Rectal Cancer

Back

Practice Essentials

Rectal cancer is a disease in which cancer cells form in the tissues of the rectum; colorectal cancer occurs in the colon or rectum. Adenocarcinomas comprise the vast majority (98%) of colon and rectal cancers; more rare rectal cancers include lymphoma (1.3%), carcinoid (0.4%), and sarcoma (0.3%).

The incidence and epidemiology, etiology, pathogenesis, and screening recommendations are common to both colon cancer and rectal cancer. The image below depicts the staging and workup of rectal cancer.



View Image

Diagnostics. Staging and workup of rectal cancer patients.

Signs and symptoms

Bleeding is the most common symptom of rectal cancer, occurring in 60% of patients. However, many rectal cancers produce no symptoms and are discovered during digital or proctoscopic screening examinations.

Other signs and symptoms of rectal cancer may include the following:

See Clinical Presentation for more detail.

Diagnosis

Perform physical examination with specific attention to the size and location of the rectal tumor in addition to possible metastatic lesions, including enlarged lymph nodes or hepatomegaly. In addition, evaluate the remainder of the colon.

Examination includes the use of the following:

Laboratory tests

Routine laboratory studies in patients with suspected rectal cancer include the following:

Screening tests may include the following:

Imaging studies

If metastatic (local or systemic) rectal cancer is suspected, the following radiologic studies may be obtained:

See Workup for more detail.

Management

A multidisciplinary approach that includes surgery, medical oncology, and radiation oncology is required for optimal treatment of patients with rectal cancer. Surgical technique, use of radiotherapy, and method of administering chemotherapy are important factors. 

Strong considerations should be given to the intent of surgery, possible functional outcome, and preservation of anal continence and genitourinary functions. The first step involves achievement of cure, because the risk of pelvic recurrence is high in patients with rectal cancer, and locally recurrent rectal cancer has a poor prognosis.

Surgery

Radical resection of the rectum is the mainstay of therapy. The timing of surgical resection is dependent on the size, location, extent, and grade of the rectal carcinoma. Operative management of rectal cancer may include the following:

Adjuvant medical management

Adjuvant medical therapy may include the following:

Pharmacotherapy

The National Comprehensive Cancer Network guidelines recommend the use of as many chemotherapy drugs as possible to maximize the effect of adjuvant therapies for colon and rectal cancer.

The following agents may be used in the management of rectal cancer:

See Treatment and Medication for more detail.

Background

Colon and rectal cancer incidence was negligible before 1900. The incidence of colorectal cancer rose dramatically following economic development and industrialization. Currently, colorectal cancer is the third most common cancer and the third leading cause of cancer deaths in both males and females in the United States.[1]

Adenocarcinomas comprise the vast majority (98%) of colon and rectal cancers. Other rare rectal cancers, including carcinoid (0.4%), lymphoma (1.3%), and sarcoma (0.3%), are not discussed in this article. Squamous cell carcinomas may develop in the transition area from the rectum to the anal verge and are considered anal carcinomas. Very rare cases of squamous cell carcinoma of the rectum have been reported.[2, 3]

Approximately 20% of colon cancers develop in the cecum, another 20% in the rectum, and an additional 10% in the rectosigmoid junction. Approximately 25% of colon cancers develop in the sigmoid colon.[2]

The incidence and epidemiology, etiology, pathogenesis, and screening recommendations are common to both colon cancer and rectal cancer. These areas are addressed together.

Anatomy

The surgical definition of the rectum differs from the anatomical definition; surgeons define the rectum as starting at the level of the sacral promontory, while anatomists define the rectum as starting at the level of the 3rd sacral vertebra. Therefore, the measured length of the rectum varies from 12 cm to 15 cm. The rectum differs from the rest of the colon in that the outer layer consists of longitudinal muscle. The rectum contains three folds, namely the valves of Houston. The superior (at 10 cm to 12 cm) and inferior (at 4 cm to 7 cm) folds are located on the left side and the middle fold (at 8 cm to 10 cm) is located at the right side.

National Comprehensive Cancer Network guidelines define rectal cancer as cancer located within 12 cm of the anal verge by rigid proctoscopy.[4] This definition was developed by the Dutch Colorectal Cancer Group study, which found that the risk of recurrence of rectal cancer depends on the location of the cancer. Univariate sub-group analyses showed that the treatment effect for surgery alone vs preoperative radiotherapy plus surgery was not significant in patients whose cancer (TNM stage I to IV) was located between 10.1 cm and 15 cm from the anal verge.[5]

Pathophysiology

The mucosa in the large intestine regenerates approximately every 6 days. Crypt cells migrate from the base of the crypt to the surface, where they undergo differentiation and maturation, and ultimately lose the ability to replicate.

The significant portions of colorectal carcinomas are adenocarcinomas. The adenoma-carcinoma sequence is well described in the medical literature.[2]  Colonic adenomas precede adenocarcinomas. Approximately 10% of adenomas will eventually develop into adenocarcinomas. This process may take up to 10 years.[2]

Three pathways to colon and rectal carcinoma have been described:

The APC adenoma carcinoma pathway involves several genetic mutations, starting with inactivation of the APC gene, which allows unchecked cellular replication at the crypt surface. With the increase in cell division, further mutations occur, resulting in activation of the K-ras oncogene in the early stages and p53 mutations in later stages. These cumulative losses in tumor suppressor gene function prevent apoptosis and prolong the cell's lifespan indefinitely. If the APC mutation is inherited, it will result in familial adenomatous polyposis syndrome.

Histologically, adenomas are classified in three groups: tubular, tubulovillous, and villous adenomas. K-ras mutations and microsatellite instability have been identified in hyperplastic polyps. Therefore, hyperplastic polyps may also have malignant potential in varying degrees.[6]

The other common carcinogenic pathway involves mutation in DNA mismatch repair genes. Many of these mismatched repair genes have been identified, including hMLH1, hMSH2, hPMS1, hPMS2, and hMSH6. Mutation in mismatched repair genes negatively affects the DNA repair. This replication error is found in approximately 90% of HNPCC and 15% of sporadic colon and rectal cancers.[2, 7] A separate carcinogenic pathway is also described in inflammatory bowel disease (IBD). Chronic inflammation such as in ulcerative colitis can result in genetic alterations which then lead into dysplasia and carcinoma formation.[2]

Epidemiology

United States

Colon and rectal cancer is the third most common cancer in both females and males. The American Cancer Society (ACS) estimates that 104,610 new cases of colon cancer and 43,340 new cases of rectal cancer will occur in 2020; 25,960 cases of rectal cancer are expected in men and 17,380 in women. For estimates of deaths, the ACS combines colon and rectal cancers; approximately 53,200 deaths from colorectal cancer are expected to occur in 2020.[1]

Overall, the colorectal cancer death rate decreased by 54% from 1970 to 2017—from 29.2 to 13.5 per 100,000, respectively—because of changing patterns in risk factors, increased screening, and improvements in treatment. However, the overall trend is driven by older adults (who have the highest rates) and masks increasing incidence in younger age groups. From 2008 to 2017, the death rate declined by 2.6% per year in those ages 55 and older but increased by 1% per year in adults younger than age 55..[1]  Currently, adults born circa 1990 have quadruple the risk of rectal cancer compared with those born circa 1950.[8]  

Tumor site tends to vary by patient age. In those aged younger than 65 years, the rectum is the most common site of colorectal cancer, accounting for 37% of cases in those under age 50 years and 36% in those 50 to 64 years of age. In individuals age 65 and older, rectal cancer accounts for 23% of colorectal cancer cases; the proximal colon is the most common site, accounting for 49% of cases.[9]

International

Although the incidence of colon and rectal cancer varies considerably by country, an estimated 1,849, 518 cases occurred worldwide in 2018. High incidences of colon and rectal cancer cases are identified in the US, Canada, Japan, parts of Europe, New Zealand, Israel, and Australia. Low colorectal cancer rates are identified in Algeria and India. The majority of colorectal cancers still occur in industrialized countries.[10]

Increases in colorectal cancer in persons younger than 50 years has also been in many other high‐income countries aside from the US, including Australia, Canada, Germany, and the United Kingdom. In Austria, however, where opportunistic screening has been used in individuals aged 40 years and older since the 1980s, rates of colorectal cancer are increasing in those aged 20 to 39 years but decreasing in those age 40 to 49 years.[9]

Race

The incidence of colorectal cancer tends to be higher in Western nations than in Asian and African countries; however, within the United States, minor differences in incidence exist among whites, African Americans, and Asian Americans. Five-year survival rates are lower among blacks (58%) than whites (67%).[1]   In the US, African American men have the highest mortality (23.8 per 100,000) and Asian/Pacific Islander and Hispanic women have the lowest mortality (8.4 and 8.8 per 100,000 respectively).[11]

A study by Yothers et al found that black patients with resected stage II and stage III colon cancer had worse overall and recurrence-free survival compared with white patients who underwent the same therapy.[12]

A study of racial disparities in mortality rates between black and white individuals with colorectal cancer by Robbins et al showed earlier and larger reductions in death rates for whites from 1985-2008.[13]  This racial disparity could be decreased with greater education to the black population regarding colorectal cancer prevention and access to treatment, including colonoscopies and polypectomies.

Sex

The incidence of colorectal malignancy is slightly higher in males than in females. The overall age-adjusted incidence of colorectal cancer in all races was 44.2 per 100,000 for males and 33.9 per 100,000 for females in 2012-2016, yielding a male-female ratio of 1.30:1. Mortality rates for colorectal cancer were also higher in males (16.9 per 100,000) than in females (11.9 per 100,000) in 2012-2016[11]  

Age

The incidence of colorectal cancer starts to increase after age 35 and rises rapidly after age 50, peaking in the seventh decade. More than 90% of colon cancers occur after age 50. However, cases have been reported in young children and adolescents.[2]  The incidence rates of colorectal cancer increased by about 2% per year in adults younger than age 55 from 2006 to 2015, largely because of increases in rectal cancer.[1]

Mortality/Morbidity

The American Cancer Society estimates that in 2019, colorectal cancer will account for 9% of cancer deaths in men and 8% in women. In the US, mortality rates have been decreasing in both sexes for the past 2 decades.  The 5-year relative survival rate is 6 5% for colorectal cancer; however, for the minority of rectal cancer patients who are diagnosed with localized disease, the 5-year survival rate is 90%.[1]

A review of eight trials by Rothwell et al found allocation to aspirin reduced death caused by cancer. Individual patient data were available from seven of the eight trials. Benefit was apparent after 5 years of follow-up. The 20-year risk of cancer death was also lower in the aspirin group for all solid cancers. A latent period of 5 years was observed before risk of death was decreased for esophageal, pancreatic, brain, and lung cancers. A more delayed latent period was observed for stomach, colorectal, and prostate cancer. Benefit was only seen for adenocarcinomas in lung and esophageal cancers. The overall effect on 20-year risk of cancer death was greatest for adenocarcinomas.[14]

A study by Banks et al showed the benefit of aspirin in preventing colon adenocarcinoma among patients with hereditary risk of colorectal cancer. In a study of 861 patients, 600 mg of aspirin daily for a mean of 25 months substantially reduced cancer incidence after 55.7 months among carriers of hereditary colorectal cancer. Further studies are needed to determine the ideal dosage and duration.[15]

In a review of 51 randomized controlled trials, Rothwell et al found that aspirin reduced the short-term incidence of cancer and the short- and long-term risk of cancer death. The authors conclude that their results support the use of daily aspirin for cancer prevention.[16]

Prognosis

The 5-year relative survival rates for rectal cancer based on SEER stage are as follows[11] :

A review of 111,453 patients in the National Cancer Data Base who were diagnosed with early-stage (T1N0 or T2N0) rectal cancer from 1998 to 2010 found that increasing age, male sex, higher comorbidity score, and positive or unknown final surgical margins were associated with poorer long-term adjusted overall survival.[17]

Recurrence of rectal cancer, which usually develops in the first year after surgery, carries a poor prognosis. Recurrence may be local, distant, or both; local recurrence is more common in rectal cancer than in colon cancer. Reported rates of local recurrence have ranged from 3.7% to 50%.[18]  Factors that influence the development of recurrence include the following:

Surgical therapy may be attempted for recurrence and includes pelvic exenteration or abdominal perineal resection in patients who had a sphincter-sparing procedure. Radiation therapy generally is used as palliative treatment in patients who have locally unresectable disease.

Patient Education

A study by Thong et al found that survivors of rectal cancer may benefit from increased focus, both clinical and psychological, on the possible long-term morbidity of treatment and its effects on health.[19]

For patient education resources, see the Digestive Disorders Center and Cancer Center. Also, see the patient education articles Colon Cancer, Colonoscopy, Sigmoidoscopy, Abdominal Pain in Adults, Rectal Bleeding, and Rectal Cancer.

History

All patients should undergo a complete history (including a family history) and assessment of risk factors for the development of rectal cancer. Many rectal cancers produce no symptoms and are discovered during digital or proctoscopic screening examinations.

Bleeding is the most common symptom of rectal cancer, occurring in 60% of patients. Bleeding often is attributed to other causes (eg, hemorrhoids), especially if the patient has a history of other rectal problems. Profuse bleeding and anemia are rare. Bleeding may be accompanied by the passage of mucus, which warrants further investigation.

Change in bowel habits is present in 43% of patients; change is not evident in some cases because the capacity of a rectal reservoir can mask the presence of small lesions. When change does occur it is often in the form of diarrhea, particularly if the tumor has a large villous component. These patients may have hypokalemia, as shown in laboratory studies. Some patients experience a change in the caliber of the stool. Large tumors can cause obstructive symptoms. Tumors located low in the rectum can cause a feeling of incomplete evacuation and tenesmus.

Occult bleeding is detected via a fecal occult blood test (FOBT) in 26% of all cases. Abdominal pain is present in 20% of the cases. Partial large-bowel obstruction may cause colicky abdominal pain and bloating. Back pain is usually a late sign caused by a tumor invading or compressing nerve trunks. Urinary symptoms may also occur if the tumor is invading or compressing the bladder or prostate.

Malaise is a nonspecific symptom and present in 9% of rectal cancer cases. Bowel obstruction due to a high-grade rectal lesion is rare, occurring in 9% of all cases. Pelvic pain is a late symptom, usually indicating nerve trunk involvement, and is present in 5% of all cases. Other manifestations include emergencies such as peritonitis from perforation (3%) or jaundice, which may occur with liver metastases (< 1%).

Physical

Physical examination is performed with specific attention to size and location of rectal cancer in addition to possible metastatic lesions, including enlarged lymph nodes or hepatomegaly. The remainder of the colon is also evaluated.

Digital rectal examination (DRE) provides an opportunity to readily detect abnormal lesions. The average finger can reach approximately 8 cm above the dentate line. Rectal tumors can be assessed for size, ulceration, and presence of any pararectal lymph nodes. Fixation of the tumor to surrounding structures (eg, sphincters, prostate, vagina, coccyx and sacrum) also can be assessed. DRE also permits a cursory evaluation of the patient's sphincter function. This information is necessary when determining whether a patient is a candidate for a sphincter-sparing procedure. Rigid proctoscopy is also performed to identify the exact location of the tumor in relation to the sphincter mechanism.

Causes

The etiology of colorectal cancer is unknown, but colorectal cancer appears to be multifactorial in origin and includes environmental factors and a genetic component. Diet may have an etiologic role, especially diet with high fat content. 

A cohort study by Tabung et al that followed 121,050 adults for 26 years found that in both men and women, intake of proinflammatory diets (replete in red, processed, and organ meat, for example) was associated with a significantly higher risk of developing colorectal cancer. Risk was especially high in overweight and obese men and, paradoxically, in lean women. Risk was also increased in men and women who do not drink alcohol.[20, 21]

Approximately 75% of colorectal cancers are sporadic and develop in people with no specific risk factors. The remaining 25% of cases occur in people with significant risk factors—most commonly, a family history or personal history of colorectal cancer or polyps, which are present in 15-20% of all cases. Other significant risk factors are certain genetic predispositions, such as hereditary nonpolyposis colorectal cancer (HNPCC; 4-7% of all cases) and familial adenomatous polyposis (FAP, 1%); and inflammatory bowel disease (IBD; 1% of all cases).

Environmental Factors

Diet

A high-fat, low-fiber diet is implicated in the development of colorectal cancer. Specifically, people who ingest a diet high in saturated animal fats and highly saturated vegetable oils (eg, corn, safflower) have a higher incidence of colorectal cancer. The mechanism by which these substances are related to the development of colorectal cancer is unknown.

Saturated fats from dairy products do not have the same carcinogenic effect, nor do oils containing oleic acid (eg, olive, coconut, fish oils). Omega-3 monounsaturated fatty acids and omega-6 monounsaturated fatty acids also appear to be less carcinogenic than unsaturated or polyunsaturated fats. In fact, recent epidemiologic data suggest that high fish consumption may provide a protective effect against development of colorectal cancer. Long-term diets high in red meat or processed meats appear to increase the risk of distal colon and rectal cancers.[22, 23]

The ingestion of a high-fiber diet may be protective against colorectal cancer. Fiber causes the formation of a soft, bulky stool that dilutes carcinogens; it also decreases colonic transit time, allowing less time for harmful substances to contact the mucosa. The decreased incidence of colorectal cancer in Africans is attributed to their high-fiber, low–animal-fat diet. This favorable statistic is reversed when African people adopt a western diet. Findings of a meta-analysis of 22 studies with a total of 2,876,136 subjects suggest that dietary fiber intake could be a protective factor against rectal cancer with a clinically relevant reduction in rectal cancer risk.[22]

Increased dietary intake of calcium appears to have a protective effect on colorectal mucosa by binding with bile acids and fatty acids. The resulting calcium salts may have antiproliferative effects, decreasing crypt cell production in the mucosa. A double-blind placebo-controlled study showed a statistically significant reduction in the incidence of metachronous colorectal adenomas.[24] Other dietary components, such as selenium, carotenoids, and vitamins A, C, and E, may have protective effects by scavenging free-oxygen radicals in the colon.

Alcohol

Alcohol intake of more than 30 g daily has been associated with increased risk of developing colorectal carcinoma, with risk of rectal cancer greater than that of colon cancer. Risk appears greater with beer than with wine.[25] Specifically, Kabat et al found that daily beer consumption of 32 ounces or more increases the risk of rectal cancer in men (odds ratio 3.5).[26]

Tobacco

Smoking, particularly when started at a young age, increases the risk of colorectal cancer.[27] Possible mechanisms for tumor development include the production of toxic polycyclic aromatic amines and the induction of angiogenic mechanisms due to tobacco smoke.

A study by Phipps et al found that smoking is also associated with increased mortality after colorectal cancer diagnosis, especially among patients with colorectal cancer with high microsatellite instability.[28]

Cholecystectomy

Following cholecystectomy, bile acids flow freely, increasing exposure to the degrading action of intestinal bacteria. This constant exposure increases the proportion of carcinogenic bile acid byproducts. A meta-analysis by Giovannucci et al revealed an increased risk of proximal colon carcinoma following cholecystectomy. Although a large number of studies suggest the increased risk of proximal colon cancer in patients following cholecystectomy, the data are not compelling enough to warrant enhanced screening in this patient population.[2]

Hereditary Factors

The relative risk of developing colorectal cancer is increased in the first-degree relatives of affected patients. For offspring, the relative risk is 2.42 (95% CI: 2.20-2.65); when more than one family member is affected, the relative risk increases to 4.25 (95% CI; 3.01-6.08). If the first-degree family member is younger than 45 years at the time of diagnosis, the risk increase is even higher.[29]

Regarding the personal history of colorectal cancer or polyps: Of patients with colorectal cancer, 30% have synchronous lesions, usually adenomatous polyps. Approximately 40-50% of patients have polyps on a follow-up colonoscopy. Of all patients who have adenomatous polyps discovered via a colonoscopy, 29% of them have additional polyps discovered on a repeat colonoscopy one year later. Malignancy develops in 2-5% of patients. The risk of cancer in people who have had polyps removed is 2.7-7.7 times that of the general population.[30]

Genetic Disorders

Familial adenomatous polyposis (FAP)

FAP is an autosomal dominant inherited syndrome that results in the development of more than 100 adenomatous polyps and a variety of extra-intestinal manifestations. The defect is in the APC gene, which is located on chromosome 5 at locus q21. The disease process causes the formation of hundreds of intestinal polyps, osteomas of bone, desmoid tumors, and, occasionally, brain tumors. Individually, these polyps are no more likely to undergo malignant transformation than are polyps in the general population. The increased number of polyps, however, predisposes patients to a greater risk of cancer. If left untreated, colorectal cancer develops in nearly 100% of these patients by age 40. Whenever the hereditary link is documented, approximately 20% of FAP cases are found to be caused by spontaneous mutation.

Hereditary nonpolyposis colorectal cancer

HNPCC is an autosomal dominant inherited syndrome that occurs because of defective mismatch repair genes located on chromosomes 2, 3, and 7. Patients have the same number of polyps as the general population, but their polyps are more likely to become malignant. These patients also have a higher incidence of endometrial, gastric, thyroid, and brain cancers.

The revised Amsterdam criteria are used to select at-risk patients (all criteria must apply):

The National Comprehensive Cancer Network (NCCN) guidelines discuss various strategies for identifying patients who should undergo testing for HNPCC. One approach is to test patients with any of the following[31] :

Another strategy is so-called universal screening, in which all individuals newly diagnosed with colorectal cancer have either microsatellite instability (MSI) or immunohistochemistry (IHC) testing for absence of 1 of the 4 DNA MMR proteins. An alternative is to test all patients with colorectal cancer diagnosed before age 70 years, and test those 70 years and older only if they meet the Bethesda criteria for colorectal cancer. The primary method for detecting HNPCC in tumor tissue from biopsied or surgically resected specimens is with either immunohistochemistry or microsatellite instability testing. The NCCN guidelines also indicate that genetic counseling is not necessary before “routine tumor testing” at a center.[31]

Inflammatory Bowel Disease

The malignant pathway in these patients does not involve any adenoma-carcinoma sequence. Cancer risk increases with duration of disease. After 10 years, the incidence of colorectal cancer in ulcerative colitis (UC) is approximately 1% per year. Patients should be evaluated for dysplastic changes via an annual colonoscopy. Dysplasia is a precursor of cancer and when present, the risk of cancer is 30%.

The incidence of colorectal cancer in patients with Crohn disease is 4-20 times greater than that of the general population. Cancer occurs in patients with disease of at least 10 years' duration. The average age at cancer diagnosis, 46-55 years, is younger than that of the general population. Cancers often develop in areas of strictures and in de-functionalized segments of intestine. In patients with perianal Crohn disease, malignancy is often present in fistulous tracts. Patients with Crohn colitis should undergo the same surveillance regimen as those with UC.

Laboratory Studies

Routine laboratory studies should include a complete blood count (CBC); serum chemistries, including liver and renal function tests; and a carcinoembryonic antigen (CEA) test. A cancer antigen (CA) 19-9 assay, if available, may also be useful to monitor the disease.

Screening CBC may demonstrate a hypochromic, microcytic anemia, suggesting iron deficiency. The combined presence of vitamin B-12 or folate deficiency may result in a normocytic or macrocytic anemia. All men and postmenopausal women with iron deficiency anemia require a GI evaluation.

Liver function tests are usually part of the preoperative workup. The results are often normal, even in patients with metastases to the liver.

Perform a CEA test in all patients with rectal cancer. A baseline level is obtained before surgery and a follow-up level is obtained after surgery. If a previously normalized CEA begins to rise in the postoperative period, this suggests possible recurrence. A CEA level higher than 100 ng/mL usually indicates metastatic disease and warrants a thorough investigation. The steps of the workup are outlined in Figure 1.



View Image

Diagnostics. Staging and workup of rectal cancer patients.

Fecal immunochemical tests appear to be accurate for detecting colorectal cancer. In a meta-analysis that examined 8 different brands of fecal immunochemical tests (FITs), Lee and colleagues found that FITs had high accuracy, high specificity, and moderately high sensitivity for the detection of colorectal cancers.[32, 33] The meta-analysis, which included 19 studies with sample sizes ranging from 80 to 27,860, showed that FITs had sensitivity of 0.79, specificity of 0.94, a positive likelihood ratio of 13.10, and a negative likelihood ratio of 0.23. The overall diagnostic accuracy of FITs was 95%.

Screening for Colon and Rectal Cancer

The process of malignant transformation from adenoma to carcinoma takes several years. The purpose of screening is to eradicate potential cancers while they are still in the benign stage of the adenoma-carcinoma sequence. Screening also increases the likelihood of discovering existing cancers while they are still in the early stage.

Screening techniques

Screening techniques include the following:

Average-risk screening

People who are asymptomatic, younger than 50 years, and have no other risk factors are considered at average risk for developing colorectal cancer. Screening of the average-risk population should begin at age 50 years and end at age 75 years.[37] Indications for screening in patients at average risk for colon and rectal cancer include the following:

A French study found that even in patients with no personal or family history of colorectal polyps or cancer, starting colonoscopy screening at age 45 instead of age 50 can be valuable. In a prospective study that included 6027 consecutive screening colonoscopies, Karsenti et al found that for the 515 patients age 45 to 49 years, the average polyp detection rate was 26% and the average neoplasia detection rate was nearly 4%. By comparison, for the 4438 patients older than 50 years, the average polyp detection rate exceeded 35% and the average neoplasia detection rate exceeded 5%. Both rates were markedly lower in the 1076 study patients age 44 years and younger.[38]

The US Multi-Society Task Force on Colorectal Cancer (USMSTF) has endorsed various cost-effective screening regimens. Screening options for the detection of adenomatous polyps and cancer for asymptomatic adults 50 years and older include FSIG every 5 years, colonoscopy every 10 years, DCBE every 5 years, or CTC every 5 years. Testing options that primarily detect cancer in asymptomatic adults 50 years and older include annual glucose-based FOBT with high-test sensitivity for cancer; annual FIT with high-test sensitivity for cancer; or SDNA with high-test sensitivity for cancer, although the optimal interval for SDNA is uncertain.

Each screening test has unique advantages. They have been shown to be cost-effective and have associated risks and limitations. Ultimately, patient preferences and availability of testing resources guide the selection of screening tests.

The main disadvantage of the structural tests is their requirement for bowel preparation. The primary advantage of structural tests is that they can detect polyps as well as cancer. Conscious sedation is usually used for colonoscopy. FSIG is uncomfortable, and screening benefit is limited to sigmoid colon and rectum. Risks for colonoscopy, DCBE, and CTC may rarely include perforation; colonoscopy may also be associated with bleeding. Positive findings on FSIG, DCBE, and CTC usually result in referral for colonoscopy.

The advantages of the stool tests are that they are noninvasive, do not require bowel preparation, can be done in the privacy of the patient's home, and are more readily available to patients without adequate insurance coverage or local resources.

In the United States, colon and rectal cancer screening rates have been averaging between 50% and 60%. Brounts and colleagues studied colorectal cancer screening in the Military Healthcare System. In this study, overall screening rates were lower in minority groups than in whites. Also, overall lower screening rates were identified in patients younger than 65 years. Although ethnicity-related, gender-related, and age-related disparities were observed, screening rates were improved in this equal-access health care system when compared with national averages.[39]

Screening of high-risk patients

A patient's family history or personal history may indicate increased risk for colorectal cancer. Patients at high risk for colon and rectal cancer due to family history who should be included in surveillance programs include those with the following:

Patients at high risk for colon and rectal cancer due to personal history who should be included in surveillance programs includes the following:

Screening recommendations by risk factor are as follows:

Histologic Findings

Histopathologic features such as poor differentiation, lymphovascular and/or perineural invasion, T4 tumor stage, and clinical findings such as obstruction or perforation, and elevated preoperative CEA levels are all associated with increased recurrence rates and worse survival.

Staging

Dukes Classification

In 1932, Cuthbert E. Dukes, a pathologist at St. Mark Hospital in England, introduced a staging system for rectal cancer. His system divided tumor classification into three stages, as follows:

This system was modified by others to include subdivisions of stages B and C, as follows:

Stage B was divided into B1 (ie, tumor penetration into muscularis propria) and B2 (ie, tumor penetration through muscularis propria).

Stage C was divided into C1 (ie, tumor limited to the rectal wall with nodal involvement) and C2 (ie, tumor penetrating through the rectal wall with nodal involvement).

Stage D was added to indicate distant metastases

Tumor, Node, Metastasis (TNM) System

This system was introduced in 1954 by the American Joint Committee on Cancer (AJCC) and the International Union Against Cancer (IUAC). The TNM system is a universal staging system for all solid cancers that is based on clinical and pathologic information. Each category is independent.

Neither the Dukes nor the TNM system includes prognostic information such as histologic grade, vascular or perineural invasion, or tumor DNA ploidy. TNM staging of rectal cancer correlates well with 5-year survival rates of patients with rectal cancer (see the TNM stage-dependent 5-year survival rate for rectal carcinomas).

TNM classification for cancer of the colon and rectum (AJCC)

Primary tumor (T) includes the following:

Regional lymph nodes (N) include the following:

Distant metastasis (M) include the following:

Table 1. Comparison of AJCC Definition of TNM Staging System to Dukes Classification.



View Table

See Table

The TNM stage – dependent 5-year survival rate for rectal carcinomas is as follows:

Approach Considerations

A multidisciplinary approach that includes surgery, medical oncology, and radiation oncology is required for optimal treatment of patients with rectal cancer. See the image below.



View Image

Staging and treatment. Rectal cancer treatment algorithm (surgery followed by adjuvant chemotherapy and radiotherapy). Initial stages are Endorectal u....

Determination of the optimal treatment plan for a patient with rectal cancer involves a complex decision-making process. Strong considerations should be given to the intent of surgery, possible functional outcome, and preservation of anal continence and genitourinary functions. The timing of surgical resection is dependent on the size, location, extent, and grade of the rectal carcinoma. The number of lymph nodes removed (12 or more; minimum, 10) at the time of surgery impacts staging accuracy and prognosis.

The first step involves achievement of cure, because the risk of pelvic recurrence is high in patients with rectal cancer and locally recurrent rectal cancer has a poor prognosis. Functional outcome of different treatment modalities involves restoration of bowel function with acceptable anal continence and preservation of genitourinary functions. Preservation of both anal and rectal reservoir function in treatment of rectal cancer is highly preferred by patients. Sphincter-saving procedures for rectal cancer are now considered the standard of care.[40]

Factors influencing sphincter and organ preservation in patients with rectal cancer can be described as follows[40] :

The following factors are associated with difficult sphincter preservation:

Patients with the following may be candidates for local excision:

Disadvantages of abdominoperineal resection include the following:

Transanal Excision

The local transanal excision of rectal cancer is reserved for early-stage cancers in a select group of patients. The lesions amenable for local excision are small (< 3 cm in size), occupying less than a third of a circumference of the rectum, preferably exophytic/polypoid, superficial and mobile (T1 and T2 lesions), low-grade tumors (well or moderately differentiated) that are located in low in the rectum (within 8 cm of the anal verge). There should also be no palpable or radiologic evidence of enlarged mesenteric lymph nodes. The likelihood of lymph node involvement in this type of lesion ranges from 0-12%.[40, 41]

A study by Peng et al found that local excision in early-stage rectal cancer may result in high local recurrence rates. The authors recommend only using this procedure in highly selected groups of patients, specifically those with a tumor size of 2.5 cm or smaller.[42]

Local excision is increasingly used to treat stage I rectal cancers despite its inferiority to total mesorectal excision, which is the current standard of care. In a study of all rectal cancer patients in the National Cancer Data Base from 1998 through 2010, researchers found that local excision was used to treat 46.5% of the patients with T1 tumors and 16.8% of those with T2 tumors. For patients with T1 cancer, local excision rates increased from 39.8% in 1998 to 62.0% in 2010. For patients with T2 cancers, rates increased from 12.2% to 21.4%.[43, 17]

Preoperative endorectal ultrasound should be performed. If nodes are identified as suggestive of cancer, do not perform transanal excision. The lesion is excised with the full thickness of the rectal wall, leaving a 1-cm margin of normal tissue. The defect is usually closed; however, some surgeons leave it open. Unfavorable pathologic features such as positive resection margins, lymphovascular invasion, lymph node metastasis, perineural invasions, and recurrent lesion at follow-up evaluations mandate salvage resection. Usually, an abdominal perineal resection or proctosigmoidectomy with coloanal anastomosis is performed as a salvage resection following failure of local excision.[41]

The advantages of local excision include rapid recovery, minimal effect on sphincter function, and relatively low perioperative morbidity and mortality. Recovery is usually rapid. The 5-year survival rate after transanal excision ranges from 65-100% (these figures include some patients with T2 lesions). The local recurrence rate ranges from 0-40%. Patients with lesions that display unfavorable histologic features but are excised completely may be treated with adjuvant radiation therapy.

Cancer recurrence following transanal excision of early rectal cancer has been studied by Weiser et al.[44] Failures due to transanal excision are mostly advanced local disease and are not uniformly salvageable with radical pelvic excision. These patients may require extended pelvic dissection with en bloc resection of adjacent pelvic organs such as the pelvic side wall with autonomic nerves, coccyx, prostate, seminal vesicle, bladder, vagina, ureter, ovary, and uterus. The long-term outcome in patients with recurrent rectal carcinoma who undergo radical resection is less favorable than expected, relative to the early stage of their initial rectal carcinoma.[44]

In summary, the treatment of T1 and T2 rectal cancers continues to be challenging. Local excision is associated with higher rate of recurrence, especially in T2 lesions. Ultimately, 15-20% of patients may experience recurrence. When local recurrence is detected, patients usually have advanced disease, requiring extensive pelvic excisions. Therefore, strict selection criteria are essential when considering local excision. All patients should be informed of the risk of local recurrence and lower cure rates associated with recurrence.[40, 44, 45]

Endocavitary Radiation

This radiotherapy method differs from external-beam radiation therapy in that a larger dose of radiation can be delivered to a smaller area over a shorter period. Selection criteria for this procedure are similar to those for transanal excision. The lesion can be as far as 10 cm from the anal verge and no larger than 3 cm. Endocavitary radiation is delivered via a special proctoscope and is performed in an operating room with sedation. The patient can be discharged on the same day.

A total of 6 application of high-dose (20Gy to 30 Gy), low-voltage radiation (50kV) is given over the course of 6 weeks. Each radiotherapy session produces a rapid shrinkage of the rectal cancer lesion. An additional booster dose can be given to the tumor bed. The overall survival rate is 83%, although the local recurrence rate as high as 30%.[41]

Transanal Endoscopic Microsurgery (TEM)

Transanal endoscopic microsurgery is another form of local excision that uses a special operating proctoscope that distends the rectum with insufflated carbon dioxide and allows the passage of dissecting instruments. This method can be used on lesions located higher in the rectum and even in the distal sigmoid colon. Transanal endoscopic microsurgery has not come into wide use yet because of a significant learning curve and a lack of availability.

Sphincter-Sparing Procedures

Procedures are described that use the traditional open technique. All of these procedures, except the perineal portions, can also be performed using laparoscopic techniques, with excellent results.   Laparoscopic surgery offers the advantages of faster recovery time and less pain, compared with open surgery. The nuances of the laparoscopic technique used are beyond the scope of this discussion.

A study by Li et al found that laparoscopic and open surgery for middle and lower rectal cancer are associated with similar long-term outcomes. The study shows the value of technical experience when performing laparoscopic surgery and encourages the use of this surgery by experienced teams.[46]

Long-term results from the UK Medical Research Council trial of laparoscopically assisted versus open surgery for colorectal cancer showed no differences between groups in overall or disease-free survival or recurrence rates.[47]  

In an international randomized, open-label trial (COlorectal cancer, Laparoscopic or Open Resection II [COLOR II]) involving 1044 patients with localized solitary rectal cancer located within 15 cm from the anal verge, comparison of the locoregional recurrence rate at 3 years showed no significant differences between the laparoscopic and open-surgery groups (5% in both). Disease-free-survival (74.8% and 70.8%, respectively), overall survival (86.7% and 83.6%), and rate of complications also showed no significant differences.[48]

Low anterior resection (LAR)

LAR is generally performed for lesions in the middle and upper third of the rectum and, occasionally, for lesions in the lower third. Because this is a major operation, patients who undergo LAR should be in good health. They should not have any preexisting sphincter problems or evidence of extensive local disease in the pelvis.

Patients will not have a permanent colostomy but should be informed that a temporary colostomy or ileostomy may be necessary. They also must be willing to accept the possibility of slightly less-than-perfect continence after surgery, although this is not usually a major problem.

Other possible disturbances in function include transient urinary dysfunction secondary to weakening of the detrusor muscle. This occurs in 3-15% of patients. Sexual dysfunction is more prominent and includes retrograde ejaculation and impotence. In the past, this has occurred in 5-70% of men, but recent reports indicate that the current incidence is lower.[49]

The operation entails full mobilization of the rectum, sigmoid colon, and, usually, the splenic flexure. Mobilization of the rectum requires a technique called total mesorectal excision (TME). TME involves sharp dissection in the avascular plane that is created by the envelope that separates the entire mesorectum from the surrounding structures. This includes the anterior peritoneal reflection and Denonvilliers fascia anteriorly and preserves the inferior hypogastric plexus posteriorly and laterally. TME is performed under direct visualization. Mesorectal spread can occur by direct tumor spread, tumor extension into lymph nodes, or perineural invasion of tumor.[35, 45, 49]

TME yields a lower local recurrence rate (4%) than transanal excision (20%), but it is associated with a higher rate of anastomotic leak (11%). For this reason, TME may not be necessary for lesions in the upper third of the rectum. The distal resection margin varies depending on the site of the lesion. A 2-cm margin distal to the lesion must be achieved. For the tumors of the distal rectum, less than 5 cm from the anal verge, the minimally accepted distal margin is 1 cm in the fresh specimen. Distal intra-mural spread beyond 1 cm occurs rarely. Distal spread beyond 1 cm is associated with aggressive tumor behavior or advanced tumor stage.[35]

The procedure is performed with the patient in the modified lithotomy position with the buttocks slightly over the edge of the operating table to allow easy access to the rectum.[45]   A circular stapling device is used to create the anastomosis. A double-stapled technique is performed. This entails transection of the rectum distal to the tumor from within the abdomen using a linear stapling device. The proximal resection margin is divided with a purse-string device.

After sizing the lumen, the detached anvil of the circular stapler is inserted into the proximal margin and secured with the purse-string suture. The circular stapler is inserted carefully into the rectum, and the central shaft is projected through or near the linear staple line. Then, the anvil is engaged with the central shaft, and, after completely closing the circular stapler, the device is fired. Two rings of staples create the anastomosis, and a circular rim or donut of tissue from the proximal and distal margins is removed with the stapling device.

According to a study by Maurer et al, the introduction of TME has resulted in an impressive reduction of local recurrence rate. TME appears to have improved survival in patients without systemic disease.[50]

The anastomotic leak rate with this technique ranges from 3-11% for middle-third and upper-third anastomosis and to 20% for lower-third anastomosis. For this reason, some surgeons choose to protect the lower-third anastomosis by creating a temporary diverting stoma. This is especially important when patients have received preoperative radiation therapy. The rate of stenosis is approximately 5-20%. A hand-sewn anastomosis may be performed; if preferred, the anastomosis is performed as a single-layer technique. The leak and stenosis rates are the same.

In R0 resection, the inferior mesenteric artery (IMA) should be excised at its origin, but this rule is not mandated by available supportive evidence. Patients with non–en-bloc resection, positive radial margins, positive proximal and distal margin, residual lymph node disease, and incomplete preoperative and intra-operative staging would not be considered to have complete resection of cancer (R0 resection).[35] Patients with R1 and R2 resection are considered to have an incomplete resection for cure. Incomplete R1 and R2 resection does not change the TNM stage but affects the curability.[35]

In a 2012 multicenter, randomized controlled trial, mesorectal excision with lateral lymph node dissection was associated with a significantly longer operation time and significantly greater blood loss than mesorectal excision alone.[51]

A study by Han et al analyzing factors that might be predictive of pathologic complete response (pCR) in patients with stage II and III rectal cancer undergoing TME after preoperative chemoradiation indentified high tumor location and low carcinoembryonic antigen (CEA) level after chemoradiation therapy as independent predictive factors for pCR.[52]

Colo-anal anastomosis (CAA)

Very distal rectal cancers that are located just above the sphincter occasionally can be resected without the need for a permanent colostomy. The procedure is as already described; however, the pelvic dissection is carried down to below the level of the levator ani muscles from within the abdomen. A straight-tube coloanal anastomosis (CAA) can be performed using the double-stapled technique, or a hand-sewn anastomosis can be performed transanally.[49]

The functional results of this procedure have been poor in some patients, who experience increased frequency and urgency of bowel movements, as well as some incontinence to flatus and stool. An alternative to the straight-tube CAA is creation of a colonic J pouch. The pouch is created by folding a loop of colon on itself in the shape of a J. A linear stapling or cutting device is inserted into the apex of the J, and the stapler creates an outer staple line while dividing the inner septum. The J-pouch anal anastomosis can be stapled or hand sewn.

An alternative to doing the entire dissection from within the abdomen is to begin the operation with the patient in the prone jackknife position. The perineal portion of this procedure involves an intersphincteric dissection via the anus up to the level of the levator ani muscles. After the perineal portion is complete, the patient is turned to the modified lithotomy position and the abdominal portion is performed. Either a straight-tube or colonic J-pouch anal anastomosis can be created; however, both must be hand sewn.[49]

The advantages of the J pouch include decreased frequency and urgency of bowel movements because of the increased capacity of the pouch. A temporary diverting stoma is performed routinely with any coloanal anastomosis.

Abdominal perineal resection (APR)

APR is performed in patients with lower-third rectal cancers. APR should be performed in patients in whom negative margin resection (see Table 2, below) will result in loss of anal sphincter function. This includes patients with involvement of the sphincters, preexisting significant sphincter dysfunction, or pelvic fixation, and sometimes is a matter of patient preference.

Table 2. Acceptable Minimal Distal and Proximal Resectional Margins for Rectal Cancer.[35, 53]



View Table

See Table

A 2-team approach is often used, with the patient in modified lithotomy position. The abdominal team mobilizes the colon and rectum, transects the colon proximally, and creates an end-sigmoid colostomy. The perineal team begins by closing the anus with a purse-string suture and making a generous elliptical incision. The incision is carried through the fat using electrocautery. The inferior rectal vessels are ligated and the anococcygeal ligament is divided. The dissection plane continues posteriorly, anterior to the coccyx to the level of the levator ani muscles.

Then, the surgeon breaks through the muscles and retrieves the specimen that has been placed in the pelvis. The specimen is brought out through the posterior opening, and the anterior dissection is continued carefully. Care must be taken to avoid the prostatic capsule in the male and the vagina in the female (unless posterior vaginectomy was planned). The specimen is removed through the perineum, and the wound is irrigated copiously. A closed-suction drain is left in place, and the perineal wound is closed in layers, using absorbable sutures. During this time, the abdominal team closes the pelvic peritoneum (this is not mandatory), closes the abdomen, and matures the colostomy.[49]

In patients who have rectal cancer with adjacent organ invasion, en bloc resection should be performed in order to not compromise cure. This situation is encountered in 15% of rectal cancer patients. The urinary bladder is the organ most commonly involved in locally advanced rectal carcinoma. Extended, en bloc resection may involve partial or complete cystectomy.[35, 49]  In women, rectal carcinoma also commonly invades the uterus, adnexa, and posterior vaginal wall.

Treatment of colorectal cancer with liver metastasis

Chemotherapeutic regimens for liver metastasis including systemic and intrahepatic administration have only had limited benefit. Systemic chemotherapy had 18-28% response rates.  However, one meta-analysis found that carefully selected patients with metastatic colorectal cancer may benefit from preoperative chemotherapy with curative intent.[54]

It is well accepted that liver resections in selected patients are beneficial. Overall, 5-year survival rates following surgical resection of liver metastasis vary from 20-40%. A study by Dhir et al found that among patients undergoing hepatic resection for colorectal metastasis, a negative margin of 1 cm or more had a survival advantage.[55]

Adjuvant Care

Although radical resection of rectum is the mainstay of therapy, surgery alone has a high recurrence rates. The local recurrence rate for rectal cancers treated with surgery alone is 30-50%. Rectal adenocarcinomas are sensitive to ionizing radiation. Radiation therapy can be delivered preoperatively, intraoperatively, or postoperatively and with or without chemotherapy.

Tumor stage, grade, number of lymph node metastasis, lymphovascular involvement, signet cell appearance, achievement of negative radial margins, and distance from the radial margin are important prognostic indicators of local and distant recurrences. Low anterior (LAR) or abdominal-perineal resection (APR) in conjunctions with total mesorectal excision (TME) should be performed for optimal surgical therapy.

A study by Margalit et al found that patients older than 75 years had difficulty tolerating combined modality chemotherapy to treat rectal cancer. They required early termination of treatment, treatment interruptions, and/or dose reductions.[56]

Adjuvant radiation therapy

Preoperative radiation therapy has many potential advantages, including tumor down-staging; an increase in resectability, possibly permitting the use of a sphincter-sparing procedure; and a decrease in tumor viability, which may decrease the risk of local recurrence. Preoperative radiation therapy works better in well-oxygenated tissues prior to surgery. Postoperatively, tissues are relatively hypoxic as a result of surgery and may be more resistant to radiotherapy. If patients have postoperative complications, initiation of adjuvant therapy may be delayed.

Preoperative radiation therapy also minimizes the radiation exposure of small bowel loops due to pelvic displacement and adhesions following surgery.[49, 57] In a study of patients with locally advanced rectal cancer, a higher dose of radiation delivered using an endorectal boost increased major response in T3 tumors by 50% without increasing surgical complications or toxicity.[58]

The disadvantages of preoperative radiation therapy include delay in definitive resection, possible loss of accurate pathologic staging, possible over-treatment of early-stage (stage I and II) rectal cancer, and increased postoperative complications and morbidity and mortality rates secondary to radiation injury. Preoperative radiation therapy decreases the risk of tumor recurrence in patients with stage II or III disease; however, this does not translate into a decrease in distant metastases or an increase in survival rate. Some recent reports cite an increase in survival; however, this is still the minority opinion.

In sum, preoperative radiotherapy may be effective in improving local control in localized rectal cancer but is only of marginal benefit in attainment of improved overall survival; it does not diminish the need for permanent colostomies and it may increase the incidence of postoperative surgical infections; it also does not decrease the incidence of long-term effects on rectal and sexual function.[59] The authors recommend preoperative chemoradiation therapy in patients with large bulky cancers and with obvious nodal involvement.[49]

A study by Cassidy et al found that elimination of neoadjuvant radiation therapy for select patients with stage II and III rectal adenocarcinoma is associated with worse overall survival. In their review of 21,707 patients with clinical T2N1 (cT2N1), cT3N0, or cT3N1 rectal cancers in the National Cancer Data Base, the 5‐year actuarial overall survival rate was 75% for patients who received neoadjuvant chemoradiotherapy versus 67.2% for those who received neoadjuvant multiagent chemotherapy (P < 0.01).[60]

The advantages of postoperative radiation therapy include immediate definitive resection and accurate pathologic staging information before beginning ionizing radiation. The disadvantages of postoperative radiation therapy include possible delay in adjuvant radiation therapy if postoperative complications ensue; no effect on tumor cell spread at the time of surgery; and decreased effect of radiation in tissues with surgically-induced hypoxia. Published randomized trials suggest that preoperative or postoperative radiation therapy appears to have a significant impact on local recurrence but does not increase survival rates.[49]

A study by Ng et al found that statin use during and after adjuvant chemotherapy did not result in improved disease-free survival, recurrence-free survival, or overall survival in patients with stage III colon cancer.[61]

Intraoperative radiation therapy

Intraoperative radiation therapy is recommended in patients with large, bulky, fixed, unresectable cancers. The direct delivery of high-dose radiotherapy is believed to improve local disease control. Intraoperative radiation therapy requires specialized, expensive operating room equipment, limiting its use.

Adjuvant chemotherapy

Chemotherapy options for colon and rectal cancer have greatly expanded in recent years, but the efficacy of chemotherapy remains incomplete and its toxicities remain substantial. Combination therapy with use of as many drugs as possible is needed for maximal effect against rectal cancer. See Table 3, below.

Table 3. Colorectal Chemotherapeutic Regimens



View Table

See Table

The most useful chemotherapeutic agent for colorectal carcinoma is 5-fluorouracil (5-FU), an antimetabolite. The prodrug, 2-deoxy-5-floxuridine (5-FUDR), is rapidly converted to 5-FU and is used for metastatic liver disease by continuous intrahepatic infusion. Fluorouracil is a fluorinated pyrimidine, which blocks the formation of thymidylic acid and DNA synthesis. Clinically, it offers good radiosensitization without severe side effects, although diarrhea can be dose limiting and, if severe, life-threatening. 5-FU has been used in conjunction with radiation (combined modality) therapy before surgery (neoadjuvant), as well as after surgery.

Stage I (T1-2, N0, M0) rectal cancer patients do not require adjuvant therapy due to their high cure rate with surgical resection. High-risk patients, including those with poorly differentiated tumor histology and those with lymphovascular invasion, should be considered for adjuvant chemotherapy and radiotherapy. The NCCN guidelines recommend combination therapy with infusional fluorouracil, folinic acid, and oxaliplatin (FOLFOX) or with capecitabine and oxaliplatin (CapeOx) as reasonable for patients with high-risk or intermediate-risk stage II disease; however, FOLFOX is not indicated for good- or average-risk stage II rectal cancer.[4] FOLFOX is associated with neuropathy and one long-term study confirmed that although overall neurotoxicity did not significantly increase after a median of 7 years, specific neurotoxicity (numbness and tingling of the hands and feet) remained elevated.[62]

Use of FOLFOX or the combination of folinic acid, fluorouracil, and irinotecan (FOLFIRI) is recommended in treatment of patients with stage III or IV disease.

Biologic therapy

Cetuximab should not be used in patients with the KRAS mutation.[63] A study by Maughan et al also found that cetuximab added to oxaliplatin-based chemotherapy has no confirmed benefit in patients with advanced colorectal cancer.[64]

In randomized phase III studies, panitumumab, a monoclonal antibody for EGFR, combined with FOLFOX4 (fluorouracil, leucovorin, and oxaliplatin) or FOLFIRI (fluorouracil, leucovorin, and irinotecan) significantly improved progression-free survival when compared to FOLFOX4 or FOLFIRI alone in patients with metastatic colorectal cancer and wild-type KRAS status.[65, 66]

Simkens et al found that patients with a high body mass index (BMI) had better overall survival on chemotherapy regimens than those with a low BMI. This effect was not seen in patients receiving chemotherapy and targeted therapy; the authors suggest that a possible decreased efficacy of bevacizumab in obese patients may account for the discrepancy.[67]

A study by Boisen et al in patients with metastatic colorectal cancer reported significantly better outcomes with first-line capecitabine and oxaliplatin (CAPEOX) plus bevacizumab in patients whose primary tumors originated in the rectum and sigmoid colon. For patients treated with CAPEOX only, no association between primary tumor location and outcome was found.[68]

Adjuvant chemoradiation therapy

Patients with locally advanced rectal cancer (T3-4, N0, M0 or any T, N1-2, M0) should receive primary chemotherapy and radiotherapy. The combination of preoperative radiation therapy and chemotherapy with fluorouracil improves local control, distant spread, and survival. The basis of this improvement is believed to be the activity of fluorouracil as a radiosensitizer. Surgical resection can be done 4 to 10 weeks after completion of chemotherapy and radiotherapy.

A meta-analysis of neoadjuvant long-course chemoradiotherapy followed by total mesorectal excision for locally advanced rectal cancer concluded that waiting for a minimum of 8 weeks from the end of chemoradiotherapy to surgical excision increases pathological complete response (pCR) and downstaging rates, and improves recurrence-free survival without compromising surgical morbidity. With longer intervals, the odds ratio (OR) for pCR was 1.41 (95% confidence interval [CI] 1.30-1.52; P <   0.001) and the OR for tumor downstaging was 1.18 (95% CI 1.05-1.32; P = 0.004). The increased rate of pCR translated to reduced distant metastasis and overall recurrence but not local recurrence.[69]

A study by Ebert et al of colorectal cancer genetics and treatment found a link between hypermethylation of transcription factor AP-2 epsilon (TFAP2E) and clinical nonresponsiveness to chemotherapy in colorectal cancer.[70]

Radioembolization

The US Food and Drug Administration (FDA) has approved yttrium-90 resin microspheres (SIR-Spheres) for the treatment of unresectable metastatic liver tumors from primary colorectal cancer in combination with adjuvant intra-hepatic artery chemotherapy with floxuridine.

A prospective, multicenter, randomized phase III study by Hendlisz et al compared the addition of yttrium-90 resin to a treatment regimen of fluorouracil 300 mg/m2 IV infusion (days 1-14 every 8 wk) with fluorouracil IV alone. Yttrium-90 was injected intra-arterially into the hepatic artery. The addition of radioembolization with yttrium-90 significantly improved time to liver progression and median time to tumor progression.[71]  

However, improvement in liver disease control has not translated to a benefit in overall survival. Three multicenter randomized, phase III trials—FOXFIRE, SIRFLOX, and FOXFIRE-Global—have evaluated the efficacy of combining first-line chemotherapy with selective internal radiotherapy  using yttrium-90 resin microspheres in patients with metastatic colorectal cancer with liver metastases. A combined analysis of those trials found that overall survival was not significantly different in patients who received FOLFOX chemotherapy plus selective internal radiotherapy (n=554) compared with those who received FOLFOX only (n=549).[72]

Response to treatment indicator

A retrospective study conducted by Santiago and colleagues found the split scar sign was a simple morphologic pattern visible on restaging magnetic resonance (MR) high-resolution T2-weighted imaging (T2-WI) which, although not sensitive, is very specific for the identification of sustained complete responders after neoadjuvant therapy in rectal cancer. Because the split scar sign is visible on high-resolution T2-weighted MR imaging it does not require any changes to standard protocol.  At first restaging pelvic MR imaging (mean: 9.1 weeks after the end of radiotherapy), the split scar sign identified patients who sustained a complete response with very high specificity (0.97) and positive predictive value (0.93-0.94).  The split scar sign has the potential to improve patient selection for "watch-and-wait" after neoadjuvant therapy in rectal cancer.[73]

Diet

In a prospective cohort study that included 1575 healthcare professionals with stage I to III colorectal cancer, Song et al found that rates of colorectal cancer (CRC)–specific mortality and overall mortality were lower in patients who had higher intake of dietary fiber, especially from cereals. Survival rates were higher in patients who increased their fiber intake after diagnosis from levels before diagnosis, and in patients reporting higher intake of whole grains.[74, 75]

After multivariable adjustment, each 5 g increment in daily fiber intake was associated with a 22% decrease in CRC-specific mortality and a 14% decrease in all-cause mortality. In patients who increased their fiber intake after diagnosis, each 5 g increase in daily fiber intake was associated with 18% lower CRC-specific mortality. The relationship between fiber intake after diagnosis and CRC-specific mortality  reached a maximum at approximately 24 g/d, beyond which no further mortality reduction was found.[74, 75]

Evaluation of the source of fiber showed that cereal fiber was associated with lower CRC-specific mortality (33% per 5-g/d increment) and all-cause mortality (22%); vegetable fiber was associated with 17% lower all-cause mortality but not with significantly lower CRC-specific; no association was found for fruit fiber. Whole grain intake was associated with lower CRC-specific mortality (28% decrease in risk per 20-g/day increment), although this beneficial association fell to 23% after adjusting for fiber intake.[74, 75]

Prevention

On December 22, 2010, the US Food and Drug Administration approved the use of quadrivalent human papillomavirus (HPV) vaccine (Gardasil) for prevention of anal cancer and associated precancerous lesions in people aged 9-26 years. HPV is associated with about 90% of anal cancer. Approval was based on a study of men who have sex with men in which HPV vaccine was shown to be 78% effective in prevention of HPV 16– and 18–related anal intraepithelial neoplasms.[76]   

As of October 2016, the only HPV vaccine available in the United States is Gardasil 9 (Merck, Whitehouse Station, NJ). It provides coverage of coverage of HPV types 6, 11, 16, 18, 31, 33, 45, 52, and 58. Gardasil 9 is approved for prevention of HPV-associated anal cancer and genital warts (condyloma acuminata) in males and females 9 to 26 years of age.[77]

Long-Term Monitoring

US Multi-Society Task Force on Colorectal Cancer guidelines recommend local surveillance with flexible sigmoidoscopy or endoscopic ultrasound (EUS) every 3−6 mo for the first 2−3 y after surgery in patients at increased risk for local recurrence.[78] This includes the following:

The task force also advises that all patients who have undergone curative resection of rectal cancer should receive their first surveillance colonoscopy 1 y after surgery (or 1 y after clearing perioperative colonoscopy).

The National Comprehensive Cancer Network recommends the following surveillance measures[4] :

Screening

Guidelines Contributor: Elwyn C Cabebe, MD Physician Partner, Valley Medical Oncology Consultants; Medical Director of Oncology, Clinical Liason Physician, Cancer Care Committee, Good Samaritan Hospital

Guidelines on colorectal screening have been issued by the following organizations:

While all the guidelines recommend routine screening for colorectal cancer and adenomatous polyps in asymptomatic adults starting at age 50, they differ with regard to frequency of screening and age at which to discontinue screening, as well as preferred screening method. For high-risk patients, the recommendations differ regarding the age at which to begin screening, as well as the frequency and method of screening.

American Cancer Society (ACS), US Multi-Society Task Force on Colorectal Cancer, and American College of Radiology

A joint guideline developed by the American Cancer Society, US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology recommends that screening for colorectal cancer and adenomatous polyps start at age 50 years in asymptomatic men and women.[79]

In addition, individuals with any of the following colorectal cancer risk factors should undergo colonoscopy at an earlier age and more frequently than average risk individuals:

Tests that detect adenomatous polyps and cancer, and their recommended frequency, include the following:

Tests that primarily detect cancer, and their recommended frequency, include the following:

American Cancer Society update

In May 2018 the ACS revised its colorectal screening guidelines, advising that regular screening for people at average risk start at age 45 years.[80]  Additional ACS recommendations include the following:

U.S. Preventive Services Task Force (USPSTF)

The USPSTF recommends that screening for colorectal cancer start at age 50 years and continue until age 75 years (A recommendation). For adults aged 76 to 85 years, the decision to screen should be individualized, taking into account the patient’s overall health and prior screening history (C recommendation).[37]

The USPSTF advises that screening is more likely to benefit older patients who have never been screened than those who have undergone screening, and is more likely to benefit patients who are healthy enough to undergo treatment for colorectal cancer treatment and who do not have other medical conditions limiting their life expectancy.[37]

The USPSTF does not recommend colorectal cancer screening for adults older than 85 years.[37]

The USPSTF notes that colorectal screening is substantially underused. As part of a strategy to increase screening rates, the guidelines provide a range of screening options rather than a ranking of tests.

Stool-based screening tests and intervals are as follows:

Direct visualization screening tests and intervals are as follows:

American College of Physicians (ACP)

In 2015, the American College of Physicians recommended that average-risk adults aged 50 to 75 years should be screened for colorectal cancer by one of the following strategies[81] :

Interval screening with fecal testing or flexible sigmoidoscopy in adults having 10-year screening colonoscopy is not recommended. Average-risk adults younger than 50 years, older than 75 years, or with an estimated life expectancy of less than 10 years should not be screened. 

American College of Gastroenterology (ACG)

The guidelines of the American College of Gastroenterology make a distinction between screening tests for cancer prevention and cancer detection, preferring cancer prevention tests.[82]  The specific guidelines for colorectal cancer screening are as follows:

Alternative cancer detection tests recommended in the ACG guidelines are as follows:

Alternative cancer detection tests in the ACG guidelines are as follows:

For screening purposes, patients with one first-degree relative diagnosed with colorectal cancer or advanced adenoma at age 60 years or older are considered at average risk. For patients with a single first-degree relative diagnosed with colorectal cancer or advanced adenoma before age 60 years, or those with two first-degree relatives with colorectal cancer or advanced adenomas, the guideline recommends colonoscopy every 5 years, beginning at age 40 years or at 10 years younger than the age at diagnosis of the youngest affected relative.

The National Comprehensive Cancer Network (NCCN)

The National Comprehensive Cancer Network (NCCN) has released separate guidelines for average-risk and high-risk individuals.[83, 31]  For average individuals, the recommendations are nearly identical to those of the joint American Cancer Society (ACS), US Multi-Society Task Force on Colorectal Cancer, and American College of Radiology.

The NCCN guidelines provide screening recommendations for patients at increased risk due to a personal or family history of any of the following[31] :

The guidelines also specify recommendations for patients with the following high-risk syndromes[31] :

Individuals meeting one or more of the following criteria should receive further evaluation for high-risk syndromes:

Screening for Familial Risk

Hereditary nonpolyposis colorectal cancer (HNPCC), also known as Lynch syndrome, is a common autosomal dominant syndrome characterized by early age at onset, neoplastic lesions, and microsatellite instability (MSI). Guidelines for Lynch syndrome screening have been developed by the National Cancer Institute (Bethesda guidelines) and the National Comprehensive Cancer Network (NCCN).[31]

The American Gastroenterological Association recommends testing all patients with colorectal cancer for Lynch syndrome; the tumor should be tested for MSI or with immunohistochemistry for MLH1, MSH2, MSH6, and PMS2 proteins.[84]

The European Society for Medical Oncology (ESMO) guidelines for familial risk-colorectal cancer [85] , which have been endorsed by the American Society of Clinical Oncology (ASCO) [86]  includes the following recommendations:

The American College of Gastoenterology recommendations are in general agreement with ESMO.[87]

Revised Bethesda guidelines for Lynch syndrome and microsatellite instability

Because cancers with MSI account for approximately 15% of all colorectal cancers, in 1996 the National Cancer Institute developed the Bethesda guidelines for the identification of individuals with HNPCC who should be tested for MSI. These guidelines were most recently revised in 2002.[88]

Postpolypectomy Surveillance

A 2020 update of US Multi-Society Task Force on Colorectal Cancer guidelines provides recommendations on postpolypectomy surveillance. The recommendations assume high-quality baseline colonoscopy, defined as meeting all the following criteria[89] :

Screening colonoscopy findings and recommended scheduling of surveillance colonoscopy are as follows[89] :

In 2020, the British Society of Gastroenterology (BSG), the Association of Coloproctology of Great Britain and Ireland (ACPGBI) and Public Health England (PHE) released joint guidelines for surveillance after polypectomy and colorectal cancer resection. According to the guidelines, the criteria for high-risk for future colorectal cancer (CRC) following polypectomy comprise either of the following[90] :

Patients who meet the high-risk criteria should undergo a single surveillance colonoscopy at 3 years.  Patients who have undergone CRC resection should have a colonoscopy at 1 year post-surgery and every 3 years thereafter.[90]

Patients who do not meet high-risk criteria postpolypectomy should participate in national bowel screening when invited. For patients who are more than 10 years younger than the national bowel screening lower age limit, colonoscopy may be considered after 5 or 10 years and individualized to age and other risk factors.

The American College of Gastroenterology has published guidelines for surveillance of patients who have had adenomas detected and removed at colonoscopy.[89]

These patients should have more frequent followup, as they are at increased risk for developing metachronous adenomas or colon cancer.

Colonoscopy findings and recommended scheduling of followup colonoscopy are as follows:

For serrated lesions, recommended surveillance intervals are as follows:

Familial Adenomatous Polyposis

The European Society of Medical Oncology offers the following recommendations for suveillance of patients with familial adenomatous polyposis (FAP)[85] :

Classic FAP

Attenuated FAP

The American Society of Colon and Rectal Surgeons recommends that patients with familial adenomatous polyposis (FAP) or with personal or family risk factors for FAP be referred to center registries and genetic counselors with experience in the multidisciplinary management of these individuals.[91]

Additional recommendations include:

 

Surgical Treatment

The American Society of Colon and Rectal Surgeons (ASCRS) defines rectal cancer as cancer located within 15 cm of the anal verge by rigid proctoscopy. ASCRS 2013 revised management guidelines and practice parameters recommend that patients with low-risk, early-stage rectal cancer be treated with primary surgical therapy. Treatment of locally advanced or high-risk disease should include neoadjuvant radiation or chemoradiation followed by surgery.[92]

Additional recommendations include the following[92] :

Guidelines from the European Society for Medical Oncology (ESMO) include the following recommendations on managrement of local and locoregional rectal cancer[93] :

Adjuvant Therapy

National Comprehensive Cancer Network (NCCN) guidelines advise that adjuvant therapy regimens for rectal cancer should include both concurrent chemotherapy/radiotherapy and adjuvant chemotherapy. Preferably, perioperative treatment should be given for a total of approximately 6 months.[4]

According to the NCCN,patients with stage I (T1-2, N0, M0) rectal cancer without high-risk features do not require adjuvant therapy due to the high cure rate with surgical resection. High-risk patients, including those with positive margins, sm3 invasion (submucosal invasion to the lower third of the submucosal level), poorly differentiated tumor histology and those with lymphovascular invasion, should be considered for adjuvant chemotherapy and radiotherapy.[4]

The NCCN guidelines recommend combination therapy with infusional fluorouracil, folinic acid, and oxaliplatin (FOLFOX) as reasonable for patients with high-risk or intermediate-risk stage II disease; however, FOLFOX is not indicated for good- or average-risk stage II rectal cancer. Adjuvant chemotherapy is encouraged for eligible patients with stage III disease.

For the majority of patients with stage II or stage III rectal cancer, the NCCN recommends the use of ionizing radiation to the pelvis along with adjuvant chemotherapy. Either of the two following sequences of therapy may be used:

Follow-up Care in Stage II and III Colorectal Cancer

Guidelines on follow-up care for survivors of stage II and stage III colorectal cancer were issued by the following organizations:

All four guidelines agree that patients with resected colon cancer (stage II and III) should undergo regular surveillance for at least 5 years following resection, and that surveillance should include regular reviews of medical history, physical examination, and carcinoembryonic antigen assays, as well as colonoscopy and abdominal and chest computed tomography (CT).[94, 95, 4, 96] The frequency of the surveillance testing differs as shown in the table below.

Table. 1



View Table

See Table

In 2016, the US Multi-Society Task Force on Colorectal Cancer issued guidelines on colonoscopy after colorectal cancer resection, which included the following recommendations[78] :

Molecular Testing in Metastatic Disease

 In 2015, the American Society for Clinical Pathology (ASCP), the College of American Pathologists (CAP), the Association for Molecular Pathology (AMP), and the American Society of Clinical Oncology (ASCO) issued a provisional clinical opinion regarding gene mutation testing to predict response to anti–epidermal growth factor receptor (EGFR)  monoclonal antibody (MoAb) therapyin patients with metastatic colorectal carcinoma (mCRC). Among the recommendations are the following[97] :

The 2016 European Society of Medical Oncology (ESMO) guidelines for the management of patients with mCRC concur with the ASCP/CAP/AMP/ASCO RAS mutational testing recommendations above. Additional recommendations include[98] :

 

Medication Summary

The goals of pharmacotherapy are to down-stage the tumor, induce remission, reduce morbidity, and prevent complications.

Fluorouracil (5-FU, Fluorouracil, Adrucil)

Clinical Context:  Blocks methylation of deoxyuridylic acid to thymidylic acid, thereby interfering with DNA synthesis. Dose is body-weight dependent and varies with specific protocol in which patient is involved.

Vincristine (Vincasar PFS, Oncovin)

Clinical Context:  Mechanism of action uncertain. May involve decrease in reticuloendothelial cell function or increase in platelet production. It is mitotic spindle inhibitor.

Leucovorin (Wellcovorin)

Clinical Context:  Potentiates effects of fluorouracil. Reduced form of folic acid that does not require enzymatic reduction reaction for activation. Allows for purine and pyrimidine synthesis, both of which are needed for normal erythropoiesis.

Given just prior to fluorouracil.

Irinotecan (Camptosar, Camptothecin-11, CPT-11)

Clinical Context:  Inhibits topoisomerase I, inhibiting DNA replication and, consequently, cell proliferation.

Oxaliplatin (Eloxatin)

Clinical Context:  A platinum-based antineoplastic agent used in combination with an infusion of 5-fluorouracil (5-FU) and leucovorin for the treatment of metastatic colorectal cancer in patients with recurrence or progression following initial treatment with irinotecan, 5-FU, and leucovorin. It forms interstrand and intrastrand Pt-DNA crosslinks that inhibit DNA replication and transcription. The cytotoxicity is cell-cycle nonspecific.

Cetuximab (Erbitux)

Clinical Context:  Recombinant human/mouse chimeric monoclonal antibody that specifically binds to the extracellular domain of human epidermal growth factor receptors (EGFR, HER1, c-ErbB-1). Cetuximab-bound EGF receptor inhibits activation of receptor-associated kinases, resulting in inhibition of cell growth, induction of apoptosis, and decreased production of matrix metalloproteinase and vascular endothelial growth factor. Indicated for treating irinotecan-refractory, EGFR-expressed, metastatic colorectal carcinoma. Treatment is preferably combined with irinotecan. May be administered as monotherapy if irinotecan is not tolerated.

Bevacizumab (Avastin)

Clinical Context:  Indicated as a first-line treatment for metastatic colorectal cancer. Murine-derived monoclonal antibody that inhibits angiogenesis by targeting and inhibiting vascular endothelial growth factor (VEGF). Inhibiting new blood vessel formation denies blood, oxygen, and other nutrients needed for tumor growth. Used in combination with standard chemotherapy.

Panitumumab (Vectibix)

Clinical Context:  Recombinant human IgG2 kappa monoclonal antibody that binds to human epidermal growth factor receptor (EGFR). Indicated to treat colorectal cancer that has metastasized following standard chemotherapy.

Class Summary

The recommendations from National Comprehensive Cancer Network 13th Annual Conference are outlined as follows. The use of as many chemotherapy drugs as possible is recommended to maximize the effect of adjuvant therapies for colon and rectal cancer. Bevacizumab in combination with chemotherapy is indicated in patients with positive or negative resectable synchronous metastases. For colon and rectal cancer, bevacizumab in combination with chemotherapy is also indicated in patients with unresectable synchronous metastases. FOLFOX —a combination of folinic acid, fluorouracil, and oxaliplatin — is reasonable to use for high-risk or intermediate-risk stage II patients (see Table 5).

FOLFOX is not indicated for good-risk or average-risk stage II patients. In patients in whom 5-fluorouracil treatment has failed, capecitabine should be avoided. Patients who experience no benefit from bevacizumab regimens should avoid continuing the therapy. Cetuximab should not be replaced with panitumumab. Patients with KRAS mutations should not be treated with cetuximab or panitumumab, as these mutations confer resistance to epidermal growth factor receptor (EGFR) inhibitors.

Papillomavirus vaccine, quadrivalent (Gardasil)

Clinical Context:  Quadrivalent human papillomavirus (HPV) recombinant vaccine. Vaccine efficacy mediated by humoral immune responses following immunization series. Indicated for prevention of anal cancer and associated precancerous lesions caused by HPV types 6, 11, 16, and 18 people aged 9-26 years.

Class Summary

HPV is associated with about 90% of anal cancer. In a study of homosexual males, HPV vaccine was shown to be 78% effective in prevention of HPV 16- and 18-related anal intraepithelial neoplasms. A study by Nielsen et al examined trends in incidence of anal cancer and high-grade anal intraepithelial neoplasia in Denmark from 1978-2008 and found that HPV vaccines may be a vital factor in the prevention of anal cancer and its precursor lesions.[99]

What is rectal cancer?What are the common signs and symptoms of rectal cancer?How is a physical exam performed in suspected rectal cancer?Which lab studies are indicated in the workup of suspected rectal cancer?Which screening tests are used in suspected rectal cancer?Which imaging studies are used in the workup of metastatic rectal cancer?What is the approach to the treatment of rectal cancer?What are the surgical options for the treatment of rectal cancer?What are the options for medical management of rectal cancer?Which drugs are recommended by the NCCN for the treatment of rectal cancer?How common is rectal cancer and what are the most common types?What is the anatomy of the rectum in relation to rectal cancer?What is the pathophysiology of rectal cancer?What are the pathways to colon and rectal cancer?What is the epidemiology of rectal cancer in the US?What is the international incidence of rectal cancer?What is the mortality of rectal cancer?What is the role of aspirin in the reduction of cancer death rates?What is the incidence of rectal cancer by race?Is rectal cancer more common in men or women?What is the incidence of rectal cancer by age?What is the clinical history of rectal cancer?How is a physical exam conducted in suspected rectal cancer?What causes rectal cancer?Which dietary habits contribute to the development or prevention of rectal cancer?What is the role of alcohol in the development of rectal cancer?What is the role of tobacco in the development of rectal cancer?What is the relationship between cholecystectomy and rectal cancer?Which hereditary factors increase the risk of rectal cancer?What is the role of familial adenomatous polyposis (FAP) in the development of rectal cancer?What causes hereditary nonpolyposis colorectal cancer (HNPCC) (Lynch syndrome), and who is at risk?What is the malignant pathway of rectal cancer in patients with ulcerative colitis or Crohn disease?Which lab studies are indicated in the workup of rectal cancer?When is screening for colon and rectal cancer indicated in high-risk patients?What is the purpose of screening for colon and rectal cancer?What are the screening options for colon and rectal cancer?When is screening for colon and rectal cancer indicated in average-risk patients?What are the screening options and regimens for colon and rectal cancer?What are the advantages of the different colon and rectal cancer screening tests?When is surveillance for colon and rectal cancer indicated?What are the screening recommendations for rectal cancer by risk factor?Which histopathologic features of rectal cancer are associated with a poor outcome?What are the 5-year survival rates for rectal cancer according to the tumor, node, metastasis (TNM) system?What are the stages of rectal cancer according to the Dukes Classification?What are the subdivisions of Dukes Classification stages B and C in rectal cancer?What is the role of the tumor, node, metastasis (TNM) system in rectal cancer?What are the TNM system tumor classifications in rectal cancer?What are the TNM system node classifications in rectal cancer?What are the TNM system metastasis classifications in rectal cancer?What are the approach considerations in the treatment of rectal cancer?Which factors influence sphincter and organ preservation in patients with rectal cancer?When is sphincter preservation difficult in the treatment of rectal cancer?When is local excision indicated in the treatment of rectal cancer?What are disadvantages of abdominoperineal resection in the treatment of rectal cancer?What is the role of transanal excision in the treatment of rectal cancer?What is the role of endocavitary radiation in the treatment of rectal cancer?What is the role of transanal endoscopic microsurgery (TEM) in the treatment of rectal cancer?What are the sphincter-sparing surgical options the treatment of in rectal cancer?What are indications for low anterior resection (LAR) in the treatment of rectal cancer?What are indications for total mesorectal excision (TME) in the treatment of rectal cancer?How effective is mesorectal excision (TME) in the treatment of rectal cancer?When is coloanal anastomosis (CAA) indicated in the treatment of rectal cancer?When is abdominal perineal resection (APR) indicated in the treatment of rectal cancer?How is an abdominal perineal resection (APR) performed in the treatment of rectal cancer?How is liver metastasis treated in patients with rectal cancer?What are the options for adjuvant care in the treatment of rectal cancer?What is the role of adjuvant radiation therapy in the treatment of rectal cancer?How effective is postoperative radiation therapy in the treatment of rectal cancer?What is the role of intraoperative radiation therapy in the treatment of rectal cancer?What is the role of adjuvant chemotherapy in the management of rectal cancer?Which chemotherapeutic agents are used in the treatment of rectal cancer?How effective is chemotherapy in the treatment of rectal cancer?What is the role of biologic therapy in the treatment of rectal cancer?What is the role of adjuvant chemoradiation in the treatment of rectal cancer?What is the role of radioembolization in the treatment of rectal cancer?What dietary recommendations are indicated in the treatment of rectal cancer?What are the USMSTF guidelines for long-term monitoring of patients with rectal cancer?What are the NCCN guidelines for long-term monitoring of patients with rectal cancer?Which organizations have issued guidelines on rectal cancer screening?What are the risk factors and ACS/USMSTF/ACR guidelines for rectal cancer screening?Which tests are recommended to detect adenomatous polyps and cancer?Which tests are recommended to detect rectal cancer?What are the updated 2018 ACS colorectal screening guidelines?What are USPSTF guidelines on rectal cancer screening?What are the stool-based tests used in rectal cancer screening and their recommended intervals?What are the direct visualization tests used in rectal cancer screening and their recommended intervals?What are the ACP recommended strategies for rectal cancer screening?What are the ACG guidelines for rectal cancer screening?What are NCCN screening recommendations for patients at increased risk of rectal cancer?Which syndromes associated with rectal cancer are addressed in the NCCN guidelines?When is further evaluation indicated for high-risk syndromes in rectal cancer screening?When is testing for Lynch syndrome (HNPCC) indicated in patients with rectal cancer?What are the guidelines for testing in familial risk-rectal cancer?What are the colonoscopy findings and associated recommended follow up in patients at risk for rectal cancer?What are recommended surveillance intervals for patients with serrated lesions and risk for rectal cancer?What are the guidelines for surveillance of patients with familial adenomatous polyposis (FAP) and risk for rectal cancer?What are the recommendations for surveillance of patients with attenuated familial adenomatous polyposis (FAP) and risk for rectal cancer?What is the role of genetic testing in patients with familial adenomatous polyposis (FAP) and risk for rectal cancer?In addition to genetic counseling, what are the recommendations for surveillance and treatment of patients with familial adenomatous polyposis (FAP) and risk for rectal cancer?What are the NCCN guidelines on adjuvant therapy regimens for rectal cancer?What are the NCCN guidelines on adjuvant therapy for stage II or stage III rectal cancer?What are the guidelines on follow-up care for survivors of stage II and stage III rectal cancer?What are the USMSTF guidelines on colonoscopy after rectal cancer resection?What are the recommendations on gene mutation testing in metastatic colorectal carcinoma (mCRC)?What are the ASCRS guidelines on surgery for rectal cancer?What are the goals of drug treatment in rectal cancer?Which medications in the drug class Vaccine are used in the treatment of Rectal Cancer?Which medications in the drug class Antineoplastic agents are used in the treatment of Rectal Cancer?

Author

Burt Cagir, MD, FACS, Clinical Professor of Surgery, Geisinger Commonwealth School of Medicine; Director, General Surgery Residency Program, Guthrie Robert Packer Hospital; Attending Surgeon, Guthrie Robert Packer Hospital and Corning Hospital

Disclosure: Nothing to disclose.

Coauthor(s)

Douglas R Trostle, MD, MBA, FACS, Chairman of Surgery, The Guthrie Clinic

Disclosure: Nothing to disclose.

Gabriel O Ologun, MD, Resident Physician, Department of Surgery, Guthrie Robert Packer Hospital

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

N Joseph Espat, MD, MS, FACS, Harold J Wanebo Professor of Surgery, Assistant Dean of Clinical Affairs, Boston University School of Medicine; Chairman, Department of Surgery, Director, Adele R Decof Cancer Center, Roger Williams Medical Center

Disclosure: Nothing to disclose.

Additional Contributors

Elwyn C Cabebe, MD, Physician Partner, Valley Medical Oncology Consultants; Medical Director of Oncology, Clinical Liason Physician, Cancer Care Committee, Good Samaritan Hospital

Disclosure: Nothing to disclose.

Philip Schulman, MD, Chief, Medical Oncology, Department of Medicine, Memorial Sloan-Kettering Cancer Center

Disclosure: Nothing to disclose.

Acknowledgements

Elizabeth Cirincione, MD Director of Colon and Rectal Surgery, Department of Surgery, Nassau University Medical Center

Disclosure: Nothing to disclose.

Wendy Hu, MD Consulting Staff, Department of Hematology/Oncology and Bone Marrow Transplantation, Huntington Memorial Medical Center

Wendy Hu, MD is a member of the following medical societies: American Association for the Advancement of Science, American College of Physicians, American Society for Blood and Marrow Transplantation, American Society of Hematology, and Physicians for Social Responsibility

Disclosure: Nothing to disclose.

References

  1. American Cancer Society. Cancer Facts & Figures 2020. American Cancer Society. Available at https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2020/cancer-facts-and-figures-2020.pdf. Accessed: March 11, 2020.
  2. Giovannucci E, Wu K. Cancers of the colon and rectum. Schottenfeld D, Fraumeni J, eds. Cancer. Epidemiology and Prevention. 3rd ed. Oxford University Press; 2006.
  3. Anagnostopoulos G, Sakorafas GH, Kostopoulos P, et al. Squamous cell carcinoma of the rectum: a case report and review of the literature. Eur J Cancer Care (Engl). 2005 Mar. 14(1):70-4. [View Abstract]
  4. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Rectal Cancer. Available at http://www.nccn.org/professionals/physician_gls/PDF/rectal.pdf. Version 2.2020 — March 3, 2020; Accessed: March 11, 2020.
  5. Kapiteijn E, Marijnen CA, Nagtegaal ID, Putter H, Steup WH, Wiggers T. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer. N Engl J Med. 2001 Aug 30. 345(9):638-46. [View Abstract]
  6. Leggett BA, Devereaux B, Biden K, Searle J, Young J, Jass J. Hyperplastic polyposis: association with colorectal cancer. Am J Surg Pathol. 2001 Feb. 25(2):177-84. [View Abstract]
  7. Wheeler JM, Bodmer WF, Mortensen NJ. DNA mismatch repair genes and colorectal cancer. Gut. 2000 Jul. 47(1):148-53. [View Abstract]
  8. Siegel RL, Fedewa SA, Anderson WF, Miller KD, Ma J, Rosenberg PS, et al. Colorectal Cancer Incidence Patterns in the United States, 1974–2013. J Natl Cancer Inst. 28 February 2017. 109:
  9. Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal cancer statistics, 2020. CA Cancer J Clin. 2020 Mar 5. [View Abstract]
  10. World Health Organization. Colorectal Cancer. WHO. Available at http://gco.iarc.fr/today/data/factsheets/cancers/10_8_9-Colorectum-fact-sheet.pdf. 2018; Accessed: March 11, 2020.
  11. Surveillance, Epidemiology, and End Results Program. SEER Stat Fact Sheets: Colon and Rectum Cancer. National Cancer Institute. Available at http://seer.cancer.gov/statfacts/html/colorect.html. Accessed: March 11, 2020.
  12. Yothers G, Sargent DJ, Wolmark N, et al. Outcomes Among Black Patients With Stage II and III Colon Cancer Receiving Chemotherapy: An Analysis of ACCENT Adjuvant Trials. J Natl Cancer Inst. 2011 Oct 19. 103(20):1498-1506. [View Abstract]
  13. Robbins AS, Siegel RL, Jemal A. Racial disparities in stage-specific colorectal cancer mortality rates from 1985 to 2008. J Clin Oncol. 2012 Feb 1. 30(4):401-5. [View Abstract]
  14. Rothwell PM, Fowkes GR, Belch JF, Ogawa H, Warlow CP, Meade TW. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomized trials. Lancet. Dec 7/2010; Early online publication.
  15. Burn J, Gerdes AM, Macrae F, et al. Long-term effect of aspirin on cancer risk in carriers of hereditary colorectal cancer: an analysis from the CAPP2 randomised controlled trial. Lancet. 2011 Dec 17. 378(9809):2081-7. [View Abstract]
  16. Rothwell PM, Price JF, Fowkes FG, Zanchetti A, Roncaglioni MC, Tognoni G, et al. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012 Apr 28. 379(9826):1602-12. [View Abstract]
  17. Stitzenberg KB, Sanoff HK, Penn DC, Meyers MO, Tepper JE. Practice patterns and long-term survival for early-stage rectal cancer. J Clin Oncol. 2013 Dec 1. 31(34):4276-82. [View Abstract]
  18. Eu KW, Seow-Choen F, Ho JM, Ho YH, Leong AF. Local recurrence following rectal resection for cancer. J R Coll Surg Edinb. 1998 Dec. 43(6):393-6. [View Abstract]
  19. Thong MS, Mols F, Lemmens VE, et al. Impact of preoperative radiotherapy on general and disease-specific health status of rectal cancer survivors: a population-based study. Int J Radiat Oncol Biol Phys. 2011 Nov 1. 81(3):e49-58. [View Abstract]
  20. Harrison P. Proinflammatory Diet Contributes to CRC Risk in Both Sexes. Medscape Medical News. Available at https://www.medscape.com/viewarticle/891665. January 23, 2018; Accessed: January 30, 2018.
  21. Tabung FK, Liu L, Wang W, Fung TT, Wu K, Smith-Warner SA, et al. Association of Dietary Inflammatory Potential With Colorectal Cancer Risk in Men and Women. JAMA Oncol. 2018 Jan 18. [View Abstract]
  22. Gianfredi V, Nucci D, Salvatori T, Dallagiacoma G, Fatigoni C, Moretti M, et al. Rectal Cancer: 20% Risk Reduction Thanks to Dietary Fibre Intake. Systematic Review and Meta-Analysis. Nutrients. 2019 Jul 12. 11 (7):916-32. [View Abstract]
  23. Chao A, Thun MJ, Connell CJ, et al. Meat consumption and risk of colorectal cancer. JAMA. 2005 Jan 12. 293(2):172-82. [View Abstract]
  24. Baron JA, Beach M, Mandel JS, van Stolk RU, Haile RW, Sandler RS, et al. Calcium supplements for the prevention of colorectal adenomas. Calcium Polyp Prevention Study Group. N Engl J Med. 1999 Jan 14. 340(2):101-7. [View Abstract]
  25. Ferrari P, Jenab M, Norat T, Moskal A, Slimani N, Olsen A, et al. Lifetime and baseline alcohol intake and risk of colon and rectal cancers in the European prospective investigation into cancer and nutrition (EPIC). Int J Cancer. 2007 Nov 1. 121(9):2065-72. [View Abstract]
  26. Kabat GC, Howson CP, Wynder EL. Beer consumption and rectal cancer. Int J Epidemiol. 1986 Dec. 15(4):494-501. [View Abstract]
  27. Tsoi KK, Pau CY, Wu WK, Chan FK, Griffiths S, Sung JJ. Cigarette smoking and the risk of colorectal cancer: a meta-analysis of prospective cohort studies. Clin Gastroenterol Hepatol. 2009 Jun. 7(6):682-688.e1-5. [View Abstract]
  28. Phipps AI, Baron J, Newcomb PA. Prediagnostic smoking history, alcohol consumption, and colorectal cancer survival: The Seattle Colon Cancer Family Registry. Cancer. 2011 Nov 1. 117(21):4948-57. [View Abstract]
  29. Johns LE, Houlston RS. A systematic review and meta-analysis of familial colorectal cancer risk. Am J Gastroenterol. 2001 Oct. 96(10):2992-3003. [View Abstract]
  30. Burt RW. Familial risk and colorectal cancer. Gastroenterol Clin North Am. 1996 Dec. 25(4):793-803. [View Abstract]
  31. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High Risk Assessment: Colorectal. NCCN. Available at http://www.nccn.org/professionals/physician_gls/pdf/genetics_colon.pdf. Version 3.2019 — December 13, 2019; Accessed: March 11, 2020.
  32. Henderson D. Home-Based Stool Test Detects 79% of Colorectal Cancers. Medscape Medical News. Available at http://www.medscape.com/viewarticle/820194. Accessed: February 10, 2014.
  33. Lee JK, Liles EG, Bent S, Levin TR, Corley DA. Accuracy of Fecal Immunochemical Tests for Colorectal Cancer: Systematic Review and Meta-analysis. Ann Intern Med. 2014. 160:171-181.
  34. de Wijkerslooth TR, Stoop EM, Bossuyt PM, Meijer GA, van Ballegooijen M, van Roon AH, et al. Immunochemical Fecal Occult Blood Testing Is Equally Sensitive for Proximal and Distal Advanced Neoplasia. Am J Gastroenterol. 2012 Jul 31. [View Abstract]
  35. Nelson H, Petrelli N, Carlin A, Couture J, Fleshman J, Guillem J, et al. Guidelines 2000 for colon and rectal cancer surgery. J Natl Cancer Inst. 2001 Apr 18. 93(8):583-96. [View Abstract]
  36. Schoen RE, Pinsky PF, Weissfeld JL, Yokochi LA, Church T, Laiyemo AO, et al. Colorectal-Cancer Incidence and Mortality with Screening Flexible Sigmoidoscopy. N Engl J Med. 2012 May 21. [View Abstract]
  37. [Guideline] US Preventive Services Task Force, Bibbins-Domingo K, Grossman DC, Curry SJ, Davidson KW, Epling JW Jr, et al. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2016 Jun 21. 315 (23):2564-75. [View Abstract]
  38. McNamara D. Evidence Backs Younger Screening for Colorectal Cancer. Medscape Medical News. Available at https://www.medscape.com/viewarticle/887768. October 30, 2017; Accessed: November 15, 2017.
  39. Brounts LR, Lehmann RK, Lesperance KE, Brown TA, Steele SR. Improved rates of colorectal cancer screening in an equal access population. Am J Surg. 2009 May. 197(5):609-12; discussion 612-3. [View Abstract]
  40. Baxter NN, Garcia-Aguilar J. Organ preservation for rectal cancer. J Clin Oncol. 2007 Mar 10. 25(8):1014-20. [View Abstract]
  41. Rothenberger D, Garcia-Aquilar J. Rectal cancer, local treatment. Current Therapy in Colon and Rectal Surgery. 2nd ed. Philadelphia, Pa: Mosby; 2005.
  42. Peng J, Chen W, Venook AP, et al. Long-term outcome of early-stage rectal cancer undergoing standard resection and local excision. Clin Colorectal Cancer. 2011 Mar 1. 10(1):37-41. [View Abstract]
  43. Brooks M. Local Excision for Rectal Cancer Increasing, but Inferior. Medscape Medical News. Available at http://www.medscape.com/viewarticle/815376. Accessed: December 23, 2013.
  44. Weiser MR, Landmann RG, Wong WD, Shia J, Guillem JG, Temple LK, et al. Surgical salvage of recurrent rectal cancer after transanal excision. Dis Colon Rectum. 2005 Jun. 48(6):1169-75. [View Abstract]
  45. Bullard KM, Rothenberger DA. Colon, rectum, and anus. Schwartz SE, ed. Principles of Surgery. 8th ed. New York, NY: McGraw Hill; 2005.
  46. Li S, Chi P, Lin H, Lu X, Huang Y. Long-term outcomes of laparoscopic surgery versus open resection for middle and lower rectal cancer: an NTCLES study. Surg Endosc. 2011 Oct. 25(10):3175-82. [View Abstract]
  47. Green BL, Marshall HC, Collinson F, Quirke P, Guillou P, Jayne DG, et al. Long-term follow-up of the Medical Research Council CLASICC trial of conventional versus laparoscopically assisted resection in colorectal cancer. Br J Surg. 2012 Nov 6. [View Abstract]
  48. Bonjer HJ, Deijen CL, Abis GA, Cuesta MA, van der Pas MH, de Lange-de Klerk ES, et al. A randomized trial of laparoscopic versus open surgery for rectal cancer. N Engl J Med. 2015 Apr 2. 372(14):1324-32. [View Abstract]
  49. van Helmond J, Beart RW. Cancer of the rectum: Operative management and adjuvant therapy. Current Therapy in Colon and Rectal Surgery. 2nd ed. Philadelphia, Pa: Mosby; 2005.
  50. Maurer CA, Renzulli P, Kull C, et al. The impact of the introduction of total mesorectal excision on local recurrence rate and survival in rectal cancer: long-term results. Ann Surg Oncol. 2011 Jul. 18(7):1899-906. [View Abstract]
  51. Fujita S, Akasu T, Mizusawa J, Saito N, Kinugasa Y, Kanemitsu Y, et al. Postoperative morbidity and mortality after mesorectal excision with and without lateral lymph node dissection for clinical stage II or stage III lower rectal cancer (JCOG0212): results from a multicentre, randomised controlled, non-inferiority trial. Lancet Oncol. 2012 Jun. 13(6):616-21. [View Abstract]
  52. Han YD, Kim WR, Park SW, Cho MS, Hur H, Min BS, et al. Predictors of Pathologic Complete Response in Rectal Cancer Patients Undergoing Total Mesorectal Excision After Preoperative Chemoradiation. Medicine (Baltimore). Nov 2015. 94(45):e1971. [View Abstract]
  53. Bujko K, Rutkowski A, Chang GJ, Michalski W, Chmielik E, Kusnierz J. Is the 1-cm Rule of Distal Bowel Resection Margin in Rectal Cancer Based on Clinical Evidence? A Systematic Review. Indian J Surg Oncol. 2012 Jun. 3 (2):139-46. [View Abstract]
  54. Quan D, Gallinger S, Nhan C, Auer RA, Biagi JJ, Fletcher GG, et al. The role of liver resection for colorectal cancer metastases in an era of multimodality treatment: A systematic review. Surgery. 2012 Jun. 151(6):860-70. [View Abstract]
  55. Dhir M, Lyden ER, Wang A, et al. Influence of margins on overall survival after hepatic resection for colorectal metastasis: a meta-analysis. Ann Surg. 2011 Aug. 254(2):234-42. [View Abstract]
  56. Margalit DN, Mamon HJ, Ancukiewicz M, et al. Tolerability of combined modality therapy for rectal cancer in elderly patients aged 75 years and older. Int J Radiat Oncol Biol Phys. 2011 Dec 1. 81(5):e735-41. [View Abstract]
  57. Ceelen WP, Van Nieuwenhove Y, Fierens K. Preoperative chemoradiation versus radiation alone for stage II and III resectable rectal cancer. Cochrane Database Syst Rev. 2009 Jan 21. CD006041. [View Abstract]
  58. Jakobsen A, Ploen J, Vuong T, Appelt A, Lindebjerg J, Rafaelsen SR. Dose-Effect Relationship in Chemoradiotherapy for Locally Advanced Rectal Cancer: A Randomized Trial Comparing Two Radiation Doses. Int J Radiat Oncol Biol Phys. 2012 May 15. [View Abstract]
  59. Wong RK, Tandan V, De Silva S, Figueredo A. Pre-operative radiotherapy and curative surgery for the management of localized rectal carcinoma. Cochrane Database Syst Rev. 2007 Apr 18. CD002102. [View Abstract]
  60. Cassidy RJ, Liu Y, Patel K, Zhong J, Steuer CE, Kooby DA, et al. Can we eliminate neoadjuvant chemoradiotherapy in favor of neoadjuvant multiagent chemotherapy for select stage II/III rectal adenocarcinomas: Analysis of the National Cancer Database. Cancer. March 2017. 123(5):783-793. [View Abstract]
  61. Ng K, Ogino S, Meyerhardt JA, et al. Relationship Between Statin Use and Colon Cancer Recurrence and Survival: Results From CALGB 89803. J Natl Cancer Inst. 2011 Oct 19. 103(20):1540-51. [View Abstract]
  62. Kidwell KM, Yothers G, Ganz PA, Land SR, Ko CY, Cecchini RS, et al. Long-term neurotoxicity effects of oxaliplatin added to fluorouracil and leucovorin as adjuvant therapy for colon cancer: Results from National Surgical Adjuvant Breast and Bowel Project trials C-07 and LTS-01. Cancer. 2012 May 8. [View Abstract]
  63. Cao S, Bhattacharya A, Durrani FA, Fakih M. Irinotecan, oxaliplatin and raltitrexed for the treatment of advanced colorectal cancer. Expert Opinion on Pharmacotherapy. 2006. 7(6):687-703.
  64. Maughan TS, Adams RA, Smith CG, et al. Addition of cetuximab to oxaliplatin-based first-line combination chemotherapy for treatment of advanced colorectal cancer: results of the randomised phase 3 MRC COIN trial. Lancet. 2011 Jun 18. 377(9783):2103-14. [View Abstract]
  65. Douillard JY, Siena S, Cassidy J, Tabernero J, Burkes R, Barugel M, et al. Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study. J Clin Oncol. 2010 Nov 1. 28(31):4697-705. [View Abstract]
  66. Peeters M, Price TJ, Cervantes A, Sobrero AF, Ducreux M, Hotko Y. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. J Clin Oncol. 2010 Nov 1. 28(31):4706-13. [View Abstract]
  67. Simkens LH, Koopman M, Mol L, et al. Influence of body mass index on outcome in advanced colorectal cancer patients receiving chemotherapy with or without targeted therapy. Eur J Cancer. 2011 Nov. 47(17):2560-7. [View Abstract]
  68. Boisen MK, Johansen JS, Dehlendorff C, Larsen JS, Osterlind K, Hansen J, et al. Primary tumor location and bevacizumab effectiveness in patients with metastatic colorectal cancer. Ann Oncol. 2013 Jul 17. [View Abstract]
  69. Ryan ÉJ, O'Sullivan DP, Kelly ME, Syed AZ, Neary PC, O'Connell PR, et al. Meta-analysis of the effect of extending the interval after long-course chemoradiotherapy before surgery in locally advanced rectal cancer. Br J Surg. 2019 Jun 19. 10(9):e0138720. [View Abstract]
  70. Ebert MP, Tanzer M, Balluff B, et al. TFAP2E-DKK4 and chemoresistance in colorectal cancer. N Engl J Med. 2012 Jan 5. 366(1):44-53. [View Abstract]
  71. Hendlisz A, Van den Eynde M, Peeters M, Maleux G, Lambert B, Vannoote J, et al. Phase III trial comparing protracted intravenous fluorouracil infusion alone or with yttrium-90 resin microspheres radioembolization for liver-limited metastatic colorectal cancer refractory to standard chemotherapy. J Clin Oncol. 2010 Aug 10. 28(23):3687-94. [View Abstract]
  72. Wasan HS, et al; FOXFIRE trial investigators., SIRFLOX trial investigators., FOXFIRE-Global trial investigators., van Hazel G, Sharma RA. First-line selective internal radiotherapy plus chemotherapy versus chemotherapy alone in patients with liver metastases from colorectal cancer (FOXFIRE, SIRFLOX, and FOXFIRE-Global): a combined analysis of three multicentre, randomised, phase 3 trials. Lancet Oncol. 2017 Sep. 18 (9):1159-1171. [View Abstract]
  73. Santiago I, Barata M, Figueiredo N, Parés O, Henriques V, Galzerano A, et al. The split scar sign as an indicator of sustained complete response after neoadjuvant therapy in rectal cancer. Eur Radiol. 2019 Jul 26. [View Abstract]
  74. Song M, Wu K, Meyerhardt JA, Ogino S, Wang M, Fuchs CS, et al. Fiber Intake and Survival After Colorectal Cancer Diagnosis. JAMA Oncol. 2017 Nov 2. [View Abstract]
  75. Davenport L. High Fiber Intake Tied to Improved Colon Cancer Survival. Medscape Medical News. Available at https://www.medscape.com/viewarticle/888006. November 3, 2017; Accessed: November 8, 2017.
  76. FDA: Gardasil approved to prevent anal cancer. U.S. Food and Drug Administration. Available at https://wayback.archive-it.org/7993/20161022094130/http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2010/ucm237941.htm. December 22, 2010; Accessed: November 8, 2017.
  77. Mulcahy N. GSK’s HPV Vaccine, Cervarix, No Longer Available in US. Medscape Medical News. Available at https://www.medscape.com/viewarticle/870853. October 24, 2016; Accessed: November 8, 2017.
  78. [Guideline] Kahi CJ, Boland CR, Dominitz JA, Giardiello FM, Johnson DA, Kaltenbach T, et al. Colonoscopy Surveillance After Colorectal Cancer Resection: Recommendations of the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology. 2016 Mar. 150 (3):758-768.e11. [View Abstract]
  79. [Guideline] Levin B, Lieberman DA, McFarland B, et al. Screening and surveillance for the early detection of colorectal cancer and adenomatous polyps, 2008: a joint guideline from the American Cancer Society, the US Multi-Society Task Force on Colorectal Cancer, and the American College of Radiology. CA Cancer J Clin. 2008 May-Jun. 58 (3):130-60. [View Abstract]
  80. [Guideline] American Cancer Society Guideline for Colorectal Cancer Screening. American Cancer Society. Available at https://www.cancer.org/cancer/colon-rectal-cancer/detection-diagnosis-staging/acs-recommendations.html. May 30, 2018; Accessed: March 11, 2020.
  81. Wilt TJ, Harris RP, Qaseem A, High Value Care Task Force of the American College of Physicians. Screening for cancer: advice for high-value care from the American College of Physicians. Ann Intern Med. 2015 May 19. 162 (10):718-25. [View Abstract]
  82. [Guideline] Rex DK, Johnson DA, Anderson JC, Schoenfeld PS, Burke CA, Inadomi JM, et al. American College of Gastroenterology guidelines for colorectal cancer screening 2009 [corrected]. Am J Gastroenterol. 2009 Mar. 104 (3):739-50. [View Abstract]
  83. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Colorectal Cancer Screening. NCCN. Available at http://www.nccn.org/professionals/physician_gls/pdf/colorectal_screening.pdf. Version 2.2019 — August 2, 2019; Accessed: March 11, 2020.
  84. [Guideline] Rubenstein JH, Enns R, Heidelbaugh J, Barkun A, Clinical Guidelines Committee. American Gastroenterological Association Institute Guideline on the Diagnosis and Management of Lynch Syndrome. Gastroenterology. 2015 Sep. 149 (3):777-82; quiz e16-7. [View Abstract]
  85. [Guideline] Balmaña J, Balaguer F, Cervantes A, Arnold D, ESMO Guidelines Working Group. Familial risk-colorectal cancer: ESMO Clinical Practice Guidelines. Ann Oncol. 2013 Oct. 24 Suppl 6:vi73-80. [View Abstract]
  86. [Guideline] Stoffel EM, Mangu PB, Gruber SB, Hamilton SR, Kalady MF, Lau MW, et al. Hereditary colorectal cancer syndromes: American Society of Clinical Oncology Clinical Practice Guideline endorsement of the familial risk-colorectal cancer: European Society for Medical Oncology Clinical Practice Guidelines. J Clin Oncol. 2015 Jan 10. 33 (2):209-17. [View Abstract]
  87. [Guideline] Syngal S, Brand RE, Church JM, Giardiello FM, Hampel HL, Burt RW, et al. ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. Am J Gastroenterol. 2015 Feb. 110 (2):223-62; quiz 263. [View Abstract]
  88. Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004 Feb 18. 96 (4):261-8. [View Abstract]
  89. [Guideline] Gupta S, Lieberman D, Anderson JC, Burke CA, Dominitz JA, Kaltenbach T, et al. Recommendations for Follow-Up After Colonoscopy and Polypectomy: A Consensus Update by the US Multi-Society Task Force on Colorectal Cancer. Gastroenterology. 2020 Mar. 158 (4):1131-1153.e5. [View Abstract]
  90. [Guideline] Rutter MD, East J, Rees CJ, Cripps N, Docherty J, Dolwani S, et al. British Society of Gastroenterology/Association of Coloproctology of Great Britain and Ireland/Public Health England post-polypectomy and post-colorectal cancer resection surveillance guidelines. Gut. 2020 Feb. 69 (2):201-223. [View Abstract]
  91. [Guideline] Church J, Simmang C, Standards Task Force, American Society of Colon and Rectal Surgeons, Collaborative Group of the Americas on Inherited Colorectal Cancer and the Standards Committee of The American Society of Colon and Rectal Surgeons. Practice parameters for the treatment of patients with dominantly inherited colorectal cancer (familial adenomatous polyposis and hereditary nonpolyposis colorectal cancer). Dis Colon Rectum. 2003 Aug. 46 (8):1001-12. [View Abstract]
  92. [Guideline] Monson JR, Weiser MR, Buie WD, Chang GJ, Rafferty JF, Buie WD, et al. Practice parameters for the management of rectal cancer (revised). Dis Colon Rectum. 2013 May. 56 (5):535-50. [View Abstract]
  93. [Guideline] Glynne-Jones R, Wyrwicz L, Tiret E, Brown G, Rödel C, Cervantes A, et al. Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018 Oct 1. 29 (Supplement_4):iv263. [View Abstract]
  94. [Guideline] Meyerhardt JA, Mangu PB, Flynn PJ, Korde L, Loprinzi CL, Minsky BD, et al. Follow-up care, surveillance protocol, and secondary prevention measures for survivors of colorectal cancer: American Society of Clinical Oncology clinical practice guideline endorsement. J Clin Oncol. 2013 Dec 10. 31 (35):4465-70. [View Abstract]
  95. [Guideline] Labianca R, Nordlinger B, Beretta GD, Mosconi S, Mandalà M, Cervantes A, et al. Early colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013 Oct. 24 Suppl 6:vi64-72. [View Abstract]
  96. [Guideline] Steele SR, Chang GJ, Hendren S, Weiser M, Irani J, Buie WD, et al. Practice Guideline for the Surveillance of Patients After Curative Treatment of Colon and Rectal Cancer. Dis Colon Rectum. 2015 Aug. 58 (8):713-25. [View Abstract]
  97. [Guideline] Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, et al. Extended RAS Gene Mutation Testing in Metastatic Colorectal Carcinoma to Predict Response to Anti-Epidermal Growth Factor Receptor Monoclonal Antibody Therapy: American Society of Clinical Oncology Provisional Clinical Opinion Update 2015. J Clin Oncol. 2016 Jan 10. 34 (2):179-85. [View Abstract]
  98. [Guideline] Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016 Aug. 27 (8):1386-422. [View Abstract]
  99. Nielsen A, Munk C, Kjaer SK. Trends in incidence of anal cancer and high-grade anal intraepithelial neoplasia in Denmark, 1978-2008. Int J Cancer. 2012 Mar 1. 130(5):1168-73. [View Abstract]
  100. Mulcahy N. NCCN: Test all colorectal cancers for Lynch syndrome. Medscape Medical News. March 14, 2014.
  101. Cheng X, Chen VW, Steele B, Ruiz B, Fulton J, Liu L, et al. Subsite-specific incidence rate and stage of disease in colorectal cancer by race, gender, and age group in the United States, 1992-1997. Cancer. 2001 Nov 15. 92(10):2547-54. [View Abstract]

Diagnostics. Staging and workup of rectal cancer patients.

Diagnostics. Staging and workup of rectal cancer patients.

Staging and treatment. Rectal cancer treatment algorithm (surgery followed by adjuvant chemotherapy and radiotherapy). Initial stages are Endorectal ultrasound staging (uT).

Diagnostics. Staging and workup of rectal cancer patients.

Staging and treatment. Rectal cancer treatment algorithm (surgery followed by adjuvant chemotherapy and radiotherapy). Initial stages are Endorectal ultrasound staging (uT).

Rectal Cancer Stages TNM Staging Duke Staging 5-Year Survival
Stage IT1-2 N0 M0A>90%
Stage IIAT3 N0 M0B60%-85%
BT4 N0 M060%-85%
Stage IIIAT1-2 N1 M0C55%-60%
BT3-4 N1 M035%-42%
CT1-4 N2 M025%-27%
Stage IVT1-4 N0-2 M15%-7%
Resection Margins Proximal Resection Margin Distal Resection Margin
Ideal Margins 5 cm or more2 cm or more
Minimally acceptable margins 5 cm or more0.5-1 cm 
COLON AND RECTAL CANCER



COMMON CHEMOTHERAPY REGIMENS



FOLFOX (every 2 weeks) Oxaliplatin 85 mg/m2 day 1



Leucovorin 200 mg/m2 day 1



5-FU 400 mg/m2 IV Bolus day 1 and 2



5-FU 600 mg/m2 IV Infusion day 1 and 2 (22 hours)



FOLFOX 4



(every 2 weeks)



(4 cycles)



Oxaliplatin 85 mg/m2 day 1



Leucovorin 200 mg/m2 day 1



5-FU 400 mg/m2 IV Bolus day 1 and 2



5-FU 2400 mg/m2 IV Infusion day 1 (46 hours)



mFOLFOX 6



(Every 2 weeks)



(4 cycles)



Oxaliplatin 85 mg/m2 day 1



Leucovorin 400 mg/m2 day 1



5-FU 400 mg/m2 IV Bolus day 1 and 2



5-FU 1200 mg/m2 IV Infusion day 2 days



CapeOX



(Twice daily x 14 days)



(every 3 weeks)



Oxaliplatin 130 mg/m2 day 1



Capecitabine 850 mg/m2 PO BID for 14 days



FOLFIRI



(every 2 weeks)



Irinotecan 165 mg/m2 day 1



Leucovorin 200 mg/m2 day 1



5-FU 400 mg/m2 IV Bolus day 1 and 2



5-FU 600 mg/m2 IV Infusion day 1 and 2 (22 hours)



FOLFOXIRI



(every 2 weeks)



Irinotecan 180 mg/m2 day 1



Oxaliplatin 85 mg/m2 day 1



Leucovorin 200 mg/m2 day 1



5-FU 3200 mg/m2 IV Infusion day (48 hours)



Bevacizumab 5-10 mg/kg IV every 2 weeks with chemotherapy
Cetuximab 400 mg/m2 IV day 1, then 250 mg/m2 IV weekly
Parameter Organization
ESMO (2013)[95] ASCO (2013)[94] NCCN (2019)[4] ASCRS (2015)[96]
History and physical examEvery 3-6 mo for 3 y, then every 6 -12 mo at 4 and 5 yEvery 3-6 mo for 3 y, then every 6 mo to 5 yEvery 3-6 mo for 2 y, then every 6 mo to 5 yEvery 3-6 mo for 2 y, then every 6 mo to 5 y
CEAEvery 3-6 mo for 3 y, then every 6 -12 mo at 4 and 5 yEvery 3 mo for 3 y*Every 3-6 mo for 2 y, then every 6 mo to 5 yEvery 3-6 mo for 2 y, then every 6 mo to 5 y
Chest CT*Every 6-12 mo for first 3 yEvery 1 y for 3 yEvery 6-12 mo to 5 yEvery 1 y for 5 y
Colonoscopy**At y 1 after surgery, and every 3-5 y thereafterAt 1 y, then every 5 y, dictated by the findings on the previous colonoscopyAt 1 y, 3 y, and 5 y if negativeAt y 1 after surgery, and every 3-5 y dictated by the findings on the first postoperative examination.
Abdominal CT*Every 6-12 mo for first 3 yEvery 1 y for 3 yEvery 6-12 mo to 5 yEvery 1 y for 5 y
ESMO = European Society of Medical Oncology; ASCO = American Society of Clinical Oncology; NCCN = National Comprehensive Cancer Network; American Society of Colon and Rectal Surgeons = ASCRS CEA = carcinoembryonic antigen; CT = computed tomography * For patients at high risk for recurrence (eg, lymphatic or venous invasion, or poorly differentiated tumors). **Colonoscopy should be performed 3-6 mo postoperatively if preoperative colonoscopy was not done, due to an obstructing lesion; otherwise, colonoscopy in 1 y; if abnormal, repeat in 1 year; if no advanced adenoma (ie, villous polyp, polyp >1 cm, or high-grade dysplasia), repeat in 3 y, then every 5 y.