and 10-year recurrence-free rates were 65% and 45% in smokers and 81% and 64% in nonsmokers. The recurrence rates were higher in heavy smokers (³ 15 cigarettes/day) than in moderate smokers.46

 

Clifford S. Cho, MD

Fellow, Department of Surgical Oncology
Memorial Sloan-Kettering Cancer Center

Layton F. Rikkers, MD, FACS

A.R. Curreri Professor and Chairman, Department of Surgery
University of Wisconsin Medical School

Clinical strategies for managing portal hypertension have undergone significant refinements over the past half-century. This evolution has been driven by advances in our understanding of the physiology of both the disease and the therapies employed against it. Today, clinical management of the portal hypertensive patient is a truly multidisciplinary endeavor, requiring the coordinated efforts of skilled intensivists, gastroenterologists, hepatologists, interventional radiologists, and surgeons. Nevertheless, portal hypertension and its manifold complications remain some of the most vexing problems encountered in modern medicine and surgery.

In this chapter, we briefly review portal venous anatomy and the pathophysiology of portal hypertension [see Sidebar Portal Hypertension: Anatomic and Physiologic Considerations]; however, our main focus is on current practical approaches to managing portal hypertension and its associated sequelae (variceal bleeding, ascites, and hepatic encephalopathy). Of particular relevance to surgeons is that the role of surgical therapy has shifted significantly. Operative treatment now occupies only the final steps in modern treatment protocols for portal hypertension—that is, it serves as a form of salvage for intractable cases that are refractory to other forms of therapy.

Clinical Evaluation


The ultimate aims of diagnostic evaluation in a patient with portal hypertension are (1) to determine the cause of portal hypertension [see Table 1], (2) to estimate hepatic functional reserve, (3) to define the portal venous anatomy and assess hemodynamic status, and (4) to identify the site of GI hemorrhage (if present). Any history of chronic alcohol abuse, hepatitis, or exposure to hepatotoxins raises the suspicion of cirrhotic liver disease. Confirmatory evidence of chronic liver disease on physical examination may be found in the form of jaundice, chest wall spider angiomata, palmar erythema, Dupuytren contractures, testicular atrophy, or gynecomastia. Ascites, splenomegaly, caput medusae, encephalopathic alterations in mental status, and asterixis are all suggestive of portal hypertension.

Investigative Studies


Laboratory studies can also provide indicators of hepatic dysfunction. The hypersplenism that often accompanies cirrhosis can produce mild to moderate pancytopenia. Anemia may also reflect variceal hemorrhage, hemolysis, or simply the chronic malnutrition or bone-marrow suppression associated with chronic alcoholism. Associated hyperaldosteronism, emesis, or diarrhea may give rise to electrolyte derangements, including hyponatremia, hypokalemia, metabolic alkalosis, and prerenal azotemia. Coagulopathy is usually attributable to chronic deficiencies in clotting factors that are normally synthesized by the liver; thus, elevation of the prothrombin time (PT) or the international normalized ratio (INR) often reflects the degree of chronic hepatic impairment. Similarly, the degree of hyperbilirubinemia can be a measure of both acute and chronic hepatic dysfunction. Hepatocellular necrosis results in marked elevations in serum aminotransferases that are readily observed in patients with chronic active viral or alcoholic hepatitis. An alanine aminotransferase (ALT)-aspartate aminotransferase (AST) ratio of 2 or higher is often seen in patients with alcoholic liver disease.

The Child-Pugh scoring system is a useful tool for quantifying hepatic functional reserve [see Table 2].1 Based on total bilirubin and albumin levels, PT (INR), and the clinical severity of ascites and hepatic encephalopathy, the Child-Pugh score predicts both the likelihood of variceal hemorrhage and its anticipated mortality. A newer assessment tool, the Model for End-Stage Liver Disease (MELD) scoring system, which takes the degree of renal impairment and the cause of hepatic dysfunction into account, has also been used to predict outcomes in cirrhotic patients.2

Management of Variceal Bleeding


The prognosis of variceal hemorrhage depends on the presence or absence of underlying cirrhosis. In noncirrhotic patients, the mortality associated with a first episode of variceal hemorrhage ranges from 5% to 10%; in cirrhotic patients, the range is from 40% to 70%. Esophagogastric varices ultimately develop in approximately one half of cirrhotic patients, and bleeding episodes occur in approximately one third of cirrhotic patients with varices. If the initial hemorrhagic episode resolves spontaneously, 30% of patients experience rebleeding within 6 weeks, and 70% experience rebleeding within 1 year. It is noteworthy that overall mortality in patients who survive 6 weeks after an episode of variceal bleeding is statistically indistinguishable from that in persons who have never experienced such an episode.

Further risk stratification is based on the extent of hepatic decompensation. The mortality associated with variceal hemorrhage is 5% for patients with Child class A cirrhosis, 25% for those with Child class B cirrhosis, and over 50% for those with Child class C cirrhosis. The likelihood of recurrent hemorrhage is 28% for patients with Child class A cirrhosis, 48% for those with Child class B cirrhosis, and 68% for those with Child class C cirrhosis.3

Treatment of Acute Variceal Hemorrhage

Figure 1. Treatment of acute variceal bleeding

Management of acute variceal hemorrhage [see Figure 1] begins with the establishment of adequate airway protection. The risk of aspiration and consequent respiratory deterioration is particularly high among patients with hepatic encephalopathy and those undergoing endoscopic therapy. Accordingly, the threshold for early endotracheal intubation should be low, particularly if endosopic therapy is considered. As with all cases of brisk hemorrhage, adequate venous access is mandatory; placement of a central venous catheter for accurate volume assessment is particularly useful in cases of major bleeding. The presence of chronic liver disease often necessitates vigorous replacement of circulatory volume and coagulation factors, often involving infusion of colloids and transfusion of fresh frozen plasma and packed red blood cells. Antibiotic prophylaxis therapy is recommended because of the propensity of bacterial infections to develop in patients with chronic liver disease after bleeding episodes.

Pharmacologic Therapy

First-line pharmacotherapy for acute variceal bleeding relies on the long-acting somatostatin analogue octreotide, which has been shown to decrease splanchnic blood flow and portal venous pressure. Octreotide is administered in a 250 µg I.V. bolus, followed by infusion of 25 to 50 µg/hr for 2 to 4 days.4 In addition, vasopressin, a strong splanchnic vasoconstrictor, has been shown to control approximately 50% of acute variceal bleeding episodes.4,5 Vasopressin is typically administered in a 20 U I.V. bolus over 20 minutes, followed by infusion of 0.2 to 0.4 U/min. The therapeutic benefits of octreotide and vasopressin appear to be similar, though the side-effect profile of octreotide appears to be much lower than that of vasopressin monotherapy.4 Adjunctive use of nitroglycerin at an initial rate of 50 µg/min (titrated according to blood pressure tolerance) effectively reduces the cardiac complications of vasopressin and thereby facilitates its administration.6 The long-acting vasopressin analogue terlipressin has been shown to be approximately as effective as octreotide.7

Endoscopic Therapy

Endoscopic treatment, in the form of sclerosant injection or band ligation, has become a standard form of therapy for acute variceal hemorrhage. Experienced endoscopists achieve initial control of hemorrhage in 74% to 95% of cases; however, rebleeding rates ranging from 20% to 50% are typically observed.

In endoscopic sclerotherapy, a sclerosant—typically either 5% sodium morrhuate (more common in the United States) or 5% ethanolamine oleate (more common in Europe and Japan)—is injected either intravariceally to obliterate the varix or paravariceally to induce submucosal fibrosis and thereby prevent variceal rupture. Three prospective, randomized, controlled trials demonstrated that endoscopic sclerotherapy, compared with traditional balloon tamponade, achieved better initial hemorrhage control, resulted in fewer episodes of rebleeding, and, in selected cohorts of patients, led to improved long-term survival.8–10 Furthermore, routine use of balloon tamponade after sclerotherapy appeared not to confer any additional therapeutic benefit.8 There are, however, significant risks associated with the use of endoscopic sclerotherapy, including pulmonary complications, transient chest pain, esophageal stricture formation with recurrent sclerotherapy, iatrogenic portal vein thrombosis, hemorrhagic esophageal ulceration, bacteremia, and esophageal perforation.11

Partially in response to the potential complications of endoscopic sclerotherapy, endoscopic variceal band ligation has been advocated as a sclerosant-free therapeutic alternative. The limited data comparing the two approaches suggest a trend toward fewer rebleeding episodes, fewer endoscopic interventions, and significantly lower procedure-related morbidity and overall mortality after variceal ligation.12

Pharmacologic versus Endoscopic Therapy

Two meta-analyses compared medical pharmacotherapy with emergency sclerotherapy as first-line treatment of acute variceal hemorrhage.13,14 No significant differences between the two approaches were demonstrated with respect to initial hemorrhage control or mortality, though treatment-related complications appeared to be significantly more common after sclerotherapy. On the basis of these studies, it has been suggested that endoscopic treatment should be reserved for cases of pharmacotherapeutic failure or that pharmacologic therapy should be initiated in situations where endoscopy is not immediately available. At present, however, it is more common for the two forms of treatment to be employed concurrently. Pharmacotherapy is often initiated in preparation for endoscopy; early mitigation or control of variceal hemorrhage can make endoscopic visualization and intervention easier, safer, and more effective. Indeed, administration of somatostatin before and after endoscopic sclerotherapy has been shown to improve treatment efficacy and decrease transfusion requirements in comparison with endoscopic sclerotherapy alone.15,16

Balloon Tamponade

Figure 2. Sengstaken-Blakemore tube

Although the devices used for balloon tamponade have evolved through numerous different forms over the years, all of them rely on the same basic principle—application of direct upward pressure against varices at the esophagogastric junction. Patients for whom balloon tamponade is considered should be intubated endotracheally to prevent airway occlusion and aspiration. The tube is inserted into the stomach, and the gastric balloon is partially inflated with 40 to 50 ml of air [see Figure 2]. An abdominal radiograph is obtained to ensure that the gastric balloon is correctly positioned within the stomach and below the diaphragm. This balloon is then further inflated until it holds 300 ml of air, and the tube is pulled upward with external traction. If hemorrhage is not controlled at this point, the esophageal balloon is inflated to a pressure of 35 to 40 mm Hg. Suction drainage is applied to both the esophageal port and the gastric port to minimize aspiration risk and monitor for recurrent hemorrhage.

When properly applied, direct tamponade therapy is 90% effective in controlling acute hemorrhage. The primary limitation of such therapy is that bleeding resumes in as many as 50% of patients after takedown and removal of the balloon. Furthermore, serious potential complications (e.g., gastric or esophageal perforation, aspiration, and airway obstruction) result in treatment-related mortalities as high as 20%.17,18 Nevertheless, in cases of brisk variceal hemorrhage refractory to pharmacologic and endoscopic therapy, balloon tamponade may have a role to play as a bridge therapy to more definitive forms of treatment, such as transjugular intrahepatic portosystemic shunting (TIPS) (see below) or operative intervention.

Transjugular Intrahepatic Portosystemic Shunting

Figure 3. Procedure for performing TIPS

A nonoperative technique for creating an intrahepatic portosystemic fistula for decompression of portal hypertension was proposed in 196919 and first performed in 1982.20 As currently practiced, TIPS is performed by (1) cannulating a hepatic vein (usually the right hepatic vein) via the internal jugular vein, (2) passing a needle from the hepatic vein through the liver parenchyma and into a portal vein branch, (3) passing a guide wire through the needle, (4) dilating the needle tract with a balloon passed over the guide wire, and (5) stenting the tract to a desired diameter, thus effectively constructing a nonselective side-to-side portosystemic shunt [see Figure 3].

Experience with TIPS in the setting of acute variceal hemorrhage is limited. However, one meta-analysis of studies comparing the efficacy of conventional endoscopic therapy (with or without pharmacotherapy) with that of TIPS in treating acute hemorrhagic episodes demonstrated a significant improvement in hemorrhage control with TIPS.21 Unfortunately, this improvement came at the cost of increased rates of hepatic encephalopathy as a consequence of the nonselective shunting of portal venous flow into the systemic venous circulation. Furthermore, the meta-analysis failed to demonstrate a significant improvement in overall mortality with TIPS.21

Given the relative paucity of data on the use of TIPS as first-line therapy for acute variceal hemorrhage, it is logical to recommend that TIPS be employed in cases of pharmacotherapeutic and endoscopic failure; the efficacy of TIPS as salvage therapy in this setting is well documented.22 Contraindications to TIPS include right heart failure and polycystic liver disease. Portal vein thrombosis is a relative contraindication.

Surgical Therapy

The role of surgical management in the treatment of acute variceal bleeding has changed considerably over the past 50 years. At present, operative intervention is reserved for cases that have proved refractory to pharmacotherapy, endoscopy, balloon tamponade, and TIPS. Numerous operations have been developed, each with its own merits and flaws.

Esophageal transection with an end-to-end anastomosis (EEA) stapler has been employed as a means of interrupting blood flow into bleeding esophageal varices. In this technique, the esophagus is mobilized, and the EEA stapler is passed into the distal esophagus through a gastrotomy. With care taken not to injure the vagus nerves and the external periesophageal veins that may be providing collateral venous drainage, a full-thickness segment of the esophagus is transected. When this technique is used on an emergency basis in a patient with acutely bleeding varices, operative mortality is as high as 76%, and the rate of operative complications (e.g., esophageal perforation, stricture, esophagitis, and infection) is approximately 26%.23 Accordingly, esophageal transection is not commonly advocated as a useful form of surgical therapy for acutely bleeding esophageal varices.

In contrast, portosystemic shunting operations have been widely used to treat acute variceal hemorrhage. The largest single body of data on this practice comes from Orloff and associates,24 who reported remarkable outcomes—71% survival at 10 years—in 400 consecutive patients undergoing emergency portacaval shunt operations (mostly side-to-side) over a 28-year period. Unfortunately, these investigators’ experience stands in stark contrast to that of most other groups, who uniformly reported operative mortalities of about 40% and 5-year survival rates of about 30%.

Another potential drawback to urgent operative shunting is the manipulation and dissection that are often necessary in the region of the porta hepatis: these measures can result in adhesions and scarring, which can complicate future orthotopic liver transplantation. For this reason, some surgeons have advocated using the mesocaval interposition shunt [see Prevention of Recurrent Variceal Hemorrhage, Surgical Therapy, Portosystemic Shunts, Nonselective Shunts, below] in the emergency setting because of its ability to lower portal pressure without complicating the hilar dissection that will be necessary if transplantation is carried out later.25 In addition, surgeons familiar with the distal splenorenal shunt (DSRS) can employ this selective shunt in some cases of acute variceal hemorrhage unaccompanied by refractory ascites.

Prevention of Recurrent Variceal Hemorrhage

Pharmacotherapy

Figure 4. Prevention of recurrent variceal bleeding

Without further treatment, the likelihood that hemorrhage will recur within 1 year after control of an acute episode of variceal bleeding is approximately 70%.26 The pharmacologic maneuver that has been used most extensively to prevent recurrent variceal bleeding [see Figure 4] is nonselective beta-adrenergic blockade, most commonly with propranolol. Although beta blockade has been shown to lower portal pressure and hepatic vein wedge pressure, its ability to induce this effect is variable and unpredictable.27 Nevertheless, a meta-analysis of multiple trials studying the effectiveness of nonselective beta blockade demonstrated a significant decline in recurrent bleeding and a trend toward improved overall survival.4 Patients with decompensated hepatic function appear to derive less benefit from beta blockade, possibly because of the downregulation of beta-adrenergic receptors associated with cirrhosis.28 Adjunctive use of the long-acting vasodilator isosorbide 5-mononitrate (ISMN) appears to potentiate the efficacy of propranolol therapy.29

Endoscopic Therapy

Repeated endoscopic therapy with sclerosant injection or band ligation has been advocated as a means of completely eradicating esophageal varices. Once the varices are eliminated, routine endoscopy is performed at 6- to 12-month intervals to prevent recurrent hemorrhage. Compared with medical treatment, long-term endoscopic therapy results in fewer rebleeding episodes.4 Nevertheless, approximately one half of endoscopically treated patients eventually experience recurrent hemorrhage, usually within the first year. Approximately one third of patients treated with repeated endoscopy ultimately must be converted to another form of therapy because of unrelenting major bleeding.30,31 For this reason, such extended endoscopic surveillance and treatment should be reserved for compliant patients who live in proximity to tertiary medical care and should be administered with the understanding that conversion to a more definitive form of therapy may be necessary if endoscopy fails.

Transjugular Intrahepatic Portosystemic Shunting

TIPS [see Figure 3] has been employed to prevent recurrent episodes of variceal hemorrhage, particularly as a form of bridge therapy for patients awaiting orthotopic liver transplantation. The potential advantage TIPS has over surgical portosystemic shunting is the ability to decompress the portal system without the risks associated with general anesthesia and without postoperative complications. The major limitation of TIPS is the shunt stenosis (caused by neointimal hyperplasia or thrombosis) that occurs in as many as 50% of patients in the first year after the procedure. Fortunately, most such episodes of stenosis are amenable to balloon dilatation or secondary shunt insertion; however, 10% to 15% of TIPS recipients experience total shunt occlusion that cannot be reversed. Furthermore, TIPS functions as a nonselective shunt, leading to hepatic encephalopathy in approximately one third of patients.32

Meta-analytic comparison of TIPS with endoscopic therapy indicates that rebleeding episodes are markedly reduced in patients treated with TIPS, but at the cost of a higher incidence of encephalopathy and a shunt malfunction rate of at least 50%. That the efficacy of TIPS is relatively short-lived makes this modality an ideal form of bridge therapy for patients who are awaiting orthotopic liver transplantation or those who have severe hepatic decompensation and thus are unlikely to live long enough to experience failure of TIPS. TIPS can reduce the number of bleeding episodes for patients on the transplant waiting list. In addition, the significant reduction in portal pressure produced by TIPS technically facilitates future liver transplantation. Finally, unlike surgical shunts, TIPS is completely removed at the time of recipient native hepatectomy.

Surgical Therapy

Surgical therapy is the most effective method of controlling portal hypertension and preventing recurrent variceal hemorrhage. The operative procedures available to the surgeon have undergone numerous modifications and become more effective over the years. Review of the surgical experience reveals that with the onset of alternative modalities (e.g., TIPS and transplantation), the risk status of patients undergoing surgical therapy (as predicted by Child’s classification) and the frequency of emergency operations have steadily declined. As a result, the incidence of postoperative hepatic encephalopathy has gradually fallen and overall survival has gradually improved.33 Surgical options for the prevention of recurrent variceal hemorrhage in patients with portal hypertension may be divided into three categories: (1) portosystemic shunt procedures, (2) esophagogastric devascularization, and (3) orthotopic liver transplantation.

Portosystemic shunts Surgical portosystemic shunting provides a means of decompressing the hypertensive portal venous system into the low-pressure systemic venous circulation. Diversion of portal blood flow from the liver also deprives the liver of important hepatotrophic hormones that are present in portal venous blood while routing cerebral toxins normally metabolized by the liver directly into the systemic circulation. As a result, the primary complications of surgical portosystemic shunting are accelerated hepatic dysfunction and hepatic encephalopathy. Primarily in an attempt to minimize these adverse sequelae, various forms of portosystemic shunting operations have evolved, which may be classified as nonselective shunts, selective shunts, or partial shunts.

Nonselective shunts

Figure 5. Portosystemic shunts

The classic nonselective portosystemic shunt is the end-to-side portacaval shunt (the so-called Eck fistula) [see Figure 5, part a]. This is the only nonselective shunt that has been rigorously compared with conventional nonoperative therapy. Several randomized, controlled trials demonstrated superior control of bleeding after operative shunting: 9% to 25% of patients experienced rebleeding after portacaval shunting (mostly related to nonvariceal hemorrhage or shunt thrombosis), whereas 65% to 98% of patients experienced rebleeding after medical therapy.34–37Markedly higher rates of spontaneous posttreatment encephalopathy were reported in the operative shunt groups; however, the overall rates of encephalopathy did not differ between the operative groups and the medical groups, because the encephalopathy seen in the medically treated patients (mainly attributable to hemorrhage and infection) eventually became equivalent to that seen in the surgically treated patients. There were trends toward improved overall survival in the surgical groups, but these trends did not attain statistical significance.

The side-to-side portacaval shunt [see Figure 5, part b] maintains the anatomic continuity of the portal vein as it passes into the liver. However, the high sinusoidal resistance typically present in the setting of cirrhosis effectively renders this shunt a nonselective one, with no measurable antegrade (i.e., hepatopedal) portal blood flow into the liver. Consequently, the encephalopathy rates are no different from those observed after end-to-side portacaval shunting. Side-to-side portacaval shunting does offer the benefit of decompressing the hepatic sinusoidal pressure via reversed (i.e., hepatofugal) flow of blood from the liver into the portal vein. Because transudation of interstitial fluid from both the liver and the intestines is thought to contribute to ascites formation, better control of ascites is achieved with a side-to-side portacaval shunt, which effectively decompresses both the splanchnic veins and the intrahepatic sinusoids, than with an end-to-side portosystemic shunt, which decompresses only the splanchnic veins. The side-to-side portacaval shunt is therefore also recommended for patients with Budd-Chiari syndrome, in whom an end-to-side portacaval shunt would not relieve intrahepatic congestion resulting from hepatic venous outflow occlusion. Otherwise, no significant outcome differences between end-to-side and side-to-side portacaval shunts have been documented. The end-to-side variant is, however, technically easier to construct.

Placement of an interposition mesocaval shunt [see Figure 5, part c] composed of prosthetic or autogenous vein grafts offers the technical advantages of avoiding hilar dissection (thereby making future liver transplantation less complicated) and permitting intentional shunt ligation in the event of refractory postoperative encephalopathy. Like the side-to-side portacaval shunt, the interposition shunt functions physiologically as a nonselective shunt because of the hepatofugal portal venous blood flow. The major drawback to the interposition shunt is shunt thrombosis, which may develop in as many as 35% of cases.

The conventional (proximal) splenorenal shunt [see Figure 5, part d] was initially advocated as a means of decompressing portal venous flow while retaining hepatopedal hepatic portal perfusion. This shunt is constructed by performing a splenectomy and anastomosing the proximal splenic vein to the left renal vein. Physiologic testing of patent conventional splenorenal shunts suggests that they eventually divert all portal flow into the renal vein and therefore effectively function as nonselective shunts. Indeed, long-term rates of hepatic encephalopathy appear to be no lower after conventional splenorenal shunting than after portacaval shunting.38 Shunt occlusion develops in about 18% of cases.39

In short, nonselective portosystemic shunts are an effective means of controlling variceal hemorrhage in cases that are refractory to other therapeutic approaches. Given the absence of any major differences in the rate of encephalopathic complications after the various nonselective shunts, the choice of a nonselective shunting procedure should be based on the surgeon’s technical familiarity with the operations and on the patient’s candidacy for future transplantation. The end-to-side portacaval shunt can be constructed relatively quickly but should be avoided in patients who have intractable ascites or Budd-Chiari syndrome and those who may subsequently undergo liver transplantation. The side-to-side portacaval shunt may provide better control of ascites but is technically more challenging to construct and should also be avoided if future transplantation is an option. The interposition mesocaval shunt is relatively easy to construct and avoids hepatic hilar dissection but is associated with a relatively high rate of shunt occlusion when a nonautogenous conduit is used. The conventional splenorenal shunt also avoids hilar dissection but is associated with a high shunt occlusion rate and is technically challenging to construct.

 

Selective shunts

Figure 6. Distal splenorenal shunt

In response to the postoperative complications seen after nonselective portosystemic shunting (hepatic encephalopathy and hepatic failure), Warren and colleagues introduced the distal splenorenal shunt in 1967.40 The DSRS has become the prototypical selective shunt, in that it selectively decompresses the esophagogastric veins while maintaining hepatopedal flow from the mesenteric veins. It is performed by anastomosing the distal splenic vein to the left renal vein and interrupting venous collaterals (e.g., the left gastric and right gastroepiploic veins) [see Figure 6]. As a result, the DSRS effectively separates the portal system into two components: (1) a decompressed esophagogastric venous circuit and (2) a persistently hypertensive mesenteric venous circuit that continues to provide hepatopedal portal flow. Thus, the DSRS does not address the mesenteric and sinusoidal hypertension that is responsible for ascites formation. Indeed, it is believed that the extensive retroperitoneal dissection required to construct this shunt may actually contribute to ascites formation through inadvertent disruption of retroperitoneal lymphatic vessels. The DSRS is contraindicated in patients who have refractory ascites or splenic vein thrombosis, those who have previously undergone splenectomy, and those with an excessively small (< 7 mm) splenic vein diameter.

Unfortunately, perfusion studies indicate that approximately one half of patients lose hepatopedal flow within 1 year after a DSRS procedure. This is a particular problem in patients with alcoholic cirrhosis. The loss of shunt selectivity is believed to result from progressive collateral diversion of portal flow into the splenic vein via a network of pancreatic and peripancreatic veins (the so-called pancreatic siphon effect). Extensive skeletonization of the splenic vein off the pancreas (so-called splenopancreatic disconnection) has been proposed as a means of minimizing this unwanted collateralization,41 but at present, the evidence is insufficient to support routine employment of this measure.

The complications of DSRS procedures are well described. Depending on patient selection, postoperative ascites formation is seen in 7% to 98% of cases; however, in only 0% to 14% of cases is ascites clinically significant and refractory to dietary sodium restriction and diuresis.23 Hepatic encephalopathy is reported in 0% to 32% of cases; several clinical trials comparing DSRS with nonselective shunting demonstrated significantly lower rates of encephalopathy after DSRS, whereas other trials found no statistically significant difference. With respect to overall survival and hemorrhage control, DSRS and nonselective shunts appear to be equivalent.42

Comparison of DSRS construction with endoscopic therapy has yielded interesting results. Two controlled trials comparing endoscopic therapy and salvage DSRS with early DSRS alone demonstrated superior hemorrhage control with early DSRS.30,31 Rates of hepatic encephalopathy did not differ between the two groups. One of the trials, conducted in an urban-suburban area where 85% of sclerotherapy failures could be rescued with salvage DSRS, found survival to be improved in patients treated with endoscopic therapy and salvage DSRS, compared with survival in patients treated with early DSRS alone.30 The other, performed in a less densely populated region where only 31% of sclerotherapy failures could be rescued with salvage DSRS, found survival to be improved in the early DSRS group.31 These data suggest that early definitive surgical intervention may be preferable for patients who are too far from a tertiary medical center to be able to reach one expeditiously in the event of uncontrollable hemorrhage.

Attention is now being turned toward comparisons between the DSRS and TIPS. One uncontrolled comparative study found that with DSRS, hemorrhage control was better, the encephalopathy rate was lower, and shunt occlusion was reduced, but the incidence of postoperative ascites was higher.43 A National Institutes of Health-sponsored randomized comparison between DSRS and TIPS is currently under way at multiple centers.

The other main form of selective portosystemic shunt is the coronary-caval shunt, initially described in Japan in 1984.44 This shunt is constructed by anastomosing an interposition graft to the left gastric (coronary) vein on one end and the inferior vena cava on the other. To date, the applicability of this procedure has been limited, and most surgeons have relatively little experience with it.

 

Partial shunts Various small-diameter interposition portosystemic shunts have been proposed as partial shunts, designed to achieve partial decompression of the entire portal venous system while maintaining a degree of hepatopedal portal flow to the liver. The most successful of these partial shunts has been the small-diameter portacaval interposition shunt. The use of a 10 mm or smaller interposition shunt, combined with extensive disruption of portosystemic collateral venous circuits, serves to maintain some degree of hepatic portal perfusion. Early experience with the 8 mm ringed polytetrafluoroethylene graft suggests that hepatic encephalopathy rates are lower with this shunt than with nonselective 16 mm grafts and that use of the smaller shunt yields comparable long-term survival.45 An early comparison of the small-diameter portacaval shunt with TIPS demonstrated lower rates of shunt occlusion and treatment failure in the operative therapy group.46

Esophagogastric devascularization

Figure 7. Modified Sugiura procedure

The most effective nonshunt operation for preventing recurrent variceal hemorrhage is esophagogastric devascularization with esophageal transection and splenectomy, as advocated by Sugiura and associates.47 Unlike simple esophageal transection, which has been used with limited success in the setting of acute hemorrhage, the Sugiura procedure and its subsequent modifications [see Figure 7] involve ligation of venous branches entering the distal esophagus and the proximal stomach from the level of the inferior pulmonary vein, combined with selective vagotomy and pyloroplasty [see 5:20 Procedures for Benign and Malignant Gastric and Duodenal Disease]. A key point is that the left gastric (coronary) vein and the paraesophageal collateral veins are preserved to permit portoazygous collateralization, which inhibits future varix formation. Initial reports from Japan cited a 5.2% operative mortality and a 6.3% rate of recurrent hemorrhage (most often from nonvariceal causes).47,48 Unfortunately, these successes have not been easily replicated in the United States, where operative mortality with this procedure has exceeded 20%, with bleeding recurring in 35% to 55% of patients.49,50 Nevertheless, modifications of the Sugiura procedure continue to be performed in patients who are unable to undergo shunting procedures because of extensive splanchnic vein thrombosis.

Orthotopic liver transplantationOrthotopic liver transplantation is the most definitive form of therapy for complications of portal hypertension. The cost of cadaveric and living-donor liver transplantation and its attendant immunosuppression, as well as the paucity of available allografts, make liver replacement an option for only a select minority of patients presenting with portal hypertensive sequelae. Accordingly, careful analysis of the outcomes of transplantation procedures in comparison with those of nontransplantation procedures is necessary for optimal allocation of this limited resource.

For patients whose portal hypertension has become refractory to nonoperative management strategies, the decision whether to employ transplantation or nontransplantation operative therapy can be based on the level of hepatic functional reserve. Patients with Child class A or mild class B cirrhosis appear to do well with nontransplantation therapy as first-line operative treatment, with the understanding that liver transplantation may remain an option for salvage therapy in the event of future hepatic functional deterioration. In contrast, patients with more advanced Child class B or Child class C cirrhosis appear to benefit from early transplantation, with nonoperative strategies employed strictly as bridge therapy for maintenance during the time spent on the allograft waiting list.51,52

 

Prophylaxis of Initial Variceal Hemorrhage

The significant mortality associated with variceal hemorrhage has prompted efforts to devise effective means of preventing the onset of initial variceal bleeding. The difficulty of identifying those 20% to 33% of cirrhotic patients who will experience bleeding episodes remains the primary challenge in the application of prophylaxis for variceal hemorrhage. Patient characteristics that predict an increased likelihood of variceal bleeding include alcoholic cirrhosis, active alcohol consumption, and severe hepatic dysfunction.53 Certain anatomic features of varices seen at the time of endoscopic examination have been shown to predict the likelihood of rupture: evidence of variceal wall thinning (cherry-red spots, red wales), variceal tortuosity, superimposition of varices on other varices, and the presence of gastric varices all appear to be correlated with a higher likelihood of hemorrhage.54

At present, pharmacologic therapy is the only measure that provides effective prophylaxis against variceal hemorrhage. Nonselective beta-adrenergic blockade, either with propranolol or the long-acting agent nadolol, reduces portal venous pressure by decreasing cardiac output and favoring splanchnic vasoconstriction. Clinical trials examining the efficacy of propranolol therapy demonstrated lowered rates of initial variceal bleeding, though the ultimate influence of beta blockade on patient survival was mixed.55–57

Endoscopic sclerotherapy has not been consistently effective in preventing initial variceal bleeding. In fact, several trials found survival to be poorer in patients treated with prophylactic sclerotherapy than in those managed with prophylactic pharmacotherapy.4,58 This difference is probably attributable to the well-documented complications associated with endoscopic sclerotherapy.

The flaws of prophylactic endoscopic sclerotherapy have led some authorities to advocate endoscopic variceal band ligation as a more effective form of prophylaxis. One trial demonstrated that variceal band ligation achieved better prophylaxis of initial variceal bleeding than propranolol therapy did.59 Clearly, this observation warrants further investigation.

Early trials comparing prophylactic portosystemic shunting with medical prophylaxis definitively showed that early operative intervention conferred no significant benefit. In fact, the significant morbidity associated with surgical shunting and the substantial risk of accelerated hepatic dysfunction and encephalopathy led to lower survival rates in patients treated with prophylactic surgical procedures.6,60 At present, the data are insufficient to recommend the use of prophylactic TIPS to prevent acute variceal hemorrhage.

Management of Ascites


The presence of ascites in a patient with portal hypertension is typically an ominous finding that is of significant prognostic importance: 1-year mortalities as high as 50% have been reported in cirrhotic patients with new-onset ascites, whereas baseline 1-year mortalities in cirrhotic patients without ascites are in the range of 10%.61 The pathogenesis of ascites formation appears to be related to the relative hypovolemia and the primary avidity of renal sodium retention that develop in patients with cirrhosis. Hypovolemia induces renin-angiotensin activation and salt and water reabsorption, which, in the setting of chronic liver dysfunction, results in excessive transudation of fluid out of the liver and the intestines and into the peritoneal cavity. The major complications of this process are spontaneous bacterial peritonitis (SBP) and hepatorenal syndrome (HRS) [see Complications, below], which account for the bulk of the morbidity and mortality associated with ascites in patients with portal hypertension.

Nonsurgical Therapy

By addressing the hyperavidity of sodium retention that drives much of ascites formation, restriction of dietary salt intake (to levels as low as 2 g of sodium a day) can resolve ascites in approximately 25% of cases. The hyperaldosteronemic state that exists can be countered by initiating diuresis with spironolactone, which, at dosages ranging from 100 to 400 mg/day, can relieve ascites in an additional 60% to 70% of patients. Although automatic addition of loop diuretics has not been proved to enhance the clinical efficacy of spironolactone, augmentation of spironolactone therapy with furosemide can be helpful for patients whose ascites is refractory to spironolactone monotherapy or who have hyperkalemia as a result of spironolactone treatment. Gradual diuresis is necessary to prevent potential complications (e.g., prerenal azotemia and HRS).62

In cases of ascites that is refractory to medical dietary restriction and diuretic therapy, large-volume paracentesis has been employed with some success. Albumin is typically infused at a dose of 6 to 8 g per liter of ascitic fluid to prevent the hypotension that results from acute volume shifts. Patients in whom ascites recurs after multiple rounds of large-volume paracentesis should be considered for TIPS. TIPS is particularly useful in patients with ascites and a history of bleeding esophageal varices; it corrects as many as 80% of medically refractory cases of ascites.63 However, the efficacy of TIPS is counterbalanced by its attendant risks (i.e., hepatic encephalopathy, shunt occlusion, and accelerated hepatic failure), especially in patients with poor hepatic functional reserve.

Surgical Therapy

Operative intervention plays only a limited role in the management of ascites. Surgically inserted peritoneovenous shunts have been compared with large-volume paracentesis in patients with ascites refractory to medical therapy. No significant differences in early control of ascites have been detected, but patients treated with peritoneovenous shunting appear to benefit from faster ascites resolution, longer palliation, and fewer hospital readmissions.64 Long-term follow-up, however, indicates that shunt occlusion occurs in 47% of patients so treated and disseminated intravascular coagulation in as many as 35%.

The morbidity and mortality associated with operative therapy make routine use of side-to-side portacaval shunts a poor option for managing ascites. The exceptions to this general statement are cases in which ascites proves refractory to medical and TIPS therapy or in which concomitant refractory variceal hemorrhage is present.

Complications

SBP is the most common form of ascitic infection. It typically is signaled by fever and abdominal tenderness and often is also accompanied by acute hepatic and renal deterioration. The diagnosis is generally made by analyzing ascitic fluid collected through paracentesis and is defined by the presence of a positive bacterial culture and a neutrophil count higher than 250/mm3 in the absence of an obvious intra-abdominal source of infection. Unlike secondary peritonitis, SBP is typically monomicrobial, and the frequency with which enteric gram-negative rods are found with SBP suggests intestinal bacterial translocation as a potential cause. SBP carries a mortality of 25% and should therefore be treated aggressively with I.V. antibiotic therapy. Given the 70% recurrence rate after an initial episode of SBP, continuation of suppressive antimicrobial therapy until ascites resolves is warranted.65

HRS, a poorly understood state characterized by progressive and refractory renal impairment, typically occurs in the setting of tense ascites and hepatic disease. Management of HRS is strictly supportive, in that the syndrome often responds only to correction of the underlying liver dysfunction. Accordingly, the only proven therapy for HRS is liver transplantation.

Management of Hepatic Encephalopathy


Hepatic encephalopathy is a complex of symptoms characterized by mental status changes ranging from impaired mentation to frank stupor. The classic neurologic finding associated with this symptom complex is asterixis. Typically, hepatic encephalopathy develops in the setting of significant portosystemic shunting or significant hepatic functional impairment. It is most commonly observed after the creation of a therapeutic nonselective portosystemic shunt. Its onset is usually precipitated by dehydration, GI hemorrhage, sepsis, or excessive protein intake; in fact, the spontaneous development of hepatic encephalopathy mandates work-up for these physiologic triggers. It has been speculated that the shunting of intestinally absorbed cerebral toxins (e.g., ammonia, mercaptans, and g-aminobutyric acid) away from hepatic metabolism is what causes hepatic encephalopathy; however, the absolute level of circulating ammonia correlates poorly with the magnitude of encephalopathic symptoms.

Correction of the triggers that cause hepatic encephalopathy often reverses the psychoneurologic disturbances. In severe cases, patients should also receive neomycin (1.5 g every 6 hours), which covers enteric urease-positive bacteria, and lactulose (20 to 30 g two to four times daily), a disaccharide GI cathartic. Both agents are believed to reduce intestinal levels of ammonia and inhibit its enteric absorption. Whereas neomycin has long-term side effects (i.e., nephrotoxicity and ototoxicity), long-term lactulose therapy is generally well tolerated. Dietary protein restriction should also be employed for long-term suppression of hepatic encephalopathy. On occasion, refractory cases of shunt-induced hepatic encephalopathy may be treated by means of intentional ligation or occlusion of the portosystemic shunt.

 

Steven M. Strasberg, MD, FACS, FRCS(C), FRCS(Ed)

Pruett Professor of Surgery
Washington University School of Medicine, St. Louis, MO

David C. Linehan, MD, FACS

Assistant Professor, Department of Surgery, and Associate Professor and Chief, Section of Hepatobiliary, Pancreatic and Gastrointestinal Surgery, Barnes Jewish Hospital
Washington University School of Medicine, St. Louis, MO

 

10.2310/7800.S05C09

Several different types of tumors affect the pancreas, the biliary tree, and the liver. Each year, hundreds of articles are published regarding pancreatic, biliary, and hepatic cancers. Accordingly, this chapter concentrates on essential principles. In particular, it focuses on common malignant tumors, addressing benign tumors and uncommon tumors only insofar as they are important in differential diagnosis.

When a patient presents with an apparent cancer of the pancreas, the biliary tree, or the liver, the surgeon must attempt to answer the following three important questions:

  1. What is the diagnosis?
  2. What is the surgical stage of the disease—that is, is the tumor resectable?
  3. What is the operative rationale that will encompass the disease and produce a margin-free resection (and, for pancreatobiliary cancers, an N1 resection)?

 

These questions form the underpinning for the process of investigation and management. In what follows, we describe our approach to each of the cancers in these terms.

Pancreatic Cancer


ductal adenocarcinoma


Ductal Adenocarcinoma of the Head of the Pancreas

Adenocarcinoma of the pancreatic head is one of the most common gastrointestinal malignancies and, because of its aggressive nature, one of the hardest cancers to cure. The safety of surgical procedures for cancers of the pancreas has improved dramatically, but 5-year actual survival rates of patients who have undergone resection are still low (about 15%).1 Cancer of the head of the pancreas is the prototypical tumor that causes painless jaundice; however, other cancers that obstruct the bile ducts also cause jaundice, including extrahepatic bile duct cancer, ampullary malignancy, duodenal cancer, and gallbladder cancer. Some of the following discussion is generalized to determining the diagnosis in patients presenting with obstructive jaundice [see 5:3 Jaundice].

Clinical evaluation

HistoryThe classic presentation of cancer of the head of the pancreas is unremitting jaundice, usually accompanied by dark urine, light stool, and pruritus. Darkening of the urine or pruritus is often the first symptom, and scleral icterus frequently is first noted by family members or coworkers. The pruritus is often severe. The jaundice sometimes is painless but more frequently is associated with epigastric pain, which is often mild. Severe acute pain is more often associated with other conditions that may cause jaundice (e.g., choledocholithiasis and pancreatitis). Back pain suggests that the tumor has invaded tissues outside the pancreas and is unresectable. Significant weight loss (> 10% of body weight) is common even when the pancreatic cancer is resectable.

In some patients, the presenting symptom is steatorrhea or diarrhea from obstruction of the pancreatic duct, weight loss, pain, or a combination of these rather than jaundice. Steatorrhea or diarrhea in the absence of jaundice is usually the result of a tumor in the uncinate process that obstructs the pancreatic duct but not the bile duct. Often these symptoms are overlooked until the tumor extends and causes jaundice. About 5% of patients have a history of diabetes of recent onset. Migratory thrombophlebitis (Trousseau sign) is uncommon and usually signifies metastatic disease. Pancreatobiliary malignancies cause biliary obstruction, but such obstruction is not commonly associated with biliary tract infection unless instruments have been employed in the biliary tree. Therefore, other diagnoses should be suspected in patients presenting with cholangitis who have not undergone biliary tract instrumentation. Patients with pancreatic cancer may also present with acute pancreatitis as the first manifestation. Vomiting and gastrointestinal (GI) bleeding are uncommon presenting symptoms and suggest the presence of advanced tumors that are obstructing or eroding the duodenum.

 

Physical examination Examination reveals scleral icterus. In some cases, the distended gallbladder may be palpable. In advanced cases, signs of metastatic disease (e.g., hepatomegaly and ascites) may be detected.

Investigative studies

Laboratory tests Liver function tests (LFTs) are of limited value in diagnosis. The serum bilirubin level is elevated in jaundiced patients, with the direct fraction exceeding 50%. The serum alkaline phosphatase level is almost always elevated when the bile duct is obstructed, and levels three to five times normal are common. Aminotransferase levels usually are moderately elevated as well. Very high aminotransferase levels suggest a hepatocellular cause of jaundice, usually viral, although impaction of a stone in the bile duct can cause transient rises in serum aspartate aminotransferase to levels higher than 1,000 IU/mL. By themselves, LFTs cannot effectively distinguish among jaundice arising from a hepatocellular cause (e.g., viral hepatitis or drug-induced cholestasis), jaundice resulting from a disease of microscopic bile ducts (e.g., primary biliary cirrhosis), or jaundice caused by any of the malignancies that obstruct the major bile ducts. To make this distinction, radiologic imaging tests are required [see Imaging, below].

Serum concentrations of the tumor marker CA 19-9 are often elevated in patients with pancreatic or biliary adenocarcinomas.2 The upper limit of the normal range is 37 U/mL. Concentrations higher than 100 U/mL are highly suggestive of malignancy, but elevations between 37 and 100 U/mL are less specific. Serum levels generally reflect the extent of the tumor: small tumors (1 cm in diameter) are rarely associated with levels higher than 100 U/mL, whereas very high concentrations (> 1,000 U/mL) suggest metastatic disease. High levels may also accompany cholangitis, but these levels should subside to normal with relief of obstruction or infection, when malignancy is absent. Measurement of CA 19-9 concentrations may be employed to detect recurrences. In patients who have elevated CA 19-9 levels that return to normal after tumor resection; a second rise in the CA 19-9 level in the follow-up period is indicative of recurrence.

 

ImagingSeveral different diagnostic imaging tests may be used in jaundiced patients, including computed tomography (CT), magnetic resonance imaging (MRI), endoscopic retrograde cholangiopancreatography (ERCP), endoscopic ultrasonography (EUS), and transabdominal ultrasonography. The technical advances in imaging achieved over the past few years are remarkable. CT and MRI can now provide high-quality images of blood vessels and ducts and their anatomic relation to tumors. These images can even be projected in three dimensions if desired, although, to date, no high level studies have shown an advantage for this type of display.

Selection of appropriate imaging tests in a jaundiced patient is influenced by patient characteristics and by the symptoms observed. For instance, the type and order of investigations appropriate for an older patient presenting with obstructive jaundice, who is likely to have a malignancy, differ from those appropriate for a young woman with severe pain, who is more likely to have choledocholithiasis. The best initial imaging test in a patient in whom malignancy is suspected is either a fine-cut (3 mm between slices) three-phase (no-contrast phase, arterial phase, and venous phase) helical (spiral) CT scan or a high-quality MRI scan. Although MRI has the advantage of being able to provide a cholangiogram (i.e., magnetic resonance cholangiopancreatography [MRCP]), small and medium-sized radiologic facilities currently tend to be more skilled at CT than at MRI; this difference should be taken into account when the first test is ordered. High-quality MRI scanners and the very latest generation of CT scanners are capable of providing cholangiograms and angiograms, as well as axial images.

Figure 1
Figure 2

The typical pancreatic cancer appears as an area of reduced attenuation (darker zone) in the pancreatic head [seeFigure 1], associated with upstream dilatation of bile ducts and the gallbladder. Often the pancreatic duct is also obstructed. As a result, the pancreatic duct may be dilated in the tail, body, and neck of the pancreas, with dilatation terminating sharply at the edge of the tumor. Pancreatic duct dilatation is often accompanied by atrophy of the body and the tail of the pancreas [seeFigure 2].

When a jaundiced patient is discovered to have a typical-appearing localized cancer of the pancreatic head on CT scanning, no further diagnostic tests are needed, and operative management should be the next step. Tissue diagnosis is unnecessary. Negative biopsy results rarely change the therapeutic approach, and in that they may be falsely negative, they are potentially misleading. Furthermore, omitting biopsy eliminates the small risk of tumor implantation in the needle tract. Selection of axial imaging as the first test often renders diagnostic ERCP, which is a more invasive test, unnecessary as well. Cholangiography also is not required for staging pancreatic head tumors [see Surgical Staging, below]. The advantages of starting with axial imaging in jaundiced patients with suspected cancer are discussed in greater detail elsewhere [see Biliary Tract Cancer, Extrahepatic Cholangiocarcinoma, Upper Duct Cholangiocarcinoma, Investigative Studies, below].

 

Additional diagnostic imaging for atypical ct or mri findings In many patients with adenocarcinoma of the pancreatic head, the typical CT findings described above are absent and additional diagnostic imaging is required. Such patients may be categorized into two groups: those with an atypical mass and those with no masson axial imaging. In either case, before ordering additional tests, it is appropriate to determine whether the CT scan is of adequate quality. The scan may have been performed without contrast, the arterial and venous phases may not have been captured appropriately, or the slice thickness may have been too great for precise visualization of the head of the pancreas. Small adenocarcinomas may be missed when the venous phase is poorly timed, especially if slice thickness is 5 mm or greater, and masses that initially appear atypical may exhibit a typical appearance when the CT scan is optimized. Neuroendocrine cancers commonly display arterial-phase enhancement, which will be missed if the scan is mistimed. In our experience, about 40% of referred patients who underwent CT scanning before arrival require a so-called pancreas protocol CT scan (i.e., a fine-cut three-phase helical scan) when they are first seen; in many of these cases, the second CT scan yields important diagnostic findings.

When no mass is present in a jaundiced patient with a periampullary tumor or another focal obstructing process (e.g., pancreatitis), the CT scan usually shows bile duct dilatation extending down to the intrapancreatic portion of the duct. The dilatation may terminate anywhere from the upper border of the pancreas to the duodenum, depending on the site of the tumor and the nature of the process obstructing the bile duct. In these conditions, ERCP is a good choice as the second test.

ERCP provides an endoscopic view of the duodenum that allows identification and biopsy of ampullary and duodenal tumors, which may be blocking the bile duct and producing jaundice. It confirms the presence of a bile duct stricture and displays its form, which is helpful in diagnosis. Focal strictures, especially those with shoulders, suggest malignancy. Long, tapering strictures limited to the intrapancreatic portion of the bile duct suggest chronic pancreatitis. Concomitant narrowing of the pancreatic duct in the head of the pancreas (the double-duct sign) suggests the presence of a small pancreatic cancer that is not visible on the CT scan. Longer or multiple pancreatic strictures suggest chronic pancreatitis. A single focal bile duct stricture in the absence of pancreatic duct abnormalities is the hallmark of cancer of the lower bile duct. Infiltrating cancers of the bile duct may cause more than one stricture along the bile duct, but when more than one stricture is present, other diagnoses (e.g., primary sclerosing cholangitis) should be considered. Both pancreatic and bile ducts may be assessed with brush cytology. This test has a 45 to 50% sensitivity for cancer3; therefore, only a positive test result is significant.

ERCP findings in a patient with no mass must be evaluated in the light of findings from other investigations. Patients with the classic double-duct sign or single focal shouldered bile duct strictures are likely to have small pancreatic or bile duct tumors. Further diagnostic support is usually not needed before laparotomy, although such support may be reassuring when the CA 19-9 concentration is lower than 100 U/mL. When doubt persists, EUS often helps resolve it. EUS may identify a small mass that was not seen on the CT scan, and biopsies may then be done. Occasionally, EUS reveals enlarged lymph nodes, which may also undergo biopsy. However, negative EUS-guided biopsy results in patients who present with painless jaundice do not exclude malignancy. When such patients have an identifiable mass on EUS and a presentation in keeping with the diagnosis of cancer, pancreaticoduodenectomy is recommended, even if EUS-guided biopsy yields negative results. If a nonoperative approach is taken, short-term follow-up at 4 to 6 weeks with repeat imaging and biopsy is mandatory. If the findings persist, laparotomy is advisable.

Occasionally, preoperative testing reveals no mass, but a mass is subsequently discovered by intraoperative palpation or intraoperative ultrasonography (IOUS). A mass palpated in the head of a pancreas that is otherwise normal or near normal in texture in the remainder of the gland is highly suggestive of malignancy and constitutes sufficient justification for resection. The same is true of a mass detected by IOUS if the mass has characteristics of malignancy (i.e., is hypoechoic). If the IOUS findings are inconclusive, biopsy with frozen-section examination is a reasonable approach. In many such cases, the whole pancreas is diffusely firm or hard, and IOUS demonstrates a diffuse change in the normal texture of the gland. When the pancreas is diffusely firm and no localized process is seen on IOUS, biopsies should be directed toward the stent in the bile duct at the point where the bile duct narrows (as seen on ultrasonography).

The ultimate diagnostic test is pancreaticoduodenectomy. If there is a strong suspicion of cancer before laparotomy or the findings at laparotomy are strongly suggestive, this procedure should be performed without preliminary biopsy. When this approach is followed, a small number of patients with suspected malignant disease will be found to have benign disease; this possibility should be explained to patients who do not undergo confirmatory tissue diagnosis before operation. Because of the limited negative predictive value of currently available tests, pancreaticoduodenectomy is sometimes still required to make a definitive diagnosis.

The finding of an atypical pancreatic head mass on a CT scan poses an additional challenge. Atypical masses may take different forms. In some cases, they exhibit attenuation that differs only slightly from that of the surrounding pancreas; in others, they have a ground-glass appearance. They may extend into the body and tail of the pancreas, or they may be localized to the head. With atypical masses, the most common problem is how to differentiate focal pancreatitis from adenocarcinoma. This differentiation can be very difficult. Pancreatitis may be present without antecedent acute attacks; without a history of alcoholism, gallstones, or hyperlipidemia; without diabetes or steatorrhea; and without calcifications in the gland. Cancer appears to be more common in patients who have had chronic pancreatitis, and the diseases may coexist. Therefore, one cannot feel confident that cancer is absent simply because chronic pancreatitis is present. Cancer should be suspected in patients with an established diagnosis of chronic pancreatitis who undergo a rapid change in status (e.g., weight loss). Diabetes is common in patients with chronic pancreatitis, but it may also be the first sign of pancreatic cancer in patients without chronic pancreatitis. Chronic pancreatitis can cause painless jaundice. A rare immune form of chronic pancreatitis, known as lymphoplasmacytic sclerosing pancreatitis, has been recognized that is particularly hard to differentiate from cancer.4 However, in these patients, the serum IgG4 level is characteristically elevated.

EUS is becoming increasingly important in the management of patients with atypical pancreatic head masses.5 When jaundice is present, EUS with or without ERCP is our usual approach; when it is absent, EUS alone is performed. EUS-guided biopsy is superior to CT-guided transabdominal biopsy in that access to the head of the pancreas is easier and the chance of needle tracking is reduced (because the biopsy is taken through the duodenal wall, which is resected if a Whipple procedure is done).

At the conclusion of all of the preceding investigations, it still may not be clear whether a malignancy is present. Clinical judgment must be exercised in deciding whether to operate or to repeat investigative studies after an interval of 2 to 3 months. Operation is favored in patients who are jaundiced, who have less pain, who have elevated CA 19-9 levels, and whose mass is suspicious for cancer. Elevation of the CA 19-9 concentration beyond 100 U/mL should be regarded as a very important finding. When EUS is inconclusive, ultrasound-guided diagnostic laparoscopy may be performed to obtain core tissue biopsies from several areas of the mass. This technique is especially useful when chronic pancreatitis is strongly suspected6 in that the multiple long core biopsies obtainable with this procedure provide a greater degree of assurance against false negative findings for cancer. Even this test, however, is not 100% accurate. The penultimate diagnostic test is laparotomy with mobilization of the pancreatic head and IOUS-guided transduodenal core biopsies of the mass. The ideal outcome with this approach is to perform pancreaticoduodenectomy in all patients who actually have cancer while reducing to a reasonable minimum resection in patients with benign disease, who in most cases are better served by biliary bypass. Fortunately, today, because of improved axial imaging and EUS, a definitive preoperative diagnosis is usually available.

 

Surgical staging

The term ‘staging’ is currently used to denote the process by which the surgeon determines whether a tumor is resectable. We prefer to use the term ‘surgical staging’ for this process so as to distinguish it from those staging classifications that define the life history and prognosis of tumors and provide the basis for comparison of results—namely, the TNM classifications developed by the American Joint Committee on Cancer (AJCC). These latter systems are also of great importance to the surgeon dealing with pancreatic tumors.

Surgical staging is started preoperatively and completed intraoperatively. Preoperative staging tests determine operability—that is, whether the tumor appears resectable after preoperative testing. However, the final decision regarding resectability is made only during the operation, on the basis of intraoperative staging. A tumor of the head of the pancreas is deemed unresectable when it is determined to have extended beyond the boundaries of a pancreaticoduodenectomy. Common reasons for unresectability include (1) vascular invasion (i.e., invasion of the superior mesenteric vein, the portal vein, the superior mesenteric artery, or, less commonly, the hepatic artery); (2) lymph node metastases that fall outside the scope of a pancreaticoduodenectomy (e.g., metastases to para-aortic and celiac lymph nodes); (3) hepatic metastases; (4) peritoneal metastases; and (5) extra-abdominal metastases (usually pulmonary). Limited vascular invasion of the superior mesenteric vein and the portal vein may be overcome by resection and reconstruction and thus is only a relative contraindication to resection. This is especially true when the tumor is small and has arisen in the vicinity of the veins. In a series from our institution (Washington University in St. Louis), about 27% of resections done for pancreatic cancer involved resection of these veins.7 Recently, the term ‘borderline’ has been introduced to describe tumors that are unresectable by standard criteria but that can be encompassed by extending the zone of resection.8 Usually, such tumors are first treated with chemoradiation. Attempts are also being made to downsize more advanced pancreatic tumors to a resectable state by chemotherapy and radiation. To date, no long-term studies have evaluated either approach. However, given the aggressive nature of this tumor, it is likely that chemotherapy and radiation will effectively downsize tumors in only a small number of patients, at least until much more effective chemotherapeutic agents are available.

The tests used to establish the diagnosis and those used to accomplish surgical staging go hand in hand. Abdominal CT scans, abdominal MRI, thoracic CT scans, and chest radiographs are obtained to detect hepatic metastases, vascular invasion, and pulmonary metastases. To assess vascular invasion, fine-cut three-phase helical CT scans or MRI scans are required. These tests may also detect enlarged lymph nodes, but it should be remembered that nodes may be enlarged for reasons other than cancer. Sometimes intraperitoneal fluid collections or peritoneal or omental nodules are identified; fluid may be sent for cytologic analysis, and omental nodules may undergo ultrasound-guided biopsy. Invasion of the mesentery, the mesocolon, or retroperitoneal tissues may also be detected by CT scanning. In the view of some surgeons, such invasion may render the tumor unresectable, but in our experience, this is rarely the case in the absence of concomitant vascular invasion: the resection may still be accomplished with clear margins by resecting the portion of the mesocolon or the mesentery that is locally invaded.

EUS may be used to guide biopsy of suspicious lymph nodes when these lie outside the planned resection zone. It has also been employed to assess vascular invasion, but in our experience, it has no advantage over CT in this regard; moreover, it is more operator dependent than CT. Staging laparoscopy is particularly effective at finding small hepatic and peritoneal nodules. About 20% of patients thought to have resectable pancreatic adenocarcinoma of the head of the pancreas before staging laparoscopy are found to have liver or peritoneal metastases at laparoscopy.9 Staging is completed intraoperatively by carefully inspecting the intra-abdominal contents, including the lesser sac, mobilizing the head of the pancreas, performing biopsies of suspicious nodules or nodes outside the planned resection zone, and attempting dissection of the superior mesenteric vein or the portal vein. Formal clinicopathologic staging according to the AJCC’s TNM system is useful for establishing the prognosis and planning additional treatment [seeTable 1 and Table 2].

All authorities agree that axial imaging of the abdomen and chest (or roentgenography of the chest) is standard practice for staging pancreatic cancer; however, not all agree on the value of other staging tests. Many authorities advocate omission of staging laparoscopy or EUS-guided biopsy of nodes on the grounds that patients are better served by palliative surgery than by endoscopic stenting of the bile duct. There is no advantage in knowing preoperatively whether small liver metastases or celiac node metastases are present if laparotomy is to be undertaken anyway. Trials examining whether better palliation is offered by surgical bypass or endoscopic stenting report conflicting results; on balance, surgical bypass is still a very good treatment for localized unresectable tumors in younger patients without serious comorbidities. We no longer perform staging laparoscopy in patients with adenocarcinoma of the pancreas except in patients who are suspected to have liver or peritoneal metastases on axial imaging. Finally, 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) may be useful in staging pancreatic cancer. Given that inflammation is frequently confused with cancer, the major role of this modality will probably be in the detection of distant metastases.

Management

Preoperative preparation All jaundiced pati-ents should receive vitamin K, a fat-soluble vitamin whose absorption is reduced by biliary or pancreatic duct obstruction. Routine preoperative bile duct decompression is unnecessary, except when jaundice has been prolonged or operative treatment will be delayed (e.g., for correction of cardiac or other comorbid conditions). Several studies have shown that surgical outcome is not improved by routine preoperative decompression in jaundiced patients. In fact, stent placement may increase the incidence of postoperative infection.10

Rationale for pancreaticoduodenectomy The technical details of pancreaticoduodenectomy are discussed more fully elsewhere [see 5:24 Procedures for Benign and Malignant Pancreatic Disease]. Therapeutic decision making necessarily includes consideration of the extent of the procedure. The operative goal is to remove the tumor with clear margins, as well as the N1 regional lymph nodes. Attempts have been made to improve results by extending the operation, either through more extensive lymph node dissections11 or through resections of superior mesenteric, hepatic, or celiac arteries.12Four randomized trials have now shown no advantage for extended lymph node dissection. Resection of celiac or superior mesenteric arteries has also not been successful in improving overall survival. The lesson seems to be that invasion of additional lymph node regions (N2) or the superior mesenteric or celiac arteries signals an aggressive tumor biology that is unlikely to be overcome by wider resections. Except for resection of the portal vein or the superior mesenteric vein and perhaps short segments of the hepatic artery to address invasion of these structures by otherwise favorable tumors, extended resections are generally unsuccessful. Even these recommended vascular resections are probably best restricted to tumors that have arisen close to the particular vessels and involved them while still small; resections of large adenocarcinomas that have grown over time to involve long stretches of the veins are best avoided. These comments do not apply to tumors that initially might have required resection of the celiac or superior mesenteric arteries but that have been downsized by chemotherapy of chemoradiation so that they can be resected without such vascular resections.

There is also continuing controversy regarding the respective merits of the standard version of the operation and its pylorus-preserving variant. There is no evidence that the two procedures differ with respect to overall survival. Pylorus preservation is associated with gastric-emptying problems in the postoperative period, but, overall, it may be associated with slightly less postoperative GI dysfunction.13 We employ pylorus preservation selectively in older, thinner patients, with the aim of minimizing disruption of GI function.

Adjuvant therapy is often given in resected patients; however, there is controversy regarding the role of chemotherapy versus chemoradiation. Randomized trials have shown that chemotherapy is beneficial and have questioned the role of radiotherapy. The latter may be helpful when an R1 resection has been performed.

 

Adenocarcinoma of Body and Tail of the Pancreas

Adenocarcinoma of the body and tail of the pancreas is less common than adenocarcinoma of the head. Because it does not produce jaundice, it tends to be recognized relatively late. Accordingly, patients are often in an advanced stage of disease at presentation. Tumors of the midbody tend to invade posteriorly to involve the superior mesenteric artery or the celiac axis, even when these lesions are only 2 to 3 cm in diameter. As a result, tumors of the tail are more likely to be resectable than tumors of the midbody when they are discovered. Many resectable tumors are discovered incidentally; by the time the tumors give rise to symptoms, they are frequently unresectable.

Clinical evaluation

Symptoms are nonspecific, consisting of abdominal and back pain (which is usually relieved by sitting up and leaning forward), weight loss, and diabetes of recent onset.

Investigative studies

Figure 3

The CA 19-9 concentration may be elevated. CT usually shows a lucent (hypoattenuating) mass [seeFigure 3], often with extension outside the pancreas and dilatation of the distal pancreatic duct, when the tumor is proximal to the tail of the gland. EUS is very useful for assessing indeterminate lesions.

Surgical staging

Surgical staging of cancers of the body and the tail is similar to that of cancers of the pancreatic head and is based primarily on CT scanning of the abdomen and the thorax. Unresectability by reason of local invasion is usually attributable to the involvement of the superior mesenteric artery, hepatic artery, or the celiac artery and less commonly to the involvement of the portal vein, the superior mesenteric vein, or the aorta. Another indicator of unresectability is enlarged para-aortic nodes. Invasion of the spleen, the stomach, the left adrenal gland, the mesocolon, the colon, the retroperitoneum, or even the left kidney is not a contraindication to resection provided that clear margins may be expected.14 Staging laparoscopy is of great value: between 20 and 50% of patients with these tumors are found to have unresectable tumors with this modality, usually due to small liver or peritoneal implants.15 Cancer of the body and the tail differs from cancer of the head in that there is no effective palliation and therefore no rationale for laparotomy if the lesion is unresectable. Celiac nerve block, which is very helpful in reducing the use of narcotics for pain control, may also be performed laparoscopically or endoscopically.

Management

Rationale for radical antegrade modular pancreatosplenectomy

Figure 4

Logically, the goal of resection of tumors of the body and tail should be the same as that of resection of tumors of the head—namely, excision of the tumor with clear margins, along with the N1 lymph nodes. In practice, this goal generally is not achieved by the traditional retrograde distal pancreatectomy, in which the spleen is taken first and which is not based on the lymph node drainage of the pancreas. Lymph node counts have been low with the traditional procedure, and positive posterior margin rates have been high. As an alternative, we have developed a technique referred to as radical antegrade modular pancreatosplenectomy (RAMPS), which accomplishes the desired goals by performing the resection in an antegrade manner from right to left and which is based on the established lymph node drainage of the gland [seeFigure 4].16 Negative tangential margin rates of 90% have been achieved with this approach. RAMPS also allows early control of the vasculature.

Mucinous Adenocarcinoma

Mucin-producing cancers are special variants of adenocarcinoma of the pancreas that often arise in preexisting lesions. The two main types of premalignant lesions are mucinous cystic neoplasm (MCN) and intraductal papillary mucinous neoplasm (IPMN) (also referred to as intraductal papillary mucinous tumor).17 A complete discussion of pancreatic cyst disease is beyond the scope of this chapter. Accordingly, we briefly address such disease as it relates to cancer of the pancreas, omitting discussion of less common cystic malignancies of the pancreas.

Mucinous cystic neoplasm

MCN occurs most often in middle-aged women, typically in the body or tail of the pancreas. MCNs are unilocular or septated cysts whose diameter ranges from subcentimeter size to 15 cm or larger. Occasionally, calcium is present in the wall. Excrescences may be present on the inner wall; if so, malignancy is more likely. Most symptomatic MCNs are between 4 and 7 cm in diameter.

Clinical evaluation and investigative studiesPatients with MCNs typically present with left-sided abdominal pain, often in the flank and the back. These lesions also are frequently discovered incidentally. Pancreatitis is rare and jaundice is uncommon, even when the lesions are situated in the head of the pancreas. MCNs must be differentiated from pseudocysts and from serous cystadenomas (SCAs), which are benign cysts. Differentiation between MCNs and pseudocysts is based on the history, imaging studies, and cyst fluid analysis. The diagnosis of pseudocyst is supported by a history of pancreatitis, a thick-walled, uncalcified cyst with associated radiologic signs of pancreatitis and often cholelithiasis, as well as cyst fluid containing high levels of amylase and lipase and a relatively low level of carcinoembryonic antigen (CEA) (< 500 ng/mL).

SCAs have the same clinical presentation as MCNs. SCAs are more frequently polycystic than MCNs, but this difference is not a certain means of discriminating between the two. In a minority (25%) of cases, SCAs have a pathognomonic central calcification with radiating arms ringed by multiple grape-sized cysts. When the individual cysts are tiny (honeycomb pattern), there are many cyst walls within the lesion, and then SCAs may be mistaken for solid tumors. Unlike pseudocysts and IPMNs, neither MCNs nor SCAs communicate with the pancreatic duct, although they may compress it. Measurement of the CEA level in cyst fluid is a good means of distinguishing MCN from SCA. SCAs have very low levels of CEA, with the cutoff being 5 ng/mL.18 The cyst fluid obtained from MCNs is often mucinous, and cytologic assessment may show mucin-producing cells; typically, the fluid is high in CEA. The CA 19-9 concentration may also be used to distinguish MCNs from SCAs, but it is not as reliable as the CEA concentration for this purpose.18

 

Surgical staging Surgical staging is required when investigations suggest that MCNs are malignant. Essentially, the same methods are used as for any pancreatic adenocarcinoma (see above). Malignancy is suggested by a solid intracystic or extramural component. Sometimes a mucinous tumor is frankly malignant with a large or dominant solid component. Such a tumor is better termed a mucinous cystadenocarcinoma, and it should be evaluated and treated from the outset in the same manner as other adenocarcinomas of the pancreas.

Management In symptomatic patients, preoperative differentiation between MCNs and SCAs is unnecessary because resection is the treatment for both. In asymptomatic patients, MCNs more than 4 cm in diameter should be excised because of the possibility of malignant degeneration.19 MCNs associated with internal excrescenses or associated with a local solid mass should be removed independent of the size of the cyst.19 The standard procedure has been open distal pancreatectomy with splenectomy, although lesser procedures, such as spleen-sparing distal pancreatectomy, central pancreatectomy, and enucleation, have all been used as well. Recently, laparoscopic distal pancreatectomy with or without sparing of the spleen has emerged as the procedure of choice because of the advantages of reduced postoperative pain and shortened length of stay.20These procedures appear to be reasonable choices provided that there is no suggestion of invasive malignancy on imaging (i.e., that there are no excrescences on the inner lining and that the surrounding pancreas appears normal). In these cases, a more formal resection that obeys oncologic goals such as a RAMPS procedure should be employed. Enucleation may be associated with a higher incidence of postoperative fistula. If invasive cancer is not detected in the resected specimen, the chances that the malignancy will recur are small; in fact, we have never seen such a recurrence.

The 4 cm cutoff for surgical treatment of MCNs in asymptomatic patients19 without other signs associated with malignant degeneration is reasonable, but recommendations could change based on additional studies of natural history. Furthermore, it is still possible that malignant degeneration could occur in smaller cysts, although the probability is low. Many cysts smaller than 2 cm are found in the course of axial imaging performed for other reasons. Such cysts are difficult to diagnose because of the small volume of cyst fluid present, and the benefit to be gained from performing a large number of pancreatectomies for these small cysts is questionable, even when they are diagnosable as MCNs. Occasionally, MCNs or symptomatic SCAs are located in the head of the pancreas and require pancreaticoduodenectomy. Some authorities feel that very large asymptomatic SCAs should also be excised because of rare instances of malignant degeneration.

 

Intraductal papillary mucinous neoplasm

Figure 5

IPMN begins as a metaplastic change in the cells lining the pancreatic ducts altering from a low cuboidal serous type of cell to a mucin-producing type. These cells are prone to progresss to dysplasia and eventual malignant transformation. Overall, IPMNs appear to undergo malignant transformation more frequently than MCNs. There are two recognized types of IPMN, which may occur either separately or together.19,21 The more common type affects the main pancreatic duct [seeFigure 5] and is called ‘main duct IPMN.’ In this type, the main pancreatic duct becomes dilated and filled with mucin. As the disease progresses toward malignancy, papillary processes may project into the lumen. The less common type, called ‘side-branch IPMN,’ affects the smaller ducts and presents as multiple (usually small) pancreatic cysts. In either type of IPMN, the disease may be either diffuse or focal; when it is focal, the head of the pancreas is the site of disease in the majority (60%) of cases. About 20% of IPMN patients have a malignancy at the time of diagnosis, although the cancer may not be evident until the specimen is examined pathologically.

Clinical evaluation and investigative studies19,21IPMN occurs predominantly in males and usually affects patients in their sixties. Pain (usually attributable to pancreatitis arising as a result of mucous obstruction of the pancreatic duct) is a common presenting symptom. Another common presentation is pancreatic insufficiency with diabetes or steatorrhea. Accordingly, it is not surprising that, formerly, many IPMN patients were diagnosed as having chronic pancreatitis. IPMN may also be discovered incidentally or may present as a cancer with signs and symptoms similar to those of other pancreatic cancers, depending on the part of the gland in which they arise. On rare occasions, cholangitis from obstruction of the common channel by mucus is the presenting problem.

The diagnosis is made on the basis of the presentation and the findings from axial imaging and ERCP. The characteristic ERCP findings are a dilated papillary orifice (‘fish mouth’) with mucus bulging from the orifice and a dilated pancreatic duct when contrast is injected. Sometimes, mucus prevents complete filling of the duct with dye. In this situation, CT scans or MRI with MRCP may be quite useful for detecting ductal dilatation and atrophy of the pancreas. MRCP is best at detecting excrescences projecting from the wall of the duct into the lumen. These signal progression of the disease toward neoplasia. In side-branch IPMN, ERCP (and often MRCP) typically demonstrates communication between the cysts and the main duct, which is often normal in size; this finding is not present in MCN or SCA and is very useful for distinguishing side-branch IPMN from these other types of cysts.

 

Management Knowledge of this entity is evolving, and recommendations are the current ones.19,21–23

Large duct IPMN

As this entity is highly premalignant, resection is indicated once the diagnosis is made. Pancreatoduodenectomy is usually required as the disease affects the duct in the head of the pancreas in most cases. Although the duct in the body and tail of the gland may be dilated, this does not mean that it is involved in dysplastic changes as dilation may be secondary to obstruction caused by the viscous mucus in the proximal duct.21 Therefore, the decision of whether to perform a complete pancreatectomy is made at the time of pancreatoduodenectomy, based on frozen section of the transected pancreatic neck. In most cases, the frozen section will show normal ductal epithelium or only mild or moderate dysplasia. Such patients do not require more pancreas to be resected. Occasionally, carcinoma, carcinoma in situ, or severe dysplasia is found, necessitating the resection of part or all of the remaining pancreas.21

Small duct IPMN

This entity is less likely to degenerate into malignancy, and the probability of malignant degeneration is related to size.22 Current indications for resection are cysts exceeding 3 cm in diameter or smaller cysts that are associated with symptoms, internal excrescences, pericyst solid tumor, or pancreatic duct obstruction manifested as a dilation of the duct distal to the cyst.

With either type of IPMN, lifelong follow-up with axial imaging is needed.21 This problem should be thought of as a field defect in the pancreas. Large duct and small duct IPMNs may coexist, and pancreatic intraepithelial neoplasia (PanIN) lesions (dyslastic lesions of the duct) may coexist with IPMN.

Most patients who require total pancreatectomy tolerate the procedure well when they are enrolled in a program keyed to this operation. Frank mucinous cancers may appear in patients with IPMN as well; they should be managed in much the same fashion as other adenocarcinomas, with the additional requirement that the resection should encompass the entire IPMN-bearing portion of the pancreas. The mucinous cancers associated with IPMN have a better prognosis than ductal adenocarcinomas do.21,23

neuroendocrine cancers


Neuroendocrine cancers account for fewer than 5% of surgically treated pancreatic malignancies. Some of these cancers are functional tumors, which produce hormones that lead to paraneoplastic syndromes. Examples include insulinoma, gastrinoma, glucagonoma, and vasoactive intestinal polypeptide–secreting tumor (VIPoma), all of which are associated with characteristic clinical syndromes. These syndromes are often produced while the tumors are still small. A detailed discussion of functional neuroendocrine tumors is beyond the scope of this chapter.

Other neuroendocrine tumors are nonfunctional and, as a result, reach a larger size before giving rise to symptoms. These lesions present with symptoms caused by mass effect and must be differentiated from ductal adenocarcinomas. Nonfunctional neuroendocrine cancers are relatively slow-growing tumors that tend to push rather than invade structures but are capable of metastasizing to lymph nodes, as well as to the liver and other organs. Pain is the most common presenting symptom. Jaundice, pancreatitis, and systemic symptoms (e.g., weight loss) are less common with these tumors than with adenocarcinoma of the pancreas. Because of the propensity of neuroendocrine tumors to deflect rather than invade the bile duct, jaundice may be absent even when tumors are located in the head of the gland.

Diagnosis, surgical staging, and treatment rationale are essentially the same for neuroendocrine cancers as for ductal adenocarcinomas. On CT scans, these lesions characteristically show enhancement in the arterial phase and are seen to push on bile ducts and vascular structures rather than encase them. Complete resection by means of pancreatoduodenectomy or distal pancreatectomy [see 5:24 Procedures for Benign and Malignant Pancreatic Disease] is indicated. Given the slow growth rate of neuroendocrine cancers and their relatively favorable prognosis (a 50 to 60% 5-year survival rate), removal of the primary lesion and any hepatic secondary lesions is justified if all tumor tissue can be removed with clear margins.

Biliary Tract Cancer


Cancers of the biliary tract (cholangiocarcinomas) may arise at any level of the biliary tree from the smallest, most peripheral intrahepatic bile duct to the termination of the common bile duct. The lesions may be subdivided into intrahepatic and extrahepatic cholangiocarcinomas. The former are described below under ‘Liver Tumors.’ Cholangiocarcinomas may grow in one of three phenotypic forms, a mass, a stricture, or an intraluminal polyp or excrescence, although combinations of these forms in different parts of the tumor may be present. The formal designations are ‘mass forming’ (MF), periductal infiltrating (PI), and intraductal growth (IG). The type of growth pattern may affect the clinical presentation.

extrahepatic cholangiocarcinoma


Extrahepatic cholangiocarcinoma (CCA) may be subdivided into lower duct CCA and upper duct CCA, with the former arising in the intrapancreatic or retroduodenal portion of the bile duct and the latter arising above it. In practice, most upper duct CCAs (also referred to as hilar CCAs or Klatskin tumors) arise just below the union of the right and left hepatic ducts, at the union of the ducts, or in the main right or left hepatic ducts. Cancer of the midportion of the bile duct at the usual insertion point of the cystic duct is more likely to be an extension of a gallbladder cancer than a primary CCA. AJCC staging criteria for these tumors are useful for establishing the prognosis and planning further treatment [seeTable 3andTable 4].

Lower Duct Cholangiocarcinoma

Clinical evaluation and investigative studies

Much of what the surgeon needs to know about lower duct CCA has already been addressed elsewhere [see Pancreatic Cancer, Adenocarcinoma of the Head of the Pancreas, above]. By far the most common presentation is painless jaundice with its constellation of associated symptoms (especially pruritus). Laboratory tests reveal the characteristic pattern of obstructive jaundice. A serum CA 19-9 concentration higher than 100 U/mL facilitates the diagnosis. Axial imaging reveals dilation of the intrahepatic bile ducts, the gallbladder (in most cases), and the extrahepatic bile ducts down to the level of the pancreatic head, where the dilatation terminates abruptly. Usually, no mass is visible. ERCP or MRCP shows a focal stricture, and ERCP brushings are positive in about 50% of cases. EUS may be helpful in that it is more sensitive for small tumors than CT. Needle biopsy is directed toward the mass or, if no mass is visible, toward the narrowest segment of the bile duct. A negative biopsy result does not rule out a small bile duct cancer. ‘Spyglass’ cholangioscopy, in which a small endoscope is directed into the biliary tree along a catheter placed by duodenoscopy, facilitates direct biopsy of the bile duct wall and lesions that project into the lumen.

The differential diagnosis includes other potential causes of focal strictures of the bile duct.24 The most common cause of a benign stricture of the intrapancreatic bile duct is pancreatitis, which may be diffuse or focal. Other causes of benign stricture include iatrogenic injury, choledocholithiasis, sclerosing cholangitis, and benign inflammatory pseudotumors [see Upper Duct Cholangiocarcinoma, Investigative Studies, Imaging, below]. Iatrogenic injuries rarely involve the intrapancreatic portion of the bile duct, although such injuries can occur in this area as a consequence of forceful instrumentation. Sclerosing cholangitis may affect this section of the bile duct but usually affects other areas of the biliary tree as well. The diagnostic steps for differentiating benign neoplasms from malignant tumors are essentially the same for lower duct CCA as for pancreatic cancer. As noted, resection may be required to make the diagnosis. In any patient presenting with jaundice and a focal stricture of the bile duct, lower duct CCA should be strongly suspected. Mass-forming bile duct cancers may also arise in the lower bile duct, but, for obvious reasons, they are difficult to differentiate from pancreatic adenocarcinomas. Perhaps in the future genetic profiling will permit this to be done. The surgical workup and management are identical.

Surgical staging

Surgical staging of lower duct CCAs is usually straightforward. These tumors are usually remote from major vascular structures and thus are not subject to the same local staging considerations as adenocarcinomas of the pancreatic head are. The exception is a tumor that extends to the top of the retroduodenal portion of the bile duct. At this point, the bile duct is apposed to the portal vein and the hepatic artery, and these structures may be invaded by bile duct tumors in this location.

Management

The treatment for resectable lesions is pancreaticoduodenectomy, and the rationale for the extent of the resection is the same as for adenocarcinoma of the head of the pancreas.

Upper Duct Cholangiocarcinoma

Figure 6

Upper duct (or hilar) CCA is a sporadically occurring tumor that may also be seen in patients with primary sclerosing cholangitis, ulcerative colitis, or parasitic infestation. It is characteristically slow growing and locally invasive, and it metastasizes more readily to lymph nodes than systemically, although intrahepatic and peritoneal metastases are not uncommon. Most hilar CCAs are cicatrizing diffusely infiltrating cancers (PI tumors), but some form masses (MF type), and others present as papillary ingrowths (IG type). These tumors are also subdivided by the upper level of the tumor in the biliary tree according to the Bismuth classification [seeFigure 6].25

When the CCA originates in one of the hepatic ducts, that duct may be obstructed for a considerable period before the tumor causes jaundice by growing into the other hepatic duct or the common bile duct. Such prolonged unilateral obstruction before the onset of the presenting symptom of jaundice may result in atrophy of the obstructed side of the liver, which may affect subsequent management. For example, because the disease is more advanced on the obstructed side, the atrophied half of the liver will be removed in almost all cases in which resection is indicated. In addition, when one side of the liver undergoes atrophy, the other side undergoes hypertrophy. These changes may lead to rotation of the liver, which, in turn, may cause the structures in the hepatoduodenal ligament to be rotated out of their normal anatomic location. For instance, if hypertrophy of the left hemiliver develops, the hepatic artery may come to lie directly in front of the bile duct.

Clinical evaluation

The usual presentation of hilar CCA consists of painless jaundice with its accompanying symptoms (especially pruritus), although some pain may be present. Cholangitis before instrumentation of the bile duct is uncommon. In patients who present in the late stages of the disease, general manifestations of cancer (e.g., malaise, weight loss, or ascites) may be noted. In patients with primary sclerosing cholangitis, the presence of CCA is often suggested by a rapid deterioration in the patient’s general condition. It is not unusual for patients with hilar CCA to have undergone a cholecystectomy in the recent past; the symptoms of pain and jaundice may be mistaken for symptoms of gallbladder disease in patients who happen also to have gallstones.

Investigative studies

Laboratory tests Laboratory testing follows the pattern previously described for obstructive jaundice [see Pancreatic Cancer, above]. Again, the most helpful diagnostic laboratory test is the serum CA 19-9 concentration: levels higher than 100 U/mL are strongly suggestive of cancer.

Imaging

Figure 7

Earlier [see Pancreatic Cancer, Ductal Adenocarcinoma, Adenocarcinoma of the Head of the Pancreas, above], the point was made that it is preferable to employ axial imaging rather than ERCP as the first imaging test in the jaundiced patient because doing so will often render ERCP, an invasive test, unnecessary. This point carries even more force in the setting of hilar CCA. Injection of dye above the malignant stricture is an integral part of ERCP. Once the dye has been injected, stents must be placed to prevent post-ERCP cholangitis. This process may involve insertion of bilateral stents, including a stent in the atrophic hemiliver. Bilateral stenting is disadvantageous because the aim is to encourage atrophy of the hemiliver to be resected and hypertrophy of the hemiliver to be retained, and insertion of a stent in the atrophic side negates that aim. Starting with CT or MRI rather than with ERCP allows detection of any hilar CCA present simultaneously with detection of atrophy. At this point, the patient can be evaluated by a multidisciplinary team with expertise in this disease, and a decision can be made regarding which side of the biliary tree to decompress (if either). Whether stents should be employed in treating hilar CCA is debatable, but if a stent is inserted, only the side to be retained should be intubated. MRCP now provides resolution that is close to that obtained with direct cholangiography [seeFigure 7].

ERCP does have one significant advantage in that it allows brushings to be obtained. Standard brushing techniques at this high level in the biliary tree are even less sensitive than those at lower levels, but spyglass technology (see above) has opened this area of the biliary tree to direct biopsy on a routine basis. EUS has been employed to obtain diagnostic tissue, with some degree of success; however, because the biopsy needle passes through the peritoneal cavity, concerns have been expressed regarding possible tumor seeding. Such seeding has not been an issue with lower duct cancers, because the biopsy tract is entirely within the future resection specimen. In many cases, a tissue diagnosis cannot be obtained preoperatively, and the diagnosis is based on the presence of a focal hilar stricture that causes jaundice.

Focal strictures of the upper bile ducts are strongly suggestive of cancer, but CCAs must also be differentiated from benign inflammatory tumors (also referred to as hepatic inflammatory pseudotumors and benign fibrosing disease).26 These inflammatory masses mimic upper duct CCAs but consist of chronic inflammatory cells and fibrous material. One distinguishing characteristic is that they do not involve blood vesels. Even today, they are very difficult to distinguish from cancers before pathologic examination of a resected specimen. Benign inflammatory tumors appear to occur most frequently in extrahepatic upper ducts, but they also occur intrahepatically and, less commonly, in lower ducts.

Gallbladder cancer may invade the porta hepatis and appear as a CCA, especially on ERCP. Gallstones are usually present. Axial imaging usually shows thickening of the gallbladder wall or the presence of a mass involving the infundibulum. Mirizzi syndrome is another cause of a focal stricture of the middle or upper bile duct. This syndrome results from compression of the bile duct by a large gallstone in the infundibulum and is usually associated with severe inflammation of the gallbladder and the characteristic signs and symptoms of acute cholecystitis. The duct is typically bowed to the left rather than focally narrowed, as in cancer. Iatrogenic causes should be considered if the patient has had a cholecystectomy. On occasion, a stricture appears years after the operation. In these cases, the probable cause of the stricture is ischemic injury to the bile duct. The presence of clips close to or indenting the duct is a clue that such injury is a possibility. Choledocholithiasis may also cause strictures, especially if cholangitis has occurred. Strictures are also frequent with recurrent pyogenic (oriental) cholangitis. Other rare tumors of the bile duct (e.g., neuroendocrine tumors) may mimic cholangiocarcinoma.

 

Surgical staging

Often the first axial imaging test reveals only the presence of intrahepatic bile duct dilatation, which stops abruptly as the ducts merge in the hepatic hilum. This finding, however, leads to MRI or CT aimed at providing high-quality cholangiograms and angiograms of the hepatic arteries and the portal veins. Surgical staging of hilar CCA, unlike that of lower duct CCA, requires exact knowledge of the macroscopic upper extent of the tumor in the bile duct. Furthermore, invasion of hepatic arteries and portal veins is common and frequently affects resectability. Thus, surgical staging also requires accurate determination of the extent of hepatic arterial or portal venous invasion and assessment of the degree of atrophy.

Bismuth type IV tumors are not resectable, except by liver transplantation. Type I through III tumors are resectable provided that the main portal vein and the proper hepatic artery, as well as the portal vein and the hepatic artery to the side of the liver to be retained, are not invaded by tumor and that the side to be retained is not atrophic. Involvement of the main portal vein or the hepatic artery is a relative rather than an absolute contraindication; lesser degrees of involvement can be handled by means of vascular resection and reconstruction in specialized centers. Unusual combinations of events may preclude resection (e.g., atrophy on one side of the liver and invasion of the hepatic artery supplying the other side, or invasion of the portal vein to one side and the bile duct on the other side to the level of the secondary biliary branches).

MRI (with MRCP and magnetic resonance angiography [MRA]) or CT with the latest generation of scanners can provide complete information regarding the extent of bile duct involvement and the degree of vascular invasion. Doppler ultrasonography is also excellent for evaluating vascular invasion. In our experience, the combination of these two investigations usually provides more usful staging information than either individually. Both are dependent on highly experienced radiologists. ERCP may be used for additional assessment of the extent of the tumor on the side to be retained if a stent on that side is deemed necessary. The use of percutaneous cholangiography is controversial, the main concern being the risk of tumor seeding along the tube, into the peritoneal cavity, and onto the surface of the liver or the abdominal wall. Nevertheless, this procedure is used extensively in Japan, where surgeons have considerable experience with selective decompression of parts of the liver as a preoperative strategy.27

Assessment of distant metastases is achieved by means of axial imaging of the chest and the abdomen. Staging laparoscopy identifies 10 to 15% of cancers that are unresectable because of peritoneal or liver metastases. FDG-PET identifies about 15% of patients with distant metastases. At present, neither of these tests is routinely employed in this setting. Staging laparoscopy has provided a good yield of unresectable cases in some studies but not in others.

Management

Preoperative preparation Unlike cancers of the lower bile duct, cancers of the upper bile duct usually necessitate major liver resection [see 5:22 Procedures for Benign and Malignant Biliary Tract Disease]. Consequently, it has been argued that the risk of postoperative hepatic failure may be lowered by preoperative decompression, especially decompression of the side to be retained, which has the dual purpose of allowing that side to recover function and of actually encouraging hypertrophy. On the other hand, stents may introduce bacteria and cause cholangitis. As noted (see above), selective percutaneous decompression is an accepted strategy in Japan; often multiple stents are inserted.28

A reasonable strategy is to proceed to operation if (1) the patient is relatively young (< 70 years), (2) there are no serious comorbid conditions, (3) the jaundice has been present for less than 4 weeks, (4) the serum bilirubin concentration is lower than 10 mg/dL, (5) the future remnant liver will include more than 35% of the total liver mass, and (6) the patient has not undergone biliary instrumentation (which always contaminates the obstructed biliary tract). In all other cases, we routinely decompress the side of the liver to be retained and wait until the serum bilirubin concentration falls to 3 mg/dL. When the future remnant liver will include less than 30 to 35% of the total liver mass, portal vein embolization (PVE) of the side to be resected may be performed to induce hypertrophy of the remnant. Because resection for hilar CCA is a major procedure in a somewhat compromised liver, it is contraindicated in patients who are in poor general condition or who have major organ dysfunction.

 

Rationale for surgery Patients with upper duct CCA are candidates for resection if they have no distant metastases (including intrahepatic metastases) and if the tumor can be removed in its entirety by means of bile duct resection [see 5:22 Procedures for Benign and Malignant Biliary Tract Disease] combined with liver resection [see 5:23 Hepatic Resection]. The goal of resection of upper duct CCA is to achieve clear resection margins by removing the tumor, the portal and celiac lymph nodes, the side of the liver in which the ductal involvement is greater (via hemihepatectomy or trisectionectomy), and the caudate lobe. (The caudate lobe is resected because cholangiocarcinomas tend to invade along the short caudate bile ducts, which enter the posterior surfaces of the main right and left bile ducts at the bifurcation of the common hepatic duct.) Recently, some surgeons have extended the portal dissection to include the portal vein when the right side of the liver is to be resected. This provides a wide resection zone and removes an area that is frequently involved by microscopic tumor.28

Liver transplantation has been used successfully to manage Bismuth type IV tumors and is usually performed after neoadjuvant chemoradiation therapy and staging laparotomy in highly selected patients.29 Liver transplantation for Bismuth tumors of lesser Bismuth grades is highly controversial.

 

Gallbladder Cancer


The incidence of gallbladder cancer in the United States is about 9,000 cases a year. This cancer almost always arises in patients with preexisting gallstones and is most often seen in elderly patients. Like ductal adenocarcinoma of the pancreas, it is highly malignant and tends to spread at an early stage to lymph nodes, to peritoneal surfaces, and to areas of the liver distant from the gallbladder fossa. AJCC staging criteria are helpful for planning management of this cancer [seeTable 5andTable 6].


Clinical Evaluation

Gallbladder cancer is discovered either incidentally during performance of cholecystectomy for symptomatic cholelithiasis or when the tumor causes symptoms related to invasion of the bile duct or to the effects of metastatic disease. In early stages of the disease in which the tumor is confined to the wall of the gallbladder, the symptoms are usually those of the associated stones—that is, the patient has biliary colic, and the cancer is silent. In later stages of disease, jaundice, weight loss, a palpable right upper quadrant mass, hepatomegaly, or ascites may develop. Jaundice occurs in about 50% of patients. It is a poor prognostic sign because it signifies extension of the tumor beyond the gallbladder and obstruction of the extrahepatic bile ducts. Consequently, most gallbladder cancer patients with jaundice have unresectable tumors. Because the signs and symptoms of gallbladder cancer are nonspecific, delays in diagnosis are common. As a result, most gallbladder cancers are not diagnosed until they have reached stage III or IV; thus, most of these aggressive tumors are unresectable at presentation, even when the patient is not jaundiced.

Investigative Studies

Laboratory tests

In stages I and II, LFTs usually yield normal results. In later stages, laboratory test abnormalities may be noted that are not diagnostic but are consistent with bile duct obstruction. Elevated alkaline phosphatase and bilirubin levels are common. An elevation in the serum CA 19-9 concentration is the most helpful diagnostic indicator.

Imaging

Because gallbladder cancer is most curable in its early stages and because the symptoms in those stages are those of cholelithiasis, it is important for surgeons to be aware of subtle signs of gallbladder cancer that are occasionally present on sonograms. These signs include asymmetrical thickening of the gallbladder wall, thickening of the wall in a patient without a history of biliary colic, a mass projecting into the lumen, multiple masses or a fixed mass in the gallbladder, calcification of the gallbladder wall (so-called porcelain gallbladder), and an extracholecystic mass. Displacement of a stone to one side of the gallbladder should also be viewed with suspicion.

In later stages of disease, CT scans usually show a gallbladder mass with or without invasion of the liver or other adjacent organs. Obstruction of the bile duct produces the usual features associated with obstructive jaundice. Percutaneous CT-guided biopsy is a useful technique for confirming the diagnosis in patients with unresectable tumors.

Porcelain gallbladder is a premalignant condition, although there is some evidence that the incidence of cancer depends on the pattern of calcification: selective mucosal calcification apparently carries a significant risk of cancer, whereas diffuse intramural calcification does not.30 It seems reasonable to resect only tumors with the former pattern, but whenever there is a question about the pattern of calcification, one should err on the side of resection.

Surgical Staging

Staging of gallbladder cancer requires knowledge of the extent of direct invasion into the liver and other adjacent organs and tissues (especially the bile duct, the portal veins, and the hepatic arteries). As in hilar CCA, this information may be obtained by means of MRCP and MRA or CT with the latest generation of scanners. Staging laparoscopy is very helpful in managing gallbladder cancer. As many as 50% of patients with this disease are found to have peritoneal or liver metastases on staging laparoscopy,8 and as with carcinoma of the body of the pancreas, no useful palliative measures can be undertaken at laparotomy.

Management

Rationale for surgery

When early-stage gallbladder cancer is suspected on the basis of diagnostic imaging, open cholecystectomy, rather than laparoscopic cholecystectomy, is probably the procedure of choice [see 5:21 Cholecystectomy and Common Bile Duct Explorationand 5:22 Procedures for Benign and Malignant Biliary Tract Disease]. Intraoperatively, if there is no evidence of spread outside the gallbladder, we recommend performing an extraserosal cholecystectomy, in which the fibrous liver plate is excised along with the gallbladder so that bare liver is exposed. It is possible to perform an extraserosal resection laparoscopically; however, in our opinion, this should not be attempted, because gallbladder perforation and bile spillage are more common with the laparoscopic version of the procedure. The negative consequences of tumor implantation or incomplete excision far outweigh any benefit that a minimally invasive approach might confer.

The excised specimen should be inked and a frozen section obtained. If there is gallbladder cancer in the specimen but the resection margins are clear and the tumor is a T1 lesion (i.e., has not penetrated the muscularis), the procedure is considered complete in that lymph node metastases are uncommon with T1 tumors (incidence < 10%). However, lymph node metastases are present in 50% of patients with T2 lesions (i.e., tumors that have invaded the muscularis). Therefore, if margins are positive or the tumor is a T2 lesion, resection of segments 4b and 5 of the liver and dissection of the portal and celiac lymph nodes are recommended. Resection of the extrahepatic bile duct and hepaticojejunostomy may be needed in some cases to obtain a complete node clearance, but this is being done less frequently as experience with portal node clearance has been obtained. If it is already clear at the commencement of the operation that the tumor is T2, one should proceed directly to liver, lymph node, and bile duct resection.

In more advanced stages of disease (T3 and T4), the aim is still excision with clear margins and resection of portal and celiac lymph nodes. To obtain clear local margins with these tumors, in addition to what is required for T2 tumors, more extensive hepatic resections—up to a trisectionectomy (resection of segments 4 through 8) [see Liver Cancer, Anatomic Considerations, below]31—may be necessary, as well as resection of adjacent organs.

Incidentally discovered gallbladder cancer

Gallbladder cancer may be an incidental finding at laparoscopic cholecystectomy, as it has been at open cholecystectomy. The incidence of this finding ranges from 0.3 to 1.0%. A concern that has arisen in the current era, in which the laparoscopic approach to cholecystectomy is dominant, is the risk of port-site implantation of tumor. Port-site implantation is the result of contact between the malignancy and the tissues surrounding the port site at the time of gallbladder extraction. Therefore, when evidence of gallbladder wall thickening is noted intraoperatively, the gallbladder should be extracted in a sac. The gallbladder should be inspected at the time of extraction, and any questionable areas should undergo biopsy.

If a gallbladder cancer is discovered at the time of operation, it should be treated without delay according to the principles stated earlier (i.e., depending on whether the margins on the excised gallbladder are clear and on the T stage of the tumor). From an oncologic viewpoint, it would seem ideal to resect the tissue around all trocar port sites. From a technical viewpoint, however, it would be very difficult and impractical to excise the full thickness of the abdominal wall circumferentially around four port sites, especially because the tract of the port site often is not at a 90° angle to the abdominal wall. If the gallbladder was extracted through a port site without having been placed into a bag, it is reasonable to attempt excision of that one port site.

Sometimes cancer is suspected, but frozen-section examination is inconclusive, and the definitive diagnosis of cancer is not made until the early postoperative period. More often, cancer is not suspected intraoperatively, and the diagnosis is made only when permanent sections of the gallbladder are examined. In these situations, patients with completely excised T1 lesions require no further therapy, and patients with higher-stage lesions should undergo reoperation in accordance with the principles outlined earlier (see above). Other appropriate reasons for not performing the additional surgery at the time of the cholecystectomy are (1) the desire to discuss the management scheme with the patient and (2) a lack of experience with the procedure for T2 tumors. Not infrequently, patients are referred to hepatic-pancreatic-biliary centers 10 to 14 days after surgery, which is an inopportune time for reoperation, especially if the first procedure was difficult. Surgery may then be delayed for 3 to 4 weeks. We restage patients with abdominal CT scans when they are referred with this diagnosis, and it is not unusual to find hepatic metastases when this is done. The survival rate is much higher after radical resection than after cholecystectomy, even when cholecystectomy was the first procedure.32

Gallbladder polyps

Benign gallbladder poyps may be adenomas or more commonly a focal form of cholesterolosis. Multiple polyps are most often due to cholesterolosis. Gallbladder polyps are discovered incidentally on ultrasonograms or CT scans or are diagnosed when they cause biliary colic. They may be malignant but are rarely so when less than 1 cm in diameter, especially when they are multiple. Most gallbladder polyps are less than 0.5 cm in diameter; these are almost always benign cholesterol polyps and may be followed if they are not giving rise to symptoms. Single polyps between 0.5 and 1 cm in diameter should probably be removed by means of cholecystectomy. Multiple asymptomatic polyps in this size range should be followed because they are more likely to be due to cholesterolosis. About one quarter of all single gallbladder polyps more than 1 cm in diameter are malignant, and such polyps should be treated as malignant as a matter of policy. Almost all polyps more than 1.8 cm in diameter are malignant.33 Gallbladder sludge and stones may be mistaken for polyps. They are differentiated by using Doppler ultrasonography to determine if the lesion has internal blood flow. If internal blood flow is absent, the lesion is unlikely to be a polyp.

Liver Cancer


anatomic considerations


A long-standing problem in discussing any surgical liver disease, especially liver cancer, has been the confusing terminology applied to liver anatomy and the various hepatic resections. Fortunately, a lucid and cogent terminology has emerged that is sanctioned by both the International Hepato-Pancreato-Biliary Association (IHPBA) and the American Hepato-Pancreato-Biliary Association (AHPBA).31 This terminology has been widely adopted around the world and translated into many languages. It may be briefly summarized as follows.

Figure 8

The fundamental principle is that the anatomic divisions of the liver are based on vascular and biliary anatomy rather than on surface markings [seeFigure 8]. This is an important point because surgical resection is a process of isolating specific liver volumes serviced by specific vascular and biliary structures. The anatomic ramifications of the hepatic artery and the bile duct are regular and virtually identical. Liver anatomy is best understood by first following these structures through a series of orderly divisions. The branching of the portal vein on the right side is similar to that of the bile duct and the hepatic artery, but its branching on the left side is unusual as a result of the need in the fetus that the umbilical portion of the portal vein acts as a conduit carrying blood in the reverse direction to that postnatally.

Figure 8

The first-order division of the proper hepatic artery and the common hepatic duct into the right and left hepatic arteries and the right and left hepatic ducts, respectively, results in division of the liver into two parts (or volumes), referred to as the right and left hemilivers (or the right and left livers) [seeFigure 8andTable 7]. In this system of terminology, the term ‘lobe’ is never used to denote a hemiliver, because it bears no relation to the internal vascular anatomy. The right hepatic artery supplies the right hemiliver, and the left hepatic artery supplies the left hemiliver. The right and left hepatic ducts drain the corresponding hemilivers. The plane between these two zones of vascular supply is called a watershed. The border or watershed of the first-order division is a plane that intersects the gallbladder fossa and the fossa for the inferior vena cava and is called ‘the midplane of the liver.’

Figure 8

The second-order division divides each of the hemilivers into two parts [seeFigure 8andTable 7], referred to as sections. The right hemiliver comprises the right anterior section and the right posterior section. These sections are supplied by a right anterior sectional hepatic artery and a right posterior sectional hepatic artery and are drained by a right anterior sectional hepatic duct and a right posterior sectional hepatic duct. The left hemiliver comprises the left medial section and the left lateral section. These sections are supplied by a left medial sectional hepatic artery and a left lateral sectional hepatic artery and are drained by a left medial sectional hepatic duct and a left lateral sectional hepatic duct.

Figure 8

The third-order division divides the liver into nine segments, each of which has its own segmental artery and bile duct [seeFigure 8andTable 7]. The caudate lobe, a unique portion of the liver that is separate from the right and left hemilivers, is also referred to as segment 1. The left lateral section comprises segments 2 and 3; the left medial section comprises segment 4 (which is sometimes further divided into segments 4a and 4b); the right anterior section comprises segments 5 and 8; and the right posterior section comprises segments 6 and 7.

primary cancers


Hepatocellular Cancer

Hepatocellular cancer (HCC), or hepatoma, is the fifth most common cancer in the world. About 90% of cases arise in patients with chronic liver disease, especially when the disease has progressed to cirrhosis. Although any condition that produces cirrhosis may lead to HCC, the most common cause is viral hepatitis. In the United States, some 3 million people are infected with hepatitis C virus (HCV), and more than 1 million people have liver disease associated with hepatitis B virus (HBV). HCV infection is much more likely to lead to HCC than HBV infection is. AJCC staging criteria are useful for planning the management of liver cancer [seeTable 8andTable 9].

Clinical evaluation

The usual presentation of sporadic HCC consists of pain, mass, and systemic symptoms of cancer, although the disease may also be discovered incidentally. HCC occurring as a complication of liver disease may present similarly, but it is often manifested first as a deterioration of liver function with the onset of jaundice, ascites, or encephalopathy.

Investigative studies

Screening programs are employed in high-risk populations. These programs, which use a-fetoprotein (AFP) levels and ultrasonographic examination of the liver to detect early HCC, may detect asymptomatic tumors.

Figure 9

The diagnosis of sporadic HCC is based on elevation of AFP levels (an indicator with 50 to 60% sensitivity) and the presence of a hepatic mass on axial images. HCCs typically demonstrate hypervascularity, which is best seen on arterial-phase images [seeFigure 9]. A pseudocapsule is often visualized, which is best seen on portal venous–phase images. MRI has been found to be more sensitive than CT in the detecton of HCC. The characteristic findings are those of a hypervascular tumor on the arterial phase with washout of contrast in the venous phase. These findings also help differentiate HCC from cirrhotic nodules, which typically do not show washout. Multifocality is also common in HCC, unlike other hepatic neoplasms. Routine biopsy is not indicated in patients with a characteristic mass, those who have a mass and an elevated AFP level, or those who are symptomatic and require treatment for pain. HCC may be very well differentiated and difficult to distinguish from hepatic adenoma and focal nodular hyperplasia on biopsy. It may also be hard to distinguish from cirrhotic nodules. Biopsy is associated with a small risk of bleeding or tumor seeding.

Surgical staging

Staging of sporadic HCC requires axial imaging of the abdomen and imaging of the chest. FDG-PET scanning is only marginally useful: HCCs are typically well differentiated, and, as a result, only 50% of the tumors are visualized. Staging laparoscopy is helpful: additional tumors are found in about 15% of patients.34

Staging also requires evaluation of the extent of liver disease. The Child-Pugh classification is used to determine operability. With few exceptions, resection is limited to Child-Pugh class A patients with near-normal bilirubin levels (< 1.5 mg/dL), normal or marginally raised prothrombin time, and no or minimal portal hypertension. The extent of resection must be tailored to the severity of the liver disease. For instance, resection of more than two segments is limited to patients with normal liver function. Too extensive resection puts the patients at risk for liver failure in the postoperative period. In Japan and China, indocyanine green clearance is used in Child-Pugh class A patients to determine the possible extent of resection.

Management

Rationale for surgeryThe rationale for surgery is clear in patients without liver disease or in Child-Pugh class B or C patients with chronic liver disease. The rationale for surgery in Child-Pugh class A patients, however, remains controversial.

Partial liver resection [see 5:23 Hepatic Resection] is the procedure of choice for sporadic HCC in patients with normal livers. In Child-Pugh class B or C patients with chronic liver disease, liver resection can be hazardous, and orthotopic liver transplantation (OLT) is the procedure of choice. To justify the use of donor organs, however, it is necessary to select patients with HCC so that the long-term outcome of OLT for HCC is similar to that of OLT for benign conditions. To achieve this goal, OLT is restricted to patients with a single tumor less than 5 cm in diameter or to patients with as many as three tumors, none of which are more than 3 cm in diameter (the Milan criteria). These criteria have been shown to be associated with OLT outcomes comparable to those for benign conditions.35

In Child-Pugh class A patients with liver disease, hepatic resection and OLT are options if the Milan criteria are met. The optimal therapeutic approach in this situation has been the subject of considerable debate, with proponents arguing for one of two strategies—namely, (1) primary OLT or (2) resection followed by OLT if HCC recurs,36,37 provided that the patients still meet the criteria for OLT (so-called salvage OLT). A complete discussion of this controversy is beyond the scope of this chapter. Currently, it would seem that the best strategy in patients who meet the criteria for OLT is to perform resection followed by transplanation if disease recurs.36,37 There is a trend toward liberalizing the OLT criteria to include single tumors 6 or 7 cm in diameter, especially if the source of the organ is a living donor.

When OLT is to be performed, it is important that the waiting time be short; these tumors progress over a timescale of a few months, and when viewed on an intention-to-treat basis, the results of OLT deteriorate significantly if the waiting time is long.38 In the United States, this concern has been dealt with by the introduction of the Model for End-stage Liver Disease (MELD) scoring system, which gives priority to recipients with HCC. It is common in the United States—and even more usual in countries with longer waiting times—to inhibit the growth of the HCC with various bridging-to-transplantation strategies during the waiting period for OLT. Such strategies include systemic chemotherapy, local treatments (e.g., radiofrequency [RF] ablation and alcohol injection), transarterial chemoembolization (TACE), and even resection of the HCC (so-called bridge resection). TACE may also be used to downsize tumors so that they meet transplant criteria.

In patients with nondiseased livers, the extent of resection depends on the size and position of the tumor. As much as 75% of the liver may be safely excised when normal liver function is present. The size of the future hepatic remnant may be determined by means of imaging. PVE of the side of the liver to be resected may be performed preoperatively to increase the size of the future remnant. It may also be used for this purpose in patients with liver disease. In these patients, PVE functions as a test of the liver’s ability to regenerate. Failure to respond to PVE is itself a contraindication to surgery in patients with chronic liver disease.

As a rule, liver resections for HCC should be anatomic [see 5:23 Hepatic Resection]. Recurrence rates are higher with nonanatomic resections because HCCs grow along portal veins and metastasize locally within segments, sections, or hemilivers, depending on how far they reach back along the portal veins. When a nonanatomic resection is performed, the resection margings should be 2 cm or greater. When HCC reaches the main portal vein, resection is generally contraindicated; the results are very poor in this situation.

 

Intrahepatic Cholangiocarcinoma

Clinical evaluation

Intrahepatic CCAs arise from intrahepatic bile ducts. The three phenotypic types, MF, PI, and IG, are described above. The MF type is by far the most common. Intrahepatic CCA tumor usually occurs in normal livers. The presentation is similar to that of sporadic HCC.

Investigative studies

Figure 10

The appearance of intrahepatic CCA on CT is suggestive of a secondary tumor [seeFigure 10]. Unlike HCC, diagnosis of intrahepatic CCA often requires biopsy, which reveals an adenocarcinoma that is indistinguishable from a hepatic metastasis. Special stains may be helpful in differentiating this tumor from a true secondary malignancy, but the differentiation is rarely certain. An elevated CA 19-9 concentration is strongly suggestive of this diagnosis if it is higher than 100 U/mL. To make the diagnosis of intrahepatic CCA, primary tumors in other sites must be excluded by means of axial imaging of the chest, the abdomen, and the pelvis; upper and lower GI endoscopy; and mammography. FDG-PET scanning is another helpful method by which an extrahepatic primary tumor may be identified.

Surgical staging

FDG-PET scanning appears to be a promising staging tool for identifying portal lymph node and distant metastases when the primary tumor is actually an intrahepatic CCA. Portal lymph node metastases are a relative contraindication to resection in patients with MF tumors; the results of resection in this situation are poor. Left-side tumors may metastasize to lymph nodes at the cardia of the stomach and along the lesser curvature or to the mediastinum.

Management

The considerations related to resection for intrahepatic CCA are similar to those for sporadic HCC (see above). Liver transplantation generally is not performed for this tumor, because of the typically poor results.

secondary cancers


Colorectal Metastases

Clinical evaluation and investigative studies

About 50% of the 150,000 patients who are diagnosed with colorectal cancer annually in the United States either have or will have liver metastases. About 10% of patients with these colorectal metastases (CRMs) are eligible for liver resection. CRMs may be diagnosed either at the time of treatment of the primary colorectal cancer (synchronous tumors) or at a later stage (metachronous tumors).

Figure 11

Synchronous tumors are diagnosed by means of either preoperative CT scanning [seeFigure 11] or intraoperative palpation. LFTs may show elevations (especially of the serum alkaline phosphatase level), but these results are not specific. CEA levels are not helpful as long as the primary tumor is in place. Metachronous tumors are most often diagnosed in the course of a postcolectomy surveillance program, either by imaging the liver with CT or FDG-PET scans or by detecting a rise in the CEA level. When synchronous metastases are discovered preoperatively, a FDG-PET scan should be done to complete the staging.

Surgical staging

In about 25% of patients, FDG-PET scans change management as a result of detecting unsuspected extrahepatic or intrahepatic disease. Sometimes it demonstrates that apparent metastases are actually benign lesions. Second primary tumors are not uncommon in patients with metachronous lesions; accordingly, such patients should also be staged by means of colonoscopy, if this procedure was not done in the preceding 6 months, as well as FDG-PET scanning. Staging laparoscopy adds little to staging if an FDG-PET scan has been done.

Intraoperative staging consists of careful palpation of intra-abdominal structures, including hepatic and portal venous lymph nodes. In patients with metachronous lesions, however, palpation of the entire abdomen may be limited by adhesions from previous operations. IOUS of the liver may also detect unsuspected lesions, although this is less likely if the patient has already been staged by means of FDG-PET.

The main value of FDG-PET in this setting is its ability to discover unsuspected extrahepatic disease. In so doing, it helps eliminate futile hepatic resections. If a patient with extrahepatic disease is treated with hepatic resection, a ‘recurrence’ is inevitable. Elimination of pointless resections has a positive effect on survival: a 2004 study from our institution found that the overall 5-year survival rate after FDG-PET was about 60%, compared with 40% after conventional imaging.39 Furthermore, after FDG-PET scanning has been done, the most important prognostic factors determining long-term outcome are the grade of the primary colorectal tumor and whether the primary tumor had metastasized to lymph nodes.40 FDG-PET–scanned patients with poorly differentiated primary tumors do poorly in terms of overall survival after hepatic resection.39 In recent years, standard PET scanners have been replaced with CT-PET scanners, which fuse the images and provide superior diagnosis and staging. For planning surgical extirpation, however, the level of detail provided by high-quality contrast-enhanced CT or MRI is also required.

Management

Rationale for surgeryThe classic criteria that determined eligibility for resection are (1) that the primary tumor has been or can be completely resected, (2) that (with uncommon exceptions) there is no extrahepatic tumor (other than the primary tumor), and (3) that it is possible to resect all tumors in the liver while leaving enough of a hepatic remnant to ensure that hepatic failure does not develop postoperatively. The considerations governing the extent of the resection and the use of PVE are similar to those for sporadic HCC.

Treatment of multiple tumors is much more common with CRMs than with HCC. However, nonanatomic resections are as effective as anatomic resections as long as the resection margin is microscopically clear. The traditional view has been that resection margins of 1 cm are mandatory and lesser margins lead to poorer results. This view has been challenged in several studies that claim that margins as narrow as 1 mm are satisfactory and are probably as effective as traditional margins provided that they are free of microscopic and gross cancer. This issue must be considered to still be unsettled, and, certainly, 1 cm margins should be the goal whenever feasible. When close margins are expected, transection of the liver with a saline-linked RF ablation device may be useful in that this device leaves a margin of devitalized tissue in the patient and in the specimen.41 When the margin is very close, it may be extended by painting the cut surface of the hepatic remnant with the RF device.

Synchronous resection of the primary tumor and the liver metastases has proved to be safe42 and is desired by many patients. The decision to proceed with hepatic resection should not be made until resection of the primary tumor has been completed and it has been determined that the margins are clear and the patient is stable. Some patients with a small number of lung lesions in addition to liver lesions have been cured by resection.

The classic criteria for resection have been challenged and extended by new surgical approaches and the advent of much improved chemotherapy, including oxaliplatin and irinotecan, as well as the targeted monoclonal antibodies bevacizumab and cetuximab. These agents have demonstrated the ability to downsize colorectal tumors both in the liver and at extrahepatic sites. Thus, many formerly unresectable patients can now have liver resection after downsizing with chemotherapy. It is generally agreed that the best time for surgery is when the size and the position of tumors are such that liver resection can be done safely.43 Continuing chemotherapy until maximal effect is undesirable because irinotecan has been associated with the development of steatohepatitis44 and oxaliplatin with endotheliolitis.45 These injuries can make liver resection hazardous. Furthermore, continued downsizing may make tumors difficult to locate. This is important because although tumors may show a complete radiologic response on CT and FDG-PET scans, this is infrequently associated with a complete pathologic response. The ability to resect multiple large tumors from the liver has been enhanced by PVE and two-stage hepatectomy. In this approach, one side of the liver is cleared of tumor and the portal vein on the other side is occluded. At a second stage, the atrophied side containing the bulk of the disease (usually the right hemiliver) is excised.

 

Extrahepatic disease in patients with hepatic metastases Good results have been obtained in patients with recurrent disease in the primary colorectal site and liver metastases, as well as liver and lung metastases.46 With the advent of the new chemotherapy, it may be warranted to extend this approach to patients with liver and portal lymph node metastases but not with positive lymph nodes in the celiac or para-aortic regions.47 Peritoneal metastases have been treated successfully by resection combined with hyperthermic intraperitoneal chemotherapy. Whether this approach combined with liver resection would be effective in patients with liver and peritoneal metastases is uncertain. Its application seems warranted in selected well-followed cases.

Ablation of colorectal metastases In situ destruction of tumors with cryotherapy or RF ablation may expand the surgeon’s ability to eradicate CRMs localized to the liver.48 RF ablation has largely supplanted cryotherapy in this context as a result of its lower incidence of complications and greater ease of use. Ablation may be used either as an adjunct to operative management or as the sole treatment when there are many metastases (but usually < 10). The efficacy of RF ablation as an adjunct to surgery remains to be determined. It is doubtful, however, that using this modality alone to eradicate multiple lesions will improve overall survival significantly because the tumor biology in such cases is likely to be that of an aggressive tumor. Recent data suggest that the preceeding statement is correct.49 FDG-PET scans should be performed in all such patients; the likelihood of discovering extrahepatic tumors increases as the number of hepatic tumors increases.39

RF ablation is not recommended for treatment of resectable metastases: it is not approved for this purpose, and using it in this way would mean substituting an unproven therapy of unknown efficacy for a proven therapy of known value. Again, based on recent publications, it is highly likely that RF ablation results in poorer long-term survival than liver resection.48 If a consenting patient with resectable metastases nevertheless insists on this less invasive therapy, the surgeon should document that the preceding considerations have been explained. RF ablation may be applied by means of open, laparoscopic, or percutaneous methods. There is good reason to believe that targeting ability is degraded as one moves to less invasive methods. This consideration should also be explained to patients, although, undoubtedly, there are some patients who, because of comorbid conditions, are candidates only for percutaneous or laparoscopic approaches.

 

Neuroendocrine Metastases

Neuroendocrine metastases are characteristically slow growing. Some are functional, especially if they arise from the ileum; metastatic liver disease from this source may produce carcinoid syndrome. 111In-pentetreotide imaging (OctreoScan, Mallinckrodt Inc., Hazelwood, Missouri) provides staging information comparable to that provided by FDG-PET in patients with CRMs.

The aims of surgical treatment are (1) to eradicate the cancer and (2) to reduce hormonal symptoms. The considerations regarding tumor eradication for neuroendocrine metastases are similar to those for CRMs—that is, resection should be performed if all cancer can be removed and no extrahepatic cancer is detectable. In highly symptomatic patients in whom conservative therapy with octreotide has failed, debulking the tumor by means of either chemoembolization or surgery may provide relief. The former is more suitable for patients with multiple small, diffuse metastases, whereas the latter is preferred for patients with large localized tumors. RF ablation may also be employed, either combined with surgical treatment or alone; this is an excellent use of this procedure in that the aim is cytoreduction rather than eradication. Debulking tumors in asymptomatic patients with the intention of extending survival is controversial. It is not recommended when more than 10% of the tumor wil remain.

Noncolorectal, Non-neuroendocrine Metastases

Occasionally, liver metastases from other primary sites behave like CRMs in that they are localized to part of the liver in the absence of extrahepatic disease. Such patients can be managed according to the same approach employed for CRMs, although the outcome is somewhat less satisfactory. Tumors that have been treated in this way with acceptable results include breast cancers, renal cell cancers, gastric cancers, acinar cell cancers of the pancreas, and ovarian cancers. Liver resection for more aggressive malignancies (e.g., metastases from gallbladder cancer and pancreatic ductal adenocarcinomas) can be expected to yield very poor results.

incidentally discovered asymptomatic hepatic mass


Now that transaxial imaging of the abdomen is commonly performed for a variety of complaints, the problem of the incidentally discovered asymptomatic hepatic mass is being encountered with increased frequency. Generally, cysts are easily distinguished from solid tumors; the main diagnostic issue is differentiation of the various solid lesions.

The differential diagnosis of the benign solid hepatic mass includes hepatic adenoma, focal nodular hyperplasia (FNH), focal fatty infiltration, cavernous hemangioma, and other rare neoplasms (e.g., mesenchymal hamartoma and teratoma)—all of which must be distinguished not only from one another but also from malignant tumors. In the past, several diagnostic tests (e.g., ultrasonography, CT, sulfur colloid scanning, and angiography) were used to differentiate these neoplasms. Currently, our usual practice is to perform MRI with gadolinium contrast enhancement, which generally allows accurate differentiation among benign tumors with a single test. Cavernous hemangiomas are usually easy to distinguish because they have a characteristic appearance on MRI (hypointense on T1-weighted images, very intense on T2-weighted images, and filling in from the periphery with gadolinium injection); if they are asymptomatic, they need not be resected. It is important to distinguish asymptomatic FNHs from hepatic adenomas: whereas resection is recommended for adenomas because of their potential for hemorrhage or malignant degeneration, asymptomatic FNHs can safely be observed. An FNH is nearly isointense on T1– and T2-weighted images; it shows slightly more enhancement than normal liver parenchyma in the early phase after contrast injection and then becomes isointense. A central scar is often, but not always, seen. Conversely, a hepatic adenoma exhibits strong early-phase enhancement with contrast administration and tends to be hyperintense on T1-weighted images.

Given that a symptomatic hepatic mass is usually treated with resection, preoperative biopsy for tissue diagnosis is rarely necessary or desirable. Modern noninvasive radiologic tests, in conjunction with a careful patient history, are often quite accurate in predicting histologic diagnosis. Biopsy of hepatic lesions should not be performed indiscriminately, because there is a small risk of complications or tumor tracking and because biopsy results often do not change management. As a rule, biopsies should be performed when definitive surgical intervention is not planned and when pathologic confirmation is necessary for institution of nonsurgical therapy.

 

Jeffrey D. Wayne, MD, FACS

Assistant Professor of Surgery
Northwestern University Feinberg School of Medicine
Staff Physician
Northwestern Memorial Hospital

Mark S. Talamonti, MD, FACS

Professor, Surgical Oncology
Northwestern University Feinberg School of Medicine
Chief, Division of Surgical Oncology
Northwestern Memorial Hospital

Gastric Adenocarcinoma


The incidence of gastric carcinoma exhibits significant geographic variability. The disease is most common in Japan and China, and high rates of occurrence have also been reported in Central and South America, Eastern Europe, and parts of the Middle East.1 In most of the more developed nations, however, gastric carcinoma is relatively uncommon. The overall incidence of this condition has decreased in the past few decades, but gastric carcinoma remains the second leading cause of cancer death worldwide. The reported reductions in gastric cancer mortality may be linked to better refrigeration and a concomitant decrease in the intake of salted, pickled, smoked, and chemically preserved foods; however, this link remains controversial. An inverse association with the consumption of fresh fruits and vegetables has also been noted.2

Gastric cancer occurs 1.5 to 2.5 times more frequently in males than in females. It is rarely diagnosed before the age of 40, and its incidence peaks in the seventh decade of life. African Americans, Hispanic Americans, and Native Americans are two times more likely to have gastric cancer than white Americans are.3

In the United States in particular, the incidence of stomach cancer has fallen substantially over the past 70 years.4 Whereas this disease was once a leading cause of cancer-related death in the United States, it now ranks 13th among major causes. Unfortunately, the decline in incidence has not translated into an improvement in the 5-year survival rate.5 Across all races, the 5-year relative survival was 23% for the period extending from 1992 to 1999.3 This result is probably related to the advanced stage at which most patients present. A 1995 study from the Commission on Cancer of the American College of Surgeons (ACS) found that 66% of patients with gastric cancer presented with locally advanced or metastatic disease.6 Resection rates ranged from 30% to 50%, and 5-year survival rates after resection with curative intent were directly related to stage at presentation. For stage I disease, the survival rate was 43%; for stage II, 37%; for stage III, 18%; and for stage IV, 20%.

Another relevant change in the epidemiology of gastric cancer is a shift in the distribution of primary lesion sites within the stomach. In the first quarter of the 20th century, two thirds of gastric cancers were located in the antrum and the prepyloric area, and only 10% arose in the cardia or the esophagogastric junction. Since the 1970s, however, adenocarcinoma of the proximal stomach has become increasingly common. In one study, the incidence of adenocarcinoma of the gastric cardia rose from 29.1% to 52.2% in the period between 1984 and 1993.7 In another, which included 18,365 gastric cancer patients from ACS-approved hospitals, a full 31% of tumors were found to be in the proximal stomach, compared with only 26% in the distal third.8 In the United States, carcinoma of the cardia occurs primarily in whites, with a male-to-female ratio of approximately 2:1. Cancer of the cardia appears to be distinct from adenocarcinoma of the distal esophagus, which frequently arises in the setting of Barrett’s esophagus.9 Associations have also been reported between cancer of the gastric cardia and infection with Helicobacter pylori or Epstein-Barr virus.10,11

Classification

Adenocarcinoma of the stomach may be divided into two histologic subtypes, intestinal and diffuse.12 Each subtype has unique pathologic, epidemiologic, etiologic, and prognostic features. The intestinal (or glandular) subtype usually arises in the distal stomach (often after a long precancerous phase), is more common in elderly patients, and has been closely associated with atrophic gastritis and diets high in nitrates and nitrose compounds.13 The characteristic histologic finding is cohesive neoplastic cells that form glandlike tubular structures. The diffuse subtype occurs more frequently in younger patients and has no identifiable precursor lesion. It may develop in any part of the stomach but shows a predilection for the cardia. Cell cohesion is absent; thus, individual cancer cells infiltrate and thicken the stomach wall without forming a discrete ulcer or mass.

In general, the prognosis for the diffuse subtype is worse than that for the intestinal subtype. Whereas intestinal lesions are seen more frequently in regions with a high incidence of gastric cancer, the incidence of diffuse lesions is constant among various populations throughout the world.14 Accordingly, the overall decline in gastric cancer over the past century has been attributed to a decline in intestinal lesions and to a decline in the incidence of H. pylori infection (see below).

Risk Factors

Historical studies of specimens obtained during operation or at autopsy suggest that gastric carcinoma, especially of the intestinal subtype, frequently develops in the presence of chronic atrophic gastritis and associated intestinal metaplasia. It has generally been assumed that adenocarcinoma of the distal stomach progresses from chronic gastritis to metaplasia through the teratogenic influence of environmental factors. The most commonly studied environmental factors are the nitrates and nitrose compounds present in high levels in salted, smoked, or pickled foods consumed in areas where gastric cancer is endemic.15 To date, however, no prospective studies have conclusively demonstrated that modern refrigeration practices and the subsequent decline in the salting, smoking, and pickling of food have been responsible for the relative decline in intestinal gastric cancer. Furthermore, the intestinal subtype may arise in the absence of metaplasia. Finally, the emergence of chronic infection with H. pylori as the dominant risk factor for gastric adenocarcinoma has challenged the paradigm of the atrophic gastritis—intestinal metaplasiaµgastric cancer sequence.

Epidemiologic studies across various populations worldwide have consistently demonstrated a strong association between H. pylori infection and gastric cancer.16 Prospective serologic studies have confirmed that persons with evidence of such infection are three to six times more likely to have gastric cancer than persons who are seronegative.17 Still, only a very small fraction of infected persons have gastric cancer. It has been estimated that more than half of the world’s inhabitants may be infected with H. pylori—a number that dwarfs the actual incidence of gastric cancer. What is clear is that H. pylori infection of the gastric mucosa leads to a state of chronic active inflammation that lasts for decades. This inflammatory process appears to be modulated by multiple forces, including genetic and environmental factors.18 Inherited traits may confer susceptibility or resistance to carcinogenesis. Indeed, first-degree relatives of gastric cancer patients have a two to three times higher relative risk of contracting the disease.19 Gastric irritants may act as promoters, and antioxidants may have a protective effect (which may be part of the reason for the reduced risk of gastric cancer associated with diets rich in fruits and vegetables).20

Unlike intestinal cancers, diffuse cancers appear not to be associated with H. pylori infection. Diffuse adenocarcinoma of the stomach is more common in young patients and has no known precursor lesion.9 The incidence of genetically associated diffuse cancers is estimated to be in the range of 5% to 10%.19 Familial cases of diffuse gastric cancer occur at an average age of 38 years and are inherited in an autosomal dominant fashion with 70% penetrance.21 Patients with blood group A have a 16% to 20% increased risk of gastric cancer.22

Clinical Evaluation

In high-risk areas (e.g., Japan), mass screening programs have been successful in identifying early gastric cancer, which is generally amenable to surgical cure.23 In fact, in some Japanese studies, as many as 40% of newly diagnosed patients had early gastric cancer. Unfortunately, in Western countries, the disease is almost always diagnosed relatively late, when it is locally advanced or metastatic. When it is superficial, gastric cancer typically produces no symptoms. As it progresses, however, a constellation of vague, nonspecific symptoms may develop, including anorexia, fatigue, weight loss, and epigastric discomfort. Dysphagia, early satiety, vomiting, and hematemesis also are seen, albeit rarely; when present, they often indicate advanced disease. Indeed, early gastric cancer has no characteristic physical findings, and many patients are not diagnosed until they present with jaundice, ascites, or a palpable mass, all of which signal incurable disease.

Investigative Studies

Until comparatively recently, an upper gastrointestinal series was often the first diagnostic test ordered to evaluate symptoms related to the upper GI tract. However, even with double-contrast techniques, which allow improved visualization of mucosal detail, false negative rates as high as 25% were reported, especially with small lesions (i.e., 5 to 310 mm).24 Accordingly, in most large series, fiberoptic endoscopy with biopsy has replaced contrast radiography as the primary diagnostic technique.25 Upper GI endoscopy with biopsy has been reported to have a diagnostic accuracy of 95%.20 However, false negatives have been re port ed, especially in the context of inadequate biopsies. Thus, it is re commended that at least four biopsy specimens taken from the region of any atypical findings.26

Staging

Two major classification systems are available for staging gastric cancer. The first is the one used in Japan, where gastric cancer is staged according to the general rules for gastric study in surgery and pathology published by the Japanese Research Society for Gastric Cancer (JRSGC).27 This elaborate system focuses on the anatomic involvement of specifically numbered lymph node stations. The second system is the one generally used in Western countries—namely, the familiar tumor-node-metastasis (TNM) system developed by the American Joint Committee on Cancer (AJCC) and the International Union Against Cancer (UICC) [see Tables 1 and 2].28 The AJCC/UICC staging system is based on a gastric cancer database and classifies lesions according to the depth to which the primary tumor penetrates the gastric wall, the extent of lymph node involvement, and the presence or absence of distant metastases.

The primary goal in the evaluation of gastric cancer patients is to stratify them into two clinical stage groups: those with loco regional disease (AJCC stages I to III) and those with systemic disease (AJCC stage IV).29 The National Comprehensive Cancer Network (NCCN) has developed consensus guidelines for the clinical evaluation and staging of patients with possible gastric cancer. These guidelines are accessible to any practitioner via the Internet (http://www.nccn.org/professionals/physician_gls/PDF/gastric.pdf) and are updated annually. Multidisciplinary evaluation is recommended for all patients. A careful history is obtained and a thorough physical examination performed, with special attention paid to comorbid conditions that might preclude operative intervention. Initial laboratory studies include a complete blood cell count with a platelet count; determination of serum electrolyte, blood urea nitrogen, creatinine, and glucose concentrations; and a liver function panel. Chest radiography is performed, along with computed tomography of the abdomen and pelvis.

Figure 1. CT scan: carcinoma of the gastric antum

Whereas CT is invaluable for detecting ascites, bulky adenopathy, and significant visceral metastases, its overall accuracy in staging tumors is modest: only 70% for advanced lesions and 44% for early lesions.30 CT assesses lymph node involvement primarily on the basis of node size. Thus, its sensitivity for N1 and N2 disease is low, ranging from 24% to 43%; however, its specificity is high, approaching 100%. Technical advances, such as spiral (helical) CT with intravenous contrast plus appropriate gastric distention with 600 to 800 ml of water (a negative contrast agent), have allowed modest improvements in overall staging with CT [see Figure 1]. Nevertheless, CT is still limited in its ability to evaluate peritoneal disease and liver metastases smaller than 5 mm.31

Figure 2. EUS image of gastric neoplasm

Given the limitations of CT, we believe that in the absence of obvious metastatic disease, locoregional staging with endoscopic ultrasonography (EUS) is vital for accurately assessing tumor penetration through the gastric wall (T stage) and ascertaining whether regional nodes (N stage) or even mediastinal or para-aortic lymph nodes may be involved (which would be considered M1 disease) [see Figure 2]. EUS is unique among imaging modalities in its ability to image the gastric wall as a five-layer structure, with each layer correlating with an actual histologic layer.32 The overall accuracy of EUS in determining the extent of infiltration ranges from 67% to 92%.33 EUS features that suggest lymph node metastasis include a rounded shape, hypoechoic patterns, and a size larger than 1 cm. In one study comparing preoperative findings from EUS with pathologic findings at operation, EUS was 100% sensitive for N0 disease and 66.7% sensitive for N1 disease.34 EUS also allows identification and aspiration of small-volume ascites. If cytologic study of the ascitic fluid so obtained confirms the presence of malignant cells, the patient is considered to have metastatic disease and therefore is not eligible for curative-intent surgery. For all of these reasons, EUS is now widely accepted as superior to conventional CT in the regional staging of gastric cancer.9

Role of Laparoscopy

The ultimate goal of any staging evaluation is to ensure that patients with metastatic disease are not treated with nontherapeutic laparotomy or other local therapies (e.g., radiation therapy), which are generally ineffective against advanced disease. Even small-volume metastatic disease identified on the surface of the liver or the peritoneum at laparotomy is associated with poor survival: in one study, patients with such disease had a life expectancy of only 6 to 9 months.35 In these situations, there is little to be gained from attempts at palliative resection.

Staging laparoscopy [see 5:20 Procedures for Benign and Malignant Gastric and Duodenal Disease] has proved to be highly relevant to the evaluation of patients with gastric cancer. In a study from the Memorial Sloan-Kettering Cancer Center (MSKCC), the investigators performed laparoscopic exploration on 110 of 111 patients with newly diagnosed gastric cancer.36 Of these 110 patients, 94% were accurately staged, with a sensitivity of 84% and a specificity of 100%, and 37% were found to have subclinical metastatic disease. Hospital stay was substantially shorter in the 24 patients who underwent diagnostic laparoscopy with biopsy only (average, 1.4 days) than in comparable patients who underwent exploratory laparotomy without resection (average, 6.5 days). Finally, at the time the data were reported, none of the patients who underwent laparoscopy had required palliative surgery. Subsequent single-institution series confirmed the utility of staging laparoscopy, reporting accuracy rates ranging from 95% to 97% and occult M1 disease rates approaching 30%.37,38 Taken as a whole, the data, though derived from relatively small single-institution experiences, are compelling, and they have led the NCCN to encourage laparoscopic staging strongly, either before or at the time of the planned resection.39

Management

Surgical Therapy

Figure 3. Workup and treatment of gastric carcinoma

Surgical resection [see 5:20 Procedures for Benign and Malignant Gastric and Duodenal Disease] remains the only potentially curative therapy for localized gastric cancer [see Figure 3]. Cure requires removal of all gross and microscopic disease. More specifically, a margin-negative (R0) resection entails wide local excision of the primary tumor with en bloc removal of all associated lymphatic vessels and any local or regional extension of disease. The downside of surgical resection as a sole modality of therapy is that it is associated with a high rate of relapse. Consequently, several areas of surgical treatment of stomach cancer remain subject to controversy. In particular, the extent of gastric re section, the extent of lymph node dissection, the optimal approach to proximal stomach lesions, and the role of splenectomy and adjacent organ resection continue to generate significant debate.

Extent of gastric resection R0 resection (i.e., resection of all gross disease with microscopically negative margins) has been shown to have a clear impact on overall survival after potentially curative surgery. In the German Gastric Cancer Study, a prospective multicenter observational trial, the calculated 10-year survival rate in the entire population was 26.3%, compared with 36.1% in patients who underwent an R0 resection.40 In a large multi-institutional adjuvant therapy trial, 19% of patients underwent an R1 resection (i.e., had resection-line involvement); only 9% of patients with stage I, II, or III disease and resection-line involvement survived beyond 5 years, compared with 27% of those who underwent an R0 resection.41 Given the propensity of tumor for submucosal spreading, many authors consider proximal margins of 5 to 6 cm, with routine frozen-section analysis, to be optimal.42,43

In an effort to lower the positive margin rate, some surgeons have proposed that total gastrectomy be considered the operation of choice for all operable gastric cancers. This approach, originally based on historical data from single institutions, has been tested in three clinical trials. In the first trial, elective total gastrectomy was compared with subtotal gastrectomy as curative-intent therapy for adenocarcinoma of the antrum.44 Elective total gastrectomy did not increase mortality, but it also did not improve 5-year survival (which was 48% in both treatment arms). In the second trial, patients with antral cancer were randomly assigned to undergo either subtotal gastrectomy or total gastrectomy with extended lymph node dissection (ELND) and en bloc distal pancreatectomy and splenectomy.45 Total gastrectomy was associated with increased operative time, greater transfusion requirements, and longer hospital stay; however, median survival was significantly better in the subtotal gastrectomy group (1,511 days versus 922 days). In the third trial, the investigators concluded that sub total gastrectomy should be the procedure of choice for cancer of the distal half of the stomach, provided that an adequate negative proximal margin could be achieved.46 This conclusion was based on their finding that 5-year survival probabilities were essentially equivalent in the two groups studied (65.3% in the subtotal gastrectomy group versus 62.4% in the total gastrectomy group).

 

Options for proximal gastric cancer As noted (see above), adenocarcinoma of the gastric cardia and the esophagogastric junction appears to be clinically distinct from adenocarcinoma of the distal stomach,47 and its incidence is currently escalating across all races and age groups. Accordingly, it is imperative that surgeons understand the surgical options for treatment of proximal gastric cancer.48

For tumors originating from the distal esophagus, esophagectomy—either transhiatal esophagectomy with a cervical anastomosis or transthoracic (Ivor-Lewis) esophagectomy with a thoracic anastomosis—is clearly the procedure of choice [see 4:7 Open Esophageal Procedures]. For tumors of the cardia, it has been suggested that esophagogastrectomy might offer a survival advantage over total gastrectomy with an esophagojejunal anastomosis. This suggestion was evaluated in a study of 1,002 patients with adenocarcinoma of the esophagogastric junction.49 The investigators divided tumors into three types on the basis of the location of the tumor center—cancers of the distal esophagus (type I), cancers of the cardia (type II), and cancers of the subcardial fundus (type III)—and analyzed the demographic and long-term survival data. Operative mortality proved to be higher with esophagogastrectomy than with ex tended total gastrectomy. Furthermore, R0 resection and lymph node status were found to be the dominant prognostic factors influencing survival. Finally, in patients with type II lesions, the pattern of lymphatic spread was primarily to paracardial, lesser curvature, and left gastric node groups. These data, taken together, led the authors to conclude that total gastrectomy is preferable to esophagogastrectomy in this setting if a margin-negative resection can be achieved.

An alternative approach to treating proximal gastric cancer is to perform a proximal subtotal gastrectomy. To date, no prospective studies have compared this method with total gastrectomy or trans hiatal esophagogastrectomy for esophagogastric junction tumors, but surgeons from MSKCC have published their retrospective experience with 98 patients who underwent either total gastrectomy or proximal subtotal gastrectomy for proximal gastric cancer over a 10-year period.50 There were no significant differences be tween the groups with respect to morbidity, mortality, or 5-year survival. It remains to be seen whether such excellent results can be achieved at other centers.

Thus, the evidence at present does not support routine performance of total gastrectomy for lesions of the distal fundus or antrum, provided that histologically negative margins are achievable without compromise of the gastric inlet. Our current practice is to perform a subtotal gastrectomy with Billroth II reconstruction for tumors of the distal stomach, a total gastrectomy with Roux-en-Y esophagojejunostomy for most cancers of the fundus and the proximal stomach [see 5:20 Procedures for Benign and Malignant Gastric and Duodenal Disease].

 

Extent of lymph node dissection Over the past decade, few topics in the surgical literature have generated more debate than the optimal extent of regional lymphadenectomy for gastric cancer. In Japan, where radical surgery for gastric cancer is now universally accepted, the JRSGC has codified the extent of lymphatic dissection according to the level of nodes dissected.51 A D1 lymph node dissection involves resection of the perigastric lymph nodes along the greater and lesser curvature of the stomach. A D0 dissection is anything less than a D1 dissection. A D2 dissection entails resection of the D1 nodes along with nodes along the common hepatic artery, the left gastric artery, the celiac axis, and the splenic artery. A D3 lymph node dissection adds resection of nodes in the hepatoduodenal ligament and the root of the mesentery. Finally, a D4 resection calls for a D3 dissection plus resection of the retroperitoneal para-aortic and paracolic lymph nodes.52 The JRSGC defines a curative operation as a gastric resection that includes lymph nodes one level beyond the level of pathologic nodal involvement. Thus, in Japan, a D2 lymph node dissection is considered the standard resection for even relatively early cancers, and numerous studies have cited the benefits of D3 and even D4 lymphadenectomy for advanced carcinoma.53–55

Western surgeons have been reluctant to embrace radical lymphadenectomy, arguing that it has yet to demonstrate an unequivocal survival advantage in any prospective, randomized trial from a Western institution or cooperative group. Detractors further argue that the survival advantage associated with more radical procedures simply reflects stage migration, a higher incidence of early gastric cancers, and differences in tumor biology and body habitus between Japanese and Western populations, and they point to the increases in operating time and morbidity that often accompany extended gastric resections. One retrospective review of the tumor registries of over 2,000 hospitals in the United States found that D2 lymph node dissection had no survival advantage over D1 lymph node dissection in terms of either the median survival time or the 5-year survival rate.56

Two prospective trials from Western Europe examined this issue further in an effort to evaluate the safety and efficacy of ELND. In the Dutch Gastric Cancer Group trial, 711 patients were randomly assigned to undergo either D1 or D2 lymphadenectomy as part of a potentially curative gastrectomy for biopsy-proven adenocarcinoma.57 This trial was unique in its use of extensive quality control measures, which included instruction and operative supervision by an expert gastric cancer surgeon from Japan (who also assisted with the processing and pathologic examination of the surgical specimens). Patients without evidence of disseminated metastases underwent either total gastrectomy or, if 5 cm proximal margins could be obtained, distal gastrectomy. In this study, a D2 lymph node dissection entailed distal pancreatectomy and splenectomy. Both morbidity and mortality were significantly high er in the D2 group than in the D1 group, and D2 dissection conferred no demonstrable survival advantage at a median follow-up of 72 months.

In a trial from the Medical Research Council in the United Kingdom, 400 patients with stage I to IIB disease were randomly assigned to undergo either a D1 or a D2 lymph node dissection.58 There was no significant difference in overall 5-year survival between the two arms, but multivariate analysis demonstrated that clinical stages II and III, advanced age, male sex, and removal of the pancreas and the spleen were independently associated with poor outcome. The authors concluded that the classic Japanese D2 dissection offered no survival advantage over D1 dissection. However, they hypothesized that D2 dissection with preservation of the distal pancreas and the spleen might lead to decreased morbidity and mortality within the extended resection group and thus potentially to superior outcomes.

Further support for this hypothesis was provided by two nonrandomized trials from specialized centers. The Italian Gastric Cancer Study Group (IGCSG) completed a phase II multicenter trial designed to evaluate the safety and efficacy of pancreas-preserving D2 lymph node dissection.59 Quality control measures included supervision by a surgeon who had studied the technique of D2 lymph node dissection at the National Cancer Center Hospital in Tokyo. At a median follow-up time of 4.38 years, the overall morbidity rate for D2 dissection in the 191 patients enrol led was 20.9%, and the in-hospital mortality was 3.1%. The 5-year survival rate for eligible patients was 55%. In a prospective series of 125 patients undergoing standardized D2 lymph node dissection at a single Western center, the investigators reported a mortality of 1.37% and an overall morbidity of 33.5%.60 As in the IGCSG study, distal pancreatectomy was avoided in all cases, except when direct extension was suspected on the basis of macroscopic findings (5.5% of cases). Overall 5- and 10-year survival rates for this highly selected cohort were 52.3% and 40%, respectively. These studies suggest D2 lymph node dissection may be safely performed in Western centers, when accompanied by careful selection of patients, strict standardization of technique, and a strategy of pancreatic preservation.

Current AJCC guidelines state that pathologic examination of at least 15 lymph nodes is required for adequate staging.28 In an effort to confirm the benefit of this staging system, investigators from MSKCC reviewed their experience with 1,038 patients who underwent R0 resection for gastric cancer.61 The location of positive lymph nodes (within 3 cm of the primary tumor versus more than 3 cm away) did not significantly affect median survival; however, the number of positive lymph nodes had a profound effect on survival. Furthermore, in cases in which at least 15 nodes were examined (27% of the total), the median survival for patients with N1 (metastasis in one to six regional lymph nodes), N2 (metastasis in seven to 15 regional lymph nodes), and N3 disease (metastasis in more than 15 regional lymph nodes) was significantly longer than the median survival reported in cases in which 14 or fewer nodes were resected with the specimens. These findings are consistent with published data from our own institution (North western University Feinberg School of Medicine), which indicate that the number of positive lymph nodes is a highly significant predictor of survival.62 In our series of 110 patients, those with N2 or N3 disease (seven or more positive lymph nodes) had a median disease-free survival (DFS) of 17.6 months, whereas those with N0 or N1 disease (six or fewer positive nodes) had a median DFS of 44 months. Data from other centers support this view as well [see Table 3].

It is our current practice to perform a D2 lymph node dissection, with resection of all perigastric lymph nodes along the greater and lesser curvatures of the stomach, as well as those along the common hepatic artery, the left gastric artery, the celiac axis, and the splenic artery [see 5:20 Procedures for Benign and Malignant Gastric and Duodenal Disease]. We make every attempt to preserve the tail of the pancreas and spleen, with multivisceral resection reserved for cases of overt direct extension of malignant disease in the absence of disseminated metastasis. This strategy should provide adequate staging in terms of the AJCC guidelines, minimize morbidity, and possibly confer a survival advantage on certain patient subgroups, as suggested by the results of the trials mentioned.

 

Role of splenectomyRoutine splenectomy has been proposed as a means of facilitating clearance of metastatic nodes along the splenic artery and in the splenic hilum, but there is little evidence to support this practice in the treatment of proximal gastric cancers. Indeed, numerous studies have documented the deleterious effect of splenectomy when it is performed as part of an extended gastric resection.

In a retrospective study of 392 patients who underwent curative gastrectomy at a high-volume cancer center, the impact of splenectomy on survival and postoperative morbidity was evaluated.63 Splenectomy was not predictive of death on multivariate analysis, but complications were far more frequent in patients who underwent splenectomy as part of surgical treatment than in those who did not (45% versus 21%). Specifically, the incidence of infectious complications was far higher in the splenectomy group than in the nonsplenectomy group (75% versus 47%).

In a review of data from an American College of Surgeons Pattern of Care Study, the investigators reported that the operative mortality was 9.8% in patients who underwent splenectomy during gastric resection, compared with 8.6% for those who did not.64 More significantly, the 5-year observed survival rate was 20.9% in the splenectomy group, compared with 31% in the nonsplenectomy group.

In a randomized, prospective trial, early and late results of total gastrectomy alone were compared with those of total gastrectomy plus splenectomy in patients being treated for cancers of the upper third of the stomach.65 All patients underwent a D2 lymph node dissection. The operative mortalities and the 5-year survival rates were similar in the two groups, but the splenectomy group had more infectious complications. Specifically, the splenectomy group had higher incidences of pulmonary complications, postoperative fever higher than 38° C (100° F), and subphrenic abscess formation. We agree with the conclusions of the authors of this study: routine splenectomy does not increase survival, and it should be reserved for situations in which the gastric tumor directly invades the splenic hilum or there is evidence of gross nodal metastases along the splenic artery.

 

Nonsurgical Therapy

Adjuvant therapyAs noted (see above), the majority of patients who present with gastric carcinoma and undergo potentially curative surgical treatment will experience locoregional failure, distant metastasis, or both and will succumb to their disease. Accordingly, numerous adjuvant approaches—including chemo therapy, radiotherapy, chemoradiation, immuno chemotherapy, and intraperitoneal chemotherapy—have been tried in gastric cancer patients with the aim of improving overall survival and DFS. The results, for the most part, have been disappointing.

Results from prospective, randomized, controlled trials of adjuvant radiation therapy in this setting have failed to establish a survival benefit. In a multi-institutional trial from 1994, patients were randomly assigned to undergo surgery alone, surgery plus adjuvant radiation, or surgery plus adjuvant multiagent chemotherapy.66 There was no significant benefit to either adjuvant regimen: overall 5-year survival was 20% for surgery alone, compared with 12% for surgery plus radiation therapy and 19% for surgery plus chemotherapy.

Results from trials of chemotherapy alone have been equally un satisfactory. Because of the established inefficacy of single-agent 5-fluorouracil (5-FU) therapy, combination chemotherapy regimens have been employed. Such regimens have included nitro sourea compounds, mitomycin-C, anthracyclines, and members of the cisplatin family.23 In a meta-analysis of 13 trials comparing adjuvant chemotherapy with observation in non-Asian countries, the odds ratio for death in the treated group was 0.8, corresponding to a relative risk of 0.94.67 This result did not, however, reflect a statistically significant improvement. Most oncologists have now abandoned the use of chemotherapy by itself in the adjuvant setting.

In an effort to derive greater therapeutic benefit than can be achieved with either radiation therapy or chemotherapy alone, combinations of the two have been used in the adjuvant setting. In Intergroup Trial 0116, 556 patients who had undergone R0 resection of adenocarcinoma of the stomach or the esophagogastric junction were randomly assigned to treatment with either surgery alone or surgery plus postoperative chemoradiotherapy.68 Patients with tumors ranging from stage IB to stage IVM0 were included; the majority had T3 tumors and node-positive disease. The therapeutic regimen consisted of 5-FU and leucovorin administered concomitantly with 45 Gy of external-beam irradiation over a period of 5 weeks. Median overall survival in the surgery-only group was 27 months, compared with 36 months in the surgery-chemoradiation group. In addition, the 3-year survival rate was 41% in the surgery-only group, compared with 50% in the surgery-chemoradiation group. The hazard ratio for death in the surgery-only group as compared with the surgery-chemoradiation group was 1.35.

In the United States, the results of Intergroup Trial 0116 have led to the acceptance of chemoradiotherapy as standard adjuvant therapy for patients who have undergone curative-intent resection of gastric cancer. Nonetheless, numerous criticisms of this trial have been expressed. Specifically, a review of the operative and pathology reports of 453 of the patients revealed a lack of surgical standardization.69 When the extent of lymphadenectomy was categorized, the majority (54.2%) of the patients were found to have undergone a D0 dissection; 38.1% underwent a D1 dissection, and only 7.5% underwent a D2 or D3 dissection. These findings suggest that the main effect of the chemoradiation therapy may have been simply to compensate for inadequate surgery. This suggestion is supported by the observation that the number of patients with local and regional recurrences was higher in the surgery-only group (178 versus 101), whereas the number of patients with distant failure was slightly higher in the adjuvant-therapy arm (40 versus 32). Furthermore, when the Maruyama Index of Un resected Disease (a computer model developed for accurate prediction of nodal station involvement in gastric cancer) was ap plied to the 556 patients eligible for the Intergroup Trial, the median Maruyama Index was 70.70 This value was far above the level considered to represent optimal surgical therapy (i.e., Maruyama Index < 5) and led the authors to conclude that the vast majority of patients in the trial had been surgically undertreated.

Currently, physicians, especially in Europe, generally eschew adjuvant therapy after R0 resection of gastric cancer, except under the auspices of a clinical trial.9 The Radiation Therapy Oncology Group has initiated a phase II trial of adjuvant chemoradiotherapy using 45 Gy of external beam radiation with cisplatin and paxitaxel, with or without 5-FU. If promising results are found, a phase III trial will follow. It is to be hoped that ongoing trials will shed further light on this complex management issue.

 

Neoadjuvant therapy As a response to the disappointing results of adjuvant therapy and the inability of many patients to regain adequate performance status after radical gastric surgery, neoadjuvant therapy protocols have been proposed.71 The theoretical benefits of a neoadjuvant treatment strategy include treatment-induced tumor downstaging, which may enhance resectability, and early administration of systemic therapy, which allows almost all patients to receive and complete the prescribed treatment. Furthermore, because treatment is administered when measurable disease is present, response to therapy may be assessed and continued only in patients who are likely to benefit. Finally, patients who are found to have rapidly progressive disease during preoperative chemotherapy may be spared having to undergo a nontherapeutic gastrectomy.72

In a report of three phase II trials from the M. D. Anderson Cancer Center, encompassing 83 patients who received neoadjuvant chemotherapy before planned surgical resection, clinical response rates ranged from 24% to 38%, with three patients (4%) exhibiting a complete pathologic response.72 Sixty-one patients (73%) were able to undergo a curative-intent resection, and the response to chemotherapy was the only significant predictor of survival on multivariate analysis.

Preoperative chemoradiation therapy has also been shown to be feasible in phase II trials. In a 2001 trial that included 23 patients, 96% of the study population received combined-modality therapy.71 Nineteen patients (83%) were able to undergo surgical resection with D2 lymphadenectomy; four patients (17%) had progressive disease and did not undergo resection. Morbidity and death rates were acceptable (32% and 5%, respectively), and 11% of patients exhibited complete pathologic responses. Over all, 63% of patients showed pathologic evidence of a significant treatment effect.

Newer neoadjuvant treatment strategies employ multiagent induction chemotherapy followed by chemoradiotherapy and planned gastric resection in patients with locally advanced but potentially resectable gastric cancer.73

 

Follow-up and Management of Recurrent Disease

Even after gross resection of all disease with microscopically negative margins (R0 resection), recurrence of gastric carcinoma is common. Adenocarcinoma of the stomach may spread through direct extension, via lymphatic channels to regional and distant lymph nodes, or via the bloodstream to distant sites. Furthermore, once tumors have penetrated the serosa (T3), peritoneal metastasis becomes a possibility. Through autopsy series and clinical studies, certain definite patterns of locoregional failure and distant metastasis have been established. Locoregional recurrences are common in the gastric bed and the adjacent lymph nodes. Clinical and reoperative evaluation have documented recurrent disease at the anastomosis, in the retroperitoneum, or in the regional lymph nodes in 3% to 69% of patients; the incidence of recurrence may vary, depending on whether the patients had received adjuvant therapy.23 One autopsy series documented a locoregional recurrence rate of 94% in patients treated with surgery alone. The peritoneum is ultimately involved in 17% to 50% of all patients. The most common sites of visceral metastases are the liver and the lungs.

In view of the high recurrence rates, all patients who have undergone resection should be seen for routine surveillance examinations. Currently, the NCCN recommends that a complete history and physical examination be conducted every 4 months for 1 year, then every 6 months for 1 year, and then yearly thereafter.39 A complete blood count, serum electrolyte concentrations, and liver function studies should also be considered. Imaging studies (e.g., CT and endoscopy) are ordered as indicated, usually in response to new symptoms. In addition, long-term vitamin B12 supplementation should be initiated for patients who have undergone a proximal or subtotal gastrectomy.

Other Gastric Malignancies


Gastric Lymphoma

Gastric lymphoma is the second most common malignancy of the stomach, accounting for 2% to 9% of gastric tumors in the United States. Lymphomas of the stomach are of the non-Hodgkin type. The stomach is the most common site of extra nodal involvement of non-Hodgkin lymphoma (NHL) and ac counts for nearly 50% of all such cases.74

Clinical Evaluation

The presenting symptoms of gastric lymphoma, like those of gastric adenocarcinoma, are nonspecific and include loss of ap petite, weight loss, vomiting, and bleeding. Overt clinical symptoms (e.g., fever and night sweats) are relatively rare: in one multicenter trial concerned with primary gastric lymphoma, they occurred in fewer than 12% of patients enrolled.75 Risk factors for gastric lymphoma include H. pylori infection, immunosuppression after solid-organ transplantation, celiac disease, inflammatory bowel disease, and HIV infection.76

Investigative Studies

The diagnosis of gastric lymphoma is most frequently established by means of endoscopy with biopsy. Staging studies include a comprehensive blood count, a lactate dehydrogenase (LDH) level, and a comprehensive chemistry panel; CT of the chest, the abdomen, and the pelvis; and, often, a bone marrow biopsy. All pathology slides should be reviewed by an experienced hematopathologist.77

Staging and Prognosis

Numerous staging systems have been employed to stage NHL of the GI tract. Of these, the one most commonly applied is a modification of the Ann Arbor staging system for lymphoma.76 For surgeons, the most important determination is often whether the NHL (1) is confined to the stomach and the perigastric nodes (stage I and II disease), (2) involves other intra-abdominal nodes and organs (stage III), or (3) extends outside the abdomen (stage IV).78

Management

Over the past decade, the management of patients with gastric lymphoma has undergone significant changes. Generally, there has been a shift away from surgical management, even in relatively localized cases (stages I and II).79 This shift is the result not only of the documented success of chemotherapy alone for more advanced cases (stages III and IV) but also of a better understanding of the etiology of gastric lymphoma.80 Approximately 45% of all gastric lymphomas are low-grade mucosa-associated lymphoid tissue (MALT) lymphomas.75 The gastric mucosa is normally devoid of lymphoid tissue. It is hypothesized that MALT develops in the stomach in response to chronic H. pylori infection.81

Nonsurgical therapy Low-grade MALT lymphoma usually presents as stage I or II disease and has an indolent course. Since 1993, when regression of low-grade MALT lymphoma after eradication of H. pylori was first reported, numerous trials have documented the efficacy of anti-H. pylori therapy, with complete remission rates ranging from 50% to 100%.79 In the German MALT Lymphoma Study, the complete remission rate was 81%; 9% of patients exhibited partial responses, and 10% showed no response.82 Low-grade lymphomas that are more advanced or do not regress with antibiotic therapy may be treated with H. pylori eradication and radiation (with or without chemotherapy).83For localized persistent disease, modest doses of radiation, on the order of 30 Gy, may be employed. When chemotherapy is required, multiagent regimens, such as cyclophosphamide-vincristine-prednisolone (COP), are often used.

Approximately 55% of gastric lymphomas are high-grade lesions, which can occur with or without a low-grade MALT component.75 These lymphomas are treated with chemotherapy and radiation therapy according to the extent of the disease. The cyclophosphamide-doxorubicin-vincristine-prednisolone (CHOP) regimen is the one most frequently employed. In some studies, the anti-CD20 monoclonal antibody rituximab has been either added to standard therapy or used alone, with encouraging results.84

 

Surgical therapy Surgical resection, once thought to be essential for the diagnosis, staging, and treatment of early-stage gastric lymphoma, now is used mainly in patients who experience bleeding or perforation. In the German Multicenter Study Group trial, 185 patients with stage I or II gastric lymphoma were treated either with gastrectomy followed by radiation or (in the case of high-grade lesions) chemotherapy plus radiation or with chemo therapy and radiotherapy alone.75 There was no significant difference in survival between the group receiving surgical treatment and the group receiving nonoperative therapy: overall 5-year survival rates were 82.5% and 84%, respectively. There were no perforations, and there was only one hemorrhage (in a patient treated with chemotherapy alone). Similarly, in a single-institution, prospective, randomized trial comparing chemotherapy alone with chemotherapy plus surgery for stage I and II lymphoma, there were no instances of perforation and only three instances of GI bleeding in the chemotherapy group, compared with two bleeding episodes in the surgery plus chemotherapy group.79

Currently, patients with early-stage high-grade gastric lymphomas are treated primarily with chemotherapy or radiation therapy; only rarely do they require surgical intervention for complications encountered during therapy. Patients with locally advanced (stage III) or disseminated (stage IV) gastric lymphoma are clearly best treated with chemotherapy, with or without radiation. Occasionally, surgery is indicated in such patients to treat residual disease confined to the stomach or to palliate bleeding or obstruction that does not resolve with nonoperative therapy. Primary surgical therapy is to be avoided in these patients because of the significant risk of complications and the delay in initiating systemic therapy.

 

Gastrointestinal Stromal Tumor

Gastrointestinal stromal tumor (GIST), though relatively rare in absolute terms, is the most common sarcoma of the GI tract,85 with approximately 6,000 cases reported each year in the United States alone. The stomach is the most common site of involvement, accounting for 60% to 70% of cases86; the small intestine (25%), the rectum (5%), the esophagus (2%), and a variety of other locations account for the remainder. On the basis of their appearance on light microscopy, GISTs were once thought to be of smooth muscle origin, and most were classified as leiomyosarcomas.87 Thus, extended gastric resection, often including contiguous organs, was advised. Recurrence developed after R0 resection in approximately 50% of cases.88 With the advent of immunohistochemistry and electron microscopy, it became clear that GIST has both smooth muscle and neural elements, and the cell of origin is now believed to be the interstitial cell of Cajal, an intestinal pacemaker cell.89 The diagnosis of GIST is secured by immunohistochemical staining for the tyrosine kinase receptor KIT (CD117), which highlights the presence of interstitial cells of Cajal. More than 95% of GISTs exhibit unequivocal staining for KIT.86 Approximately two thirds of GISTs also express CD34. Histologically, these tumors may exhibit a spindle cell pattern, an epithelioid pattern, or a mixed subtype.

Clinical Evaluation

The median age of incidence is 63 years, and tumors are generally between 0.5 and 44 cm in diameter at the time of diagnosis (median diameter, 6 cm).86 Mass-related symptoms (e.g., abdominal pain, bloating, and early satiety) may be present. Melena or anemia from overlying mucosal ulceration may be present as well. A small subset of patients have peritonitis as a consequence of tumor rupture and subsequent hemorrhage. Finally, many GISTs are discovered incidentally during operation, abdominal imaging, or endoscopy.

Investigative Studies

When a GIST is suspected, abdominal and pelvic imaging with either CT or MRI is indicated. Chest imaging is performed as well. Endoscopy, with or without EUS, may occasionally help with surgical planning, but because of the infrequency of mucosal involvement, it is rarely diagnostic.90 Surgical consultation should be obtained to determine whether the lesion can be resected. If the tumor is resectable, biopsy should not be performed, because of the risk of tumor rupture and intra-abdominal dissemination. Biopsy may be required, however, if the patient has widespread disease or may be enrolling in a trial of neoadjuvant therapy. In such cases, biopsy may be performed percutaneously or at the time of EUS.

Staging and Prognosis

Although the majority of gastric GISTs have a benign course, a wide spectrum of biologic behavior has been observed. Of the prognostic factors examined to date, tumor size and mitotic rate appear to be the most valuable. If the tumor is less than 2 cm in diameter and the mitotic count is lower than five per high-power field (HPF), the risk of an aggressive disease course is considered to be very low. Conversely, if the tumor is larger than 10 cm, if the mitotic count is higher than 10/HPF, or if the tumor is larger than 5 cm with a mitotic count higher than 5/HPF, the risk of aggressive clinical behavior is considered to be high. For all other tumors, the risk of aggressive disease is considered to be intermediate.86

Management

Surgical therapy The role of surgery in the treatment of a GIST is to resect the tumor with grossly negative margins and an intact pseudocapsule. Lymph node involvement is rare with GISTs, and thus, no effort is made to perform ELND. The tumor must be handled with care to prevent intra-abdominal rupture. Formal gastric resection is rarely required: as a rule, it is indicated only for lesions in close proximity to the pylorus or the esophagogastric junction.

Nonsurgical therapy If the tumor has metastasized or has advanced locally to the point where surgical therapy would result in excessive morbidity, the patient is treated with the tyrosine kinase inhibitor imatinibmesylate. Imatinib is a selective inhibitor of a family of protein kinases that includes the KIT-receptor tyrosine kinase, which is expressed in the majority of GISTs. Originally indicated for the treatment of chronic myelocytic leukemia, imatinib was approved for the treatment of KIT-positive GIST in 2002, when phase II clinical trials documented sustained objective responses in a majority of patients with advanced unresectable or metastatic GIST.91 Patients with borderline resectable lesions should be treated with imatinib until they exhibit a maximal response as documented by CT and positron emission tomography (PET); surgery may then be undertaken to resect any residual foci of disease. Similarly, whereas patients with metastatic disease are unlikely to manifest a complete response to imatinib therapy, they should be periodically reevaluated and considered for resection should surgical treatment become technically feasible.90

After an R0 resection of a GIST, no adjuvant therapy is indicated unless the patient is participating in a clinical trial. The American College of Surgeons Oncology Group is currently conducting two trials of imatinib in the postoperative setting. A phase II trial (Z9000) of imatinib, 400 mg/day, for patients with high-risk GIST, has reached accrual, and a phase III trial (Z9001) comparing 1 year of imatinib, 400 mg/day, with placebo in patients with intermediate-risk GIST is currently under way.

 

Gastric Carcinoid

Gastric carcinoid tumors are rare, accounting for fewer than 11% to 30% of all GI carcinoids and fewer than 1% of all gastric tumors.92 The median age at diagnosis is 62, and tumors are equally distributed between men and women.

Clinical Evaluation and Investigative Studies

Gastric carcinoid tumors are often discovered during endoscopic examination of patients experiencing chronic abdominal pain; patients may also complain of vomiting and diarrhea. These tumors are rarely associated with symptoms of the carcinoid syndrome. Diagnosis is usually confirmed by endoscopic biopsy, and EUS is helpful in determining the extent of gastric wall penetration and the degree of regional lymph node involvement.

Gastric carcinoid tumors have been divided into three types, primarily on the basis of their association (or lack thereof) with hypergastrinemia. Type I tumors are associated with chronic atrophic gastritis, are generally small (< 1 cm), and are often multiple and polypoid. They grow slowly and only rarely metastasize to regional nodes or distant sites. Type II tumors are associated with the Zollinger-Ellison syndrome and multiple endocrine neoplasia type I (MEN I) and, like type I tumors, are usually small and multiple. They also grow slowly, but they are more likely to metastasize than type I gastric carcinoids are. Type III (sporadic) gastric carcinoid tumors are the most biologically aggressive type. They are often large (> 1 cm) at the time of diagnosis and are not associated with hypergastrinemia. Type III lesions frequently metastasize to regional nodes (54%) or the liver (24%).92

Management

For patients with small, solitary type I tumors, endoscopic polypectomy [see 5:18 Gastrointestinal Endoscopy] is indicated to remove the source of the hypergastrinemia. For patients with type II lesions, treatment is similar to that for patients with type I lesions, with the extent of gastric resection determined by the size and number of lesions. For patients with type III lesions, however, either distal or total gastrectomy with ELND is required.93 All patients undergoing a less than total gastrectomy should be followed with serial endoscopy at regular intervals.94

Small Bowel Malignancies


Malignant tumors of the small intestine are rare, accounting for fewer than 5% of all GI tract malignancies. In the United States, only a few thousand new cases of small bowel cancer are reported each year.95 The majority of small bowel malignancies are adenocarcinomas, lymphomas, or carcinoid tumors,96 though GISTs are being noted with increasing frequency in the small intestine. Treatment of lymphomas, carcinoid tumors, and GISTs in the small bowel is nearly identical to treatment of the same lesions in the stomach [see Other Gastric Malignancies\, above] and thus will not be covered further in this chapter. Our focus here is on the presentation, diagnosis, and treatment of adenocarcinoma of the small bowel. Like gastric adenocarcinoma, small bowel adenocarcinoma is usually staged according to the AJCC/UICC TNM classification system [see Tables 4 and 5].

Clinical Evaluation

Between 46% and 55% of small bowel adenocarcinomas occur in the duodenum.96,97 Patients frequently present with nausea, vomiting, abdominal pain, weight loss, and GI bleeding98; occasionally, they present with iron deficiency anemia or a positive fecal occult blood test result. In rare cases, small bowel obstruction, often with the tumor serving as a lead point for intussusception, is the first manifestation of the disease.97

Investigative Studies

When an adenocarcinoma is located in the duodenum, the diagnosis is often made by means of esophagogastroduodenoscopy (EGD). Lesions within the first 100 cm of the small bowel may be evaluated with push enteroscopy. When the adenocarcinoma is situated elsewhere in the small bowel, it is localized with small bowel radiographs. Some authors consider enteroclysis to be superior to the more commonly used small bowel follow-through in this setting, in that enteroclysis is better able to demonstrate fine mucosal detail.99 In experienced hands, enteroclysis may therefore be more sensitive.100 Some lesions are identified when CT or MRI is performed to evaluate complaints of abdominal pain. Further more, abdominal imaging may yield complementary staging information (e.g., the presence of regional adenopathy or metastatic disease). One promising new method for the identification of small bowel tumors is wireless capsule endoscopy.101 This minimally invasive technique may be particularly useful in identifying small lesions in the distal jejunum and ileum that cannot be identified radiographically.

Management

Aggressive surgical resection remains the cornerstone of therapy for adenocarcinoma of the small intestine.102 For periam pullary lesions, pancreaticoduodenectomy is typically required to achieve a margin-negative resection. For lesions in the distal duodenum, a segmental sleeve resection with a duodenojejunostomy is appropriate. For lesions in the jejunum or the ileum, segmental resection may be performed with a wide mesenteric resection to encompass potentially involved regional lymph nodes. Contiguous organs are resected en bloc as necessary.98

Because the presenting signs and symptoms are often vague and nonspecific, diagnosis is often delayed. In one series, only 6 (11%) of the 53 patients were suspected of having a small bowel tumor at admission.102 In a retrospective review of patients with small bowel tumors treated at our institution, the mean duration of symptoms before surgical management was 110 months, and more than 50% of the patients were found to have stage III or IV disease.98

The 5-year survival rate continues to be low (24% to 37%).98,103,104 Significant predictors of good overall survival include complete (R0) resection and low AJCC tumor stage.98,103,104 The available evidence indicates that all patients with small bowel neoplasms should be offered an oncologically sound surgical resection. In one series, curative (R0) resection was accomplished in 51% of cases.103

 

Eric J. DeMaria, MD, FACS

Professor of Surgery, Duke University School of Medicine, and Vice Chair and Chief of Network General Surgery and Director of EndoSurgery and Bariatric Surgery, Duke University Medical Center

Christopher J. Myers, MD

Minimally Invasive Surgery Fellow, Duke University School of Medicine

 

10.2310/7800.2008.S05C07

It is clear that severe obesity is associated with a significant increase in morbidity1 and a decreased life expectancy.2 Morbid obesity—defined as (a) a body weight that exceeds the ideal body weight by 100 lb or more or (b) a body mass index (BMI) greater than 35 kg/m2—has been shown to have a significant genetic basis.34 To date, attempts to manage morbid obesity with medical weight reduction programs have met with an unacceptably high incidence of recidivism.5 The approach that has had the greatest and longest-lasting success in achieving weight loss is bariatric surgery.

Preoperative Evaluation


Many surgeons are afraid to operate on the morbidly obese patient because they presuppose a marked increase in perioperative morbidity and mortality. It is now possible, however, to stratify the mortality risk for patients undergoing gastric bypass (GBP) by using a scoring system known as the Obesity Surgery Mortality Risk Score (OS-MRS), which includes five independent variables that can be identified preoperatively: (1) BMI greater than or equal to 50 kg/m2, (2) male gender, (3) hypertension, (4) pulmonary embolus risk (including previous thrombosis, pulmonary embolus, inferior vena cava [IVC] filter, right-side heart failure, and obesity hypoventilation syndrome [OHS]), and (5) patient age greater than or equal to 45 years. These factors were associated with a greater 90-day mortality in a prospective study of 2,075 patients who underwent GBP at a single institution,6 which was the basis for the initial proposal of this scoring system. The OS-MRS was subsequently validated in a multicenter study involving four institutions and 4,431 patients.7 With the presence of each variable equal to 1 point, each patient’s potential score ranged from 0 to 5. Patients with a score of 0 or 1 had a low mortality risk (group A; mortality, 0.2%); those with a score of 2 or 3 had an intermediate mortality risk (group B; mortality, 1.1%); and those with a score of 4 or 5 had a high mortality risk (group C; mortality, 2.4%). These findings suggest that the OS-MRS is a valuable tool that can be effectively used to stratify risk and facilitate surgical decision making and patient discussion regarding bariatric surgery.

Although the morbidly obese patient is certainly at greater risk, this risk can be markedly reduced by paying careful attention to detail in preoperative and postoperative care. The increased risks encountered in these patients include wound infection, dehiscence, thrombophlebitis, pulmonary embolism (PE), anesthetic calamities, acute postoperative asphyxia in patients with obstructive sleep apnea syndrome (SAS), acute respiratory failure, right ventricular or biventricular cardiac failure, and missed acute catastrophes of the abdomen (e.g., anastomotic leakage). The ensuing discussion begins by focusing on issues that the surgeon should carefully consider when operating on an extremely overweight patient.

Morbidity Associated with Central Fat Deposition


Much has been written about the increased health risks inherent in central (android) fat deposition as compared with peripheral (gynoid) fat deposition. It is thought that in the former, the increased metabolic activity of mesenteric fat is associated with increased metabolism of amino acids to sugar, which leads to hyperglycemia and hyperinsulinism. Hyperinsulinism gives rise to increased sodium absorption and hypertension. Furthermore, central obesity has been linked to hypercholesterolemia. Hence, these patients have a significantly higher incidence of diabetes, hypertension, hypercholesterolemia, and gallstones8—which explains the higher mortality of the apple distribution of body fat in comparison with the pear distribution. In the past, fat distribution was measured on the basis of the waist-to-hip ratio; however, computed tomographic scanning has shown that abdominal circumference is a more accurate measurement of central fat distribution.9 Morbidly obese women have significantly increased intra-abdominal pressure (IAP), and this increase is associated with stress and urge overflow urinary incontinence.10 With weight loss comes a significant decrease in bladder pressure and correction of incontinence. IAP, as reflected in bladder pressure, appears to be closely correlated with sagittal abdominal diameter and waist circumference but not with waist-to-hip ratio (many morbidly obese patients have both central and peripheral obesity). The increased IAP associated with central obesity may give rise to other comorbid factors as well, including venous stasis ulcers, OHS [see Respiratory Insufficiency of Obesity, below], gastroesophageal reflux, and inguinal and incisional hernias.

Respiratory Insufficiency of Obesity


Obese patients are at risk for respiratory difficulties, which may be present before operation or may be exacerbated by an operation. The term pickwickian syndrome (which derives from The Posthumous Papers of the Pickwick Club, by Charles Dickens) was resurrected from the late 1800s to describe a morbidly obese 52-year-old man who fell asleep in a poker game while holding a hand containing a full house.11 He was taken to the hospital by friends who presumed he was ill. The pickwickian syndrome is now known to comprise two pulmonary syndromes associated with morbid obesity: SAS and OHS.12

Sleep Apnea Syndrome

SAS is a potentially fatal complication of morbid obesity. A diagnosis of SAS should be suspected when there is a history of loud snoring, frequent nocturnal awakening with shortness of breath, and daytime somnolence. It is estimated that 2% of middle-aged women and 4% of middle-aged men in the US workforce have SAS, and the incidence is markedly higher in the severely obese.13 Patients will often admit to falling asleep while driving and waking up with their car on the road’s median strip or bumping its guardrail. It is extremely important that trauma surgeons be aware of the relation between obesity and somnolence should a morbidly obese patient be seen in the emergency department after an automobile accident in which he or she fell asleep at the wheel. Patients with SAS suffer from repeated attacks of upper airway obstruction during sleep. The cause is probably related to a large, fat tongue, as well as to excessive fat deposition in the uvula, pharynx, and hypopharynx. The normal genioglossus reflex is depressed, but this depression may be secondary to the excessive weight of the tongue. These patients are notorious snorers. As a result of inadequate stage IV and rapid eye movement (REM) sleep, they are markedly somnolent during the day.

Patients with SAS are at high risk for acute upper airway obstruction and respiratory arrest when undergoing an operation and general anesthesia. Therefore, any patients with suspected SAS should undergo preoperative polysomnography at a sleep center to confirm the diagnosis. Medications are usually ineffective. Stimulants, such as methylphenidate hydrochloride (Ritalin), should not be used. If a patient has a respiratory disturbance index (RDI) greater than 25—indicating more than 25 apneic or hypopneic episodes per hour of sleep—or has cardiac dysrhythmias in association with apnea, treatment by nocturnal nasal continuous positive airway pressure (nasal CPAP) should be provided. With this technique, air flowing through a nasal mask against a constant airway resistance enters the nasal pharynx and pushes the tongue forward to prevent recurrent obstruction.14 The pressure can be adjusted for each patient. Unfortunately, many patients cannot tolerate the device, because it is cumbersome and noisy and tends to dry out the upper airway, though dryness can be prevented with an inexpensive room humidifier. If the patient has severe SAS with an RDI greater than 40 and does not respond with elimination of the apneic episodes or cannot tolerate nasal CPAP, a tracheostomy should be considered. An extra-long tracheostomy tube is usually necessary because of the depth of the trachea in the morbidly obese patient.

Obesity Hypoventilation Syndrome

Figure 1.

OHS is a condition associated with morbid obesity in which a person suffers from hypoxemia and hypercapnia when breathing room air while awake but resting.15 Spirometry reveals decreases in forced vital capacity, residual lung volume, expiratory reserve volume, functional residual capacity, and maximum minute volume ventilation, usually without obstruction of airflow [see Figure 1]. The most profound decrease is that in expiratory reserve volume; it is probably secondary to increased IAP and a high-riding diaphragm. Thus, these patients have a restrictive rather than an obstructive pulmonary disease. The decreased expiratory reserve volume implies that many alveolar units are collapsed at end-expiration, which leads to perfusion of unventilated alveoli, or shunting. Patients with OHS often are heavy smokers or have additional pulmonary problems, such as asthma, sarcoidosis, idiopathic pulmonary fibrosis, or recurrent PE. One study of patients who underwent operation for morbid obesity showed no statistically significant difference in weight between those who had OHS and those who did not.12

Chronic, severe hypoxemia is associated with three complications that put patients with OHS at risk: polycythemia, pulmonary arterial vasoconstriction, and pulmonary hypertension. The polycythemia further increases the already significant risk of venous thrombosis and PE. If the hemoglobin (Hb) concentration is 16 g/dl or greater, phlebotomy to a concentration of 15 g/dl should be performed to reduce the postoperative risk of venous thrombosis. If the pulmonary arterial pressure (PAP) is 40 mm Hg or higher, consideration should be given to prophylactic insertion of an IVC filter because of the high risk of a fatal pulmonary embolism in these patients.16 Placement of an IVC filter can be a challenge because the appropriate landmarks cannot be identified in the operating room with fluoroscopy. It is necessary, before operation, to tape a quarter to the patient’s back over the second lumbar vertebra with the aid of fixed radiographs and then, during operation, to aim for the quarter with the insertion catheter, using fluoroscopy. Because these patients are usually too heavy for angiography tables, the filter usually cannot be inserted percutaneously in the radiology department.

Chronic hypoxemia also leads to pulmonary arterial vasoconstriction and severe pulmonary hypertension and eventually to right-side heart failure or cor pulmonale with neck vein distention, tricuspid valvular insufficiency, right upper quadrant tenderness secondary to acute hepatic engorgement, and massive peripheral edema.1718 Such patients may also have a significantly elevated pulmonary artery wedge pressure (PAWP), which suggests left ventricular dysfunction.17 Morbidly obese patients with a history of pulmonary disease or a BMI greater than 50 kg/m2 should have preoperative determinations of blood gas values. If arterial blood gas (ABG) measurement reveals severe hypoxemia (i.e., arterial oxygen tension [Pao2] f55 mm Hg), severe hypercapnia (arterial carbon dioxide tension [Paco2] g47 mm Hg), or both, the patient should undergo Swan-Ganz catheterization. If the PAWP is 18 mm Hg or greater, intravenous furosemide should be administered for diuresis before elective operation. However, some patients may require a high ventricular filling pressure. A low cardiac output and hypotension may follow diuresis, necessitating volume reexpansion.

Figure 2.

It is highly probable that some of the elevated PAP and PAWP measurements are caused by the increased IAP in the morbidly obese patient [see Figure 2]).1920 The high IAP leads to an elevated diaphragm, which in turn increases intrapleural pressure and thereby PAP and PAWP; if the pleural pressure is measured with an esophageal transducer, the transmyocardial pressure can be estimated. For this reason, these patients may require a markedly elevated PAWP to maintain an adequate cardiac output, and excessive diuresis may lead to hypotension. The same reasoning may be applied to a patient with a distended abdomen resulting from peritonitis and pancreatitis in whom what seem to be unusually high cardiac filling pressures are necessary. Therefore, one must rely on relative changes in cardiac output in response to either volume challenge or diuresis to determine the optimal PAWP in morbidly obese patients.

Patients with OHS respond rapidly to supplemental oxygen. However, oxygen administration is occasionally associated with significant CO2 retention, which necessitates intubation and mechanical ventilation. Because their pulmonary disease is restrictive rather than obstructive, these patients are usually easy to ventilate without high peak airway pressures. ABG measurements need not return to normal values before extubation; it is only necessary that they return to their preoperative values. These values are achieved early after laparoscopic procedures and, on average, 4 days after major open upper abdominal operations, when patients experience a decrease in incisional pain.21

It is important to emphasize that morbidly obese patients, especially those with respiratory insufficiency, should be placed in the reverse Trendelenburg position to maximize diaphragmatic excursion and to increase residual lung volume.22 These patients will often complain of air hunger and respiratory distress when they lie supine. So-called breaking of the bed at the waist may exacerbate the problem by pushing the abdominal contents into the chest, thereby raising the diaphragm and further reducing lung volumes. Placing these patients in the leg-down position may predispose them to venous stasis, phlebitis, and PE; this tendency should be offset with intermittent venous compression boots [see Thrombophlebitis, Venous Stasis Ulcers, and Pulmonary Embolism, below].23

Both SAS and OHS can be completely corrected with weight reduction after gastric operation for morbid obesity: the nocturnal apneas resolve, the Pao2 rises, and the Paco2 falls to normal as lung volumes improve.12

Cardiac Dysfunction


Morbidly obese patients are at significant risk for coronary artery disease as a result of an increased incidence of systemic hypertension, hypercholesterolemia, and diabetes. Because of this increased risk for cardiac dysfunction, preoperative electrocardiography should be performed on all obese patients 30 years of age or older.

Figure 3.

Cardiac dysfunction in the morbidly obese patient is usually associated with respiratory insufficiency of obesity, especially OHS.11 An elevated PAP in these patients may be secondary to hypoxemia-induced pulmonary arterial vasoconstriction, to elevated left atrial pressures secondary to left ventricular dysfunction, or to a combination of these; it may also be secondary to the increased pleural pressures arising from an elevated diaphragm secondary to increased IAP.172023 It is unusual for morbidly obese patients without respiratory insufficiency to experience significant cardiac dysfunction in the absence of severe coronary artery disease. Morbidly obese patients often have systemic hypertension, which can aggravate left ventricular dysfunction; however, mild left ventricular dysfunction can be documented in many morbidly obese patients in the absence of systemic hypertension.2425 Circulating blood volume, plasma volume, and cardiac output increase in proportion to body weight.25 Massively obese patients may occasionally present with acute heart failure: it is reasonable to assume that the enormous metabolic requirements of such patients can present a greater demand for blood flow than the heart can provide. Vigorous diuresis often corrects such acute heart failure. Significant weight loss corrects pulmonary hypertension [see Figure 3], as well as the left ventricular dysfunction associated with respiratory insufficiency.1726

Thrombophlebitis, Venous Stasis Ulcers, and Pulmonary Embolism


Morbidly obese individuals have difficulty walking, tend to be sedentary, have a large amount of abdominal weight resting on their IVC, and have increased intrapleural pressure (which impedes venous return).1820 All of these conditions increase the tendency toward phlebothrombosis. Patients are most at risk when immobilized in the supine position for long periods in the OR. These patients have been shown to have low levels of antithrombin, which may increase their tendency toward venous thrombosis.27 It has also been suggested that starvation, particularly in the postoperative period, may be associated with high levels of free fatty acids, which may predispose to perioperative thrombotic complications.28 Patients with severe OHS often have a noticeably elevated PAP, which can lead to right-side heart failure and can increase the risk of venous stasis and thrombosis. Investigators have noted that patients with primary idiopathic pulmonary hypertension are at significant risk for fatal PE.13

The risk of deep vein thrombosis (DVT) increases with a prolonged operation or a postoperative period of immobilization, and it increases even further in the morbidly obese patient. Standard or low-molecular-weight heparin should be administered subcutaneously 30 minutes before operation and at appropriate intervals thereafter (depending on the type of heparin used) for at least 2 days or until the patient is ambulatory. Because respiratory function in the morbidly obese patient is greatly enhanced with the reverse Trendelenburg position, intermittent sequential venous compression boots should be used to counteract the increased venous stasis and the propensity for clotting. It is important that the intermittent venous compression boots be used before induction of anesthesia and throughout the operative procedure. Such boots are usually part of a standard preoperative protocol in gastric procedures for weight control; their use should not be unintentionally neglected in preparation for other elective or emergency procedures on morbidly obese patients. Patients with severe venous stasis disease (e.g., pretibial stasis ulcers or bronze edema) are at significantly increased risk for fatal PE.29 Prophylactic insertion of an IVC filter should be considered in these patients (as for patients with OHS and a high PAP). All patients should make every attempt to walk during the evening after operation. Bariatric surgery–induced weight loss will correct the venous stasis disease in most cases.29

Figure 4.

Venous stasis ulcers can be quite difficult to treat in a thin person; they are almost impossible to cure in a patient with morbid obesity [see Figure 4]. The most important goal in the management of these ulcers is weight loss, which almost invariably leads to healing of the ulcer, probably as a result of decreased IAP.29

Gallstones


Approximately one third of morbidly obese patients either have had a cholecystectomy or may have had gallstones noted at the time of another intra-abdominal operative procedure (e.g., a gastric operation for morbid obesity). Preoperative evaluation of the gallbladder may be technically quite difficult in morbidly obese patients because ultrasonography may fail to visualize gallstones. Intraoperative ultrasonography is probably much more accurate. Should symptomatic gallstones be present in a patient undergoing a gastric procedure for obesity, the gallbladder should be removed if the surgeon judges it safe to perform this additional procedure. If placement of an adjustable gastric band is contemplated, the cholecystectomy should be undertaken first and the indwelling device placed only in the absence of intra-abdominal bile spillage during the procedure.

In past studies, rapid weight loss led to the development of gallstones in 25 to 40% of patients who underwent GBP. The risk of cholelithiasis in this setting can be reduced to 2% by administering ursodeoxycholic acid, 300 mg orally twice daily.30 Laparoscopic cholecsytectomy at the time of laparoscopic gastric bypass can be technically challenging; consequently, many surgeons prefer to take an expectant approach to the gallbladder rather than complicate the bariatric procedure with a simultaneous cholecystectomy (unless cholecystectomy is clearly indicated in a particular patient).

Pseudotumor Cerebri


Pseudotumor cerebri is an unusual complication of morbid obesity that is associated with benign intracranial hypertension, papilledema, blurred vision, headache, and elevated cerebrospinal fluid pressures.31 It has been our experience that patients with pseudotumor cerebri are not at any additional perioperative risk and that CSF need not be removed before anesthesia and major abdominal operations. There is some theoretical concern that gastrointestinal contamination during GBP may cause shunt infection in patients who have been previously treated with indwelling shunts to relieve elevated CSF pressures. Successful weight reduction cures pseudotumor cerebri.3233

Degenerative Osteoarthritis


Degenerative osteoarthritis of the knees, hips, and back is a common complication of morbid obesity. Weight reduction alone may greatly reduce the pain and immobility that afflict these patients. In some cases, the damage may be so extensive that a total joint replacement is desirable; however, joint replacement in patients who weigh more than 250 lb is associated with an unacceptable incidence of loosening.34 Weight reduction by means of a gastric bariatric operation may be the most sensible initial approach, to be followed by joint replacement after weight loss if pain and dysfunction persist.

Operative Planning


Anesthesia in Patients with Respiratory Insufficiency


Morbidly obese patients can be intimidating to the anesthesiologist because they are at significant risk for complications from anesthesia, especially during induction. The risk is particularly great for obese patients with respiratory insufficiency. An obese patient often has a short, fat neck and a heavy chest wall, which make intubation and ventilation a challenge. If endotracheal intubation proves difficult, however, such a patient can usually be well ventilated with a mask. Awake intubation can be performed, with or without fiberoptic aids, but is quite unpleasant and rarely necessary.

It is extremely important that at least two anesthesia personnel be present during induction and intubation for patients with respiratory insufficiency of obesity. An oral airway is inserted after muscle paralysis with succinylcholine and sodium pentobarbital induction. One person elevates the jaw, hyperextends the neck, and ensures a tight fit of the mask, using both hands. To ensure adequate oxygen delivery, a second person compresses the ventilation reservoir bag, using two hands because of the resistance to air flow from the poorly compliant, heavy chest wall. After ventilation with 100% oxygen for several minutes, intubation is attempted. If difficulties are encountered within 30 seconds, the steps above should be repeated until the patient has been successfully intubated. A volume ventilator is required during operation. Placing the patient in the reverse Trendelenburg position expands total lung volume and facilitates ventilation22; however, the reverse Trendelenburg position increases lower-extremity venous pressure and therefore mandates the use of intermittent sequential venous compression boots. It is helpful to monitor blood gases through a radial arterial line or a digital pulse oximeter.

Laparoscopic Versus Open Approach to Bariatric Surgery


Bariatric surgical procedures, like most other general surgical procedures, have undergone a transition from an open approach to one that places more emphasis on minimally invasive or laparoscopic techniques. Laparoscopic GBP was first described in 1994 and became widely accepted in 1999, though it was not until 2004 that, according to a national audit of bariatric surgery performed at academic centers, the number of laparoscopic GBP procedures performed exceeded the number of open GBP procedures.

At present, the laparoscopic approach to GBP is favored because it achieves a comparable degree of weight loss while possessing some notable advantages over its open counterpart.21 Open GBP is performed through an upper midline incision, whereas laparoscopic GBP is performed through five or six small incisions. Abdominal wall retractors and mechanical retraction of the abdominal viscera, which are necessary for adequate exposure during an open procedure, are not required during a laparoscopic procedure, which makes use of gas insufflation (for pneumoperitoneum) and the effects of body positioning and gravity to facilitate intraoperative exposure. With the elimination of the large surgical incision and mechanical retraction, the laparoscopic GBP patient experiences less operative trauma, less postoperative pain, and fewer wound-related complications. In addition, laparoscopic GBP yields less impairment of immediate postoperative pulmonary function and a lower systemic stress response. A 2007 study of 22,422 patients who underwent Roux-en-Y GBP for treatment of morbid obesity compared the outcomes of laparoscopic procedures (n=16,357) with those of open procedures (n=6,065).35 The mean length of hospital stay was significantly lower in the laparoscopic group (2.7 days versus 4.0 days), as were the overall complication rate (7.4% versus 13.0%), the 30-day readmission rate (2.6% versus 4.7%), the in-hospital mortality (0.1% versus 0.3%), and the mean cost ($13,743 versus $14,585 [US]).

Choice of Surgical Procedure


The gastric operations performed for morbid obesity include both GBP procedures and gastric restrictive procedures (i.e., gastroplasty and gastric banding). Randomized, prospective trials have conclusively shown that GBP is as effective for weight control as the malabsorptive jejunoileal (JI) bypass is, while resulting in significantly fewer complications.3637 JI bypass is associated with a substantial incidence of both early complications (e.g., acute cirrhosis, electrolyte imbalance, and fulminant diarrhea)38 and late complications (e.g., cirrhosis, interstitial nephritis, arthritis, enteritis, nephrocalcinosis, and recurrent oxalate renal stones).39 If evidence of cirrhosis, renal failure secondary to interstitial nephritis, or other complications mandates reversal of a JI bypass, the patient, if not extremely ill, should be converted to a GBP; otherwise, all the lost weight is sure to be regained, and the obesity-related comorbidity will return. Admittedly, however, some patients have done well after JI bypass and do not need to have the operation reversed.

Several randomized, prospective trials have found that horizontal gastroplasty yields poorer results than GBP.40-42 Failure of horizontal gastroplasty has generally been attributed to technical causes (e.g., enlargement of the proximal pouch or the stoma or disruption of the staple line). Vertical banded gastroplasty (VBG) was developed in the hope that it would solve these technical problems and yield weight loss comparable to that seen after GBP without incurring the significant risk of iron, calcium, and vitamin B12 deficiencies associated with GBP. In the 1990s, a procedure known as adjustable silicone gastric banding was developed, which involved placement of a restrictive ring around the proximal stomach to create a small gastric pouch. In this restrictive procedure, which can be done laparoscopically in the vast majority of patients, weight loss can be enhanced and vomiting minimized by adjusting the ring diameter via transcutaneous access to the subcutaneous reservoir.

Although VBG and, presumably, other restrictive procedures appear to be excellent from a technical point of view,43 multiple randomized, prospective trials have found such approaches to be significantly less effective than standard GBP. In one comparison trial, patients addicted to sweets lost much more weight after GBP than after VBG because they experienced symptoms of dumping syndrome when ingesting sweets.44 The failure rate was high after VBG because these patients experienced no difficulties when eating candy or drinking nondietetic sodas. Subsequent randomized, prospective trials confirmed the superiority of GBP.4546 Furthermore, maintenance of successful weight loss after GBP appears to continue for as long as 14 years after operation: in the average patient, weight loss amounts to about two-thirds of excess weight at 1 to 3 years after operation, three-fifths at 5 years, and more than half in years 5 through 10.4748 It has been suggested that standard (i.e., proximal) GBP will fail in 10 to 15% of patients because these patients will frequently nibble on high-fat snacks (e.g., corn chips, potato chips, and buttered popcorn). Such patients may have to be converted to a combined restrictive and malabsorptive procedure, such as partial biliopancreatic diversion (BPD).49

The original BPD procedure involves hemigastrectomy and anastomosis of the distal 250 cm of intestine to the stomach; the bypassed small intestine is reanastomosed to the ileum 50 cm from the ileocecal valve. BPD with duodenal switch is a variant of the original procedure in which a linear gastric tube based on the lesser curvature is created (sleeve gastrectomy), with the pylorus left intact, and an ileal Roux limb is brought up for anastomosis to the proximal duodenum. BPD has been associated with a high incidence of deficiencies of fat-soluble vitamins, hypocalcemia-induced osteoporosis, and protein-calorie malnutrition.50 These nutritional deficiencies may be more common in the United States, where fat intake is high, than in many other countries. In Italy, for example, starch intake (as in pasta) probably outstrips fat intake; still, a number of Italian patients have had to be readmitted for parenteral nutrition and extension of the common absorptive intestinal tract because of refractory malnutrition. In some patients, it might be possible to convert a failed proximal GBP into a modified BPD with a 150 cm absorptive ileal limb (a procedure often referred to as distal GBP); however, these patients must also be monitored carefully for deficiencies of fat-soluble vitamins, for osteoporosis, and for malnutrition.

Superobese patients—defined as those whose weight is 225% of ideal body weight or greater or whose body mass index (BMI) is 50 kg/m2 or higher—will lose, on average, only about half of their excess weight, rather than two-thirds, after standard GBP. In these patients, a 150 cm proximal Roux-en-Y procedure (so-called long-limb GBP [see Open Proximal Gastric Bypass, Operative Technique, below]) may increase weight loss in the first few years after operation without causing an increase in nutritional complications.51

In choosing the appropriate surgical approach, it is important to take into account the tremendous surgical revolution that laparoscopy has brought about in the treatment of morbid obesity. Now that every operation performed to treat obesity can be done laparoscopically, laparoscopic bariatric surgery is not only common but, in many centers, predominant. For this reason, as well as because laparoscopic obesity treatment requires advanced technical skills, minimally invasive bariatric procedures have become a cornerstone of training for surgeons now learning laparoscopic surgery.

Vertical Banded Gastroplasty


Operative Technique


The first step in VBG is to make a circular stapled opening in the stomach 5 cm from the esophagogastric junction. A 90 mm bariatric stapler with four parallel rows of staples is then applied once between this opening and the angle of His. (At this point, according to Mason, the originator of the procedure, the volume of the pouch should be measured by means of an Ewald tube placed by the anesthetist; ideally, pouch volume should be 15 ml.)

Figure 5.

Next, a strip of polypropylene mesh is wrapped around the gastrogastric outlet on the lesser curvature and sutured to itself—but not to the stomach—in such a way as to create an outlet with a circumference of 5 cm for the small upper gastric pouch [see Figure 5a]. Some surgeons have used a stomal outlet 4.5 cm in circumference, but this smaller outlet has not led to better weight loss; in fact, many patients with the 4.5 cm outlet exhibit maladaptive eating behavior, drinking high-calorie liquids because meat tends to get caught in the small stoma.

Figure 5.

Silastic ring gastroplasty [see Figure 5b] is a variant of VBG that uses a vertical staple line and a stoma reinforced with Silastic tubing.

Complications


Complications of VBG include erosion of the polypropylene mesh used to restrict the gastroplasty stoma into the gastric lumen, enlargement of the pouch, stomal stenosis, reflux esophagitis, and mild vitamin deficiencies.52 To date, mesh erosion has been infrequently observed after VBG. Pouch enlargement is fairly common with horizontal gastroplasty but is much less likely to occur with VBG, in which the vertical staple line is placed in the thicker, more muscular part of the stomach. In addition, stomal diameter remains fixed with the mesh band. If mesh erosion, pouch enlargement, stomal stenosis, disabling GI reflux, or recurrent vomiting occurs, it is probably best to convert the patient to GBP. In particular, patients with a Silastic ring VBG may exhibit intractable vomiting of solid foods with no evidence of mechanical obstruction. In our experience, conversion of these patients to GBP yields good results and eliminates the vomiting problem. Finally, vitamin deficiencies can usually be prevented by having VBG patients take a standard multivitamin daily for life.

Laparoscopic Adjustable Gastric Banding


Gastric banding is another form of gastroplasty, in which a synthetic band is placed around the stomach just below the esophagogastric junction. In several series, gastric banding has yielded markedly variable results with respect to achievement of weight loss. Furthermore, it has been associated with slipping or kinking of the banded stoma, obstruction at the band, and intractable vomiting.

Figure 6.

Laparoscopic adjustable gastric banding (LAGB) is significant advance over open gastric banding procedures, primarily because of the adjustability of the band. Open gastric banding procedures have used a variety of materials to constrict the gastric lumen and carry a recognized risk of postoperative nausea and vomiting that do not respond to any treatment short of reoperation. The adjustable gastric bands available for use in LAGB [see Figure 6] are silicone devices with an inflatable reservoir that can be inflated or deflated postoperatively through a subcutaneous port placed deep in the abdominal wall for percutaneous access. Saline is injected into or withdrawn from the reservoir to adjust gastric luminal diameter. These diameter changes can be measured by means of barium contrast evaluation, but currently, most adjustments are made without x-ray guidance. If intractable vomiting develops, saline can be removed from the band to alleviate the problem; similarly, if the patient fails to lose weight after operation, additional saline may be injected into the band to narrow the gastric lumen further.

Use of the laparoscopically placed adjustable gastric band (Lap-Band, Allergan Corp., Irvine, CA) was approved by the US Food and Drug Administration in June 2001. Key data on safety and effectiveness were provided by a prospective, single-arm trial involving 299 patients at eight centers in the United States. In this study, patients who completed 36 months of follow-up achieved a mean reduction in BMI of 39% and a mean overall loss of 18% of baseline body weight. However, 28% of patients lost less than 10% of their initial body weight (a clear definition of failed weight loss). More than half (62%) of these patients lost more than 25% of their excess weight. Most patients (76%) experienced at least one adverse event, and 33% of patients required removal of the banding system.

Subsequent studies have yielded similar results. A 2007 review of two multicenter prospective, single-arm surgical trials evaluating a total of 485 patients who underwent placement of a gastric band (92% laparoscopically) between June 1995 and June 2001 suggested that the procedure was as effective as was previously believed.53 The change in mean BMI (kg/m2) was 38 to 47% at 1 year and 39% at 3 years. The percentage of initial body weight lost was 17 to 18% at 1 year and 18% at 3 years. Similarly, most patients (66 to 76%) experienced upper GI symptoms at 1 year. In one of the trials, 33% of the patients (96/292 patients) had had their bands removed at 9 years, either because of complications or because of inadequate weight loss.53

In a 2006 study comparing outcomes, LAGB proved to be just as safe as, cheaper than, and almost as effective as laparoscopic Roux-en-Y gastric bypass (LRYGB).54 This retrospective review of 590 bariatric procedures (120 LRYGB, 470 LAGB) performed between November 2000 and July 2004 suggested that both operating time and duration of hospitalization were significantly shorter in LAGB patients. Complication rates and reoperation rates were similar in the two groups. Patients who underwent LRYGB initially lost weight more rapidly: their mean percentage of excess body weight lost (%EBWL) was 65% during postoperative year 1, compared with 39% for LAGB patients. Thereafter, weight loss slowed, remaining nearly unchanged at 3 years (63%). Patients who underwent LAGB initially lost weight more slowly, but the ongoing weight loss was continuous, eventually approaching that of LRYGB. At 3 years, the %EBWL for LAGB patients was 55%.

Operative Technique


LAGB is performed by using a five-port technique. Initial abdominal access is obtained via a supraumbilical trocar, and the remaining ports are placed sequentially along the right and left costal margins. The liver is retracted via the subxiphoid port, and the proximal stomach is visualized with a laparoscope inserted through the umbilical port.

Subsequent steps are done according to the pars flaccida technique. A retrogastric tunnel for band insertion is created at the posterior confluence of the diaphragmatic crura in a plane of dissection that is easily developed with minimal blunt dissection after electrocauterization of the peritoneal membrane. This tunnel is placed cephalad to the posterior peritoneal reflection, so that the free space of the lesser sac posterior to the stomach is not entered. Additional dissection is then carried out laterally at the angle of His to open the peritoneum and start clearing a plane behind the proximal stomach.

Figure 7.

A specially designed implement is inserted behind the stomach from the lesser curvature to the angle of His and used to grasp the tubing of the banding device and pull it around the stomach. The banding device is then locked into place at the chosen location on the proximal stomach [see Figure 7]. Gastrogastric sutures are placed to create a tunnel of stomach overlying the banding device—but not the buckle—so as to hold the device in position. The band tubing is brought through the left midclavicular trocar port, which is placed via the left midclavicular line subcostal trocar incision and fixed to the abdominal wall fascia with sutures. The tubing is connected to the reservoir, which is filled with saline.

Troubleshooting


It is essential to place the band properly during the initial procedure. The results to date suggest that the proximal pouch must be very small to optimize weight loss. In addition, proper placement minimizes—though it does not eliminate—the risk of band slippage and the complications thereof.

Several techniques have been suggested for posterior fixation of the band, but they are more difficult than anterior fixation techniques. With the pars flaccida technique, posterior fixation of the band is not necessary to prevent band slippage. Anterior fixation, however, is routinely performed, with interrupted sutures of nonabsorbable material placed between the distal and the proximal stomach to allow tissue to be apposed over the band and held in place.

Although LAGB appears easier than many of the procedures done to treat obesity, there is a definite learning curve. A number of surgical misadventures have been reported, including gastric perforation, splenic injury, and malpositioning of the band.

Complications


Figure 8.

Band slippage (anterior, posterior, or concentric) may occur even after proper placement, resulting in intolerance of oral intake and vomiting. Such complaints are an indication for an upper GI series, which usually reveals dilatation of the proximal pouch and rotation of the band [see Figure 8]. Initial treatment consists of evacuating all saline from the band. Frequently, however, the proximal pouch does not return to its normal size, and symptoms recur or fail to resolve. Laparoscopic or open revision of the banding procedure is then required; if the patient also has not lost a sufficient amount of weight, conversion to GBP may be recommended. It is noteworthy that band erosion into the stomach, a not infrequent complication of the use of mesh in VBG or in the Angelchik prosthesis for gastroesophageal reflux treatment, has not been frequently reported. Longer follow-up is necessary to evaluate the true extent of this risk.

As after any form of gastroplasty, the patient may fail to lose weight or may regain lost weight. Inappropriate eating behaviors (e.g., intake of high-calorie sweets) are the most likely cause. If obesity-related comorbid conditions persist, conversion to proximal GBP is appropriate.

Outcome Evaluation


How successful LAGB is at achieving weight loss over the long term remains unclear. The adjustability and reversibility of the operation, as well as the decreased disability that results, make it attractive to both patients and physicians. The procedure appears to avoid some of the major postoperative complications associated with open GBP (e.g., incisional hernia, marginal ulcer, and stomal stenosis). Band slippage remains a major postoperative concern, however, though the incidence of slippage does appear to decrease as the surgeon’s experience with the procedure increases. More significant, there appears to be a high frequency of failed weight loss—as high as 15 to 20% of all patients undergoing the procedure and possibly even higher. European data confirm that there is a significant failure rate but also suggest that the remaining patients achieve a degree of weight loss approaching that seen with proximal GBP. Whether these reports will withstand the scrutiny of long-term follow-up remains to be seen.

Open Proximal Gastric Bypass


Figure 9.

Proximal GBP results in greater weight loss than the gastric restrictive procedures (see above) and carries a lower incidence of weight regain; consequently, it is often considered the gold standard for bariatric surgery. Compared with the version of GBP performed at our institution, the original GBP created a much larger proximal gastric pouch and a much wider anastomotic opening, and it was often associated with inadequate weight loss. In the later version, three superimposed 55 or 90 mm staple lines are placed across the proximal stomach in such a way as to create a gastric pouch no larger than 30 ml with a Roux limb at least 45 cm long and a stoma no larger than 1 cm [see Figure 9]. This anatomic situation is largely replicated when GBP is done laparoscopically, but an isolated gastric pouch is created with stapled transection of the stomach.

Operative Technique


Step 1: Initial Incision and Abdominal Exploration

Once the patient is anesthetized, the abdomen receives a thorough, careful cleansing with povidone-iodine and is draped in a sterile fashion. An upper midline incision is made and extended through the fascia alongside the xiphoid process to facilitate cephalad exposure. The incision is routinely carried down to the supraumbilical area. The deep layer of subcutaneous fat can often be separated bluntly with aggressive lateral traction applied by the surgeon and the assistant, and the midline usually can then be identified for fascial incision. The electrocautery is used to enter the abdominal cavity, and a thick layer of subfascial preperitoneal fat is often encountered before entry into the peritoneal cavity. Abdominal exploration is undertaken in every patient, including examination of the liver for possible signs of liver disease. Other incidental findings may become apparent as well.

Troubleshooting

Unexpected significant liver disease is occasionally discovered at the time of operation. If the patient has cirrhosis without portal hypertension, one should perhaps proceed with bypass if the patient’s comorbid conditions make it mandatory; liver transplantation carries increased risk in morbidly obese patients. The gallbladder should be palpated for gallstones, which, if found, may be an indication for cholecystectomy at the time of the bypass procedure. If there are no visual or palpable gallbladder abnormalities, intraoperative ultrasonography may be used to examine the gallbladder.

It is not unusual to discover other previously unrecognized conditions during GBP, primarily because symptoms may not be obvious in morbidly obese patients and because their large size tends to make radiologic imaging difficult or even impossible. For example, intraoperative discovery of pelvic cysts and tumors is not uncommon in obese female patients. Such lesions may be excised during GBP; on occasion, if they appear benign and their location prevents safe excision, they may be managed with careful follow-up.

Step 2: Mobilization of Esophagus

The bypass procedure itself is begun by mobilizing the distal esophagus and encircling it with a soft rubber drain 0.5 in. in diameter. The gastrohepatic omentum is bluntly entered at a point overlying the caudate lobe, with care taken to look for and avoid injury to an aberrant left hepatic artery. The phrenoesophageal ligament overlying the anterior and lateral distal esophagus is sharply incised to facilitate subsequent blunt mobilization of the distal esophagus. To prevent esophageal injury, the nasogastric tube is carefully palpated within the lumen of the esophagus during mobilization, and blunt dissection proceeds widely around this important landmark. Laterally, dissection must be at the level of the esophagus or higher.

Troubleshooting

If dissection is too low laterally, it may result in blunt injury to the short gastric vessels, bleeding, and the need for urgent splenectomy, which is no easy task in a morbidly obese patient. In addition, it may lead to creation of an inappropriately large pouch by keeping the surgeon from recognizing that some of the stomach is above the level at which the encircling rubber drain is placed.

Step 3: Division of Mesentery and Dissection around Stomach

Once the esophagus is mobilized, the assistant’s left hand is placed through the gastrohepatic omental opening behind the stomach wall on the lesser curvature. The space between the first and second branches of the left gastric artery is then identified as a landmark for location of the gastric staple line, both to ensure that the pouch created is no larger than 30 ml and to prevent injury to the left gastric artery, which usually runs cephalad to this location. With the surgeon’s posterior finger pressing anteriorly to place tension on the tissue, a fine-tip right-angle clamp and the electrocautery pencil are used to divide the mesentery carefully at this level immediately alongside the stomach wall so as to create a mesenteric opening that will admit a large right-angle clamp.

Figure 10.

The avascular tissue on the posterior wall of the stomach is then bluntly dissected between the opening in the gastrohepatic omentum and the lateral angle of His, which is identified by the encircling rubber drain. The blunt tip of a large 28 French red rubber tube is placed behind the stomach in a medial-to-lateral direction along this dissected path to encircle the stomach [see Figure 10]. The open end of the red rubber tube is subsequently brought through the previously created mesenteric opening with a large right-angle clamp. The stomach is now ready for stapling, and the red rubber tube serves as a guide for introduction of the stapler. At this point, all intraluminal tubes and devices (e.g., the nasogastric tube and the esophageal stethoscope) are removed from the esophagus by the anesthetist.

Troubleshooting

When a tube is inadvertently stapled within the stomach, excising it from the nontransected gastric staple line can be a technical challenge. To remove the stapled tube, it is generally necessary to use a stapler to transect the stomach, thereby creating the potential for significant injury to the gastric tissue unless the transection is precisely superimposed over the previous staple line. The tube can then be excised from each side (proximal and distal) of the divided gastric staple lines.

Step 4: Creation and Mobilization of Roux Limb and Jejunojejunostomy

The ligament of Treitz is identified, and the jejunum is measured to a point 45 cm beyond the ligament—or somewhat more distally to enhance mobilization of what will become the Roux limb if the mesentery appears foreshortened—at which point the jejunum is divided with a stapler. An 8 to 12 cm segment of jejunum may be resected at this point to create a larger mesenteric defect, which should facilitate mobilization of the limb to the proximal stomach. Mesenteric dissection is carried posteriorly in fat with the sequential application of clamps until further dissection appears either unnecessary for mobilization or unwise (i.e., likely to cause mesenteric vascular injury or ischemia of the Roux limb).

A side-to-side jejunojejunostomy is then created with a 60 mm linear stapler at least 45 cm beyond the initial point of jejunal division for standard proximal GBP. Some surgeons perform this anastomosis 150 cm downstream for the long-limb modification of the procedure used in superobese patients [see Operative Planning, Choice of Surgical Procedure, above] or even further distally for the distal GBP modification, which greatly enhances malabsorption. It is important not to narrow the efferent lumen at the jejunojejunostomy site, particularly with the longer-limb modifications, in which the lumen at the distal end of the Roux limb may be quite small. The enterotomies made to allow placement of the stapler can usually be closed with a 55 mm stapler loaded with 3.5 mm staples; however, if stapling would cause undue narrowing of the lumen, the closures should be handsewn instead.

Troubleshooting

It may be preferable to mobilize the Roux limb before committing to stapling the stomach so that it can be determined whether the limb can be extended to reach the proximal stomach without being placed under tension. In those rare cases in which the mesentery is too foreshortened to permit the limb to reach the proximal stomach, it is advisable to change the procedure to VBG or gastric banding rather than create a gastrojejunal anastomosis under tension and thereby incur the increased risk of leakage.

Step 5: Gastric Stapling and Gastrojejunostomy

Figure 11.

The Roux limb is brought through the mesentery of the transverse colon with blunt dissection and then brought up to the proximal stomach. The 55 or 90 mm stapler, loaded with 4.8 mm staples, is guided behind the stomach by inserting its open-mouthed end into the lumen of the previously positioned red rubber tube. Once it is determined that the staple line will reach completely across the stomach and that the stomach is not folded on itself, the stomach is stapled three times in such a way that the three staple lines are superimposed [see Figure 11].

Figure 12.

A 1 cm anastomosis is created between the proximal stomach pouch and the Roux limb. We prefer a handsewn anastomosis for this procedure, using an outer layer composed of interrupted 2-0 or 3-0 silk sutures and an inner layer composed of a continuous absorbable 3-0 polyglycolic acid (Dexon) suture. When the posterior aspect of the anastomosis is complete, a 30 French dilator is placed orally by the anesthetist and is guided through the anastomosis by the surgeon to ensure that the stoma has the appropriate diameter [see Figure 12]. The anterior aspect of the anastomosis is then completed.

Troubleshooting

A significant concern for many bariatric surgeons has been a high incidence of staple line disruption causing failed weight loss or weight regain; in one series, the incidence of such disruption was 35%. To minimize this risk, some surgeons advocate transecting the stomach. Currently, this step is routinely carried out as part of a laparoscopic GBP. In an open GBP, one may transect the stomach either by applying a linear cutting stapler with 3.5 or 4.8 mm staples (depending on the estimated gastric wall thickness) or by inserting two parallel noncutting transverse anastomosis (TA)–90 staplers and cutting between them with a scalpel after the staplers are fired. Other surgeons, however, prefer to leave the stomach undivided and oversew the staple line. In our version of open GBP, we find that placing three or four precisely superimposed staple lines reduces the incidence of staple line disruption to less than 2%.

Another advantage of gastric transection besides reduction of staple line disruption is that it allows the Roux limb to be brought up to the gastric pouch via a retrocolic and retrogastric tract, which is substantially shorter and places less tension on the limb. This approach is particularly helpful in severely obese patients with a fatty and foreshortened mesentery, in whom it is difficult to free the Roux limb sufficiently to reach the proximal stomach without tension. The possibility that gastric transection may prove helpful in a specific patient is another reason why it is advisable to delay stapling the stomach until the Roux limb has been mobilized.

Step 6: Assessment of Anastomosis

When the entire anastomosis is complete, the dilator is removed and an 18 French nasogastric tube is advanced by the anesthetist while the tip is carefully guided through the anastomosis by the surgeon. The Roux limb is occluded with the assistant’s left hand or with an atraumatic intestinal clamp, and the esophagus is occluded by placing tension on the rubber drain surrounding it while the anesthetist injects a series of 10 ml aliquots of methylene blue dye through the nasogastric tube to determine whether the anastomosis is leaking. A total of 30 to 60 ml of methylene blue must usually be injected; lesser amounts will not stress the suture line enough to constitute an adequate test. Alternatively, the anastomosis can be tested by performing intraoperative gastroscopy. If leakage is present, the air insufflated during the procedure will be visible bubbling from the leaking areas when the air-distended anastomosis is submerged in irrigation fluid.

Troubleshooting

When an intraoperative leak is identified, the area of leakage should be oversewn with silk sutures until injection of additional methylene blue dye via the nasogastric tube yields no further leakage. The most difficult area to repair is the posterior suture line, which is quite close to the gastric staple line. Posterior leaks are usually repaired by reinforcing the posterior suture line with additional sutures between the excluded stomach and the jejunal limb; often, the entire posterior suture line is oversewn. In addition, a viable pedicle of omentum may be mobilized and placed around the anastomosis for further reinforcement. Closed suction drains may also be placed in this area, both to detect possible postoperative leakage and to control a postoperative leak or fistula.

Finally, a gastrostomy tube may be placed in the excluded portion of the stomach. This measure provides postoperative decompression, which should prevent the development of undue tension on the Roux limb as a result of gastric distention. In addition, it establishes a route for enteral feeding if a fistula develops. Fortunately, such fistulas are rare. When they do occur, they often heal if (1) they are well drained, (2) there is no distal obstruction or local abscess, and (3) the patient is receiving nutritional support with no oral intake. A gastrostomy tube should also be placed in the distal gastric pouch when extensive adhesions from a previous procedure or a difficult gastric reoperation increase the risk of postoperative gastric distention.

Step 7: Closure

When the absence of leakage is confirmed or when any leaks identified have been controlled, the tip of the nasogastric tube may be positioned further down in the Roux limb and left to continuous suction overnight. All mesenteric defects—at the jejunojejunostomy, at the mesocolon, and behind the Roux limb (Peterson hernia)—are then closed to prevent an internal hernia. The abdominal fascia is reapproximated with a continuous double-looped No. 2 suture, subcutaneous tissues are irrigated with a crystalloid solution, and the skin is closed with skin staples. No subcutaneous sutures or drains are used in routine cases.

Complications


Proximal GBP is associated with a significant incidence of stomal stenosis and with marginal ulcer.55 The former responds to endoscopic stomal dilatation, and the latter usually responds to proton pump inhibitor (PPI) therapy. Addition of sucralfate to this regimen may be helpful. The risk of marginal ulcer appears to be increased in smokers and in patients who consume nonsteroidal anti-inflammatory drugs (NSAIDs). We routinely discourage NSAID use after GBP. Perforation of the proximal gastric pouch, probably arising from perforation of a deep ulcer, has been seen with administration of high-dose NSAIDs or with untreated ulcer diathesis.

Iron, vitamin B12, and folic acid deficiencies may occur but can usually be corrected with oral supplementation52; accordingly, GBP patients, like VBG and gastric banding patients, should be advised to take a multivitamin daily for life. Compared with gastroplasty and gastric banding, GBP results in significantly lower serum hemoglobin and iron concentrations. This is primarily a problem in menstruating women. All menstruating women who have undergone GBP should be treated prophylactically with supplemental oral ferrous sulfate, 325 mg/day. As many as six iron tablets a day may be required if menstrual bleeding is heavy. Hormonal therapy to control or temporarily eliminate menses may be helpful. On occasion, intramuscular iron injections or, rarely, hysterectomy may be necessary. The risk of vitamin B12 deficiency is higher after GBP than after gastroplasty or gastric banding, but this condition can be prevented with supplemental oral vitamin B12, 500 mg/day. A few patients may require (or prefer) monthly B12 injections, which they can learn to administer themselves.

Concerns have been expressed that GBP can lead to other divalent cation deficiencies. Our group has not encountered zinc deficiencies 5 to 9 years after GBP, though we have observed calcium deficiencies leading to osteoporosis, which may take many years to become manifest and may not be biochemically evident because of normal serum calcium levels. It is therefore recommended that all GBP patients take oral calcium supplements. Some may require vitamin D supplementation. Magnesium deficiencies should be treated with MgSO4 supplementation.

Although nutritional deficiencies do not appear to be a greater problem with long-limb GBP than with standard proximal GBP, monitoring patients for possible malabsorption of the fat-soluble vitamins A, D, and E after long-limb GBP is advisable.

BPD may be associated with all of the complications seen after GBP. In addition, patients who undergo BPD may experience diarrhea, severe protein malnutrition (manifested as hypoalbuminemia), and deficiencies of vitamins A (manifested as severe night blindness), D (manifested as severe osteoporosis), and E.50 Hypoalbuminemia may respond to oral pancreatic enzymes but often must be treated with total parenteral nutrition. In some patients, it may prove necessary to lengthen the absorptive intestinal tract from 50 cm to 200 cm.

Outcome Evaluation


A series of 672 open proximal GBP procedures reported a 1.2% incidence of anastomotic leakage with peritonitis, a 4.4% incidence of severe wound infections (defined as infections serious enough to delay hospital discharge), an 11.4% incidence of minor wound infections and seromas (which were easily treated at home), a less than 1% incidence of gastric staple line disruption with the use of three superimposed applications of a 90 mm linear stapler, a 15% incidence of stomal stenosis, a 13% incidence of marginal ulcer, a 16.9% incidence of incisional hernia, and a 10% incidence of cholecystitis necessitating cholecystectomy.47 Gallstones developed in 32% of the GBP patients who had a normal intraoperative gallbladder sonogram within 6 months of surgery, and sludge was observed in another 10%. In a multicenter randomized, prospective trial, the incidence of gallstones within 6 months of GBP was reduced from 32% to 2% by giving patients ursodeoxycholic acid, 300 mg twice daily.56 Gallstone formation beyond 6 months is uncommon. The operative mortality in this series was less than 1%. Patients with respiratory insufficiency of obesity had an operative mortality of 2.2%, whereas those without pulmonary dysfunction had an operative mortality of 0.4%.

Neither the data from this randomized, prospective trial nor the data from selective studies support the contention that VBG is safer than GBP. Although GBP includes one more anastomosis than VBG, complications such as leaks and peritonitis occur with both operations. A common criticism of GBP is that it is difficult to evaluate the distal gastric pouch and duodenum after the operation. Such evaluation, however, can be done in 75% of patients by means of retrograde passage of an endoscope into the duodenum and the stomach and in other patients by means of percutaneous distal distention gastrography. Bleeding from either the distal gastric pouch or a duodenal ulcer is rare. Gastric mucosal metaplasia of the bypassed portion of the stomach may occur in some 5% of patients after retrograde endoscopy, a finding that has raised concerns regarding the risk of carcinoma arising at that location. To date, however, although many thousands of these procedures have been performed over the past four decades, few cases of cancer in the bypassed stomach have been reported.

Laparoscopic Gastric Bypass


Laparoscopic GBP is currently the most popular bariatric procedure in the United States, both because of the rapid weight loss it achieves and because of the strong overall surgical trend toward minimally invasive approaches. As noted [see Operative Planning, Laparoscopic versus Open Approach to Bariatric Surgery, above], laparoscopic GBP achieves the same weight-loss results as open GBP but yields less pain, reduced disability, and a shorter duration of hospitalization. Physiologically, laparoscopic GBP results in less operative trauma than open GBP, less impairment of pulmonary function, and a less pronounced stress response. In addition, the laparoscopic technique is associated with lower incidences of major wound infections and incisional hernias. Accordingly, we recommend laparoscopic GBP over any other bariatric procedure.

Laparoscopic GBP poses significant technical challenges, even for surgeons with advanced laparoscopic skills. Most of the variations seen at different institutions are related to various techniques for creation of the gastrojejunal anastomosis, with some groups using a circular stapler, others a linear stapler, and still others a handsewn technique. The anvil of the circular stapler may be placed within the proximal gastric pouch either by means of flexible upper GI endoscopy, through an approach similar to the snare-and-wire technique used for placement of a percutaneous endoscopic gastrostomy (PEG) tube [see 5:18 Gastrointestinal Endoscopy], or by means of a gastrotomy of the stomach before pouch creation for intra-abdominal anvil placement, followed by staple closure of the gastrotomy. Peroral placement of the stapler’s anvil can be problematic: even the small 21 mm anvil is hard to pass through the proximal esophagus in some patients. To facilitate esophageal passage, a ‘flip-top’ anvil design has been introduced. With this design, a 25 mm anvil can generally be passed without undue difficulty, thereby lowering the risk of postoperative stenosis. We routinely use the linear stapling method to create the gastrojejunal anastomosis; it is easier than circular stapling in this setting, and there is no risk of esophageal trauma from anvil passage.

Operative Technique


Step 1: Initial Access and Trocar Placement

Figure 13.

Initial access to the abdomen is obtained through a small left subcostal incision. Gas is insufflated into the abdomen via a Veress needle to a pressure of 15 mm Hg; on occasion, a pressure of 18 to 20 mm Hg may be necessary. A dilating 5 mm trocar is then placed in this location. Many surgeons use commercially available trocars that allow direct vision through the scope during passage of a 12 mm trocar. We encourage preinsufflation of the abdominal cavity before such a device is employed so as to enhance identification of the peritoneal sac and avoid organ injury. Additional trocars are placed in specific locations [see Figure 13]. The liver is retracted with a metal Nathanson liver retraction device anchored to the bed, which is inserted after a 5 mm sharp trocar is used to develop a tract into the abdominal cavity in the subxiphoid position and removed. If the left lateral section of the liver is very large (as in patients with steatosis), additional liver retractors may be necessary.

On occasion, the enlarged fatty falciform ligament must be dissected from the anterior abdominal wall with an ultrasonic scalpel or a bipolar cautery device. The advent of long laparoscopic graspers, staplers, and energy dissectors has facilitated operating on the proximal stomach and allows the use of trocar locations away from the abdominal midline. A 12 mm port placed in the right paramedian position serves as the surgeon’s primary operative port; two lateral subcostal 5 mm ports allow both surgeon and assistant to employ two-handed techniques for the entire procedure. In most patients, a 45° angled viewing 5 mm laparoscope is inserted via a supraumbilical 5 mm trocar. A longer 10 mm scope may enhance visibility, particularly for especially heavy or tall patients, in whom a 5 mm scope may be subject to damage from excessive torque. High-definition imaging improves visualization during the procedure.

Step 2: Creation of Roux Limb and Jejunojejunostomy

In most reports of laparoscopic GBP, regardless of how the gastrojejunostomy is done, the approach to creating the Roux limb is essentially the same. The patient is placed in a supine position, and graspers are used to bring the omentum upward into the upper abdomen so that the transverse colon and the underlying mesocolon are exposed. Graspers are then placed on the transverse mesocolon and used to elevate it anteriorly so that the ligament of Treitz is exposed. The position of the ligament of Treitz is confirmed by careful manipulation and verification that the bowel is attached to the retroperitoneum at this location; this may be a more difficult task in patients who have previously undergone abdominal procedures.

With the help of a measuring instrument, the small bowel is measured to a point 50 cm from the ligament of Treitz, where it is transected with the endoscopic gastrointestinal anastomosis (GIA) stapler, loaded with 2.5 mm staples. A 0.5 in. Penrose rubber drain may be sutured to the cut end of the Roux limb so that it is not confused with the other cut end of the bowel. The Roux limb is then measured to a length of 45 to 60 cm. (As in open GBP, the Roux limb should be significantly longer—up to 150 cm—if the long-limb modification is being performed.) The afferent side of the previously transected small bowel is then attached with a simple absorbable suture to the proposed jejunojejunostomy site on the Roux limb. Intracorporeal suturing is facilitated by using an automatic suturing device. Positioning is important: the afferent limb should be kept toward the mesocolon (toward the left side of the surgeon’s field of view), with the Roux limb brought toward the surgeon (visualized on the right side of the operative field of view).

Two holding sutures are placed, one proximal to the proposed site of the enterotomy for the anastomosis and the other distal to this site, to help manipulate the bowel onto the stapler. A small (1 cm) enterotomy is made with the electrocautery in each of the two adjacent bowel limbs in preparation for passage of the linear stapler, loaded with 2.5 mm staples in a 60 mm cartridge, into the bowel. The stapler is then fired, creating a 60 mm side-to-side anastomosis. A suture is placed at the staple line to elevate the corner of the anastomosis for subsequent stapled closure of the defect, and a third holding suture is placed at the midpoint of the enterotomy. The three sutures are grasped to facilitate closure of the enterotomy by another application of the linear stapler, with great care taken to avoid narrowing at the junction of the Roux limb and the large common channel of the anastomosis. To facilitate closure without obstruction, all three knots should be visible above the stapler, but tissue amputation should be minimal. Once completed, the anastomosis is inspected both for integrity and for possible narrowing. At this point, cephalad traction is applied to the remaining suture (in the medial position) to facilitate placement of a continuous suture to close the mesenteric defect.

Some surgeons prefer to create a loop gastrojejunostomy, thereby avoiding the technical challenges associated with creating a Roux limb laparoscopically. This approach was abandoned years ago in open GBP because of the unacceptably high incidence of postoperative bile reflux and the increased severity of complications resulting from high output of digestive juices when leakage occurred at the gastrojejunal anastomosis; it should be abandoned in laparoscopic GBP as well. The practice of using such suboptimal methods for the purpose of performing this complex operation more expeditiously is poorly conceived.

Step 3: Passage of Roux Limb

Next, the greater omentum is divided from its free edge to its junction with the transverse colon so that the limb can be brought up in an antecolic fashion between the divided halves of the omental ‘apron,’ which reduces tension on the limb. Alternatively, a retrocolic, retrogastric tunnel may be created so that the Roux limb can be advanced to the proximal stomach for anastomosis. The retrocolic, retrogastric approach is superior if the antecolic approach appears to be placing undue tension on the limb as it is advanced to the proximal pouch. For retrocolic passage, the ligament of Treitz is identified by lifting the transverse mesocolon anteriorly, and a spot 1 to 2 cm anterior and to the left of the ligament is chosen as the starting point for electrocautery dissection. The middle colic artery should be medial to this point of entry into the mesocolon. The goal of the dissection is to identify the posterior wall of the stomach, which may be difficult in patients who are extremely obese, have a fatty, foreshortened mesocolon, or have previously undergone abdominal surgery.

Once the stomach is visible, it is grasped and elevated through the mesocolic window, and the end of the Penrose drain is grasped and brought through the mesocolic defect into the lesser sac. When the Penrose is in place in the lesser sac, the omentum is pulled down from the epigastrium to allow visualization of the stomach, and the patient is placed into a steep reverse Trendelenburg position. The Penrose drain will be exposed when the cutting stapler is employed to create the proximal gastric pouch, and care must be taken to avoid stapling the drain.

There has been some controversy regarding the relative merits and deficiencies of antecolic and retrocolic techniques for passage of the Roux limb. The original open GBP often made use of a retrocolic, antegastric approach. The subsequent evolution of gastric transection methods led to a retrocolic, retrogastric approach to limb passage, which decreased the distance the limb had to travel to reach the proximal stomach and reduced the tension placed on the gastrojejunal anastomosis. Eventually, the antecolic, antegastric technique was introduced; this approach became popular with laparoscopic surgeons because it usually made the procedure faster to perform. The advantages of this technique were confirmed by a study that prospectively compared 33 consecutive patients who underwent retrocolic LRYGB with 33 who underwent antecolic LRYGB over a 3-year follow-up period.57 The results suggested a similar decrease in BMI (from 45 kg/m2 before operation to 34 kg/m2 at 36 months) and a similar incidence of complications (e.g., internal hernias and strictures). Operating time was significantly shorter for the antecolic procedure (mean, 188 minutes) than for the retrocolic procedure (mean, 219 minutes).

Other issues that have been raised in comparing the two techniques include the risk of internal hernias through mesenteric defects and the various risks inherent in an anastomosis performed under tension (e.g., leakage, ischemic stricture, and ulceration). In addition, retrocolic passage of the Roux limb during LRYGB leaves a short segment of the limb that is not easily visualized; unrecognized twisting of this segment may occur and lead to postoperative obstruction. A 2007 study that evaluated 754 LRYGB patients (300 retrocolic, 454 antecolic) over a median follow-up period of 16 months found that the rate of complications (e.g., surgical exploration for intestinal obstruction and internal hernias) was higher after the retrocolic procedure.58 Another 2007 study that evaluated 353 LRYGB patients (218 retrocolic, 135 antecolic) over a mean follow-up period of 28 weeks found that the incidence of postoperative gastrojejunal leakage was higher after the antecolic procedure.59 These varying study outcomes suggest that a prospective, randomized trial will ultimately be required to determine whether antecolic Roux limb passage is superior or inferior to retrocolic passage with respect to outcomes or complication rates.

Both techniques continue to be employed today, and the current evidence is not sufficient to support a blanket recommendation favoring one over the other. We would maintain, however, that if undue tension is evident when the Roux limb is passed via the antecolic pathway, it is advisable to switch to the shorter route available via the retrocolic, retrogastric pathway.

Step 4: Dissection around Stomach and Creation of Gastric Pouch

Figure 14.

The mesentery of the lesser curvature is transected with a bipolar electrocautery device to provide hemostasis. Despite the potential for problems with transection of the neurovascular bundle, no clear evidence of such problems has been found in hundreds of procedures performed with this access technique. Dissection posterior to the stomach is performed in the avascular free plane of the lesser sac. Additional dissection along the lesser curvature is not recommended, because it may increase the devascularization of the pouch. Further dissection is done at the angle of His to create a connection with the posterior gastric space. A linear endoscopic GIA stapler loaded with 3.5 mm staples in 60 mm cartridges is then employed in sequential firings to transect the stomach and create the proximal gastric pouch; three or four firings are usually necessary [see Figure 14]. When revisional bariatric surgery is performed on the previously operated-on stomach or when buttressing materials are used to reinforce the staple line, a staple height of 4.8 mm is usually required.

Step 5 (Circular Stapling): Placement of Stapler in Gastric Pouch

As noted (see above), surgeons use several different techniques to complete the gastrojejunal anastomosis. If a linear stapler is used, the next step is gastrojejunostomy [see Step 6b (Linear Stapling), below]. With the technique reported in Wittgrove’s original description of the procedure,60 however, in which the anvil of a circular stapler is passed down the patient’s esophagus, the next step in the procedure is to have an assistant perform flexible upper GI endoscopy of the gastric pouch. The pouch wall is transilluminated by the endoscope light, and a site is chosen for the gastrojejunostomy. The endoscopist then places an endoscopic snare, which can be seen pressing against the tissue of the pouch wall. A small opening is made in the pouch with the electrocautery scissors so that the snare can be pushed through the gastric wall at this location. The snare is then used to grasp a wire placed through a needle across the abdominal wall in the left abdomen, and snare and wire are withdrawn through the mouth along with the scope.

The anvil of the stapler is attached to the end of the wire that was drawn through the mouth, and the surgeon pulls on the other end of the wire to deliver the anvil through the mouth, down the esophagus, and into the gastric pouch, where it can be visualized. The electrocautery is used to enlarge the opening in the gastric wall slightly, and the stem of the stapler is then brought through the gastrotomy. To prevent anastomotic leakage after stapling, the gastrotomy should be no larger than the diameter of the stem; if it is too large, it can be closed around the stem by placing one or two simple sutures.

An alternative technique for anvil placement in the pouch involves making a gastrotomy distal to the planned staple line before transection of the stomach. After the anvil is introduced into the abdomen via a dilated trocar site, a long sharpened dissector is employed to grasp a suture tied to the anvil stem. The dissector is then passed into the lumen of the stomach and used to perforate the future pouch in location that is appropriate for the eventual anastomosis. Once the anvil stem has been advanced through the perforation, the dissector is withdrawn. Stapling of the stomach may include amputation of the gastrotomy site. The pouch is created by stapling around the anvil and thus can be made very small.

Troubleshooting

Difficulty in passing the anvil perorally may arise at the level where the trachea separates from the esophagus in the deep pharynx. This difficulty can usually be overcome either by having the endoscopist perform a jaw-thrust maneuver or by placing a large laryngoscope blade deep in the pharynx to make the anvil visible in the proximal esophagus and then nudging the anvil forward with either the tip of the blade or a McGill forceps. Transiently deflating the endotracheal tube balloon may also help. Occasionally, a large, blunt esophageal dilator is used to place pressure on the top of the anvil. Given the potential for esophageal injury if the anvil will not advance, it is important not to apply excessive force. In one case from our experience, the anvil became lodged in the proximal esophagus beyond the laryngoscopic view, and the long suture holding it broke; retrograde passage of an esophageal dilator placed via a gastrotomy in the pouch was required to dislodge the anvil. Other surgeons who use wire to draw the anvil down the esophagus have identified nontransmural esophageal injuries on postoperative contrast studies or have seen subcutaneous emphysema in the neck after operation. There is at least one case report describing an esophageal perforation with this technique.

Step 6a (Circular Stapling): Gastrojejunostomy

In the circular stapling technique, the stapled end of the Roux limb is opened to permit introduction of the stapler. A 12 mm trocar site in the left upper quadrant is dilated so that the circular stapler can be inserted through the abdominal wall without the need for a trocar. Lubrication is helpful for this step, and covering the device with a sterile plastic bag helps to protect the wound from contamination when the device is extracted. The transabdominal stapler then cannulates the Roux limb. The stapler is advanced 3 to 4 cm down the limb, and the spike on the body of the device is brought through the antimesenteric portion of the bowel by unscrewing the stapler under direct vision. Once the stapler has been opened completely, it is laparoscopically joined to the anvil protruding from the gastric pouch, then closed and fired.

Once fired, the stapler is removed from the abdominal wall, and a balloon trocar device is used to close the dilated abdominal wall opening. A single firing of the endoscopic GIA stapler, loaded with 2.5 mm staples, is often all that is required to close the enterotomy left from passage of the stapler. Interrupted or continuous absorbable sutures are then placed around the stapled anastomosis to provide added security and reduce tension on the staple line.

Step 6b (Linear Stapling): Gastrojejunostomy

Figure 15.
Figure 16.

Initially, a continuous posterior row of nonabsorbable suture material is placed to secure the Roux limb to the posterior gastric pouch. An enterotomy and a gastrotomy are performed that are large enough to admit the jaws of the endoscopic GIA stapler, which is loaded with 3.5 mm staples in a 45 mm cartridge for a side-to-side anastomosis [see Figure 15] by means of the same techniques employed in creating the jejunojejunostomy. The cartridge is inserted to its full length and fired to create an inner stapled layer [see Figure 16]. We use the full length of the staple cartridge because we have found that our technique (a complete two-layer anastomosis that includes 360° oversewing of the anastomosis over a 30 French dilator or a flexible 9 to 11 mm gastroscope) can lead to obstruction if the full cartridge length is not employed. The remaining gastroenterotomy defect is closed in two layers with continuous sutures (absorbable for the inner layer, nonabsorbable for the outer).

In our experience, using the linear stapler for the gastrojejunostomy simplifies the procedure, reduces operating time, and eliminates the concerns about injury to the body of the esophagus that arise with passage of the circular stapler.

Step 6c (Handsewn Anastomosis): Gastrojejunostomy

Some surgeons employ laparoscopic suturing techniques to create a handsewn gastrojejunostomy, usually with a single layer of continuous nonabsorbable suture material. In particular, robotic surgery techniques usually include hand suturing, using the robotic effectors to grasp the tissue and the needle and, ultimately, to tie the suture. Robotic techniques are hampered by the setup time required for placement of the robotic arms, but they have been touted as being particularly useful for operating on superobese persons, in whom manipulation of traditional laparoscopic instruments is made difficult by the extreme thickness of the abdominal wall, which interferes with the trocar’s fulcrum effect. In such cases, use of a robotic technique appears to improve the ergonomics of the procedure and relieve the surgeon’s hand and arm fatigue.

Step 7: Assessment of Anastomosis

In every case, regardless of which anastomotic technique is employed, flexible upper GI endoscopy is performed to assess the anastomosis. The Roux limb is occluded with an intestinal clamp to prevent excessive bowel and distal gastric dilatation. The patient is placed in the supine position, and the area around the anastomosis is irrigated with saline; the presence of air bubbles, easily detectable in the irrigant, indicates that the anastomosis is leaking. In most cases, the pouch and the Roux limb can be distended tightly, and even tiny air leaks are reinforced with additional sutures. The anastomosis and the staple lines are visualized, and the endoscope is navigated through the anastomosis into the Roux limb whenever possible. After adequate visualization and testing, the gas is suctioned from the intestine, the Roux limb is unclamped, and the endoscope is removed. Alternatively, some surgeons prefer to distend the anastomosis with methylene blue dye to exclude leakage.

Step 8: Closure

Figure 17.

Finally, a 10 mm closed suction drainage tube may be placed adjacent to the anastomosis to permit monitoring for postoperative leaks. In our program, we no longer do this on a routine basis. If a drain is placed, it is usually removed after the patient begins oral intake. Selected patients undergo upper GI series to evaluate for postoperative leakage [see Figure 17]. This radiographic study is considered mandatory in patients with equivocal postoperative signs and symptoms of leakage, though surgical reexploration may be preferable and is a more definitive assessment in such patients.

In antecolic Roux-en-Y procedures involving division of the greater omentum, the lateral tongue of greater omentum may be wrapped in a lateral-to-medial direction posteriorly around the gastrojejunostomy, then sutured to the anterior pouch tissue so as to reinforce the anastomosis. We do not do this routinely—only when we have technical concerns about the integrity of the anastomosis. We also sometimes use the distal gastric remnant as a live tissue patch over areas of concern in a difficult anastomosis. The liver retractor is removed under direct vision to ensure that no bleeding occurs. When dilating trocars have been employed, the trocar sites are not routinely closed. Trocar sites manually dilated to permit extraction of a specimen (e.g., gallbladder) or passage of a circular stapler should be closed with sutures to avoid risk of hernia formation; we have found that such closure increases postoperative incisional pain at these sites. Skin wounds are closed with minimal subcuticular sutures, and the epidermis is reapproximated with commercially available skin closure adhesives.

Hand-Assisted Laparoscopic Gastric Bypass


Because total intracorporeal laparoscopic GBP is such a challenging technical adventure, a hand-assisted version of the procedure was developed.61 This technique served as a bridge to the total intracorporeal approach, in that it made it possible to learn the technical aspects of a difficult, highly advanced laparoscopic procedure while enjoying the security provided by the presence of a hand within the abdomen for palpation and manipulation during the procedure. This added security is the major advantage of the hand-assisted approach. The major disadvantage is the potential for complications at the incision used for manual access. The complications seen at this site are reminiscent of those seen after open GBP, including major wound infection, dehiscence, and hernia formation.

In a series of hand-assisted laparoscopic GBP procedures from our institution, there was one major wound infection in the hand incision and one instance of postoperative fascial dehiscence. Subsequent study suggested that the hand-assisted procedure did not reduce the incidence of wound-related complications and incisional hernias and that it significantly increased the cost of surgical treatment when compared with open GBP. It appears, therefore, that the primary role of hand-assisted laparoscopic GBP may be in helping surgeons to negotiate the steep learning curve associated with total laparoscopic GBP. Some surgeons, however, have persisted with hand-assisted techniques, and large series have been published.

Complications


The complications observed to date after laparoscopic GBP include the usual problems that occur in some patients after open GBP, including marginal ulcer and stenosis at the gastrojejunal anastomosis necessitating dilatation. On rare occasions, a gastrogastric fistula may lead to a treatment-resistant marginal ulcer. The major advantage of laparoscopic GBP over open GBP is likely to be reduced wound complications (e.g., major wound infection and incisional hernia). We have seen several relatively minor trocar site infections after laparoscopic GBP but none that carried the long wound-care disability characteristic of a major wound infection after open GBP. Intermediate-term weight loss with laparoscopic GBP appears to be identical to that with open GBP.

Postoperative Management


After operation, the obese patient should be kept in the reverse Trendelenburg position and should not be extubated until he or she is fully alert and showing evidence of adequate ventilatory effort [see 8:6 Mechanical Ventilation]. In the absence of respiratory insufficiency, most obese patients can be extubated in the OR or the recovery room and returned to a standard hospital room.

Patients with obstructive SAS may have to be managed with overnight mechanical ventilation in the ICU, particularly if an open bariatric procedure was performed. Patients who were receiving ventilatory support with nasal CPAP before operation should have this treatment reinstituted after operation; monitoring for prolonged apnea should be continued in the ICU or in a stepdown unit with digital oximetry. If apnea occurs, simply waking the patient should correct the problem.

Patients with OHS may require prolonged mechanical ventilation until the pain of breathing resolves, particularly after open procedures. Such patients cannot be expected to manifest normal ABG values, and they should be weaned to their preoperative values—hence the value of obtaining. baseline ABG values before operation in these patients. The weaning process may require several days. It is important that these patients remain in the reverse Trendelenburg position to maximize diaphragmatic excursion by reducing the pressure on the chest cavity that may result from the often very large and heavy abdominal cavity. Swan-Ganz catheters, inserted preoperatively in patients with severe OHS, are useful in monitoring postoperative intravascular volume and oxygen delivery status. Excessive diuresis or restriction of fluids should be avoided.

It is extremely important to encourage early postoperative ambulation for the morbidly obese patient. These patients often experience less pain than one might expect, and it is frequently possible to get motivated patients up and walking in the afternoon or early evening after a major abdominal procedure. If the patients have been advised preoperatively of the merits of early postoperative ambulation and know it is for their own welfare, they may be more willing to cooperate.

These basic principles of postoperative management generally apply to laparoscopic cases as well, but with some differences. Unlike patients who have undergone open GBP, those who have undergone laparoscopic GBP usually do not have a nasogastric tube left in place. The bariatric surgical patient may begin to drink small amounts of liquids on postoperative day 1 and may be kept on a liquid diet with liquid protein supplementation for several weeks. Alternatively, many programs advance postoperative patients to a pureed diet with no sugar or concentrated sweets as soon as they can tolerate it. We have found that when patients consume only liquids during the edematous healing phase of the anastomosis, there is less need for urgent dietary counseling during the first few weeks after surgery. Discharge usually takes place between 1 and 3 days after operation. Vitamin administration should be initiated early after operation; chewable tablets may be used if necessary. Thiamine deficiency is very uncommon but can be quite serious if it progresses to the Wernicke-Korsakoff syndrome of neurologic damage. Such deficiency may develop within weeks in patients who are kept on a liquid-only diet without multivitamins but appears to be more common in patients who experience persistent vomiting. Any postoperative bariatric surgical patient with unexplained neurologic symptoms (e.g., visual changes, confusion, sensory changes in the extremities, or gait disturbance) should be considered for evaluation of potential nutritional causes, including thiamine deficiency and vitamin B12 deficiency.

The basic principles of postoperative management apply to both laparoscopic and open cases.

General Complications of Surgery in the Morbidly Obese Patient


The following are some of the main complications that may be associated with any abdominal operation (open or laparoscopic GBP, VBG, or LAGB) in a severely obese patient.

Abdominal Catastrophe


It may be very difficult to recognize an abdominal catastrophe in patients who are very young, very old, or morbidly obese or who are receiving high doses of steroids. For example, the obese patient may present in the ED with a perforated duodenal ulcer or a ruptured diverticulum, complaining of abdominal pain, and yet on abdominal examination have no evidence of peritoneal irritation (i.e., no guarding, tenderness, or rigidity). This situation has been well documented in patients in whom an anastomotic or gastric leak has developed after operation for morbid obesity.62 Symptoms include shoulder pain, pelvic or scrotal pain, back pain, tenesmus, urinary frequency, and, of great importance, marked anxiety. Signs of infection (e.g., fever, tachypnea, and tachycardia) may be absent, though tachycardia is often the first sign of a significant problem. Patients with peritonitis often have clinical symptoms and signs suggesting a massive pulmonary embolus: severe tachypnea, tachycardia, and sudden hypotension. Such acute pulmonary failure is probably secondary to sepsis-induced acute respiratory distress syndrome (ARDS). Thus, peritonitis must be suspected in any morbidly obese patient with acute respiratory failure. One advantage of the laparoscopic approach to bariatric surgery is that if there is concern about a possible leak, reexploration can be carried out without the need to reopen a large abdominal incision.

Because a high index of suspicion of peritonitis is required to detect the condition in morbidly obese patients, radiographic contrast studies with water-soluble agents such as diatrizoate meglumine may be indicated even when there are few clinical signs. If a perforated viscus is suspected, exploratory laparoscopy or laparotomy may be necessary despite normal findings on radiographic contrast study.

Internal Hernia


GBP places patients at risk for internal hernia with a closed-loop obstruction, leading to bowel strangulation. There are three typical locations for these internal hernias: the jejunojejunostomy anastomosis, the opening in the transverse mesocolon through which the retrocolic Roux limb is brought, and the Petersen hernia (located posterior to the Roux limb between the Roux mesentery and the transverse mesocolon). The primary symptom of an internal hernia is periumbilical pain, usually in the form of cramping consistent with visceral colic. These internal hernias may be very difficult to diagnose. Upper GI radiographic series and abdominal CT scans are often normal, providing a false sense of security. The resulting assumption that no problem exists may be devastating for the patient should bowel infarction occur as a consequence of closed-loop obstruction. The plain abdominal radiograph should always be carefully inspected for abnormal placement or spreading of the staples in the Roux-en-Y anastomosis. The safest course of action in patients with recurrent attacks of cramping periumbilical pain is abdominal surgical exploration. The frequency of this complication seems to have increased with the advent of laparoscopic GBP, presumably because of the difficulty of closing the potential hernia spaces completely. Some attribute the problem to the decreased tendency toward adhesion formation after laparoscopic surgery or to reduction of the intra-abdominal fatty tissue in the mesentery after successful weight loss surgery. We have seen one patient who had a patent mesenteric defect despite an intact suture; the apparent cause of the defect was a dramatic reduction in the mesenteric fat incorporated in the closure.

Acute Gastric Distention


After GBP, massive gaseous distention occasionally develops in the distal bypassed stomach, sometimes leading to a gastric perforation or disruption of the gastrojejunostomy. The primary symptoms of this complication are hiccups and a bloated sensation reported by the patient. Massive gastric dilatation can lead to severe left shoulder pain and shock. During the era when open GBP was predominant, gastric paralytic ileus was a known cause of this complication, particularly in patients requiring extensive adhesiolysis at the time of the bypass. Currently, with the advent of laparoscopic GBP, the problem is usually secondary to transient edema or to mechanical obstruction at the jejunojejunostomy anastomosis. The diagnosis is made by means of an urgent upright abdominal radiograph, which reveals the markedly dilated and air-filled bypassed stomach. Occasionally, the stomach is filled with fluid, and the diagnosis may be more difficult; in such cases, CT scanning may be helpful. Percutaneous transabdominal skinny-needle decompression can aid in the management of this complication and allow time for tissue edema at the jejunojejunostomy to resolve. If the dilatation recurs or the patient experiences serious difficulty, an emergency laparotomy should be performed, a gastrostomy tube inserted, and the jejunojejunostomy evaluated.

Altered Insulin Requirements in Diabetic Patients


Patients with type 2 diabetes may require large amounts of insulin for blood glucose control because of significant insulin resistance. It is not unusual, however, to note a complete absence of the requirement for insulin in the immediate postoperative period in morbidly obese patients. Therefore, insulin should be withheld on the morning of operation. In morbidly obese patients who have undergone GBP, there is often a marked reduction in the requirement for insulin throughout the postoperative period and even at discharge, possibly because of increased release of gastric inhibitory peptide (GIP) from the proximal small bowel. Therefore, regular subcutaneous insulin should be administered to GBP patients according to a sliding scale after operation until insulin requirements can be determined. Before discharge, the patient should be taking an appropriate dose of insulin, but afterward, he or she must perform frequent finger-stick blood glucose determinations, given that the need for insulin will decrease progressively with weight loss.

In one study of 23 patients with diabetes mellitus who underwent gastric bariatric operation, the average requirement for insulin decreased from 74 units/day before operation to 8 units/day after operation.63 Fourteen of the 23 patients were able to discontinue insulin completely, 11 by the time of discharge from the hospital 1 week after operation. These benefits were maintained during long-term follow-up to 39 months and were a result of a major decrease in insulin resistance that was associated with decreased food intake, as well as with weight loss.

Failed Weight Loss and Weight Regain


A postoperative problem that deserves special mention is the risk of failed weight loss or weight regain. This is one of the most difficult problems associated with bariatric surgery and may arise after any gastric procedure for morbid obesity. Patients who have undergone gastroplasty or gastric banding may have difficulty with solid foods and come to exhibit maladaptive eating behavior involving frequent ingestion of high-calorie liquid carbohydrates (a common reason for failure of a bariatric procedure). Conversion to GBP appears to be a successful management strategy for these patients. In a study in which 53 VBG patients were converted to GBP, the average loss of excess weight after VBG was 31 P5%; 2 years after conversion to GBP, the average loss of excess weight increased to 67 P2%, a value virtually identical to that in the primary GBP group.64

Inadequate weight loss is also seen in GBP patients. In some, stomal dilatation eventually develops after the procedure; however, no correlation between stomal size and weight loss has been demonstrated for GBP patients, and reoperation to make the pouch or the stoma smaller has not yielded any benefit over time when the initial procedure has failed. New techniques for narrowing the dilated gastrojejunostomy stoma endoscopically are currently being tested, but there remains the larger question as to whether such interventions are of any real value for long-term weight control. In a small percentage of patients, the failure of GBP may be attributable to either the loss or the absence of dumping syndrome symptoms, which leads to resumed ingestion of high-calorie sweets or, more commonly, to frequent ingestion of high-fat junk foods (e.g., potato or corn chips, microwave popcorn, and peanut butter crackers) that crumble easily and empty quickly from the pouch, thereby keeping the patient from feeling full. Repeated dietary counseling over a period of years is required to educate patients to eat low-calorie, high-fiber foods (e.g., raw carrots, broccoli, cauliflower, apples, and oranges) that stay in the small gastric pouch longer and provide a sensation of early satiety.

We make clear to patients, well in advance of the operation, that bariatric surgery is designed to provide them with a tool that will assist them in behavior modification and thereby help them help themselves. Obesity can be beaten by surgical treatment, but patients must continue to make good food choices. Patients who begin to ingest more than 1,100 kcal/day often begin to gain weight; even if the weight gain is only 0.5 lb/month, this amounts to 6 lb/year, or 60 lb in 10 years. Bariatric surgical patients may need lifelong nutritional counseling to optimize the results of surgical management of morbid obesity. In addition, appropriate regular physical exercise is an important adjunct to surgical treatment, and patients who comply with exercise treatment not only lose more weight but also are better able to maintain their weight loss. Accordingly, we recommend that patients follow a suitable exercise regimen for the rest of their lives