Home » Articles posted by UtahGastroBlog (Page 2)

Author Archives: UtahGastroBlog

The Diagnosis and Management of Focal Liver Lesions

Please review the excellent ACG Clinical Guideline on “The Diagnosis and Management of Focal Liver Lesions“.
Focal liver lesions (FLL) have been a common reason for consultation faced by gastroenterologists and hepatologists. The increasing and widespread use of imaging studies has led to an increase in detection of incidental FLL. It is important to consider not only malignant liver lesions, but also benign solid and cystic liver lesions such as hemangioma, focal nodular hyperplasia, hepatocellular adenoma, and hepatic cysts, in the differential diagnosis.
      
Solid Focal Liver Lesions (FLL)
Suspected hepatocellular carcinoma
  • An MRI or triple-phase CT should be obtained in patients with cirrhosis with an ultrasound showing a lesion of >1cm.
  • Patients with chronic liver disease, especially with cirrhosis, who present with a solid FLL are at a very high risk for having HCC and must be considered to have HCC until otherwise proven.
  • A diagnosis of HCC can be made with CT or MRI if the typical characteristics are present: a solid FLL with enhancement in the arterial phase with washout in the delayed venous phase should be considered to have HCC until otherwise proven.
  • If an FLL in a patient with cirrhosis does not have typical characteristics of HCC, then a biopsy should be performed in order to make the diagnosis.
Suspected cholangiocarcinoma
  • MRI or CT should be obtained if CCA is suspected clinically or by ultrasound.
  • A liver biopsy should be obtained to establish the diagnosis of CCA if the patient is non operable.

Suspected hepatocellular adenoma

  • Oral contraceptives, hormone-containing IUDs, and anabolic steroids are to be avoided in patients with hepatocellular adenoma.
  • Obtaining a biopsy should be reserved for cases in which imaging is inconclusive and biopsy is deemed necessary to make treatment decisions.
  • Pregnancy is not generally contraindicated in cases of hepatocellular adenoma < 5 cm and an individualized approach is advocated for these patients.
  • In hepatocellular adenoma ≥ 5 cm, intervention through surgical or nonsurgical modalities is recommended, as there is a risk of rupture and malignancy.
  • If no therapeutic intervention is pursued, lesions suspected of being hepatocellular adenoma require follow-up CT or MRI at 6- to 12-month intervals. The duration of monitoring is based on the growth patterns and stability of the lesion over time.
  • Clinical variants of hepatocellular adenoma
    • Liver adenomatosis.
      • Multiple adenomas, defined as between > 3 and ≥ 10 lesions, are collectively referred to as liver adenomatosis. These multiple lesions have identical clinical, histological, and radiographic features as hepatocellular adenomas and are managed in the same manner.
    • Telangiectatic hepatocellular adenoma
      • Previously known as telangiectatic focal nodular hyperplasia, telangiectatic hepatocellular adenoma (THCA) has recently been reclassified as a subcategory of inflammatory hepatocellular adenoma. THCA should be managed as aggressively as hepatocellular adenomas as they are likely to be symptomatic, prone to hemorrhage, and may contain focal areas of necrosis. The high likelihood of hemorrhage combined with an unknown potential of transformation to HCC makes surgery the recommended treatment.

Suspected hemangioma

  • An MRI or CT scan should be obtained to confirm a diagnosis of hemangioma.
  • Liver biopsy should be avoided if the radiologic features of a hemangioma are present.
  • Pregnancy and the use of oral contraceptives or anabolic steroids are not contraindicated in patients with a hemangioma.
  • Regardless of the size, no intervention is required for asymptomatic hepatic hemangiomas. Symptomatic patients with impaired quality of life can be referred for surgical or nonsurgical therapeutic modalities by an experienced team.

Suspected focal nodular hyperplasia

  • An MRI or CT scan should be obtained to confirm a diagnosis of FNH. A liver biopsy is not routinely indicated to confi rm the diagnosis.
  • Pregnancy and the use of oral contraceptives or anabolic steroids are not contraindicated in patients with FNH.
  • Asymptomatic FNH does not require intervention.
  • Annual US for 2-3 years is prudent in women diagnosed with FNH who wish to continue OCP use. Individuals with a firm diagnosis of FNH who are not using OCP do not require follow-up imaging.
Suspected nodular regenerative hyperplasia
  • Liver biopsy is required to confirm the diagnosis of NRH.
  • Pregnancy and the use of oral contraceptives or anabolic steroids are not contraindicated in patients with an NRH.
  • Asymptomatic NRH does not require intervention.
  • Management of NRH is based on diagnosing and managing any underlying predisposing disease processes.
       
Cystic Focal Liver Lesions (FLL)
Suspect simple hepatic cysts
  • A hepatic cyst identified on US with septations, fenestrations, calcifications, irregular walls, or daughter cysts should prompt further evaluation with a CT or MRI.
  • Asymptomatic simple hepatic cysts should be observed with expectant management.
  • Aspiration of asymptomatic, simple hepatic cysts is not recommended.
  • Symptomatic simple hepatic cysts may be managed with laparoscopic deroofing rather than aspiration and sclerotherapy, dictated based on availability of local expertise.
Suspected biliary cystadenoma (BC) or cystadenocarcinoma (BCA)
  • Routine fluid aspiration is not recommended when BCA is suspected because of limited sensitivity and the risk of malignant dissemination.
  • Imaging characteristics suggestive of BC or BCA, such as internal septations, fenestrations, calcifications, or irregular walls, should lead to referral for surgical excision.
  • Complete surgical excision, by an experienced team, is recommended if BC or BCA is suspected.
Suspected polycystic liver disease
  • Routine medical therapy with mammalian target of rapamycin inhibitors or somatostatin analogs is not recommended.
  • Aspiration, deroofing, resection of a dominant cyst(s) can be performed based on the patient’s clinical presentation and underlying hepatic reserve.
  • Liver transplantation with or without kidney transplantation can be considered in patients with refractory symptoms and significant cyst burden.
Suspected hydatid cysts
  • MRI is preferred over CT for concomitant evaluation of the biliary tree and cystic contents.
  • Monotherapy with antihelminthic drugs is not recommended in symptomatic patients who are surgical or percutaneous treatment candidates.
  • Adjunctive therapy with antihelminthic therapy is recommended in patients undergoing PAIR or surgery, and in those with peritoneal rupture or biliary rupture.
  • Percutaneous treatment with PAIR is recommended for patients with active hydatid cysts who are not surgical candidates, who decline surgery, or who relapse after surgery.
  • Surgery, either laparoscopic or open, based on available expertise, is recommended in complicated hydatid cysts with multiple vesicles, daughter cysts, fistulas, rupture, hemorrhage, or secondary infection.

 

Am J Gastroenterol advance online publication, 19 August 2014; doi: 10.1038/ajg.2014.213

Am J Gastroenterol advance online publication, 19 August 2014; doi: 10.1038/ajg.2014.213


Imaging characteristics of solid liver lesions
HCA (hepatocellular adenoma)
US:
  • Heterogeneous; hyperechoic if steatotic but anechoic center if hemorrhage
CT:
  • Well demarcated with peripheral enhancement; homogenous more often than heterogeneous; hypodense if steatotic, hyperdense if hemorrhagic
MRI:
  • HNF1 α : signal lost on chemical shift; moderate arterial enhancement without persistent enhancement during delayed phase
  • IHCA: markedly hyperintense on T2 with stronger signal peripherally; persistent enhancement in delayed phase
  • β – Catenin: inflammatory subtype has same appearance as IHCA; noninflammatory is heterogeneous with no signal dropout on chemical shift, isointense of T1 and T2 with strong arterial enhancement and delayed washout
THCA (telangiectatic hepatocellular adenoma)
US:
  • Variable appearance
CT:
  • Hypo- to isoattenuating
MRI:
  • T1: heterogeneous and well-defined iso- to hyperintense mass. Strongly hyperintense with persistent contrast enhancement in delayed phase
Hemangioma 
US:
  • Hyperechoic with well-defined rim and with few intranodular vessels
CT:
  • Discontinuous peripheral nodular enhancement isoattenuating to aorta with progressive centripetal fi ll-in
MRI:
  • T1: hypointense; discontinuous peripheral enhancement with centripetal fi ll-in
  • T2: hyperintense relative to spleen
FNH (focal nodular hyperplasia)
US:
  • Generally isoechoic
CT:
  • Central scar. Arterial phase shows homogenous hyperdense lesion; returns to pre contrast density during portal phase that is hypo- or isodense
MRI:
  • T1: isointense or slightly hypointense. Gadolinium produces early enhancement with central scar enhancement during delayed phase
  • T2: slightly hyperintense or isointense
NRH (nodular regenerative hyperplasia)
US:
  • Isoechoic / hyperechoic
CT:
  • Nonenhancing nodules, sometimes hypodense, with variable sizes (most sub-centimeter)
MRI:
  • T1: hyperintense
  • T2: varied intensity (hypo / iso / hyperintense)

Imaging characteristics of cystic liver lesions
Simple hepatic cysts (SHCs)
US:
  • Anechoic, homogeneous,fluid filled. Smooth margins
CT:
  • Well-demarcated, water-attenuated, smooth lesion without an internal structure. No enhancement with contrast
MRI:
  • Well-defined, homogeneous lesion. No enhancement with contrast.
  • T1: hypointense signal intensity
  • T2: hyperintense signal intensity
Biliary cystadenomas (BCs) 
US:
  • Irregular walls, internal septations forming loculi
CT:
  • Heterogeneous septations, internal septations, irregular papillary growths, thickened cyst walls
MRI:
  • May appear heterogeneous.
  • T1: Hypointense signal intensity
  • T2: Hyperintense signal intensity
Polycystic liver disease (PCLD)
US:
  • Multiple hepatic cysts, similar in characteristics to SHC US findings
CT:
  • Multiple hepatic cysts, similar in characteristics to SHC CT findings
MRI:
  • Multiple hepatic cysts, similar in characteristics to SHC MRI findings
Hydatid cysts (HCs) 
US:
  • May appear similar to SHC. Progress to develop thick, calcified walls, hyperechoic / hypoechoic contents. Daughter cysts in periphery.
CT:
  • Hypodense lesion with hypervascular pericyst wall, distinct endocyst wall. Calcified walls and septa easily detected. Daughter cysts seen peripherally within mother cyst.
MRI:
  • T1: Hypointense signal intensity of cyst contents.
  • T2: Hyperintense signal intensity of cyst contents.
  • Hypointense rim on T2.
  • Daughter cysts seen peripherally within mother cyst. Collapse parasitic membranes seen as floating linear structures within cyst.
BC, biliary cystadenoma;
BCA, biliary cystadenocarcinoma;
CCA, cholangiocarcinoma;
CT, computed tomography;
FLL, focal liver lesion;
FNH, focal nodular hyperplasia;
HCA, hepatocellular adenoma;
HCC, hepatocellular carcinoma;
HNF1 α, hepatocyte nuclear factor-1 α;
IHCA, inflammatory hepatocellular adenoma;
IUD, intrauterine device;
MRI, magnetic resonance imaging;
NRH, nodular regenerative hyperplasia;
OCP, oral contraceptive;
PAIR, puncture, aspiration, injection, and reaspiration;
THCA, telangiectatic hepatocellular adenoma;
US, ultrasonography.

Genetic Testing and Management of Hereditary Gastrointestinal Cancer Syndromes

This guideline presents recommendations for the management of patients with hereditary gastrointestinal cancer syndromes and specifically discusses genetic testing and management of Lynch syndrome, familial adenomatous polyposis (FAP), attenuated familial adenomatous polyposis (AFAP), MUTYH-associated polyposis (MAP), Peutz–Jeghers syndrome, juvenile polyposis syndrome, Cowden syndrome, serrated (hyperplastic) polyposis syndrome, hereditary pancreatic cancer, and hereditary gastric cancer. Download the ACG Clinical Guideline.

 

SUMMARY STATEMENTS:
Standard for minimal cancer family history assessment in gastrointestinal (GI) practice:
  • A family history of cancer and premalignant GI conditions that provides sufficient information to develop a preliminary determination of the risk of a familial predisposition to cancer should be obtained for all patients being evaluated in outpatient gastroenterology and endoscopy practices.
  • Essential elements of a family history include presence and type of cancer diagnoses in first- and second-degree relatives, and presence and (ideally) type of polyps in first-degree relatives; age and lineage should be noted for each diagnosis.
Lynch syndrome (LS):
  • All newly diagnosed colorectal cancers (CRCs) should be evaluated for mismatch repair deficiency.
  • Analysis may be done by immunohistochemical testing for the MLH1/MSH2/MSH6/PMS2 proteins and/or testing for microsatellite instability. Tumors that demonstrate loss of MLH1 should undergo BRAF testing or analysis for MLH1 promoter hypermethylation.
  • Individuals who have a personal history of a tumor showing evidence of mismatch repair deficiency (and no demonstrated BRAF mutation or hypermethylation of MLH1), a known family mutation associated with LS, or a risk of ≥5% chance of LS based on risk prediction models should undergo genetic evaluation for LS.
  • Genetic testing of patients with suspected LS should include germline mutation genetic testing for the MLH1, MSH2, MSH6, PMS2, and/or EPCAM genes or the altered gene(s) indicated by immunohistochemical (IHC) testing.
Adenomatous polyposis syndromes:
1. Familial adenomatous polyposis (FAP)/MUTYH-associated polyposis/attenuated polyposis
  • Individuals who have a personal history of >10 cumulative colorectal adenomas, a family history of one of the adenomatous polyposis syndromes, or a history of adenomas and FAP-type extracolonic manifestations (duodenal/ampullary adenomas, desmoid tumors (abdominal>peripheral), papillary thyroid cancer, congenital hypertrophy of the retinal pigment epithelium ((CHRPE), epidermal cysts, osteomas) should undergo assessment for the adenomatous polyposis syndromes.
  • Genetic testing of patients with suspected adenomatous polyposis syndromes should include APC and MUTYH gene mutation analysis.
2. Hamartomatous polyposis syndromes
    a. Peutz–Jeghers syndrome (PJS)
  • Individuals with perioral or buccal pigmentation and/or two or more histologically characteristic gastrointestinal hamartomatous polyp(s) or a family history of PJS should be evaluated for PJS.
  • Genetic evaluation of a patient with possible PJS should include testing for STK11 mutations.
    b. Juvenile polyposis syndrome (JPS)
  • Individuals with five or more juvenile polyps in the colorectum or any juvenile polyps in other parts of the GI tract should undergo evaluation for JPS.
  • Genetic evaluation of a patient with possible JPS should include testing for SMAD4 and BMPR1A mutations.
    c. Cowden syndrome (PTEN hamartoma tumor syndrome)
  • Individuals with multiple gastrointestinal hamartomas or ganglioneuromas should be evaluated for Cowden syndrome and related conditions.
  • Genetic evaluation of a patient with possible Cowden syndrome should include testing for PTEN mutations.
    d. Serrated/hyperplastic polyposis syndrome
  • Individuals who meet at least one of the following criteria have the clinical diagnosis of serrated polyposis syndrome (SPS):
    1. at least 5 serrated polyps proximal to the sigmoid colon with ≥2 of these being >10 mm;
    2. any number of serrated polyps proximal to the sigmoid colon in an individual who has a first-degree relative (FDR) with serrated polyposis; and
    3. >20 serrated polyps of any size, distributed throughout the large intestine.
  • A clear genetic etiology has not yet been defined for SPS, and therefore genetic testing is currently not routinely recommended for SPS patients; testing for MUTYH mutations may be considered for SPS patients with concurrent adenomas and/or a family history of adenomas.
3. Hereditary pancreatic cancer
  • Individuals should be considered to be at risk for familial pancreatic adenocarcinoma if they
    1. have a known genetic syndrome associated with pancreatic cancer, including hereditary breast-ovarian cancer syndrome, familial atypical multiple melanoma and mole syndrome (FAMMM), PJS, LS, or other gene mutations associated with an increased risk of pancreatic adenocarcinoma; or
    2. have two relatives with pancreatic adenocarcinoma, where one is a FDR;
    3. have three or more relatives with pancreatic cancer; or
    4. have a history of hereditary pancreatitis.
  • Genetic testing of patients with suspected familial pancreatic cancer should include analysis of BRCA1/2, CDKN2A, PALB2, and ATM. Evaluation for PJS, LS, and hereditary pancreatitis-associated genes should be considered if other component personal and/or family history criteria are met for the syndrome.
4. Hereditary gastric cancer
    Hereditary diffuse gastric cancer (HDGC)
  • Individuals with
    1. ≥2 cases of diffuse gastric cancer, with at least one diagnosed at <50 years;
    2. ≥3 cases of documented diffuse cancer in first- or second degree relatives independent of age of onset;
    3. diffuse gastric cancer diagnosed at <40 years;
    4. a personal or family history of diffuse gastric cancer and lobular breast cancer with one diagnosed at <50 years should be evaluated for HDGC.
  • Genetic testing of individuals who fulfill HDGC clinical criteria should include analysis of CDH1 mutations.

 


 

SUMMARY OF RECOMMENDATIONS:
Lynch syndrome (LS)
  • In individuals at risk for or affected with LS, screening for colorectal cancer by colonoscopy should be performed at least every 2 years, beginning between ages 20 and 25 years. Annual colonoscopy should be considered in confirmed mutation carriers.
  • Colectomy with ileorectal anastomosis (IRA) is the preferred treatment of patients affected with LS with colon cancer or colonic neoplasia not controllable by endoscopy. Segmental colectomy is an option in patients unsuitable for total colectomy if regular postoperative surveillance is conducted.
  • Hysterectomy and bilateral salpingo-oophorectomy should be offered to women who are known LS mutation carriers and who have finished child bearing, optimally at age 40–45 years.
  • Screening for endometrial cancer and ovarian cancer should be offered to women at risk for or affected with LS by endometrial biopsy and transvaginal ultrasound annually, starting at age 30 to 35 years before undergoing surgery or if surgery is deferred.
  • Screening for gastric and duodenal cancer can be considered in individuals at risk for or affected with LS by baseline esophagogastroduodenoscopy (EGD) with gastric biopsy at age 30–35 years, and treatment of H.pylori infection when found. Data for ongoing regular surveillance are limited, but ongoing surveillance every 3–5 years may be considered if there is a family history of gastric or duodenal cancer.
  • Screening beyond population-based recommendations for cancers of the urinary tract, pancreas, prostate, and breast is not recommended unless there is a family history of the specific cancers.
  • Although data suggest that daily aspirin may decrease the risk of colorectal and extracolonic cancer in LS, currently the evidence is not sufficiently robust or mature to make a recommendation for its standard use.
Adenomatous polyposis syndromes
Familial adenomatous polyposis (FAP)/MUTYH-associated polyposis (MAP)/attenuated polyposis
  • In individuals at risk for or affected with the classic AP syndromes, screening for colorectal cancer by annual colonoscopy or flexible sigmoidoscopy should be performed, beginning at puberty. In families with attenuated familial adenomatous polyposis (AFAP) or MAP, surveillance should be by colonoscopy.
  • Absolute indications for immediate colectomy in FAP, AFAP, and MAP include: documented or suspected cancer or significant symptoms. Relative indications for surgery include the presence of multiple adenomas >6 mm, a significant increase in adenoma number, and inability to adequately survey the colon because of multiple diminutive polyps.
  • Screening for gastric and proximal small bowel tumors should be done using upper endoscopy including duodenoscopy starting at age 25–30 years. Surveillance should be repeated every 0.5–4 years depending on Spigelman stage of duodenal polyposis: 0=4 years; I=2–3 years, II=1–3 years, III=6–12 months, and IV=surgical evaluation. Examination of the stomach should include random sampling of fundic gland polyps. Low-grade dysplasia is common in fundic gland polyps, and surgery should be reserved for high-grade dysplasia or cancer.
  • Annual thyroid screening by ultrasound should be recommended to individuals affected with FAP, MAP, and attenuated polyposis.
  • Biannual screening should be offered to affected infants until age 7 years with α-fetoprotein and ultrasounds.
  • Postsurgical surveillance should include yearly endoscopy of rectum or ileal pouch, and examination of an ileostomy every 2 years.
Hamartomatous polyposis syndromes
Peutz–Jeghers syndrome (PJS)
  • Surveillance in affected or at-risk PJS patients should include monitoring for colon, stomach, small bowel, pancreas, breast, ovary, uterus, cervix, and testes cancers. Risk for lung cancer is increased, but no specific screening has been recommended. It would seem wise to consider annual chest radiograph or chest computed tomography (CT) in smokers.
Juvenile polyposis syndrome (JPS)
  • Surveillance of the gastrointestinal (GI) tract in affected or at-risk JPS patients should include screening for colon, stomach, and small bowel cancers.
  • Colectomy and ileorectal anastomosis or proctocolectomy and ileal pouch-anal anastomosis is indicated for polyp-related symptoms, or when the polyps cannot be managed endoscopically.
  • Cardiovascular examination for and evaluation for hereditary hemorrhagic telangiectasia should be considered for SMAD4 mutation carriers.
Cowden syndrome (PTEN hamartoma tumor syndrome)
  • Surveillance in affected or at-risk Cowden syndrome patients should include screening for colon, stomach, small bowel, thyroid, breast, uterine, kidney, and skin (melanoma) cancers.
Serrated/hyperplastic polyposis syndrome
  • Patients with serrated polyposis should undergo colonoscopies every 1–3 years with attempted removal of all polyps >5 mm diameter.
  • Indications for surgery for serrated polyposis syndrome (SPS) include an inability to control the growth of serrated polyps, or the development of cancer. Colectomy and ileorectal anastomosis is a reasonable option given the risks of metachronous neoplasia.
  • There is no evidence to support extracolonic cancer surveillance for SPS at this time. Screening recommendations for family members are currently unclear pending further data and should be individualized based on results of baseline evaluations in family members.
Hereditary pancreatic cancer
  • Surveillance of individuals with a genetic predisposition for pancreatic adenocarcinoma should ideally be performed in experienced centers utilizing a multidisciplinary approach and under research conditions. These individuals should be known mutation carriers from hereditary syndromes associated with increased risk of pancreatic cancer (Peutz-Jeghers, hereditary pancreatitis, familial atypical multiple melanoma and mole syndrome (FAMMM)) or members of familial pancreatic cancer kindreds with a pancreatic cancer affected first-degree relative. Because of a lower relative risk for pancreatic adenocarcinoma development in BRCA1, BRCA2, PALB2, ATM, and LS families, surveillance should be limited to mutation carriers with a first or second-degree relative affected with pancreatic cancer.
  • Surveillance for pancreatic cancer should be with endoscopic ultrasound (EUS) and/or magnetic resonance imaging (MRI) of the pancreas annually starting at age 50 years, or 10 years younger than the earliest age of pancreatic cancer in the family. Patients with PJS should start surveillance at age 35 years.
  • Because of the increased risk for pancreatic cancer development when compared with a pancreatic cyst in the sporadic setting, cystic lesion(s) of the pancreas detected during surveillance of a hereditary pancreatic cancer-prone family member requires evaluation by centers experienced in the care of these high-risk individuals. Determining when surgery is required for pancreatic lesions is difficult and is best individualized after multidisciplinary assessment.
Hereditary gastric cancer
Hereditary diffuse gastric cancer
  • Management for patients with hereditary diffuse gastric cancer should include:
    1. prophylactic gastrectomy after age 20 years (>80% risk by age 80);
    2. breast cancer surveillance in women beginning at age 35 years with annual mammography and breast MRI and clinical breast examination every 6 months; and
    3. colonoscopy beginning at age 40 years for families that include colon cancer.

 


    
Amsterdam criteria I 
  • At least three relatives with colorectal cancer (CRC); all of the following criteria should be present:
    • One should be a first-degree relative of the other two;
    • At least two successive generations must be affected;
    • At least one of the relatives with CRC must have received the diagnosis before the age of 50 years;
    • Familial adenomatous polyposis should be excluded;
    • Tumors should be verified by pathologic examination.
Amsterdam criteria II
  • At least three relatives must have a cancer associated with Lynch syndrome (colorectal, cancer of the endometrium, small bowel, ureter, or renal-pelvis); all of the following criteria should be present:
    • One must be a first-degree relative of the other two;
    • At least two successive generations must be affected;
    • At least one relative with cancer associated with Lynch syndrome (LS) should be diagnosed before age 50;
    • Familial adenomatous polyposis should be excluded in the CRC case(s) (if any);
    • Tumors should be verified whenever possible.
Revised Bethesda guidelines
  • Tumors from individuals should be tested for microsatellite instability (MSI) in the following situations:
    • CRC diagnosed in a patient who is younger than 50 years of age
    • Presence of synchronous, or metachronous, colorectal or other LS-related tumors, regardless of age
    • CRC with MSI-high histology diagnosed in a patient who is younger than 60 years of age
    • CRC diagnosed in a patient with one or more first-degree relatives with an LS-related cancer, with one of the cancers being diagnosed under age 50 years
    • CRC diagnosed in a patient with two or more first- or second-degree relatives with LS-related cancer regardless of age

 


    
Colorectal cancer risk assessment tool
(Patient who answers yes to any question should have more comprehensive family history evaluation)
  • Do you have a first-degree relative (mother, father, brother, sister, or child) with any of the following conditions diagnosed before age 50?
    • Colon or rectal cancer
    • Cancer of the uterus, ovary, stomach, small intestine, urinary tract (kidney, ureter, bladder), bile ducts, pancreas, or brain
  • Have you had any of the following conditions diagnosed before age 50 years?
    • Colon or rectal cancer
    • Colon or rectal polyps
  • Do you have three or more relatives with a history of colon or rectal cancer? (This includes parents, brothers, sisters, children, grandparents, aunts, uncles, and cousins)
    • Lynch syndrome-related cancers include colorectal, endometrial, gastric, ovarian, pancreas, ureter and renal pelvis, biliary tract, brain (usually glioblastoma), small intestinal cancers, as well as sebaceous gland adenomas and keratoacanthomas.
    • Presence of tumor-infiltrating lymphocytes, Crohn’s-like lymphocytic reaction, mucionous/signet-ring differentiation, or medullary growth pattern.

 


     
Standards for informed consent for genetic testing in gastrointestinal (GI) practice
Components of a proper informed consent for cancer genetic testing should include:
  1. Information on the specific genetic mutation(s) or genomic variant(s) being tested, including whether or not the range of risk associated with the variant will affect medical care.
  2. Implications of positive and negative results.
  3. Possibility that the test will not be informative.
  4. Options for risk estimation without genetic or genomic testing.
  5. Risk of passing a genetic variant to children.
  6. Technical accuracy of the test, including, where required by law, licensure of the testing laboratory
  7. Fees involved in testing and counseling and, for direct to consumer testing, whether the counselor is employed by the testing company.
  8. Psychological implications of test results (benefits and risks).
  9. Risks and protections against genetic discrimination by employers or insurers.
  10. Confidentiality issues, including, for direct-to-consumer testing companies, polices related to privacy and data security.
  11. Possible use of DNA testing samples in future research.
  12. Options and limitations of medical surveillance and strategies for prevention after genetic or genomic testing.
  13. Importance of sharing genetic and genomic test results with at-risk relatives so that they may benefit from this information.
  14. Plans for follow-up after testing.

 


 

Spigelman Duodenal adenomatosis staging system

Polyps         1 Point        2 Points            3 Points

Number       <4               5–20                 >20

Size             0-4mm        5-10mm            >10

Histology     Tubular       Tubulovillous    Villous

Dysplasia     Mild            Moderate          Severe

Recommended duodenal surveillance frequency

Spigelman stage      Total points          Frequency of surveillance

0                                0                         Every 4 years

I                                 ≤4                       Every 2–3 years

II                                5-6                      Every 1–3 years

III                               7-8                      Every 6–12 months

IV                               9-12                    Expert surveillance every 3- 6 months

Surgical evaluation, complete mucosectomy or duodenectomy or Whipple procedure if

duodenal papilla is involved

Management of Colon Ischemia

The American College of Gastroenterology published an excellent guideline on the management of Colon Ischemia in December 2014. This important topic was summarized by Utah Gastroenterology. For detailed information, please review the excellent guideline paper.

 

Abbreviations:

  • AMI – acute mesenteric ischemia
  • IRCI – isolated right colon ischemia
  • SMA – superior mesenteric artery
  • IMA – inferior mesenteric artery

 

Colonic Ischemia Recommendations and Best Practice Summary Statements

Clinical Presentation

  • The diagnosis of CI is usually established in the presence of symptoms including sudden cramping, mild, abdominal pain; an urgent desire to defecate; and passage within 24 h of bright red or maroon blood or bloody diarrhea.
  • A diagnosis of non-isolated right colon ischemia (non-IRCI) should be considered when patients present with hematochezia.

Imaging of CI

  • CT with intravenous and oral contrast should be the first imaging modality of choice for patients with suspected CI to assess the distribution and phase of colitis.
  • The diagnosis of CI can be suggested based on CT findings (e.g., bowel wall thickening, edema, thumbprinting).
    Multiphasic CTA should be performed on any patient with suspected IRCI or in any patient in whom the possibility of acute mesenteric ischemia (AMI) cannot be excluded.
  • CT or MRI findings of colonic pneumatosis and porto-mesenteric venous gas can be used to predict the presence of transmural colonic infarction.
  • In a patient in whom the presentation of CI may be a heralding sign of acute mesenteric ischemia (AMI) (e.g., isolated right colon ischemia (IRCI), severe pain without bleeding, atrial fibrillation), and the multiphasic CT is negative for vascular occlusive disease, traditional splanchnic angiography should be considered for further assessment.

Colonoscopy in the Diagnosis of CI

  • Early colonoscopy (within 48 h of presentation) should be performed in suspected CI to confirm the diagnosis.
  • When performing colonoscopy on a patient with suspected CI, the colon should be insufflated minimally.
  • In patients with severe CI, CT should be used to evaluate the distribution of disease. Limited colonoscopy is appropriate to confirm the nature of the CT abnormality. Colonoscopy should be halted at the distal most extent of the disease.
  • Biopsies of the colonic mucosa should be obtained except in cases of gangrene.
  • Colonoscopy should not be performed in patients who have signs of acute peritonitis or evidence of irreversible ischemic damage (i.e., gangrene and pneumatosis).

Severity and Treatment of CI

  • Most cases of CI resolve spontaneously and do not require specific therapy.
  • Surgical intervention should be considered in the presence of CI accompanied by hypotension, tachycardia, and abdominal pain without rectal bleeding; for IRCI and pan-colonic CI; and in the presence of gangrene.
  • Antimicrobial therapy should be considered for patients with moderate or severe disease.

Risk Factors

  • Comorbid cardiovascular disease and diabetes mellitus should increase consideration of CI in patients with typical clinical features.
  • A history of IBS and constipation should be sought in patients suspected to have CI.
  • Selective cardiology consultation is justified in patients with CI, particularly if a cardiac source of embolism is suspected.
  • Chronic kidney disease is associated with increased mortality from CI.
  • Evaluation for thrombophilia should be considered in young patients with CI and all patients with recurrent CI.
  • Surgical procedures in which the inferior mesenteric artery (IMA) has been sacrificed, such as abdominal aortic aneurysm repair and other abdominal operations, should increase consideration of CI in patients with typical clinical features.
  • In patients suspected of having CI, a history of medication and drug use is important, especially constipation-inducing medications, immunomodulators, and illicit drugs.

Clinical Presentation

  • IRCI is associated with higher mortality rates compared with other patterns of CI.

Laboratory Tests in CI

  • Laboratory testing should be considered to help predict CI severity.
  • Decreased hemoglobin levels, low serum albumin, and the presence of metabolic acidosis can be used to predict severity of CI.

Severity and Treatment of CI

  • When considering mortality risk for patients undergoing surgical intervention for acute CI, the Ischemic Colitis Mortality Risk (ICMR) factors should be utilized.

 


Detailed Data Summary 

1.Risk factors

  • Comorbid cardiovascular disease and diabetes mellitus should increase consideration of CI in patients with typical clinical features.
  • A history of irritable bowel syndrome (IBS) and constipation should be sought in patients suspected to have CI.
  • Selective cardiology consultation is justified in patients with CI, particularly if a cardiac source of embolism is suspected.
  • Chronic kidney disease and chronic obstructive pulmonary disease are associated with increased mortality from CI.
  • Evaluation for thrombophilia should be considered in young patients with CI and in all patients with recurrent CI.
  • Surgical procedures in which the inferior mesenteric artery (IMA) has been sacrificed, such as abdominal aortic aneurysm repair and other abdominal operations, should increase consideration of CI in patients with typical clinical features.
  • In patients suspected of having CI, a history of medication and drug use should be sought, especially constipation- inducing medications, immunomodulators, and illicit drugs.

 

2. Medical conditions and surgical history independently associated with colon ischemia in multivariate analyses of case–control studies

  • Atherosclerosis
  • Atrial fibrillation
  • Chronic obstructive pulmonary disease
  • Congestive heart failure
  • Diarrhea
  • Irritable bowel syndrome
  • Diabetes
  • Dyslipidemia
  • Rheumatoid arthritis
  • Systemic rheumatologic disorders

 

3a. Drugs proposed to predispose to CI and postulated pathogenesis  (moderate evidence)

  • Constipation-inducing drugs (Predicted CI in patients with abdominal pain)

    • All drugs: C-CS; increased risk 0.68 (0.62–1.27)
    • Opioids: C-CS; increased risk 1.96 (1.43–2.67)
    • Nonopioids: C-CS; increased risk 1.75 (1.25–2.44)
      • Reduced blood flow, increased intraluminal pressure
  • Immunomodulator drugs
    • Antitumor necrosis factor-α inhibitors for rheumatoid arthritis
    • Type 1 interferon-α for hepatitis C
    • Type 1 interferon-β for multiple sclerosis)
      • Cytokines affecting thrombogenesis
  • Illicit drugs
    • Amphetamines
    • Cocaine
      • Vasoconstriction, hypercoagulation, direct endothelial injury
3b. Drugs proposed to predispose to CI and postulated pathogenesis  (low evidence)
  • Multiple Classes
    • Antibiotics
    • Appetite suppressants
    • Bitter orange
    • Hydroxycut
    • Ma huang
    • Phentermine
    • Xenadrine (bitter orange, ma huang, caffeine, salicin)
      • Vasoconstriction
  • Chemotherapeutic drugs
    • R-CHOP
    • Taxanes
    • Vinorelbine/cisplatin
      • Direct epithelial toxicity, inhibited repair of vascular injury
  • Decongestants
    • Pseudoephedrine
    • Phenylephrine
      • Vasoconstriction
  • Diuretics
    • Extracellular volume deficit, lower peripheral vascular resistance, vasoconstriction
  • Ergot alkaloids (often combined with caffeine)
    • Vasoconstriction
  • Hormonal therapies
    • Predominance of women among young patients
    • Female hormones
    • Oral contraceptives
    • Estrogen replacement
      • Hypercoagulability, endothelial injury
  • Laxatives
    • Osmotic agents
    • Bisacodyl
    • Bisacodyl/polyethylene glycol
    •  Lubiprostone
      • Increased motility or rapid intravascular volume deficit, reduced perfusion
  • Psychotropic drugs (hypotension, constipation)
  • Serotoninergic drugs
    • 5-Hydroxytryptamine1 receptor agonists
    • 5-hydroxytryptamine3 receptor antagonist
    • 5-hydroxytryptamine4 partial agonist
      • For 5-hydroxytryptamine1 receptor agonists vasoconstriction; for other agents various factors
 3c. Drugs proposed to predispose to CI and postulated pathogenesis (very low evidence)
  • Mulitple Classes
    • Digitalis
    • Kayexalate
    • NO-Xplode
    • NSAIDs
    • Statins
    • Vasopressors
      • Vasoconstriction

 

4. Clinical Presentation

  • The diagnosis of CI is usually established because of symptoms including sudden cramping, mild, abdominal pain; an urgent desire to defecate; and passage within 24 h of bright red or maroon blood per rectum or bloody diarrhea.
  • A diagnosis of non-IRCI should be considered when patients present with hematochezia.
  • CI isolated to the right colon (IRCI) is associated with higher mortality rates compared with other patterns of CI.

 

5. Laboratory Testing

  • Laboratory testing should be considered to help predict CI severity.
  • Decreased hemoglobin levels, low serum albumin, and the presence of metabolic acidosis can be used to predict severity of CI.
  • Recommended initial serology and stool studies for suspected colon ischemia (CI):
    • Blood tests
      • Albumin
      • Amylase
      • Complete blood count
      • Comprehensive electrolyte panel
      • Creatine kinase (CK)
      • Lactate
      • Lactate dehydrogenase (LDH)
    • Stool tests
      • Clostridium difficile toxin assay
      • Culture
      • Ova and parasite

 

6. Imaging of CI

  • CT with intravenous and oral contrast should be ordered as the imaging modality of choice for patients with suspected CI, to assess the distribution and phase of colitis.
  • The diagnosis of CI can be suggested based on CT findings (e.g., bowel wall thickening, edema, and thumbprinting)
  • Multiphasic CT angiography (CTA) should be performed on any patient with suspected IRCI or in any patient in whom the possibility of AMI cannot be excluded.
  • CT or magnetic resonance imaging (MRI) findings of colonic pneumatosis and portomesenteric venous gas can be used to predict the presence of transmural colonic infarction.
  • In a patient in whom the presentation of CI may be a heralding sign of acute mesenteric ischemia (AMI; e.g., IRCI, severe pain without bleeding, and atrial fibrillation), and the multiphasic CT is negative for vascular occlusive disease, traditional splanchnic angiography should be considered for further assessment.

 

7. Classification of disease severity and management

  • Mild
    • Typical symptoms of CI with a segmental colitis not isolated to the right colon and with none of the commonly associated risk factors for poorer outcome that are seen in moderate disease.
      • Observation
      • Supportive care
  • Moderate
    • Any patient with CI and up to three of the following factors:
      • Male gender
      • Hypotension (systolic blood pressure <90mmHg)
      • Tachycardia (heart rate >100beats/min)
      • Abdominal pain without rectal bleeding
      • BUN >20mg/dl
      • Hgb <12g/dl
      • LDH >350U/l
      • Serum sodium <136mEq/l (mmol/l)
      • WBC >15 cells/cmm (×109/l)
      • Colonic mucosal ulceration identified colonoscopically
        • Correction of cardiovascular abnormalities (e.g., volume replacement)
        • Broad-spectrum antibiotic therapy
        • Surgical consultation
  • Severe
    • Any patient with CI and more than three of the criteria for moderate disease or any of the following:
      • Correction of cardiovascular abnormalities (e.g., volume replacement)
      • Broad-spectrum antibiotic therapy
      • Peritoneal signs on physical examination
      • Pneumatosis or portal venous gas on radiologic imaging
      • Gangrene on colonoscopic examination
      • Pancolonic distribution or IRCI on imaging or colonoscopy
        • Emergent surgical consultation (treatment is likely to be surgical)
        • Transfer to intensive care unit
        • Correction of cardiovascular abnormalities (e.g., volume replacement)
        • Broad-spectrum antibiotic therapy

 

8. Predictors of disease severity (factors from multiple studies that are significantly associated with the requirement for surgery and/or mortality)

  • Epidemiologic factors
    • Antibiotic treatment
    • Chronic obstructive pulmonary disease
    • Chronic kidney disease
    • Hepatitis C positivity
    • History of cancer
    • Male gender
    • Warfarin use
  • Presentation of disease
    • Abdominal pain without rectal bleeding
    • Nonbloody diarrhea
    • Peritoneal signs
    • Symptom onset after admission
  • Vital signs
    • Pulse >90-100beats/min
    • Systolic blood pressure <90mmHg
  • Serology
    • Hemoglobin <12mg/dl
    • Na <136mEq/l (mmol/l)
    • LDH >450U/l
    • BUN >28.0 mg/dl
  • Colonoscopic finding
    • Ulceration
    • Distribution of disease
    • Bilateral or right-side disease distribution
    • Isolated right colon involvement

 

9. Colonoscopy in the diagnosis of CI

  • Early colonoscopy (within 48 h of presentation) should be performed in suspected CI cases to confirm the diagnosis.
  • When performing colonoscopy on a patient with suspected CI, the colon should be insufflated minimally.
  • In patients with severe CI, CT should be used to evaluate the distribution of disease. Limited colonoscopy is appropriate to confirm the nature of the CT abnormality. The endoscopic procedure should be stopped at the distal-most extent of the disease.
  • Biopsies of the colonic mucosa should be obtained except in cases of gangrene.
  • Colonoscopy should not be performed in patients who have signs of acute peritonitis or evidence of irreversible ischemic damage.

 

10. Severity and treatment CI

  • Most cases of CI resolve spontaneously and do not require specific therapy.
  • Surgical intervention should be considered in the presence of CI accompanied by hypotension, tachycardia, and abdominal pain without rectal bleeding; for IRCI and pancolonic CI; and in the presence of gangrene.
  • Antimicrobial therapy should be considered for patients with moderate or severe disease.
  • When considering the mortality risk for patients undergoing surgical intervention for acute CI, Ischemic Colitis Mortality Risk (ICMR) factors should be utilized.

 

11. Indications for surgery in colonic ischemia

  • Acute indications
    • Peritoneal signs
    • Massive bleeding
    • Universal fulminant colitis with or without toxic megacolon
    • Portal venous gas and/or pneumatosis intestinalis on imaging
    • Deteriorating clinical condition
  • Subacute indications
    • Failure of an acute segmental ischemic colitis to respond to treatment within 2–3 weeks with continued symptoms or a protein-losing colopathy
    • Apparent healing but with recurrent bouts of sepsis
  • Chronic indications
    • Symptomatic colon stricture
    • Symptomatic segmental ischemic colitis

 

12. Risk factors for perioperative mortality

  • Low output heart failure (e.g., cardiac ejection fraction <20% on echocardiogram)
  • Acute kidney injury
  • Subtotal or total colectomy
  • Lactate >2.5mmol/l
  • Pre- and intraoperative catecholamine administration
    • Risk factors = Mortality
      • 0 = 10.5%
      • 1 = 28.9%
      • 2 = 37.1%
      • 3 = 50.0%
      • 4 = 76.7%
      • 5 = 100.0%

 

ACG Clinical Guideline: Epidemiology, Risk Factors, Patterns of Presentation, Diagnosis, and Management of Colon Ischemia (CI)

Am J Gastroenterol 2015; 110:18–44

http://gi.org
http://gi.org/clinical-guidelines/
http://gi.org/clinical-guidelines/clinical-guidelines-sortable-list/
http://gi.org/guideline/epidemiology-risk-factors-patterns-of-presentation-diagnosis-and-management-of-colon-ischemia/
http://gi.org/wp-content/uploads/2015/01/ACG_Guideline_Colon-Ischemia_January_2015.pdf

Antibiotic Prophylaxis for GI Endoscopy

The American Society for Gastrointestinal Endoscopy (ASGE) updated its guidelines on antibiotic prophylaxis for GI endoscopy in November 2014.

This important topic was summarized by Utah Gastroenterology. For detailed information, please review the excellent guideline paper.

As per the paper “..bacteremia can occur after endoscopic procedures and has been advocated as a surrogate marker for infective endocarditis (IE) risk. However, clinically significant infections are extremely rare. Despite an estimated 14.2 million colonoscopies, 2.8 million flexible sigmoidoscopies, and perhaps as many upper endoscopies performed in the United States each year, only approximately 25 cases of IE have been reported with temporal association to an endoscopic procedure. …. This document is an update of the prior ASGE document on antibiotic prophylaxis for GI endoscopy, discusses infectious adverse events related to endoscopy, and provides recommendations for periprocedural antibiotic therapy..”.

 

ASGE RECOMMENDATIONS

  1. The ASGE recommends against the routine administration of antibiotic prophylaxis solely for prevention of infective endocarditis (IE).
  2. The ASGE suggests that patients with high-risk cardiac conditions and established GI tract infections in which enterococci may be part of the infecting bacterial flora should receive antibiotic coverage.
  3. The ASGE recommends against antibiotic prophylaxis before ERCP when obstructive biliary tract disease is not suspected or complete biliary drainage is anticipated.
  4. The ASGE recommends that antibiotic prophylaxis be administered before ERCP in patients who have had liver transplantation or who have known or suspected biliary obstruction, where there is a possibility of incomplete biliary drainage. Antibiotics that cover biliary flora such as enteric gram-negative organisms and enterococci should be used and continued after the procedure if biliary drainage is incomplete.
  5. The ASGE recommends against antibiotic prophylaxis before diagnostic EUS or EUS-FNA of solid lesions of the GI tract.
  6. The ASGE suggests antibiotic administration prior to EUS-FNA of mediastinal cysts.
  7. The ASGE suggests administration of prophylactic antibiotics before EUS-FNA of pancreatic or peripancreatic cysts.
  8. The ASGE recommends administration of parenteral cefazolin (or an antibiotic with equivalent microbial coverage) to all patients before PEG/PEJ tube placement.
  9. The ASGE recommends that all patients with cirrhosis admitted with GI bleeding should have antibiotic therapy instituted at admission with intravenous ceftriaxone (or an antibiotic with equivalent microbial coverage [eg, oral norfloxacin] in patients allergic to or intolerant of ceftriaxone).
  10. The ASGE recommends against administration of antibiotic prophylaxis before GI endoscopic procedures for patients with synthetic vascular grafts or other nonvalvular cardiovascular devices (eg, implantable electronic devices).
  11. The ASGE recommends against antibiotic prophylaxis for patients with orthopedic prosthesis undergoing any GI endoscopic procedure.
  12. The ASGE suggests administration of antibiotic prophylaxis before endoscopy of the lower GI tract in patients undergoing continuous ambulatory peritoneal dialysis.

 

The 2007 American Heart Association (AHA) guidelines for prophylaxis of IE stated that the administration of prophylactic antibiotics solely to prevent IE was no longer recommended for patients undergoing GI endoscopy. The AHA based its recommendations on several lines of evidence including

  • (1) cases of IE associated with GI procedures are anecdotal
  • (2) no data demonstrate a conclusive link between GI procedures and the development of IE
  • (3) there are no data that demonstrate that antibiotic prophylaxis prevents IE after GI-tract procedures
  • (4) IE is more likely to be caused by bacteremia resulting from usual daily activities, and
  • (5) an extremely small number of cases of IE may be prevented even if antibiotic prophylaxis were 100% effective.

 

The AHA also delineated cardiac conditions associated with the highest risk of an adverse outcome from IE, including

  1. prosthetic (mechanical or bioprosthetic) cardiac valves,
  2. history of previous IE,
  3. cardiac transplant recipients who develop cardiac valvulopathy, and
  4. patients with congenital heart disease (CHD) including those with unrepaired cyanotic CHD including palliative shunts and conduits; those with completely repaired CHD with prosthetic material or devices, placed surgically or by patients with these cardiac conditions who have established infections of the GI tract in which enterococci may be part of the infecting flora (such as cholangitis) and particularly for those who are about to undergo an endoscopic procedure that may increase the risk of bacteremia (such as ERCP), the AHA suggests that inclusion of an agent active against enterococci in the concurrent antibiotic regimen may be reasonable. Although GI tract infections often are polymicrobial, antibiotic coverage for enterococci is recommended, because only enterococci are likely to cause IE. However, the AHA reiterates that no studies have demonstrated that such therapy would prevent enterococcal IE.

 
Therefore the ASGE suggests that patients with high-risk cardiac conditions and established GI tract infections in which enterococci may be part of the infecting bacterial flora should receive antibiotic coverage:

Prevention of infective endocarditis (IE) – Cardiac conditions

1) All cardiac conditions: Antibiotic prophylaxis is not indicated solely to prevent IE.
2) Cardiac conditions associated with the highest risk of an adverse outcome from IE should receive antibiotic coverage:

  • Prosthetic cardiac valve
  • History of IE
  • Cardiac transplant recipients who develop cardiac valvulopathy
  • Patients with congenital heart disease (CHD)
  • Unrepaired cyanotic CHD including palliative shunts and conduits
  • Completely repaired CHD with prosthetic material or device, placed surgically or by catheter, for the first 6 months after the procedure
  • Repaired CHD with residual defects at the site or adjacent to the site of a prosthetic patch or device

For patients with these conditions who have established infections of the GI tract (such as cholangitis) and for those who receive antibiotic therapy to prevent wound infection or sepsis associated with a GI tract procedure, it is recommended that the antibiotic regimen include an antimicrobial agent active against enterococci, such as penicillin, ampicillin, piperacillin, or vancomycin.

Diagnosis & Management of Celiac Disease

The Consensus guidelines by the British Society of Gastroenterology as published in GUT August 2014 on the diagnosis & management of celiac disease are summarized by Utah Gastroenterology. For detailed information, please review the excellent guideline paper.

A multidisciplinary panel from eight countries (Sweden, UK, Argentina, Australia, Italy, Finland, Norway and the USA) reviewed the literature on diagnosis and management of adult celiac disease (CD). These recommendations of the British Society of Gastroenterology examine the diagnosis and management of CD, the classification of CD, genetics and immunology, diagnostics, serology and endoscopy, follow-up, gluten-free diet, refractory CD and malignancies, quality of life, novel treatments, patient support, and screening for CD.

 

Background

  • CD is an immune-mediated small intestinal enteropathy triggered by exposure to dietary gluten in genetically predisposed individuals. Although the prevalence varies considerably, a large-scale study from Finland, Italy, the UK and Germany found a prevalence of CD of around 1%, with a recent US study showing a prevalence of 0.71%.
  • CD is more frequently diagnosed in women than in men with a ratio between 1.5 and 2. Traditionally patients with CD presented with malabsorption, diarrhea, steatorrhea, weight loss or failure to thrive (‘classical CD’), but the proportion of newly diagnosed patients with ‘non-classical CD and even asymptomatic CD have gained prominence.  Screening may be initiated because the individual has a CD-associated disorder or has symptoms and is a first-degree relative to a patient with CD.
  • Newly diagnosed patients with CD can present with a wide range of symptoms, including:
    • anemia
    • vague abdominal symptoms (often similar to irritable bowel syndrome (IBS))
    • neuropathy
    • ataxia
    • depression
    • short stature
    • osteomalacia and osteoporosis
    • liver disease
    • adverse pregnancy outcomes
    • lymphoma

 

Genetics, Immunology and Trigger Factors

  • Environmental factors are important in CD and ingestion of gluten is a prerequisite for the development of CD. Children breastfed at and beyond gluten introduction may be at lower risk of developing CD in childhood, although research is not consistent. Gastrointestinal infections, drugs, interferon α and surgery have also been implicated as trigger factors. A high prevalence (10%) among first-degree relatives of patients with CD and a greater concordance rate in monozygotic twins (∼75%) than in dizygotic twins indicates a strong genetic component. CD shows a very strong association with a particular HLA variant termed HLA-DQ2.5. Most patients are DR3, DQ2 positive. A smaller subset of patients with CD express a very similar DQ2.5 molecule encoded by a combination of DR5 and DR7 positive haplotypes. While the proportion of individuals with CD who are DQ2.5 positive varies in different geographical areas it is generally ≥90%. The majority of the remaining patients are HLA-DQ8 positive (DR4, DQ8 haplotype).

 

Diagnosis of CD

  • Diagnosis of CD is made by serology and duodenal biopsy, ideally with the patient on a normal gluten-containing diet.
  • Biopsy remains essential for the diagnosis of adult CD and cannot be replaced by serology. Exceptions are patients with coagulation disorders and pregnant women, in whom biopsy may not be feasible or should be postponed until postpartum.
  • The diagnosis of CD may not be straightforward, if patients are already on a GFD at the time of presentation, biopsies were not oriented correctly or show solely intraepithelial lymphocytosis (lymphocytic duodenosis) without architectural changes. In these situations, patients need to be maintained on a gluten-containing diet and additional testing or referral to a clinician with a specific interest in CD may be necessary.
  • To state a definite diagnosis of CD, villous atrophy is required. However, lesser degrees of damage (≥25 intraepithelial lymphocytes (IELs) but no villous atrophy) combined with positive serology (IgA-endomysium antigen (EMA), tissue transglutaminase (TTG) or IgG-deamidated gliadin peptide (DGP) may also represent CD (‘probable CD’), and in these circumstances a trial with a gluten free diet (GFD) may be considered to further support the diagnosis of CD. Human leucocyte antigen (HLA) status may also aid diagnosis. Differential diagnoses of lymphocytic duodenosis should be ruled out if there is no response to GFD.
  • The diagnosis of CD is readily established in those who have positive serology and a duodenal biopsy with obvious celiac histology (increased intraepithelial lymphocytosis, crypt hyperplasia and villous atrophy) while consuming a gluten-containing diet. These patients can immediately initiate a GFD with confidence.

1. Serology in CD Diagnosis

  • Serological detection depends on the presence of specific endomysial antibodies (EMAs), IgA anti-tissue transglutaminase antibodies (IgA-TG2, also called a-TTG, TTA) and/or deamidated antigliadin antibodies (DGP, either IgA or IgG isotype).7 IgG-TG2 is primarily useful in patients with known IgA deficiency.

2. Biopsy and Endoscopy in CD

  • There are endoscopic markers of villous atrophy described – scalloping or reduction of duodenal folds and modularity – but these are not sensitive enough to preclude a biopsy, and a normal endoscopic appearance may occur in the presence of villous atrophy. Therefore, the endoscopic appearance of the duodenum should not determine whether biopsy is performed.
  • Biopsy of the duodenum for a diagnosis of CD should be performed irrespective of the prior performance of serological tests, if the patient exhibits symptoms or signs of CD, such as diarrhea, weight loss or anaemia. Biopsies can be mounted on fibre-free paper to aid orientation, or alternatively biopsies could be free floated in formalin.
  • The villous atrophy may be patchy in CD; hence multiple biopsies from the bulb and the more distal duodenum are recommended. The taking of at least four biopsy specimens is associated with a doubling of the diagnostic rate compared with patients undergoing a lower number of biopsies (less than four).
  • In patients with persistently positive celiac serology but a normal mucosa, repeat small intestinal biopsy should be considered, including biopsies from the jejunum.
  • Video capsule endoscopy may support a CD diagnosis in this setting.
  • It is important that if serology has not been performed prior to the biopsy, then this must be carried out by the requesting physician on receipt of a histology report suggesting a diagnosis of CD. A biopsy finding of villous atrophy is not specific for CD. Although CD is the commonest cause of villous atrophy, there are other causes.
  • Follow-up biopsies may be considered in patients with CD, and are potentially helpful in identifying patients at increased risk of lymphoma.
  • Follow-up biopsies are not mandatory if the patient with CD is asymptomatic on a GFD, and has no other features that suggest an increased risk of complications.
  • Follow-up biopsies should be undertaken in patients with CD whose condition does not respond to a GFD.

3. Histopathology Diagnosis of CD

The following features should be stated in the report:

  1. Number of biopsies (including those from the duodenal bulb) and orientation.
  2. The architectural features (normal, partial, sub-total or total villous atrophy).
  3. Comment on the content of the lamina propria (in CD these are lymphocytes, plasma cells and eosinophils, and occasionally neutrophils, but cryptitis and crypt abscesses should suggest other pathology).
  4. Presence of Brunner’s glands.
  5. Presence of crypt hyperplasia, villous height: crypt depth ratio (3:1). The absence of plasma cells suggests common variable immunodeficiency.
  6. Evaluation of IELs (with immunocytochemical staining for T cells (CD3) in equivocal cases) is vital. The normal count <25IELs/100 enterocytes should be taken as the norm.

4. Endoscopy in Seronegative Individuals

  • The prevalence of seronegative CD is 6–22% of all diagnosed cases.
  • Individuals of white European, Middle Eastern, North African or North Indian origin who undergo upper endoscopy for anemia, weight loss or diarrhea should have duodenal biopsies performed, irrespective of whether they have had serology for CD. These features may well indicate that CD or an alternative mucosal cause of malabsorption is present.
  • It has been suggested that duodenal biopsy should be considered in any individual undergoing endoscopy, because CD is common and has many varied clinical manifestations, including reflux, a common indication for endoscopy.

5. Role of HLA in the Diagnosis of CD

  • CD is associated with specific HLA types in virtually all populations and is associated with the carriage of the gene pairs that encode DQ2.5 and DQ8. The diagnostic value of HLA genotyping in patients who may have CD revolves around its high negative predictive value, meaning that patients who lack the appropriate HLA genotype pairs described above are very unlikely to have CD. However, the positive predictive value of the HLA genotyping for CD susceptibility is very low as a large proportion of individuals without CD carry either HLA-DQ2 or HLA-DQ8.
  • HLA genotyping may be used in patients with suspected CD but who fail to respond to a GFD. A negative test in this circumstance would indicate that patients are highly unlikely to have CD (<1% of patients with CD are negative for DQ2 and DQ8) and thus the clinician can direct diagnostic efforts elsewhere.
  • HLA typing may similarly be used in self-treated patients on a GFD (without prior appropriate testing for CD) before changing their diet. HLA typing may have an adjunctive role to identifying individuals who are not genetically at risk of CD and in whom further evaluation for CD is not necessary, saving a large number of repeated tests for CD in patients who would otherwise have to undergo testing because they have symptoms and a first-degree relative with CD.

6. Other Causes of Lymphocytic Duodenosis and Villous Atrophy

  • Lymphocytic duodenosis is a common condition (3.8% of a population negative for coeliac serology) seen in association with infection (particularly Helicobacter pylori), altered immune states, for example, common variable immunodeficiency, autoimmune and chronic inflammatory disorders, drugs and neoplasia. There are other causes of villous atrophy in duodenal biopsies, including immune disorders and deficiency, food hypersensitivity, infection, drugs, neoplasia and miscellaneous disorders.

 

Dermatitis Herpetiformis (DH)

  • Dermatitis herpetiformis (DH) is the cutaneous manifestation of gluten-sensitive enteropathy precipitated by exposure to dietary gluten. It is characterized clinically by herpetiform clusters of intensely itchy urticated papules and small blisters distributed on the extensor aspects of the elbows and knees and over the buttocks and on the scalp. The commonest age of onset is between the third and fourth decade. Male patients are affected twice as often as female patients. The major diagnostic criterion for diagnosis is the presence of granular IgA deposits in the dermal papillae of uninvolved perilesional skin as shown by direct immunofluorescence, and the diagnosis should not be made unless this has been confirmed. Less than 10% of patients with DH have symptoms or signs of malabsorption but most have evidence of CD that responds to a GFD and relapses on gluten challenge. Patients with DH present with skin manifestations and are not usually troubled by the underlying small bowel problem at the time of presentation. Abnormality of the small intestinal mucosa with either total or subtotal villous atrophy is found in approximately 70% of patients with DH. A further 25% have normal villous architecture with increased IELs. DH shares with CD an increased risk of developing lymphomas but this seems to be confined to those with severe gut involvement. The risk similarly declines with time on a strict GFD. Due to rash and itch, dapsone is often initiated. More than 70% of patients on a strict GFD are however able to slowly wean off dapsone over a period of 24 months.

 

Clinical Follow-up

  • There is a paucity of data pertaining to adherence to a GFD being improved by follow-up in patients with CD. One of the key factors relating to adherence is dietetic input and regular follow-up. Patients should be encouraged to join disease-specific patient support groups. Once the disease is stable and the patients manage their diet without any problems, annual follow-ups should be initiated.
  • The physician should check on small intestinal absorption
    • full blood count
    • ferritin
    • serum folate
    • vitamin B12
    • calcium
    • alkaline phosphatase),
  • associated autoimmune conditions
    • thyroid-stimulating hormone and thyroid hormone(s)
    • serum glucose
  • liver disease
    • aspartate aminotransferase/alanine aminotransferase
  • dietary adherence
    • anti-TG2 or EMA/DGP
  • In follow-up of CD, the key endpoints are normalisation of the health of patients judged by an absence of symptoms, and mucosal healing. Lack of symptoms or negative serological markers are not reliable or responsive surrogates of mucosal response to diet. The proportion of patients who do not achieve full histological recovery on diet varies, with most reports suggesting mucosal healing in 57–76%.
  • Some experts favor repeat intestinal biopsy after 1 year of dietary therapy.

 

Assessing Adherence to the GFD, Gluten Challenge, and Medical Management during Follow-up

  • Whilst the panel of experts agree adherence to the GFD is most important for the health of a patient with CD, there are no evidence-based recommendations regarding the most useful way to assess this. There are four steps to assess dietary adherence:
    • clinical assessment of symptoms
    • dietetic review
    • serum antibodies
    • follow-up biopsy
  • Symptomatic patients should be evaluated more thoroughly than asymptomatic patients.
  • To perform a gluten challenge, a 14-day gluten intake at ≥3 g of gluten/day (two slices of wheat bread per day) to induce histological and serological changes in the majority of adults with CD is recommended. The challenge can be prolonged to 8 weeks if serology remains negative at 2 weeks.
  • Long-term medical follow-up can be in secondary care clinics or in primary care as long as the expertise is available. Prompt access to specialist or secondary care is recommended if any problems arise.
  • The need for long-term follow-up is controversial.

 

Increased Risk of Osteoporosis and Bone Fracture

  • It is recommended to measure calcium, alkaline phosphatase and vitamin D levels (and parathyroid hormone for compensatory increase) at diagnosis and replace as necessary.
  • Calcium intake should be maintained at or above 1000 mg per day.
  • Bone density should be measured in those at high risk of osteoporosis.
  • Repeat bone density investigations (generally after an interval of ≥2 years) should be considered in patients who have low bone density on index measurement following initiation of appropriate treatment, or who have evidence of ongoing villous atrophy or poor dietary adherence.
  • Postmenopausal women with CD may require supplementation in addition to the GFD.
  • Loss of bone density at a greater than expected rate should prompt measurement of vitamin D levels, dietary review of adherence, consideration of repeat intestinal mucosal biopsy and review of additional risk factors such as hypogonadism.

 

Vaccination Recommendations 

  • Hyposplenism associated with CD may result in impaired immunity to encapsulated bacteria. Hyposplenism does not seem to correlate with duration of GFD. Vaccination against Pneumococcus is therefore recommended. However, it is unclear whether vaccination with the conjugated vaccine is preferable in this setting and whether additional vaccination against Haemophilus,Meningococcus and Influenza should be considered if not previously given.
  • It should also be noted that patients with CD may have a weaker response to hepatitis B vaccination than normal.

 

Screening for CD

  • There is insufficient evidence to recommend population screening for CD, however there should be a low threshold for case finding in clinical practice as per National Institute for Health and Care Excellence guidelines. Symptomatic first-degree relatives of patients with CD should undergo CD testing.

 

Gluten-free Diet

  • The mainstay of treatment of CD and DH is a GFD. The term gluten should be used to indicate not only wheat-based proteins (gliadins), but it also includes those from barley (hordeins) and rye (secalins), and cereal hybrids such as triticale. Research indicates that oats uncontaminated by gluten are probably safe for patients with CD. Patients with CD should be educated to avoid cereals and food containing gluten (breakfast cereals, flours, pasta, cakes, biscuits, sauces etc) derived from wheat, barley or rye and food made from gluten-contaminated oats, and encouraged to eat naturally occurring gluten-free foods and alternative sources of starch (corn, rice, potatoes etc).
  • Patients must have a sufficient intake of nutrients, vitamins, fiber and calcium present in their GFD.
  • Whilst contamination of the diet by gluten may be unavoidable a clinical response and mucosal recovery can be achieved by strict adherence to a gluten-free diet. Systematic study review suggests that while the amount of tolerable gluten varies among people with CD, a daily gluten intake of <10 mg is unlikely to cause significant histological abnormalities.
  • Patients with CD complain about the limitations in their social life because of gluten-free meals or concern about the safety of food when eating out. The availability of gluten-free food is clearly limited in more rural areas and shopping for gluten-free food is time consuming. In most countries, high-quality gluten-free products are available in supermarkets or in special health food stores and on the internet, but the cost of gluten-free food is much greater than the equivalent wheat-based foods.

 

Patient Information and Support

 

Non-responsive CD (NRCD)

  • After adoption of the GFD, 4–30% of patients with CD report persisting symptoms and are considered to be affected by non-responsive CD (NRCD). Inadvertent or deliberate gluten exposure is the most frequent cause of NRCD. Assessment for ongoing enteropathy plays a central role, therefore a follow-up biopsy is needed. Small bowel imaging should be performed in any patient with abdominal pain, persisting fever, obstruction, anaemia, gastrointestinal bleeding or unexplained weight loss. If duodenal biopsy does not reveal a persistent enteropathy, symptoms are likely to be due to a second condition. While the GFD is efficacious at controlling the signs and symptoms of CD and improving intestinal histology in most patients, virtually all individuals with CD will have symptomatic exacerbations due to gluten exposure and up to 30% of patients will have symptoms severe or chronic enough to visit their treating physician.
  • NRCD may be considered in three categories.
    • First NRCD is due to continued dietary exposure to gluten.
    • Second, it may be due to a pre-existing or coincidental condition causing symptoms that resemble CD which may have led to the detection of otherwise asymptomatic CD.
    • Finally, it may be due to conditions associated with CD, such as secondary lactose intolerance, pancreatic exocrine insufficiency, small bowel bacterial overgrowth, microscopic colitis and cow’s milk protein sensitivity.

 

Refractory Celiac Disease (RCD)

  • RCD is defined as persistent or recurrent malabsorptive symptoms and/or signs with villous atrophy despite a strict GFD for more than 12 months in the absence of other causes of villous atrophy or malignant complications and after confirmation of CD. In symptomatic patients with ongoing enteropathy and RCD, celiac-related malignancies and disorders that mimic CD must be excluded.
  • RCD is subdivided into type I (RCDI) and type II (RCDII). The most important aspect of differentiating RCDI and RCDII is demonstration of a monoclonal population of T cells or aberrant T cells in the latter. Patients with RCDII have a poorer prognosis, due predominantly to nutritional complications and transformation into enteropathy-associated T-cell lymphoma (EATL). Ulcerative jejunoileitis (UJI) is a rare condition characterised by inflammatory ulceration of the small bowel that arises in RCD. The finding of UJI should raise suspicion for lymphoma.
  • There is no standard treatment for RCD. Elemental diets, systemic steroids, oral budesonide, oral thioguanines including azathioprine are used in RCDI, but have limited benefit in RCDII. In RCDII, cyclosporine, cladribine and high-dose chemotherapy with autologous stem cell support have been reported; however, therapy must be individualised and include surveillance for EATL.
  • EATL is a rare lymphoma strongly associated with RCDII, which carries a poor prognosis with a cumulative 5-year survival of less than 20%. Currently, two groups of EATL are recognised: EATL type I accounts for 80–90% of all cases and is a large cell lymphoma exclusively associated with CD. In contrast, EATL type II has not been associated with CD.
  • The poor prognosis of EATL is determined by extent of disease at diagnosis, multifocal small bowel involvement, poor general health and presence of complications including perforation that preclude chemotherapy.
  • Presence of RCDII is associated with a poor prognosis compared with isolated EATL in CD without RCDII.
  • There is no proven effective treatment in RCDII, though a number of strategies have been proposed, and patients should be referred to a tertiary centre to optimise their management.
  • Finally, numerous studies have confirmed the association between CD and B-cell lymphoma and small intestinal adenocarcinoma.

 

Novel Treatment

  • None of the available novel treatments can as yet be recommended for use outside clinical trials.

 

In Summary 

  • The treatment of CD is a lifelong and strict GFD. The goal of treatment is to relieve symptoms, achieve mucosal healing, avoid complications of CD. Follow-up of CD is needed to ensure response to symptoms, prevention of consequences, and continued maintenance of motivation to remain gluten free.
  • Adequate (more than four) biopsies should be taken, from the distal duodenum and the duodenal bulb to maximise diagnosis.
  • Duodenal biopsies are recommend  before the diagnosis of CD.
  • Ideally a combination of serology and biopsies done on a gluten-containing diet will then provide the most robust diagnosis of CD.
  • Specific serology such as IgA-TG2 and IgG-DGP with or without a strategy for determination of total IgA level should be the preferred serologic strategy for detection of CD.
  • The paper recommends testing for CD in those with suggested symptoms or syndromes, especially if they have a first-degree relative with CD.

Recommendations

  1. Diagnosis of CD requires duodenal biopsy when the patient is on a gluten-containing diet and for the vast majority of adult patients also positive serology.
  2. Biopsy remains essential for the diagnosis of adult CD and cannot be replaced by serology. Follow-up should aim at strict adherence to a gluten-free diet.
  3. In individuals undergoing an upper endoscopy in whom laboratory tests or symptoms or endoscopic features suggest CD, duodenal biopsy should be considered.
  4. Duodenal biopsy should be retained as the mainstay for the diagnosis of adult CD and cannot be replaced by serology.
  5. At endoscopy, if there is suspicion of CD, then at least four biopsy specimens should be obtained, including a duodenal bulb biopsy.
  6. In serologically negative patients showing signs of malabsorption (such as anemia or diarrhea) or a family history of CD, a duodenal biopsy should be considered.
  7. Follow-up biopsies may be considered in patients with CD, and are potentially helpful in identifying patients at increased risk of lymphoma.
  8. Follow-up biopsies are not mandatory if the patient with CD is asymptomatic on a GFD, and has no other features that suggest an increased risk of complications.
  9. Follow-up biopsies should be undertaken in patients with CD whose condition does not respond to a GFD.
  10. HLA typing should be used to rule out CD. A positive DQ2.5 or DQ8 can never confirm the diagnosis.
  11. HLA typing should be used in individuals who are self-treated on a GFD and never had appropriate testing for CD before changing their diet.
  12. HLA typing can be used to rule out CD, and minimise future testing, in high-risk individuals with CD, for example, first-degree relatives.
  13. When dietary adherence is questioned, it should be reviewed by a dietitian.
  14. Symptomatic patients should be evaluated more thoroughly than asymptomatic patients.
  15. Patients with CD require follow-up by a dietitian and/or clinician with an interest or expertise in this field.
  16. Patients should have annual haematological and biochemical profiles.
  17. Newly diagnosed patients should have vaccination for Pneumococcus.
  18. Bone density should be measured after 1 year of diet in patients who have additional risk factors for osteoporosis or if over the age of 55 years.
  19. Adult patients with CD should have a calcium intake of at least 1000 mg per day.
  20. A GFD is the core management strategy for prevention of osteoporosis.
  21. There is insufficient evidence to recommend population screening for CD, however there should be a low threshold for case finding in clinical practice as per National Institute for Health and Care Excellence guidelines.
  22. Symptomatic first-degree relatives of patients with CD should undergo CD testing.
  23. Patients should adhere to a GFD and have an intake of less than 10 mg gluten per day.
  24. Gluten challenge is not recommended in the ordinary patient with CD, but in patients in whom the diagnosis remains unclear despite a follow-up biopsy, gluten challenge should be performed.
  25. Patients may commence gluten-free oats at diagnosis.
  26. A GFD is recommended to decrease the excess risk of adverse foetal outcome and of lymphoma among patients with CD.
  27. At diagnosis, patients should be encouraged to join their national celiac support group.
  28. Patients with persistent symptoms despite a GFD should have a follow-up biopsy.
  29. In symptomatic patients with ongoing enteropathy and RCD, coeliac-related malignancies and disorders that mimic CD must be excluded.
  30. Small bowel imaging should be performed in any patient with abdominal pain, fever, obstruction, anaemia, gastrointestinal bleeding or unexplained weight loss.
  31. Patients with RCD should be referred to a tertiary centre to optimise their management.
  32. None of the available novel treatments can as yet be recommended for use outside clinical trials.

 

http://gut.bmj.com/content/63/8/1210

http://gut.bmj.com/content/63/8/1210.full

http://gut.bmj.com/content/63/8/1210.full.pdf+html

http://www.bsg.org.uk

http://gut.bmj.com

Management of Benign Anorectal Disorders – ACG Clinical Guidelines

 

The American College of Gastroenterology has released new guidelines on the management of benign disorders of anorectal function and/or structure. These guidelines summarize the definitions, diagnostic criteria, differential diagnoses, and treatments of a group of benign disorders of anorectal function and / or structure. Disorders of function include defecation disorders, fecal incontinence, and proctalgia syndromes, whereas disorders of structure include anal fissure and hemorrhoids

 

Defecatory disorders
Diagnosis should be based on clinical history of chronic constipation, abnormal balloon expulsion test (BET) and anorectal manometry (ARM) results.
Digital rectal examination (DRE) should be the initial screening method, and barium- or MR defecography should be used to rule out obstructions.
Biofeedback is the preferred treatment for adult patients.
Chronic proctalgia
Diagnosis should be based on history of rectal pain episodes lasting 20 minutes or more, abnormal DRE results and exclusion of other causes of pain. Structural causes should be eliminated by imaging study or endoscopy.
BET and ARM can identify patients likely to respond to biofeedback therapy, the preferred treatment. Electrical stimulation and digital massage are among inferior treatments.
Proctalgia fugax
Diagnosis should be based on history of intermittent anorectal pain in episodes less than 20 minutes.
Structural causes of anorectal pain should be ruled out by imaging, endoscopy or other tests.
There are no evidence-supported treatments; patients should be assured the disorder is benign.
Fecal incontinence
Predisposing conditions should be identified. Patients should be asked directly and not relied upon for spontaneously reporting these symptoms.
Symptom severity and characteristics are best determined by the Bristol stool scale and bowel diaries, respectively.
Physical examinations should rule out causal diseases. Other diagnostics include digital anorectal exam and DRE. For patients who fail to respond to conservative therapies, ARM, BET, rectal sensation testing, pelvic floor and anal canal imaging and anal EMG are recommended.
Symptom management strategies include education, diet, skin care, pharmacologic agents, antidiarrheal agents and pelvic floor rehabilitation.
The only minimally invasive procedure is injectable anal bulking agents, but more evidence of its efficacy is needed. Surgical treatments include sacral nerve stimulation, anal sphincteroplasty, dynamic graciloplasty, artificial anal sphincter, and colostomy as a last resort.
Anal fissure
Treatments for acute fissure include sitz-baths, psyllium fiber and bulking agents.
Treatments for chronic fissure include calcium channel blockers, nitrates, local injections of botulinum toxin or surgical internal anal sphincterotomy.
Hemorrhoids
Diagnosis should be based on history, physical examination or endoscopy.
Thrombosed external hemorrhoids should typically be treated with excision.
Internal hemorrhoids should initially be treated with increased fiber and fluid intake.
First- to third- degree hemorrhoids that remain symptomatic should be treated with banding, sclerotherapy, infrared coagulation or ligation.
Surgical procedures include hemorrhoidectomy, stapled hemorrhoidopexy and Doppler-assisted hemorrhoidal artery ligation.

 

DEFECATORY DISORDERS (DD)
1. DDs are defined as difficulty in evacuating stool from the rectum in a patient with chronic or recurring symptoms of constipation.
2. Gastroenterologists and other providers should not make the diagnosis of DD on the basis of a single abnormal test because none is sufficiently specific. However, confidence in the diagnosis is increased if there is a combination of a clinical history of chronic constipation and two abnormal tests, i.e., impaired ability to evacuate a 50-ml water-filled balloon or abnormal defecography and evidence from pelvic floor EMG or ARM that the patient is unable to relax pelvic floor muscles or increase rectal pressure during simulated defecation.
3. Digital rectal examination is a useful first test to screen for DD, as it has good negative predictive value.
4. Barium or MR defecography can identify structural causes of outlet obstruction if one is expected. They may also confirm or exclude the diagnosis of DD when the clinical features suggest DD but the results of ARM and BET are equivocal.
5. Biofeedback is the preferred treatment for DD in adults.
The treatment protocols used in most RCTs include the following steps:
  • Patient education – explain to patients that they unconsciously squeeze their anus when they are trying to defecate and this holds the stool in the rectum.
  • Simulated defecation training – for patients who do not increase intraabdominal pressure during simulated defecation, the use of feedback on rectal balloon pressure teaches them to tighten their abdominal wall muscles and lower their diaphragm to push stool out.
  • Training to relax pelvic floor muscles while simulating defecation — for patients who paradoxically contract their pelvic floor muscles during simulated defecation, provide visual feedback on anal canal pressure or averaged EMG activity from the anal canal to teach this skill.
  • Practicing simulated defecation – patients practice defecation of a lubricated, inflated balloon while the therapist gently pulls on the catheter to assist them. Remind the patient to relax the pelvic floor muscles, increase abdominal pressure using abdominal wall muscles, and concentrate on the sensations produced by balloon passage.

 

PROCTALGIA SYNDROMES
1. Gastroenterologists and other providers should make a diagnosis of chronic proctalgia based on a history of recurring episodes of rectal pain, each lasting at least 20 minutes, a digital rectal examination showing tenderness to palpation of the levator ani muscles, and exclusion of other causes for rectal pain by history and diagnostic testing.
2. Gastroenterologists and other providers should obtain an imaging study or endoscopy to rule out structural causes of rectal pain.
3. Gastroenterologists and other providers should obtain a BET and ARM to identify patients with chronic proctalgia and levator muscle tenderness who are likely to respond to biofeedback.
4. Biofeedback to teach relaxation of pelvic floor muscles during simulated defecation is the preferred treatment.
5. Electrical stimulation is superior to digital massage but inferior to biofeedback.
6. Gastroenterologists and other providers should make a diagnosis of proctalgia fugax on the basis of a history of intermittent bouts of severe pain in the anal canal or lower rectum lasting less than 20 minutes.
7. Gastroenterologists and other providers should exclude structural causes of anorectal pain (e.g., anal fissure, hemorrhoids, cryptitis, malignancy) by imaging, endoscopy, or other appropriate tests.
8. Gastroenterologists and other providers should assure patients that the disorder is benign. The evidence for specific treatments is no better than anecdotal.

 

FECAL INCONTINENCE (FI)
1. Gastroenterologists and other providers should ask patients about the presence of FI directly rather than relying on spontaneous reporting.
2. Gastroenterologists and other providers should identify conditions that may predispose to FI i.e.:
  • Anal sphincter weakness
  • Traumatic: obstetric, surgical (e.g., fistulotomy, internal sphincterotomy)
  • Nontraumatic: scleroderma, internal sphincter degeneration of unknown etiology
  • Neuropathy: peripheral (e.g., pudendal) or generalized (e.g., diabetes mellitus)
  • Disturbances of pelvic floor: rectal prolapse, descending perineum syndrome
  • Inflammatory conditions: radiation proctitis, Crohn’s disease, ulcerative colitis
  • Central nervous system disorders: dementia, stroke, brain tumors, multiple sclerosis, spinal cord lesions
  • Diarrhea: irritable bowel syndrome, post-cholecystectomy diarrhea Other: fecal retention with overflow, behavioral disorders
3. Gastroenterologists and other providers should determine symptom severity by quantifying stool type using the Bristol stool scale, as well as characterizing the frequency, amount of leakage, and the presence of urgency.
4. Gastroenterologists and other providers should obtain bowel diaries because they are superior to self-reports for characterizing bowel habits and FI.
5. Gastroenterologists and other providers should perform a physical examination to eliminate diseases to which FI is secondary.
6. Gastroenterologists and other providers should perform a digital anorectal examination to identify rectal masses, gauge anal sphincter tone at rest, during voluntary contraction of the anal sphincter and pelvic floor muscles, and during simulated defecation.
7. Gastroenterologists and other providers should perform a digital rectal examination before making a referral for anorectal manometry.
8. ARM, BET, and rectal sensation should be evaluated in patients who fail to respond to conservative measures.
9. Pelvic floor and anal canal imaging, as well as anal EMG, should be considered for patients with reduced anal pressures who have failed conservative therapy, particularly if surgery is being considered.
10. Gastroenterologists and other providers should manage patients with FI using education, dietary modifications, skin care, and pharmacologic agents to modify stool delivery
and liquidity before diagnostic testing, particularly when symptoms are mild and not bothersome.
11. Gastroenterologists and other providers should prescribe antidiarrheal agents for FI in patients with diarrhea.
12. Pelvic floor rehabilitative techniques are effective and superior to pelvic floor exercises alone in patients with FI who do not respond to conservative measures.
13. Minimally invasive procedures such as injectable anal bulking agents may have a role in patients with FI who do not respond to conservative therapy.
14. There is insufficient evidence to recommend radiofrequency ablation treatment to the anal sphincter at this time.
15. Sacral nerve stimulation should be considered in patients with FI who do not respond to conservative therapy.
16. Anal sphincteroplasty should be considered in patients with FI who do not respond to conservative therapy and who have an anatomic sphincter defect.
17. Dynamic graciloplasty and artificial anal sphincter, where available, may possibly allow the occasional patient with FI to avoid colostomy.
18. Colostomy is a last resort procedure that can markedly improve the quality of life in a patient with severe or intractable FI.

 

ANAL FISSURE
1. Gastroenterologists and other providers should use nonoperative treatments such as sitz-baths, psyllium fiber, and bulking agents as the first step in therapy of acute fissure.
2. Gastroenterologists and other providers should treat chronic anal fissure with topical pharmacologic agents such as a calcium channel blockers or nitrates.
3. Gastroenterologists and other providers should refer patients who do not respond to conservative or pharmacologic treatment for local injections of botulinum toxin or surgical internal anal sphincterotomy.

 

HEMORRHOIDS
1. Gastroenterologists and other providers should diagnose hemorrhoids by history and physical examination. If there is bleeding, the source often requires confirmation by endoscopic studies.
2. Most patients who present urgently (within ~ 3 days of onset) with a thrombosed external hemorrhoid benefit from excision.
3. Gastroenterologists and other providers should treat patients with symptomatic hemorrhoids first with increased fiber intake and adequate fluids.
4. Gastroenterologists and other providers should consider patients with first- to third-degree hemorrhoids that remain symptomatic after dietary modifications for office procedures
such as banding, sclerotherapy, and infrared coagulation. Ligation is probably the most effective option.
5. Gastroenterologists and other providers should refer for surgical operations (hemorrhoidectomy, stapled hemorrhoidopexy, and Doppler-assisted hemorrhoidal artery ligation) those patients who are refractory to or cannot tolerate office procedures, who have large, symptomatic external tags along with their hemorrhoids, who have large third-degree hemorrhoids, or who have fourth-degree hemorrhoids.

 

The paper can be accessed and downloaded at http://gi.org/guideline/management-of-benign-anorectal-disorders/

Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis

The World Gastroenterology Organization (WGO) Global Guidelines published guidelines on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis  in the July issue of the Journal of Clinical Gastroenterology.

Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are the leading cause of liver disease in the western countries and NAFLD and NASH play an equally important role in the Middle East, Far East, Africa, the Caribbean, and Latin America. While NAFLD and NASH lack definitive etiology, effective therapy, or evidence-based clinical guidelines, the published WGO recommendations offer needed guidance for clinicians.

 

NAFLD is defined by excessive fat accumulation (steatosis) in the liver. NAFLD does not correlate with increased short-term morbidity or mortality.

 

NASH: A subgroup of NAFLD patients develop inflammation (steatohepatitis) which is histologically indistinguishable from alcoholic steatohepatitis. NASH dramatically increases the risks of cirrhosis, liver failure, and hepatocellular carcinoma.The exact cause of NASH has not been elucidated, although it is most closely related to insulin resistance, obesity, and the metabolic syndrome. Possible other risk factors include: hypertension, type 2 diabetes, sleep apnea, a positive family history, nonblack ethnicity, obesity, hyperlipidemia, and a sedentary lifestyle.

 

WGO Recommendations for NAFLD/NASH Diagnosis:
  • Suspected NAFLD involves central obesity, diabetes mellitus, dyslipidemia, metabolic syndrome, abnormal liver function tests, and/or ultrasound changes consistent with fatty liver.
  • Minimal assessment should include
    • bilirubin
    • alanine aminotransferase
    • aspartate aminotransferase
    • g-glutamyl transferase
    • albumin
    • fasting serum lipids
    • complete blood count
    • hepatitis C virus
    • hepatitis B surface antigen
    • antinuclear antibody
    • fasting blood glucose
    • oral glucose tolerance test if fasting blood glucose level of 5.6 mmol/L or higher
    • height, weight, body mass index, waist circumference
    • blood pressure
    • abdominal ultrasound
  • Optional tests include
    • abdominal computed tomography if uninformative ultrasound
    • liver biopsy if diagnostic uncertainty or for patients at risk for advanced hepatic fibrosis.
  • Additional tests include tests for
    • hereditary hemochromatosis
    • Wilson’s disease
    • alpha-1 antitrypsin deficiency
    • polycystic ovary syndrome
    • autoimmune liver diseases

 

WGO SUMMARY STATEMENT

  • NAFLD and NASH represent a major global public health problem, which is pandemic and affects rich and poor countries alike.
  • There is insufficient evidence to justify screening for NASH/advanced liver disease in the general population.
  • The diagnosis should be sought in all patients who present with risk factors for NASH.
  • Not all patients with risk factors will have NAFLD or NASH, and not all patients with NASH will have standard risk factors.
  • Not every person with fatty liver needs aggressive therapy.

 

  • Diet and exercise should be instituted for all patients.
  • Liver biopsy should be reserved for those patients who have risk factors for NASH and/or other liver diseases.
  • Patients with NASH or risk factors for NASH should first be treated with diet and exercise. Vitamin E or pentoxifylline may be added in these patients. Experimental therapy should be considered only in appropriate hands and only in patients who fail to achieve a 5% to 10% weight reduction over 6 months to 1 year of successful lifestyle changes.
  • Bariatric surgery should be considered in patients in whom the above approaches fail, and it should be performed before the patient becomes cirrhotic.
  • Liver transplantation is successful in patients who meet the criteria for liver failure; however, NASH may recur after transplantation and is likely to be denied to patients with morbid obesity.
  • NAFLD and NASH are also becoming an increasingly serious problem in pediatric patients, including those under the age of 10.

 

Ultimately, NAFLD and NASH are diagnoses of exclusion and require careful consideration of other diagnoses. Just as the clinician cannot diagnose NASH on the basis of clinical data alone, the pathologist can document the histologic lesions of steatohepatitis, but cannot reliably distinguish those of nonalcoholic origin from those of alcoholic origin.

J Clin Gastroenterol. 2014;48:467-473.

Prevention and Treatment of Venous Thromboembolism in Patients with IBD

Guidelines for the management of venous thromboembolism (VTE) from the American College of Chest Physicians do not address patients with inflammatory bowel disease (IBD), a group with a high risk of both VTE and gastrointestinal bleeding. The current published consensus statements identify how the 9th  American College of Chest Physician (ACCP) guidelines on antithrombotic therapy and prevention of thrombosis should be applied to patients with IBD, and how patients with IBD should be managed when those guidelines are not appropriate or have not addressed issues that are specific to IBD patients.                                                                                                                                                                                                 
In Summary
  • Inflammatory Bowel Disease (IBD) patients have about a 3-fold higher risk of Venous Thromboembolism (VTE) compared with the general population.
  • Moderate to severe disease activity drives the increased risk of VTE in IBD and should be considered a provoking factor.
  • For the prevention of VTE, strong recommendations are made for 1) anticoagulant thromboprophylaxis over no prophylaxis for patients with IBD who are hospitalized with moderate to severe IBD flares without severe bleeding, for 2) anticoagulant thromboprophylaxis over no prophylaxis for inpatients with IBD who have undergone major abdominal-pelvic or general surgery, and 3) against anticoagulant thromboprophylaxis in outpatients with an IBD flare if they have not had a previous VTE.
  • For the treatment of VTE, strong recommendations are made for a minimum of 3 months of anticoagulant therapy for adult and pediatric IBD patients with a symptomatic DVT, PE, or splanchnic vein thrombosis. If anticoagulant therapy is being stopped in patients with a reversible provoking factor, it should not be stopped until the risk factor has resolved for at least 1 month.
  • Hospitalized patients with moderate-to-severe flares (without severe bleeding) should receive anticoagulant thromboprophylaxis with low-molecular-weight heparin, heparin, or fondaparinux.

  • Patients hospitalized for indications unrelated to IBD or for IBD with non-severe gastrointestinal bleeding should receive anticoagulant prophylaxis.

  • Hospitalized patients with severe IBD-related bleeding should receive mechanical prophylaxis (preferably intermittent pneumatic compression (IPC)). Once IBD-related bleeding is no longer severe, substitute anticoagulation for mechanical prophylaxis.

  • Patients should receive anticoagulant prophylaxis during hospitalization for major abdominopelvic or general surgery.

  • Outpatients with IBD flares and no prior VTE do not need prophylaxis.

  • Patients with prior VTE and moderate-to-severe flares should receive anticoagulant prophylaxis unless prior episodes occurred only after major surgery.

  • Pregnant women with IBD should receive anticoagulant prophylaxis after C-section until hospital discharge.

  • Patients with VTE who have coexisting IBD do not need testing for hereditary or acquired hypercoagulable states.

  • Patients who experience their first episode of VTE while in remission should receive indefinite anticoagulation unless there is an unrelated reversible provoking factor (then a minimum of 3 months).

  • Patients who experience their first episode of VTE in the presence of active disease should be anticoagulated until IBD is in remission for 3 months.

  • Recommendations for pediatric patients are similar to those for adults.

  • Patients with symptomatic acute splanchnic vein thrombosis (portal, mesenteric, splenic vein thrombosis) should be treated with principles similar to those with VTE in other sites.

  • Patients with asymptomatic, incidentally detected splanchnic vein thrombosis (on imaging studies) should not be anticoagulated.

Please review the original position statement as published in Gastroenterology March, 2014 by the Canadian Association of Gastroenterology.
Nguyen GC et al. Consensus statements on the risk, prevention, and treatment of venous thromboembolism in inflammatory bowel disease: Canadian Association of Gastroenterology. Gastroenterology 2014 Mar; 146:835.

Guidelines for Safety in the Gastrointestinal Endoscopy Unit

Check out this great position statement and guideline by the American Society for Gastrointestinal Endoscopy (ASGE) on the key strategies to maintain safety in the GI endoscopy unit:

From the position statement: …Over the past 2 years, surveyors have called into question accepted practices at many accredited endoscopy units. Many of these issues relate to the Ambulatory Surgical Center Conditions for Coverage set forth by CMS and the lack of distinction between the sterile operating room and the endoscopy setting. The following is a summary of issues that have been faced by endoscopy units throughout the country along with the ASGE position and accompanying rationale.

In summary:

  • Each unit should have a designated flow for the safe physical movement of dirty endoscopes and other equipment.
  • Procedure rooms vary in size, with more complex procedures requiring greater space for more specialised equipment and, in some cases, additional staff.
  • Before starting an endoscopic procedure, the patient, staff, and performing physician should verify the correct patient and procedure to be performed.
  • A specific infection prevention plan must be implemented and directed by a qualified person.
  • Gloves and an impervious gown should be worn by staff engaged in direct patient care during the procedure.
  • The unit should have a terminal cleansing plan that includes methods and chemical agents for cleansing and disinfecting the procedural space at the end of the day.
  • For patients undergoing routine endoscopy under moderate sedation, a single nurse is required in the room in addition to the performing physician.
  • Complex procedures may require additional staff for efficiency but not necessarily for safety.
  • At a minimum, patient monitoring should be performed before the procedure, after administration of sedatives, at regular intervals during the procedure, during initial recovery, and before discharge.
  • For cases in which moderate sedation is the target, the individual responsible for patient monitoring may perform brief interruptible tasks.
  • For cases in which moderate sedation is the target, there are currently inadequate data to support the routine use of capnography.

For those interested in designing or improving an endoscopy unit, this position statement is a must-read.

SOURCE: American Society for Gastrointestinal Endoscopy. The Role of Endoscopy in the evaluation and management of dysphagia. Gastrointest Endosc 2014:79:191-201

http://www.asge.org/assets/0/71542/71544/4a572112-29a4-4313-8ab8-b7801e8f84e2.pdf

Consider Food Allergy Testing in Managing Eosinophilic Esophagitis

Food antigens can induce esophageal remodeling with fibrosis, which can resolve with appropriate food antigen elimination. Addressing food allergens in the treatment of eosinophilic esophagitis (EoE) is recommended and although allergy testing is common practice for pediatric gastroenterologists, many adult gastroenterologists are unfamiliar with the practice. A recently (online published) article provides in-depth guidance, including:

  • Allergy testing for foods may be more useful in pediatric patients.

  • The negative predictive values of tests for identifiable food allergies are generally higher than positive PVs.

  • Patients with EoE are often highly atopic and have polysensitization to both food (particularly in children) and aeroallergens (particularly in adults).

  • Ample evidence supports use of skin-prick or atopy-patch testing for building an elimination diet in children, but not in adults.

  • Do not utilize food-specific panels using serum immunoglobulin E (or other immunoglobulins) in this setting.

  • EoE recurs in >90% of pediatric patients after food reintroduction, and only 8% become tolerant of all their food triggers.

  • Repeat endoscopic biopsy with histologic evaluation after sequential food reintroduction – do not rely on a positive test for food allergy.

  • Consider the possibility of concurrent allergic diatheses in patients with EoE.

Studies recommend that one food or food group be introduced every 4 to 6 weeks with observation of clinical symptoms and a subsequent endoscopy if no change in symptoms occurs. A food trigger is identified based on the recurrence of symptoms and esophageal eosinophilia (≥15 eosinophils/high-power field) after reintroduction of a specific food group. Because patients typically have multiple food triggers, the process should be continued until all foods have been reintroduced into the diet or an acceptable diet is reached. Dieticians can play an important role in identifying possible cross-contamination of specific food items. Clearly, an integrated approach involving allergists, gastroenterologists, and pathologists is warranted in evaluating the possible role of food allergy in eosinophilic esophagitis.

http://www.cghjournal.org/article/S1542-3565(13)01302-5/fulltext