Organ-Related Staging and Grading

Chapter 2 Organ-Related Staging and Grading


Oropharynx


Esophagus


Stomach


Small Intestine


Colon


Liver


Biliary System


Pancreas


Spleen


Peritoneal Cavity



Oropharynx


Anatomical Variants


Laryngotracheal-Esophageal Cleft: Grading the Laryngotracheal-Esophageal Cleft According to Pettersson


Aims

The original classification of laryngotracheal-esophageal clefts is divided into three types depending on the extent of tracheal involvement (for Type IV see Comments below).


 
















Type I


Cleft involves the larynx, inter-arytenoid muscles, and the cricoid laminae


Type II


Extends beyond the cricoid lamina up to the cervical trachea (sixth tracheal ring)


Type III


Involves the trachea including the carina


Type IV


Cleft extending beyond the carina into either one or both main stem bronchi


From Ryan DP, Muehrcke DD, Doody DP. Laryngotracheoesophageal cleft (type IV): Management and repair of lesions beyond the carina. J Pediatr Surg. 1991;26:962–970. With permission of Elsevier.


Comments

Ryan et al. added type IV in 1991. They described three cases of laryngotracheal-esophageal clefts extending into the bronchi.


References

Pettersson G. Laryngotracheal esophageal cleft. Z Kinderchir. 1969;7:43–49.


Ryan DP, Muehrcke DD, Doody DP. Laryngotracheoesophageal cleft (type IV): Management and repair of lesions beyond the carina. J Pediatr Surg. 1991;26:962–970.


Laryngotracheal-Esophageal Cleft: Grading of Laryngotracheal Cleft According to Evans


Aims

To modify Pettersson’s classification of laryngotracheal-esophageal clefts to allow a more precise definition of the milder cases in which surgery carries a reasonable prognosis.


 



















Type 1


Limited to cricoid cartilage: 1A Inter-arytenoid cleft, extending to, but not into the superior aspect of the cricoid cartilage; 1B Cleft extends partially through the cricoid; 1C Cleft extends completely through the cricoid


Type 2


Cleft includes the proximal 3 cm of the tracheoesophageal septum


Type 3


Involves the trachea including the carina


Type 4


Cleft involves the entire tracheoesophageal septum


From Pettersson G. Inhibited separation of larynx and the upper part of trachea from oesophagus in a newborn; report of a case successfully operated upon. Acta Chir Scand. 1955;110:250–254. By permission of Taylor & Francis AS.


References

Armitage EN. Laryngotracheo-esophageal cleft. A report of three cases. Anaesthesia. 1984;39:706–713.


Pettersson G. Inhibited separation of larynx and the upper part of trachea from oesophagus in a newborn; report of a case successfully operated upon. Acta Chir Scand. 1955;110: 250–254.


Laryngotracheal-Esophageal Cleft: Grading of Laryngotracheal-Esophageal Cleft According to Armitage


Aims

To review the clinical features, associated congenital abnormalities, management, and morbidity of infants presenting with posterior laryngeal and laryngotracheal clefts.


 




















Type I


31%


Clefts are limited to the inter-arytenoid region above the vocal folds. This type does not involve the cricoid cartilage


Type II


47%


This type includes the cricoid and extends into the cervical trachea


Type III


22%


This type involves the thoracic trachea


From Evans JNG. Management of the cleft larynx and tracheoesophageal clefts. Ann Otol Rhinol Laryngol. 1985;94:627–630. With permission of Annals Publishing Co.


Comments

Treatment is conservative, via primary endoscopic surgical repair or primary repair via an anterior laryngofissure. Gastroesophageal reflux is controlled by fundoplication.


References

Evans JNG. Management of the cleft larynx and tracheoesophageal clefts. Ann Otol Rhinol Laryngol. 1985;94: 627–630.


Laryngotracheal-Esophageal Cleft: Grading of Laryngotracheal-Esophageal Cleft According to Benjamin and Inglis and According to Dubois


Aims

Benjamin and Inglis and Dubois proposed a classification of laryngeal clefts with four main types.


 
















Grading according to Benjamin and Inglis

Type I


Cleft is limited to the supraglottic lumen above the vocal folds


Type II


A partial cleft of the cricoid extending below the level of the vocal folds


Type III


Involves the whole cricoid cartilage and may extend to the cervical TE septum


Type IV


Involves a major part of the TE wall in the thorax


From Benjamin B, Inglis A. Minor congenital laryngeal clefts: diagnosis and classification. Ann Otol Rhinol Laryngol. 1989;98:417–420. With permission of Annals Publishing Co.


 



















Grading according to Dubois

Type I


Cleft extends down to, but does not involve, the superior portion of the posterior cricoid plane


Type II


Cleft extends into and at times as far down as the inferior aspect of the posterior cricoid plane


Type III


Cleft extends into the cervical trachea to a variable distance


Type IV


Cleft extends into the thoracic trachea and may reach the carina or even beyond to involve one or both main-stem bronchi


From DuBois JJ, Pokorny WJ, Harberg FJ, Smith RJ. Current management of laryngeal and laryngotracheoesophageal clefts. J Pediatr Surg. 1990;25:855–860. With permission of Elsevier Inc.


Inflammation


Oral Problems in the Elderly: The Dental Screening Tool


Aims

To propose a screening instrument for oral problems in the elderly for referral and further evaluation.


 
































The dental screening tool

 


 


Score


D


Dry mouth


2


E


Eating difficulty


1


N


No dental care within past two years


1


T


Tooth loss


2


A


Alternative food selection because of masticatory problems


1


L


Lesions, scores, or lumps in mouth


1


From Bush LA, Horenkamp N, Morley JE, Spiro A 3rd. D-E-N-T-A-L: A rapid self-administered screening instrument to promote referrals for further evaluation in older adults. J Am Geriatr Soc. 1996;44:979–481. With permission of Blackwell Publishing.


image


Fig. 2.1 Laryngotracheal-esophageal clefts: grading according to Benjamin and Inglis and according to Dubois.


Comments

The cleft larynx is a rare congenital anomaly. Type II–IV require urgent airway management. Dubois classified laryngotrachealesophageal clefts on an embryological basis.


References

Benjamin B, Inglis A. Minor congenital laryngeal clefts: diagnosis and classification. Ann Otol Rhinol Laryngol. 1989; 98:417–420.


DuBois JJ, Pokorny WJ, Harberg FJ, Smith RJ. Current management of laryngeal and laryngotracheoesophageal clefts. J Pediatr Surg. 1990;25:855–860.


Comments

This score can be used to identify potential oral problems.


References

Bush LA, Horenkamp N, Morley JE, Spiro A 3rd. D-E-N-T-A-L: A rapid self-administered screening instrument to promote referrals for further evaluation in older adults. J Am Geriatr Soc. 1996;44:979–481.


Thompson WM, Brown RH, Williams SM. Dentures, prosthetic treatment needs, and mucosal health in an institutionalized elderly population. N Z Dent J. 1992;88:51.


Gastroesophageal Reflux: The Eccles and Jenkins Index of Dental Lesions


Aims

Dental and periodontal lesions in patients with gastroesophageal reflux disease are graded.


 























Rating


Erosion severity


Grade 0


No superficial enamel modification


Grade 1


Loss of superficial enamel features without reaching dentin


Grade 2


Dentin exposure less than 1/3 of surface


Grade 3


Dentin exposure more than 1/3 of surface


From Eccles JD, Jenkins WG. Dental erosion and diet. J Dent. 1974;2: 153–159. With permission of Elsevier.


Neoplastic Lesions


Pharyngeal Carcinoma: AJCC TNM Staging System for Oropharynx and Hypopharynx Malignancy


 













Primary tumor (T)

TX


Primary tumor cannot be assessed


T0


No evidence of primary tumor


Tis


Carcinoma in situ


 



















Oropharynx

T1


Tumor 2 cm or less in greatest dimension


T2


Tumor more than 2 cm but not more than 4 cm in greatest dimension


T3


Tumor more than 4 cm in greatest dimension


T4a


Tumor invades the larynx, deep/extrinsic muscle of tongue, medial pterygoid, hard palate, or mandible


T4b


Tumor invades lateral pterygoid muscle, pterygoid plates, lateral nasopharynx, or skull base or encases carotid artery


 



















Hypopharynx

T1


Tumor limited to one subsite of hypopharynx and 2 cm or less in greatest dimension


T2


Tumor invades more than one subsite of hypopharynx or an adjacent site, or measures more than 2 cm but not more than 4 cm in greatest diameter without fixation of hemilarynx


T3


Tumor measures more than 4 cm in greatest dimension or with fixation of hemilarynx


T4a


Tumor invades thyroid/cricoid cartilage, hyoid bone, thyroid gland, esophagus or central compartment soft tissue


T4b


Tumor invades prevertebral fascia, encases carotid artery, or involves mediastinal structures


Comments

Specificity is limited, especially with associated dental decay. The superficial effects of reflux on enamel are difficult to recognize. Only grade II and III can be seen with a retro mirror.


References

Eccles JD, Jenkins WG. Dental erosion and diet. J Dent. 1974;2: 153–159.


 




























Regional lymph nodes (N): oropharynx and hypopharynx

NX


Regional lymph nodes cannot be assessed


N0


No regional lymph node metastasis


N1


Metastasis in a single ipsilateral lymph node, 3 cm or less in greatest dimension


N2


Metastasis in a single ipsilateral lymph node, more than 3 cm but not more than 6 cm in greatest dimension, or multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension, or in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension


N2a


Metastasis in a single ipsilateral lymph node more than 3 cm but not more than 6 cm in greatest dimension


N2b


Metastasis in multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension


N2c


Metastasis in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension


N3


Metastasis in a lymph node more than 6 cm in greatest dimension


 













Distant metastasis (M): oropharynx and hypopharynx

MX


Presence of distant metastasis cannot be assessed


M0


No distant metastasis


M1


Distant metastasis


image


 
















Histologic grade (G)

GX


Grade cannot be assessed


G1


Well differentiated


G2


Moderately differentiated


G3


Poorly differentiated


 
















Residual tumor (R)

RX


Presence of residual tumor cannot be assessed


R0


No residual tumor


R1


Microscopic residual tumor


R2


Macroscopic residual tumor


From Greene, FL, Page, DL, Fleming ID et al. Used with the permission of the American Joint Committee on Cancer (AJCC), Chicago, Illinois. The original source for this material is the AJCC Cancer Staging Manual. 6th ed (2002) published by Springer-New York, www.springeronline.com.


Comments

The AJCC TNM staging system uses three basic descriptors that are then grouped into stage categories. The first component is “T,” which describes the extent of the primary tumor. The next component is “N,” which describes the absence or presence and extent of regional lymph node metastasis. The third component is “M,” which describes the absence or presence of distant metastasis. The final stage groupings (determined by the different permutations of “T,” “N,” and “M”) range from Stage 0 through Stage IV.


Reference

Greene, FL, Page, DL, Fleming ID et al. AJCC Cancer Staging Manual. 6th ed. New York: Springer, 2002. http://www.cancerstaging.org/.


Larynx Carcinoma: AJCC TNM Staging System of the Larynx


 













Primary tumor (T)

TX


Primary tumor cannot be assessed


T0


No evidence of primary tumor


Tis


Carcinoma in situ


 



















Supraglottis

T1


Tumor limited to one subsite of supraglottis with normal vocal cord mobility


T2


Tumor invades mucosa of more than one adjacent subsite of supraglottis or glottis or region outside the supraglottis (e.g., mucosa of base of tongue, vallecula, medial wall of pyriform sinus) without fixation of the larynx


T3


Tumor limited to larynx with vocal cord fixation and/or invades any of the following: postcricoid area, pre-epiglottic tissues, paraglottic space, and/or minor thyroid cartilage erosion (e.g., inner cortex)


T4a


Tumor invades through the thyroid cartilage and/or invades tissues beyond the larynx (e.g., trachea, soft tissues of neck including deep extrinsic muscle of the tongue, strap muscles, thyroid, or esophagus)


T4b


Tumor invades prevertebral space, encases carotid artery, or invades mediastinal structures


 

























Glottis

T1


Tumor limited to the vocal cord(s) (may involve anterior or posterior commissure) with normal mobility


T1a


Tumor limited to one vocal cord


T1b


Tumor involves both vocal cords


T2


Tumor extends to supraglottis and/or subglottis, or with impaired vocal cord mobility


T3


Tumor limited to the larynx with vocal cord fixation, and/or invades paraglottic space, and/or minor thyroid cartilage erosion (e.g., inner cortex)


T4a


Tumor invades through the thyroid cartilage and/or invades tissues beyond the larynx (e.g., trachea, soft tissues of neck including deep extrinsic muscle of the tongue, strap muscles, thyroid, or esophagus)


T4b


Tumor invades prevertebral space, encases carotid artery, or invades mediastinal structures


 



















Subglottis

T1


Tumor limited to the subglottis


T2


Tumor extends to vocal cord(s) with normal or impaired mobility


T3


Tumor limited to larynx with vocal cord fixation


T4a


Tumor invades cricoid or thyroid cartilage and/or invades tissues beyond the larynx (e.g., trachea, soft tissues of neck including deep extrinsic muscles of the tongue, strap muscles, thyroid, or esophagus)


T4b


Tumor invades prevertebral space, encases carotid artery, or invades mediastinal structures


 




























Regional lymph nodes (N)

NX


Regional lymph nodes cannot be assessed


N0


No regional lymph node metastasis


N1


Metastasis in a single ipsilateral lymph node, 3 cm or less in greatest dimension


N2


Metastasis in a single ipsilateral lymph node, more than 3 cm but not more than 6 cm in greatest dimension, or multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension, or in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension


N2a


Metastasis in a single ipsilateral lymph node more than 3 cm but not more than 6 cm in greatest dimension


N2b


Metastasis in multiple ipsilateral lymph nodes, none more than 6 cm in greatest dimension


N2c


Metastasis in bilateral or contralateral lymph nodes, none more than 6 cm in greatest dimension


N3


Metastasis in a lymph node more than 6 cm in greatest dimension


 













Distant metastasis (M)

MX


Presence of distant metastasis cannot be assessed


M0


No distant metastasis


M1


Distant metastasis


image


 
















Histologic grade (G)

GX


Grade cannot be assessed


G1


Well differentiated


G2


Moderately differentiated


G3


Poorly differentiated


 
















Residual tumor (R)

RX


Presence of residual tumor cannot be assessed


R0


No residual tumor


R1


Microscopic residual tumor


R2


Macroscopic residual tumor


From Greene, FL, Page, DL, Fleming ID et al. Used with the permission of the American Joint Committee on Cancer (AJCC), Chicago, Illinois. The original source for this material is the AJCC Cancer Staging Manual. 6th ed (2002) published by Springer-New York, www.springeronline.com.


Comments

The AJCC TNM staging system uses three basic descriptors that are then grouped into stage categories. The first component is “T,” which describes the extent of the primary tumor. The next component is “N,” which describes the absence or presence and extent of regional lymph node metastasis. The third component is “M,” which describes the absence or presence of distant metastasis. The final stage groupings (determined by the different permutations of “T,” “N,” and “M”) range from Stage 0 through Stage IV.


Reference

Greene, FL, Page, DL, Fleming ID et al. AJCC Cancer Staging Manual. 6th ed. New York: Springer, 2002. http://www.cancerstaging.org/.



Esophagus


Symptoms


Gastroesophageal Reflux Disease (GERD): The Carlsson–Dent Reflux Questionnaire


Aims

To design a reflux disease questionnaire for the diagnosis of gastroesophageal reflux disease that recognizes classic symptom patterns for reflux disease.


image


Comments

The Carlsson–Dent Questionnaire is a self-administered questionnaire with seven items (heartburn, pain in the stomach, regurgitation, discomfort in the stomach, food sticking in the gullet after swallowing, feeling sick, and vomiting) that focus on the nature of the symptom sensations and provoking, exacerbating, or relieving factors.


References

Carlsson R, Dent J, Bolling-Sternevald E et al. The usefulness of a structured questionnaire in the assessment of symptomatic gastroesophageal reflux disease. Scand J Gastroenterol. 1998; 33:1023–1029.


GERD: Symptomatic Score According to DeMeester


Aims

To be able to score GERD-related symptoms in a quantitative way.


image


Comments

Maximum score is 6 points. With such a score the reflux interferes constantly with daily activities, and there is occasional pulmonary aspiration.


References

DeMeester TR, Johnson LF. The evaluation of objective measurements of gastroesophageal reflux and their contribution to patient management. Surg Clin North Am. 1976;56: 39–53.


GERD: Mayo Clinic Gastroesophageal Reflux Questionnaire (GERQ) According to Locke


Aims

To develop a self-report questionnaire to measure gastroesophageal reflux disease in the general population. To measure symptoms experienced over the prior year and to collect the medical history.


image


Comments

This is a valid questionnaire that is applicable in population-based studies to assess gastroesophageal reflux disease.


References

Locke GR, Talley NJ, Weaver AL, Zinsmeister AR. A new questionnaire for gastroesophageal reflux disease. Mayo Clin Proc. 1994;69:539–547.


GERD: Clinical Score for Predicting Findings at Endoscopy According to Locke


Aims

The aim was to design a scoring system to predict findings on the basis of clinical information.


image


Comments

Points are assigned for specific characteristics, and then summed to give the total score. To obtain a predicted probability, a constant is subtracted from the total score and this difference multiplied by another constant (see formula).


References

Locke GR, Zinsmeister AR, Talley NJ. Can symptoms predict endoscopic findings in GERD? Gastrointest Endosc. 2003;58: 661–670.


GERD: Reflux Disease Diagnostic Questionnaire (RDQ)


Aims

To develop a brief, reliable, and valid self-administered questionnaire that can facilitate the diagnosis of gastroesophageal reflux disease in primary care.


 













Regurgitation


Acid taste severity


Movement of materials severity


Acid taste frequency


Movement of materials frequency


Heartburn


Frequency of burning behind breastbone


Severity of burning behind breastbone


Frequency of pain behind breastbone


Severity of pain behind breastbone


Dyspeptic symptoms


Upper stomach pain severity


Upper stomach pain frequency


Upper stomach burning frequency


Upper stomach burning severity


Response options are scaled as closed-ended and Likert-type, with categories ranging from 1 to 5 or with 1–7 points for frequency, severity, and duration of symptoms.


From Shaw MJ, Talley NJ, Beebe TJ et al. Initial validation of a diagnostic questionnaire for gastroesophageal reflux disease. Am J Gastroenterol. 2001;96:52–57. With permission of Blackwell Publishing.


Comments

Content and wording of the RDQ are developed with multiple iterations of expert opinion and patient interviewing. The RDQ is a reliable and valid 12-item questionnaire of three scales (regurgitation, heartburn, dyspeptic symptoms).


References

Shaw MJ, Talley NJ, Beebe TJ et al. Initial validation of a diagnostic questionnaire for gastroesophageal reflux disease. Am J Gastroenterol. 2001;96:52–57.


Shaw MJ, Fendrick AM, Kane RL, Adlis SA, Talley NJ. Self-reported effectiveness and physician consultation rate in users of over-the-counter histamine-2 receptor antagonists. Am J Gastroenterol. 2001;96:673–676.


GERD: GERD Symptom Assessment Scale (GSAS)


The GSAS is a very comprehensive evaluative symptom scale. It is a well-validated 15-item scale that includes associated symptoms as well as predominant symptoms. It also assesses different symptom dimensions (frequency of episodes, intensity of symptoms, level of distress). Subjects are first asked if they experienced the symptom (yes/no) during the past week. Asking the subject to indicate, “How many times in the past week did you have this symptom,” assesses frequency. The severity of each symptom is assessed on a 4-point scale ranging from 1 (slight) to 4 (very severe). Distress is also assessed on a 4-point scale ranging from 0 (not at all) to 3 (very much). The GSAS is self-assessed twice, once before and once after treatment. The scale does not assess nocturnal symptoms.


References

Rothman M, Farup C, Stewart W, Helbers L, Zeldis J. Symptoms associated with gastroesophageal reflux disease: development of a questionnaire for use in clinical trials. Dig Dis Sci. 2001;46:1540–1549.


Damiano A, Handley K, Adler E, Siddique R, Bhattacharyja A. Measuring symptom distress and health-related quality of life in clinical trials of gastroesophageal reflux disease treatment: further validation of the Gastroesophageal Reflux Disease Symptom Assessment Scale (GSAS). Dig Dis Sci. 2002;47:1530–1537.


GERD: A Reflux Questionnaire According to Allen


This symptom questionnaire is specifically designed for GERD. The GERD score consists of six questions on specific symptoms, but no questions on atypical symptoms. Both intensity and frequency of symptoms is monitored. The scale is applied before and 6 months after therapy.


References

Allen CJ, Parameswaran K, Belda J, Anvari M. Reproducibility, validity, and responsiveness of a disease-specific symptom questionnaire for gastroesophageal reflux disease. Dis Esophagus. 2000;13:265–270.


GERD: Ulcer Esophagitis Subjective Symptoms Scale (UESS)


This questionnaire comprises 10 items covering four dimensions (abdominal pain, reflux discomfort (acid regurgitation, heartburn), intestinal discomfort, and sleep dysfunction. The self-assessment scale is to be applied at study start and after 4 weeks of therapy.


References

Dimenas E, Glise H, Hallerback B, Hernqvist H, Svedlund J, Wiklund I. Quality of life in patients with upper gastrointestinal symptoms. An improved evaluation of treatment regimens? Scand J Gastroenterol. 1993;28:681–687.


GERD: Gastroesophageal Reflux Disease Activity Index (GRACI) According to Williford


Aims

To develop a reflux disease activity index that can be used frequently and is inexpensive as compared to frequent endoscopy and pH monitoring.


image


Comments

The index is to be administered at baseline and every 3 months thereafter.


References

Spechler SJ, Williford WO, Krol WF et al. Development and validation of a gastroesophageal reflux disease activity index (GRACI). Gastroenterology 1990;98:130.


Williford WO, Krol WF, Spechler SJ, and the VA Cooperative Study Group on Gastroesophageal Reflux Disease (GERD). Development for and results of the use of a gastroesophageal reflux disease activity index as an outcome variable in a clinical trial. Control Clinical Trials. 1994;15:335–348.


GERD ReQuest


The ReQuest (Reflux Questionnaire) comprises 7 dimensions (general well-being, acid complaints, upper abdominal/stomach complaints, lower abdominal/digestive complaints, nausea, sleep disturbances, other complaints) tested by a leading question for frequency (7-point Likert scale) and intensity (100 mm visual analogue scale), making up the short version of ReQuest. The long version also includes 67 symptom descriptions that are assigned to the 7 dimensions. The dimensions are grouped into two subscales: (1) ReQuest-GI, including dimensions with GI complaints (acid complaints, upper abdominal/stomach complaints, and nausea); and (2) ReQuest-WSO, including dimensions affecting the aspects of well-being (general well-being, sleep disturbances, and other complaints).


In a further development, the ReQuest symptom score is combined with the LA grading system of esophageal injury.


image


References

Bardhan KD, Stanghellini V, Armstrong D, Berghofer P, Gatz G, Monnikes H. Evaluation of GERD symptoms during therapy. Part I. Development of the new GERD questionnaire ReQuest. Digestion. 2004;69:229–237.


Monnikes H, Bardhan KD, Stanghellini V, Berghofer P, Bethke TD, Armstrong D. Evaluation of GERD symptoms during therapy. Part II. Psychometric evaluation and validation of the new questionnaire ReQuest in erosive GERD. Digestion. 2004;69:238–244.


Bardhan KD, Armstrong D, Fass R, Berghöfer P, Gatz G, Mönnikes H. The ReQuest/LA classification: a novel, integrated approach for the comprehensive assessment of treatment outcome of gastroesophageal reflux disease (GERD). Abstract at 2005 World Congress of Gastroenterology.


GERD: Overview of Instruments Used for Symptom and Quality of Life Assessment in Gastroesophageal Reflux Disease


Numerous symptom scales have been used in the literature to measure GERD symptom severity, but most do not fulfill the essential requirements for validity. The reader is referred to the overview presented by Stanghellini et al. and Rentz et al. (see table opposite).


References

Rentz AM, Battista C, Trudeau E. Symptom and health-related quality-of-life measures for use in selected gastrointestinal disease studies: a review and synthesis of the literature. Pharmacoeconomics. 2001;19:349–363.


Stanghellini V, Armstrong D, Monnikes H, Bardhan KD. Systematic review: do we need a new gastroesophageal reflux disease questionnaire? Aliment Pharmacol Ther. 2004; 19:463–479.


image


image


Table References

(1) Greatorex R, Thorpe JA. Clinical assessment of gastroesophageal reflux by questionnaire. Br J Clin Pract. 1983;37(4):133–5.


(2) Reidel WL, Clouse RE. Variations in clinical presentation of patients with esophageal contraction abnormalities. Dig Dis Sci. 1985;30(11):1065–71.


(3) Andersen LI, Madsen PV, Dalgaard P, Jensen G. Validity of clinical symptoms in benign esophageal disease, assessed by questionnaire. Acta Med Scand. 1987;221(2):171–7.


(4) Dimenäs E, Glise H, Hallerback B, Hernqvist H, Svedlund J, Wiklund I. Quality of life in patients with upper gastrointestinal symptoms. An improved evaluation of treatment regimens? Scand J Gastroenterol. 1993;28:681–7.


(5) Svedlund J, Sjödin I, Dotevall G. GSRS—a clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome and peptic ulcer disease. Dig Dis Sci. 1988; 33:129–34.


(6) Talley NJ, Fullerton S, Junghard O, Wiklund I. Quality of life in patients with endoscopy-negative heartburn: reliability and sensitivity of disease-specific instruments. Am J Gastroenterol. 2001;96(7):1998–2004.


(7) Revicki DA, Wood M, Wiklund I, Crawley J. Reliability and validity of the Gastrointestinal Symptom Rating Scale in patients with gastroesophageal reflux disease. Qual Life Res. 1998;7(1):75–83.


(8) Mold JW, Reed LE, Davis AB, Allen ML, Decktor DL, Robinson M. Prevalence of gastroesophageal reflux in elderly patients in a primary care setting. Am J Gastroenterol. 1991;8:965–70.


(9) Ruth M, Mansson I, Sandberg N. The prevalence of symptoms suggestive of esophageal disorders. Scand J Gastroenterol. 1991;26(1):73–81.


(10) Räihä IJ, Impivaara O, Seppala M, Sourander LB. Prevalence and characteristics of symptomatic gastroesophageal reflux disease in the elderly. J Am Geriatr Soc. 1992; 40(12):1209–11.


(11) Johnsson F, Roth Y, Damgaard Pedersen NE, Joelsson B. Cimetidine improves GERD symptoms in patients selected by a validated GERD questionnaire. Aliment Pharmacol Ther. 1993;7(1):81–6.


(12) Orenstein SR, Cohn JF, Shalaby TM, Kartan R. Reliability and validity of an infant gastroesophageal reflux questionnaire. Clin Pediatr (Phila). 1993;32(8):472–84.


(13) Orenstein SR, Shalaby TM, Cohn JF. Reflux symptoms in 100 normal infants: diagnostic validity of the infant gastroesophageal reflux questionnaire. Clin Pediatr (Phila). 1996;35(12):607–14.


(14) Locke GR, Talley NJ, Weaver AL, Zinsmeister AR. A new questionnaire for gastroesophageal reflux disease. Mayo Clin Proc. 1994;69(6):539–47.


(15) Williford WO, Krol WF, Spechler SJ. Development for and results of the use of a gastroesophageal reflux disease activity index as an outcome variable in a clinical trial. VA Cooperative Study Group on Gastroesophageal Reflux Disease (GERD). Control Clin Trials. 1994;15(5):335–48.


(16) Carlsson R, Dent J, Bolling-Sternevald E et al. The usefulness of a structured questionnaire in the assessment of symptomatic gastroesophageal reflux disease. Scand J Gastroenterol. 1998;33(10):1023–9.


(17) Shaw M, Talley NJ, Adlis S, Beebe T, Tomshine P, Healey M. Development of a digestive health status instrument: tests of scaling assumptions, structure and reliability in a primary care population. Aliment Pharmacol Ther. 1998;12:1067–78.


(18) Shaw MJ, Beebe TJ, Adlis A, Talley NJ. Reliability and validity of the digestive health status instrument in samples of community, primary care, and gastroenterology patients. Aliment Pharmacol Ther. 2001;15:981–7.


(19) Ho KY, Kang JY, Seow A. Prevalence of gastrointestinal symptoms in a multiracial Asian population, with particular reference to reflux-type symptoms. Am J Gastroenterol. 1998;93(10):1816–22.


(20) Talley NJ, Phillips SF, Melton J 3rd, Wiltgen C, Zinsmeister AR. A patient questionnaire to identify bowel disease. Ann Intern Med. 1989;11:671–4.


(21) Manterola C, Munoz S, Grande L, Riedemann P. Construction and validation of a gastroesophageal reflux symptom scale. Preliminary report. Rev Med Chil. 1999; 127(10):1213–22.


(22) Manterola C, Munos S, Grande L, Bustbos L. Initial validation of a questionnaire for detecting gastroesophageal reflux disease in epidemiological settings. J Clin Epidemiol. 2002;55:1041–5.


(23) Allen CJ, Parameswaran K, Belda J, Anvari M. Reproducibility, validity, and responsiveness of a disease-specific symptom questionnaire for gastroesophageal reflux disease. Dis Esophagus. 2000;13(4):265–70.


(24) Rothman M, Farup C, Stewart W, Helbers L, Zeldis J. Symptoms associated with gastroesophageal reflux disease: development of a questionnaire for use in clinical trials. Dig Dis Sci. 2001;46(7):1540–9.


(25) Shaw MJ, Talley NJ, Beebe TJ, et al. Initial validation of a diagnostic questionnaire for gastroesophageal reflux disease. Am J Gastroenterol. 2001;96(1):52–7.


(26) Ofman JJ, Shaw M, Sadik K, et al. Identifying patients with gastroesophageal reflux disease. Dig Dis Sci. 2002;47: 1863–9.


Dysphagia: Grading of Dysphagia


Aims

To score dysphagia of patients with malignant esophagogastric obstruction treated with the use of self-expanding metal stents.


 























Grading of dysphagia

Grade


Symptoms


0


Able to eat a normal diet


1


Able to eat some solid food


2


Able to eat some semi-solids only


3


Able to swallow liquids only


4


Complete dysphagia


From Ogilvie AL, Dronfield MW, Ferguson R, Atkinson M. Palliative intubation of oesophagogastric neoplasms at fibreoptic endoscopy. Gut. 1982;23:1060–1067. With permission of BMJ Publishing Group.


Comments

The dysphagia score improves after stent placement from a mean of 3.6 to 1.6 after two weeks.


References

Ogilvie AL, Dronfield MW, Ferguson R, Atkinson M. Palliative intubation of oesophagogastric neoplasms at fibreoptic endoscopy. Gut. 1982;23:1060–1067.


Siersema PD, Schrauwen SL, Blankenstein M van et al. Self-expanding metal stents for complicated and recurrent esophagogastric cancer. Gastrointest Endosc. 2001;54: 579–586.


Dysphagia: Grading Dysphagia According to Sugahara


Aims

To grade dysphagia in esophageal malignancies. The Sugahara dysphagia grading is divided into 6 categories, grade 1 is the least, and grade 6 the most severe:


Grade 1 is defined as eating normally.


Grade 2 patients require liquids with meals.


Grade 3 dysphagia is when a patient is able to take semi-solids but unable to take any solid food.


Grade 4 patients can take liquids only.


Grade 5 patients are unable to take liquids, but still able to swallow saliva.


Grade 6 patients are unable to swallow saliva.


Comments

Scoring system used in palliative treatment of patients with advanced esophageal cancer.


References

Sugahara S, Ohara K, Yoshioka H. Improvement of swallowing function in patients with esophageal cancer treated by radiology. J Jpn Soc Cancer Ther. 1996;31:1124–1130.


Dysphagia: Swallowing Score According to O’Rourke


Aims

To analyze the outcome of patients with strictures of the esophagus after radiotherapy for carcinoma.


 






















Swallowing score

Score


1


Normal, eats anything


2


Eats soft food only


3


Eats pureed foods only


4


Drinks liquid only


5


No swallowing at all


 



















Success score for palliation of dysphagia

Score


I


Mean swallowing score 1 and 2; dilatation required at intervals longer than 6 months


II


Mean swallowing score 1 and 2; dilatation required at intervals between 3–6 months


III


Mean swallowing score 3 or requires dilatation every 1–3 months


IV


Mean swallowing score 4 or less, or requires dilatation monthly


Reproduced from O’Rourke IC, Tiver K, Bull C, Gebski V, Langlands AO. Swallowing performance after radiation therapy for carcinoma of the esophagus. Cancer. 1988;61:2022–2026. With permission granted by John Wiley & Sons, Ltd. on behalf of the BJSS, Ltd.


Comments

Patients are treated with dilatation. Score III is the most common success score.


References

O’Rourke IC, Tiver K, Bull C, Gebski V, Langlands AO. Swallowing performance after radiation therapy for carcinoma of the esophagus. Cancer. 1988;61:2022–2026.


Achalasia: Grading Symptomatic Outcome After Pneumodilatation According to Vantrappen


Aims

To score success rates in long-term follow-up after pneumodilatation for achalasia.


 























Success of dilatation according to Vantrappen

Class


Description


1. Excellent


Completely free of symptoms


2. Good


Occasional (< once a week) dysphagia or pain of short duration; defined as retrosternal hesitancy of food lasting from a few seconds (2–3 sec) to a couple of minutes (2–3 min) and disappearing after drinking fluids


3. Moderate


Dysphagia > once/week, lasting < a few minutes and not accompanied by regurgitations or weight loss


4. Poor


Dysphagia > once/week or lasting a few minutes or accompanied by regurgitations or weight loss


From Vantrappen G, Hellemans J. Treatment of achalasia and related motor disorders. Gastroenterology. 1980;79:144–154. With permission from the American Gastroenterological Association.


Comments

Excellent and good results are considered treatment success. Patients can have subjective improvement of symptoms but still score a “poor” result because of continuing dysphagia, weight loss, or regurgitation.


References

Vantrappen G, Hellemans J. Treatment of achalasia and related motor disorders. Gastroenterology. 1980;79:144–154.


Achalasia: Symptom score according to Vaezi


Aims

To evaluate the severity of achalasia symptoms in a trial that compared botulinum toxin to pneumatic dilatation.


 




































































Symptom score according to Vaezi

Symptom


 


Score


Dysphagia


1 = once per month or less


 


 


2 = once per week, up to 3 to 4 times a month


 


 


3 = two to four times per week


 


 


4 = once per day


 


 


5 = several times per day


 


Regurgitation


1 = once per month or less


 


 


2 = once per week, up to 3 to 4 times a month


 


 


3 = two to four times per week


 


 


4 = once per day 5 = several times per day


 


Chest pain


1 = once per month or less


 


 


2 = once per week, up to 3 to 4 times a month


 


 


3 = two to four times per week


 


 


4 = once per day


 


 


5 = several times per day


 


 


                                              Total symptom score


From Vaezi MF, Richter JE, Wilcox CM et al. Botulinum toxin versus pneumatic dilatation in the treatment of achalasia: a randomized trial. Gut. 1999;44:231–239. With permission of BMJ Publishing Group.


Comments

The maximum score is 15. Clinical response is defined as a greater than 50 % improvement in the total symptom score.


References

Vaezi MF, Richter JE, Wilcox CM et al. Botulinum toxin versus pneumatic dilatation in the treatment of achalasia: a randomized trial. Gut. 1999;44:231–239.


Achalasia: Clinical Classification of Achalasia Severity—The Eckardt Score


Aims

A careful interview and the use of modern methods that concentrate on pathophysiologic aspects always allow an early diagnosis and the initiation of therapy of achalasia.


image


Comments

The final score is the sum of the individual ratings for each symptom/sign.


References

Eckardt VF, Aignherr C, Bernhard G. Predictors of outcome in patients with achalasia treated by pneumatic dilation. Gastroenterology. 1992;103:1732–38.


Eckardt VF. Clinical presentation and complications of achalasia. Gastrointest Endosc Clin N Am. 2001;11:281–292.


Anatomical Variants


Esophageal Atresia and Tracheo-Esophageal Fistula: Classification According to Waterston


Aims

To describe the anatomic characteristics of each anomaly.


 





























Type


 


A


Common type: the most common anomaly, the esophagus ends in a blind pouch and the lower esophagus joins the respiratory tract near the tracheal bifurcation.


B


Common type with an atretic fistula: as type A, with in addition no lumen of the lower esophageal portion near the trachea.


C


Atresia without tracheo-esophageal fistula: second most common anomaly. There is a (wide) gap between the esophageal ends.


D


Atresia with tracheo-esophageal fistula to the upper esophagus.


E


Atresia with fistula to upper and lower pouches.


F


Tracheo-esophageal fistula without atresia.


G


As type F, with a diaphragm partly obstructing the esophageal lumen (or atretic esophagus with esophagus with fused fistulas from each end).


 
















Risk groups

Group A


Birth weight > 2500 g and well


Group B


1. Birth weight 1800 to 2500 g and well; or 2. Birth weight > 2500 g but moderate pneumonia and other congenital anomaly


Group C


1. Birth weight < 1800 g; or 2. Birth weight > 1800 g with severe anomaly or pneumonia


From Waterston DJ, Bonham Carter RE, Aberdeen E. Oesophageal atresia: Tracheo-oesophageal fistula. A study of survival in 218 infants. The Lancet. 1962;1:819–822. With permission of Elsevier.


Comments

Type A and C are the most common types. Waterston established prognostic criteria in 1962 to distinguish between cases that could be treated during a single operative session. The criteria are no longer valid.


References

Waterston DJ, Bonham Carter RE, Aberdeen E. Oesophageal atresia: Tracheo-oesophageal fistula. A study of survival in 218 infants. Lancet. 1962;1:819–822.


image


Fig. 2.2 Esophageal atresia and tracheoesophageal fistula: classification according to Waterston.


Esophageal Malformation: Classification According to Gross


Aims

To stage common esophagotracheal malformations.


 


























Classification according to Gross

Type


 


A


Esophageal atresia without tracheoesophageal fistula


B


Atresia with proximal tracheoesophageal fistula


C


Atresia with distal tracheoesophageal fistula


D


Atresia with both proximal and distal fistula


E


Tracheoesophageal fistula without esophageal atresia


F


Esophageal stenosis


image


Fig. 2.3 Esophageal malformation: classification according to Gross.


Comments

Commonly used classification system of esophageal atresia and tracheo-esophageal fistula. Type C is most common.


References

Gross RE. The Surgery of Infancy and Childhood. Philadelphia: WB Saunders; 1953.


Small-Caliber Esophagus: Classification According to Vasilopoulos


Aims

The aim was to classify small caliber esophagus into three groups.


 
















Vasilopoulos classification of small-caliber esophagus

Type 1


Early small-caliber esophagus (eosinophilic esophagitis with dysphagia for solids but normal barium esophagogram and esophagoscopy)


Type 2


Advanced small-caliber esophagus (eosinophilic esophagitis with dysphagia for solids and a small-caliber esophagus detectable on barium esophagogram and/or esophagoscopy)


Type 3


Ringed esophagus (eosinophilic esophagitis with dysphagia for solids and a small caliber esophagus with rings or corrugated esophagus detectable on barium esophagogram and/or esophagoscopy)


Vasilopoulos S, Shaker R. Defiant dysphagia: small-caliber esophagus and refractory benign esophageal strictures. Current Gastroenterology Reports. 2001;3:225–230. With permission of Current Science, Inc.


Comments

The small caliber esophagus has a smooth, diffusely narrow esophageal lumen that can be appreciated by barium esophagography or esophagoscopy. There is no stricture or cicatrization. This is a rare esophageal disease entity of which the long-term outcome is poorly known. Type 3 especially is clinically relevant.


References

Vasilopoulos S, Shaker R. Defiant dysphagia: small-caliber esophagus and refractory benign esophageal strictures. Current Gastroenterology Reports. 2001;3:225–230.


image


Fig. 2.4 Ringed esophagus, with multiple concentric rings and normal overlying mucosa (Type 3).


Vascular Disorders


Esophageal Varices: Grading of Esophageal Varices According to Dagradi


Aims

The aim is to be able to grade esophageal varices by endoscopic evaluation.


 























Grading of esophageal varices according to Dagradi

Grading


 


1.


Diameter of 2 mm or less, which are brought out only by compressing the esophageal wall with the tip of the instrument


2.


Similar diameter, no compression needed


3.


3–4 mm


4.


5 mm or more


5.


Small on top of the larger submucosal varices (bright red and telangiectatic cherry-red spots)


From Dagradi AE, Rodiles DH, Cooper E, Stempien SJ. Endoscopic diagnosis of esophageal varices. Am J Gastroenterol. 1971;56:371–377. With permission of Blackwell Publishing.


Comments

Grades 4 and 5 are potential bleeders.


References

Dagradi AE, Rodiles DH, Cooper E, Stempien SJ. Endoscopic diagnosis of esophageal varices. Am J Gastroenterol. 1971; 56:371–377.


Dagradi AE, Ruiz RA, Weingarten ZG. Influence of emergency endoscopy on the management and outcome of patients with upper gastrointestinal hemorrhage. Analysis of 500 cases. Am J Gastroenterol. 1979;72:403–415.


Esophageal Varices: Grading of Esophageal Varices According to Tytgat


Aims

To grade esophageal varices by endoscopic evaluation.


 























Grading of esophageal varices according to Tytgat

Grade


 


I


Smaller than 2 mm, visible only on deep inspiration


II


Diameter 2–3 mm (clearly visible varices)


III


Diameter 3–4 mm (prominent, locally coil-shaped varices, partially occupying the lumen)


IV


Diameter 4–5 mm (tortuous, sometimes grape-like varices)


V


Diameter larger than 5 mm, often with superimposed cherry red spots or red wale markings


From Tytgat GN, Mulder CJ, eds. Procedures in Hepatogastroenterology. Dordrecht: Kluwer Academic Publishers; 1997. With kind permission of Springer Science and Business Media.


Comments

Grades IV and V are particularly prone to bleeding.


References

Tytgat GN, Mulder CJ, eds. Procedures in Hepatogastroenterology. Dordrecht: Kluwer Academic Publishers; 1997.


image


Fig. 2.5a–e Esophageal varices: grading according to Tytgat. a Grade I; b Grade II; c Grade III.


image


Fig. 2.5d, e d Grade IV; e Grade V.


Esophageal Varices: Grading According to Conn


Aims

To grade esophageal varices by their size in endoscopic evaluation.


 

















Grade


 


I


Flat varices, disappearing when insufflating air within the esophagus


II


Varices permanently visible in the lumen of the esophagus but appearing in well-separated bands


III


Varices that are extremely prominent in the lumen, so that they appear to almost occlude the lumen of the esophagus when not inflated


To estimate the varices diameter with the width of an open biopsy forceps.


 























Grade


 


0


Nil


I


One-fourth bite width


II


One-half bite width


III


Three-fourth bite width


IV


One or more bite widths


Comments

The height of the esophageal varices is compared with the width of biopsy forceps with a bite width of 5 mm.


References

Conn HO, Binder H, Brodoff M. Fiberoptic and conventional esophagoscopy in the diagnosis of esophageal varices. A comparison of techniques and observers. Gastroenterology. 1967;52:810–818.


Conn HO, Brodoff M. Emergency esophagoscopy in the diagnosis of upper gastrointestinal hemorrhage; a critical evaluation of its diagnostic accuracy. Gastroenterology. 1964; 47:505–512.


Esophageal Varices: Classification According to Beppu


Aims

Proposing standards for recording endoscopic findings of esophageal varices.


image


 


























Total score


Rate of bleeding (%)


>+ 1.14


0


+ 0.38~ + 1.14


20.6


0~ + 0.38


40.0


–0.38~ 0


64.5


–1.14~ –0.38


90.2


<–1.14


100


From Beppu K, Inokuchi K, Koyanagi N et al. Prediction of variceal hemorrhage by esophageal endoscopy. Gastrointest Endoscopy. 1981;27:213–218. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

The rules for recording endoscopic findings in esophageal varices were proposed by the Japanese Research Society for Portal Hypertension. The total score is calculated as follows:


S = XF + XC + XRWM + XCRS +XHCS + XE


According to Beppu, the following signs are particularly valuable for predicting rebleeding: blue varices, red wale marking, cherry-red spots, and large-sized varices.


References

Beppu K, Inokuchi K, Koyanagi N et al. Prediction of variceal hemorrhage by esophageal endoscopy. Gastrointest Endoscopy. 1981;27:213–218.


Japanese Research Society for Portal Hypertension. The general rules for recording endoscopic findings on esophageal varices. Jpn J Surg. 1980;10:84–87.


Japanese Research Committee on Portal Hypertension. The general rules for recording endoscopic findings of esophageal varices, revised edition. Acta Hepatol Jpn. 1991;33: 277–281.


Esophageal Varices: Revised General Rules for Recording Endoscopic Findings of Esophago-gastric Varices According to the Japanese Society for Portal Hypertension


Aims

General rules for recording endoscopic findings of esophageal varices were made in 1980. In 1991 the rules were revised as new modalities of endoscopic treatment were developed.


 





































































































































Category


Code


Subcategory


Location


L


Longitudinal extent


 


 


Locus superior (ls)


 


 


Locus medialis (Lm)


 


 


Locus inferior (Li)


 


 


Locus gastrica (Lg)


 


 


Gastric varices adjacent to the cardiac orifice (Lg-c)


Form


F


Shape and size


 


 


Lesions assuming varicose appearance (F0)


 


 


Straight small-caliber varices (F1)


 


 


Moderately enlarged, beady varices (F2)


 


 


Markedly enlarged, nodular, or tumor-shaped varices (F3)


Color


C


Fundamental color


 


 


White varices (Cw)


 


 


Blue varices (Cb)


Red color sign


RC


Red wale marking (RWM)


 


 


Cherry red spot (CRS)


 


 


Hematocystic spot (HCS)


 


 


RC (–)


 


 


RC (+)


 


 


RC (++)


 


 


RC (+++)


 


 


Telangiectasia [TE(+)]


Bleeding sign


B


During bleeding


 


 


Spurting


 


 


Oozing


 


 


After hemostasis


 


 


Red plug


 


 


White plug


Mucosal findings


E


Erosion (E)


 


Ul


Ulcer (U)


 


S


Scar (S)


From Idezuki Y. General rules for recording endoscopic findings of esophagogastric varices (1991). Japanese Society for Portal Hypertension. World J Surg. 1995;19:420–422. With kind permission of Springer Science and Business Media.


Comments

This scoring system is used for endoscopic evaluation of the therapeutic effect of sclerotherapy for esophageal varices.


References

Idezuki Y. General rules for recording endoscopic findings of esophagogastric varices (1991). Japanese Society for Portal Hypertension. World J Surg. 1995;19:420–422.


Esophageal Varices: Northern Italian Endoscopy Club (NIEC) Index


Aims

To identify prospectively patients with the highest risk of bleeding.


image


image


Fig. 2.6 Endoscopic views showing grading of varices by size (F1, F2, F3).


image


Fig. 2.7a–c Endoscopic views showing various red color signs. a Cherry-red spots (arrows). b Wale markings (arrow). c Hematocystic spot (arrow).


 

































Risk class


NIEC Index


Rate of bleeding (%) at 1 year


1


< 20.0


1.6


2


20.0–25.0


11.0


3


25.1–30.0


14.8


4


30.1–35.0


23.3


5


35.1–40.0


37.8


6


> 40.0


68.9


From Northern Italian Endoscopy Club for the study and treatment of esophageal varices. Prediction of the first variceal hemorrhage in patients with cirrhosis of the liver and esophageal varices: a prospective multicenter study. N Engl J Med. 1988;319:983–989. With permission of the New England Journal of Medicine.


Comments

This score uses only three variables, one of which is the Child– Pugh classification, the other two are endoscopically scored features of the varices. The grading of the size of the varices is based on the scoring of the Japanese Research Society for Portal Hypertension.


References

Northern Italian Endoscopy Club for the study and treatment of esophageal varices. Prediction of the first variceal hemorrhage in patients with cirrhosis of the liver and esophageal varices: a prospective multicenter study. N Engl J Med. 1988; 319:983–989.


Esophageal Varices: Revision the NIEC Index According to Merkel


Aims

Merkel et al. revised the NIEC index after performing a prospective study to predict the first esophageal variceal hemorrhage. The size and presence of red wale markings are more important than the Child–Pugh class for the prediction of esophageal variceal hemorrhage.


 





















 


Revised coefficient


Regression NIEC


Size of varices


1.12


0.4365


Red wale markings


0.36


0.3193


Child–Pugh class


0.04


0.645


From Merkel C, Zoli M, Siringo S et al. Prognostic indicators of risk for first variceal bleeding in cirrhosis: a multicenter study in 711 patients to validate and improve the North Italian Endoscopic Club (NIEC) index. Am J Gastroenterol. 2000;95:2915–2920. With permission of Blackwell Publishing.


Comments

This index resembles the Paquet and Sarin grading systems because of the weighting of the revised NIEC index, with a low coefficient for the Child–Pugh class.


References

Merkel C, Zoli M, Siringo S et al. Prognostic indicators of risk for first variceal bleeding in cirrhosis: a multicenter study in 711 patients to validate and improve the North Italian Endoscopic Club (NIEC) index. Am J Gastroenterol. 2000;95: 2915–2920.


Esophageal Varices: Criteria for High-Risk Patients with Esophageal Varices According to Paquet


Aims

To select high-risk patients for prophylactic sclerotherapy.


 










Criteria according to Paquet

Existence of varices III–IV bearing erosions


Varices II–IV without erosions but coagulation factors below 30%


Or both


image


Fig. 2.8 Endoscopic degree of esophageal varices according to Pacquet. The black points (degree IV) signal possible danger of bleeding.


Paquet KJ. Prophylactic endoscopic sclerosing treatment of the esophageal wall in varices—a prospective controlled randomized trial. Endoscopy. 1982;14:4–5.


Comments

The criteria are based on the size of the varices, erosions, and the presence of coagulopathy.


References

Paquet KJ. Prophylactic endoscopic sclerosing treatment of the esophageal wall in varices—a prospective controlled randomized trial. Endoscopy. 1982;14:4–5.


Esophageal Varices: Grading of Variceal Size According to Westaby


Aims

To grade varices for sclerotherapy according to the degree of protrusion into the lumen.


 













Grade 1


Varix is flush with the wall of the esophagus


Grade 2


Protrusion of varix no further than half-way down the center of the lumen


Grade 3


Protrusion more than half-way down the center of the lumen


image


Fig. 2.9 Esophageal Varices: Grading of variceal size according to Westaby.


From Westaby D, MacDougall BRD, Melia W, Theodossi A, Williams R. A prospective randomized study of two sclerotherapy techniques for esophageal varices. Hepatology. 1983;3:681–684. With permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.


References

Westaby D, MacDougall BRD, Saunders JB et al. A study of risk factors in patients with cirrhosis and variceal bleeding. In: Westaby D, MacDougall BRD, Williams R, eds. Variceal Bleeding. London: Pittman Medical; 1982.


Westaby D, MacDougall BRD, Melia W, Theodossi A, Williams R. A prospective randomized study of two sclerotherapy techniques for esophageal varices. Hepatology. 1983;3: 681–684.


Esophageal Varices: Scoring System for Endoscopic Signs of Esophageal Varices According to Snady and Feinman


Aims

To identify cirrhotic patients that have never had variceal hemorrhage and who are most prone to bleeding.


 









































































Scoring system for endoscopic signs of esophageal varices

Endoscopic sign


 


Assigned rating


Form or size


 


 


F1


Small and straight varices, not disappearing with insufflation


1


F2


Large tortuous varices, occupying less than one-third of lumen


2


F3


Largest size varices, coil shaped, occupying more than one-third of the lumen


3


Location


 


 


Li


Within the lower one-third


1


Lm


Within the lower two-thirds, below tracheal bifurcation


2


Ls


Extending above the tracheal bifurcation to the cricopharyngeus


3


Color


 


 


Cw


White varices appearing like large folds of esophageal mucosa


1


Cbw


Varices that cannot be clearly assigned to Cb or Cw


2


Cb


Dark blue varices, appearing cyanotic


3


Red color sign


 


 


+


Present but no more than 10 lesions found


1


++


Not extensive, but more than 10 lesions are easily visible


2


+++


Extensive, covering all varices in the entire esophagus


3


From Snady H, Feinman L. Prediction of variceal hemorrhage: a prospective study. Am J Gastroenterol. 1988;83:519–525. With permission of Blackwell Publishing.


Comments

A score ≥ 8 is classified as high grade. A score ≤ 7 is classified as low grade. In a prospective trial with a mean duration of 26 months, the association with bleeding for high grade was 73 % versus 7 % for low grade.


References

Snady H, Feinman L. Prediction of variceal hemorrhage: a prospective study. Am J Gastroenterol. 1988;83:519–525.


Esophageal Varices: Three-Dimensional Endoscopic Ultrasonography Classification of Variceal Blood Flow Pattern According to Nakamura


Aims

To determine the indication of endoscopic variceal ligation based on the vascular pattern classified by three-dimensional endoscopic ultrasonography.


 




















Classification of variceal blood flow pattern

Type


Description


1


Cardial inflow without para-esophageal veins


2


Cardial inflow with para-esophageal veins


3


Azygous-perforating pattern


4


Complicated pattern


image


Fig. 2.10a–d Esophageal varices: classification of vascular patterns based on 3-D endoscopic ultrasonographic findings. a Type 1: cardial-inflow type without paraesophageal veins; b Type 2: cardial-inflow type with paraesophageal veins; c Type 3: azygos-perforating pattern; d Type 4: complex pattern.


From Nakamura S, Murata Y, Mitsunaga A et al. Evaluation of endoscopic treatment for esophageal varices by three-dimensional endoscopic ultrasonography. Gastrointest Endosc. 2001;53:140. With permission from the American Society for Gastrointestinal Endoscopy. From Nakamura S, Murata Y, Mitsunaga A, Oi I, Hayashi N, Suzuki S. Hemodynamics of esophageal varices on three-dimensional endoscopic ultrasonography and indication of endoscopic variceal ligation. Digestive Endoscopy. 2000;15:289–297. With kind permission of Springer Science and Business Media.


Comments

Variceal ligation is particularly indicated for patients who have collaterals such as para-esophageal veins running parallel to the varices.


References

Nakamura S, Murata Y, Mitsunaga A et al. Evaluation of endoscopic treatment for esophageal varices by three-dimensional endoscopic ultrasonography. Gastrointest Endosc. 2001;53:140.


Nakamura S, Murata Y, Mitsunaga A, Oi I, Hayashi N, Suzuki S. Hemodynamics of esophageal varices on three-dimensional endoscopic ultrasonography and indication of endoscopic variceal ligation. Digestive Endoscopy. 2000;15:289–297.


Infectious Disorders


Esophageal Candidiasis: Endoscopic Grading System for Candida Esophagitis


Aims

To determine the spectrum of Candida esophagitis.


 




















Endoscopic grading system for Candida esophagitis

Grade


Appearance


I


A few raised whitish plaques up to 2 mm in diameter, with hyperemia but no edema or ulceration


II


Multiple raised white plaques larger than 2 mm in size, with hyperemia, edema but no ulceration


III


Confluent linear and nodular elevated plaques, with hyperemia and frank ulceration


IV


The findings for grade III plus increased friability of the mucous membrane and occasional narrowing of the lumen


image


Fig. 2.11a, b Esophageal candidiasis: endoscopic grading system. a Kodsi Grade II; b Kodsi Grade IV.


From Kodsi BE, Wickremesinghe C, Kozinn PJ, Iswara K, Goldberg PK. Candida esophagitis: a prospective study of 27 cases. Gastroenterology. 1976;71:715–719. With permission from the American Gastroenterological Association.


Comments

This grading system is used for controlled studies for the treatment of Candida esophagitis.


References

Kodsi BE, Wickremesinghe C, Kozinn PJ, Iswara K, Goldberg PK. Candida esophagitis: a prospective study of 27 cases. Gastroenterology. 1976;71:715–719.


Esophageal Candidiasis: Clinical Classification of Barbaro


Aims

To score symptoms related to Candida esophagitis.


 



















Clinical classification of Barbaro

Grade 0


Absence of symptoms


Grade I


Symptoms of mild entity not compromising the capability to swallow both solid and liquid substances


Grade II


Symptoms of moderate entity not compromising (grade IIa) or compromising (grade IIb) the capability to swallow solid substances, but not compromising the capability to swallow liquid substances


Grade III


Symptoms of severe entity, compromising either the capability to swallow solid substances only (grade IIIa) or compromising the capability to swallow both solid and liquid substances (grade IIIb)


From Barbaro G, Barbarini G, Di Lorenzo G. Fluconazole vs. itraconazole-flucytosine association in the treatment of esophageal candidiasis in AIDS patients, a double blind, multicenter placebo-controlled study. Chest. 1996;110:1507–1514. With permission from The American College of Chest Physicians.


Comments

This score is used for randomized studies for the treatment of Candida esophagitis.


References

Barbaro G, Barbarini G, Di Lorenzo G. Fluconazole vs. itraconazole-flucytosine association in the treatment of esophageal candidiasis in AIDS patients, a double blind, multicenter placebo-controlled study. Chest. 1996;110: 1507–1514.


Inflammatory Disorders: GERD


GERD: the DeMeester Score


Aims

To produce an objective measure of esophageal acid exposure based on 24-hour pH scoring.


 


























DeMeester score

24-hour pH monitoring components


Normal value


Supine period


< 1.2 % (0.286 ± 0.467)


Total period


< 4.2 % (1.47 ± 1.38)


# Episodes > 5 min


3 or less (0.64 ± 1.28)


Longest episode


< 9.2 min (3.83 ± 2.78)


Upright period


< 6.3 % (2.33 ± 1.97)


# Total episodes


< 50 (18.93 ± 13.78)


From Johnson LF, DeMeester TR. Twenty-four hour pH monitoring of the distal esophagus. A quantitative measure of gastroesophageal reflux. Am J Gastroenterol. 1974;62:325–332. With permission of Blackwell Publishing.


Comments

A 24-hour pH score is determined by calculating the number of standard deviation equivalents in each measured value of the six components. One starts at a fixed reference point placed two standard deviations below the respective measured mean value in controls. This is a commonly used scoring system for acid reflux.


References

DeMeester TR, Johnson LF. The evaluation of objective measurements of gastroesophageal reflux and their contribution to patient management. Surg Clin North Am. 1976;56:39–53.


DeMeester TR, Wang CI, Wernly JA et al. Technique, indications, and clinical use of 24-hour esophageal pH monitoring. J Thorac Cardiovasc Surg. 1980;79:656–670.


Johnson LF, DeMeester TR. Twenty-four hour pH monitoring of the distal esophagus. A quantitative measure of gastroesophageal reflux. Am J Gastroenterol. 1974;62:325–332. Johnson LF, DeMeester TR. Development of the 24-hour intraesophageal pH monitoring composite scoring system. J Clin Gastroenterol. 1986;8(Suppl 1):52–58.


GERD: Reflux Esophagitis Grading According to Savary–Miller


Aims

To classify reflux esophagitis into different grades by endoscopic evaluation.


 




















Reflux esophagitis grading according to Savary–Miller (1977)

Grade


 


I


One or more nonconfluent erythematous spots or superficial erosions


II


Confluent erosive or exudative mucosal lesions which do not extend around the entire esophageal circumference


III


Erosive or exudative mucosal lesions which cover the whole esophageal circumference and lead to inflammation of the wall without stricture


IV


Chronic mucosal lesions, interpreted as esophageal ulcer, mural fibrosis, stricture, shortening, and scarring with columnar (Barrett’s) epithelium


 




















Reflux esophagitis grading according to Savary–Miller (1981)

Grade


 


I


Erythematous or erythemato-exudative erosion (alone or multiple) which can cover several folds (as long as they are not confluent)


II


Confluent but not circumferential


III


Circumferential erosive and exudative lesions


IV


(a) Chronic lesions (ulcer, stenosis, endobrachyesophagus, cylindric cell epithelialization) with active inflammation


(b) Cicatricial stage (stenosis, brachyesophagus, cylindric cell epithelialization) without active inflammation


 























Savary–Miller–Monnier grading (1989)

Grade


 


I


Isolated (or sometimes multiple) erythematous or erythemato-exudative erosion, covering a single mucosal fold


II


Multiple erosions covering several mucosal folds partly confluent but never circumferential


III


Circular extension of erosive and exudative lesions


IV


(a) Ulcer


(b) Fibrosis (leading to stenosis and brachyesophagus)


V


Presence of cylindric cell epithelialization acquired in the form of a disc, strip, or sleeve


From Ollyo JB, Lang F, Fontolliet C, Monnier P. Savary–Miller’s new endoscopic grading of reflux-oesophagitis: a simple, reproducible, logical, complete and useful classification. Gastroenterology. 1990;98:100. With permission from the American Gastroenterological Association.


Comments

In 1977 Miller included the Savary grading of esophagitis in an atlas on digestive tract fiberscopy. Since then the grading has been known as Savary–Miller. In 1981 stage 4 was divided into two subgroups: stage 4a: associated with erosive or ulcerated lesions; stage 4b: not associated with erosive ulcerated lesions. In 1989 Monnier improved the grading system by modifying stage 1 and 2 and by adding stage 5 to include cylindric cell scars.


References

Miller G, Savary M, Monnier P. Notwendige Diagnostik: endoskopie. In: Blum AL, Siewert JR, eds. Reflux-therapie. Berlin: Springer; 1981:336–354.


Ollyo JB, Monnier P, Fontolliet C, Savary M. Classification endoscopique de l’esophagite par reflux. Paris: Résumé du Video Digest; 1989:145–155.


Ollyo JB, Lang F, Fontolliet C, Monnier P. Savary–Miller’s new endoscopic grading of reflux-oesophagitis: a simple, reproducible, logical, complete and useful classification. Gastroenterology. 1990;98:100.


Ollyo JB, Fontolliet C. Nouvelle classification Lausannoise des oesophagites de reflux. Acta Endoscopica. 1991;21:383–388 Savary M. La séméiologie endoscopique de l’incontinence gastro-oesophagienne [thesis]. Départment ORL, Université de Lausanne, Suisse, 1967.


Savary M, Miller G. L’oesophage. Manuel et atlas d’endoscopie. Soleure, Switzerland: Editions Gassmann S.A; 1977. Savary M, Miller G. The esophagus: Handbook and Atlas of Endoscopy. Soleure, Switzerland: Editions Gassmann S.A; 1978:135–144.


GERD: Endoscopic Grading According to Hetzel


Aims

To define the severity of endoscopically detectable reflux-induced injury of the squamous esophageal lining.


 

























Grading system

Grade 0


Normal mucosa (no abnormalities noted)


Grade I


Erythema, hyperemia, and/or friability present (no macroscopic erosions visible)


Grade II


Superficial ulceration or erosions involving < 10 % of the mucosal surface area on the distal 5 cm of esophageal squamous mucosa


Grade III


Superficial ulceration or erosions involving ≥ 10% but < 50% of the mucosal surface area on the distal 5 cm of esophageal squamous mucosa


Grade IV


Deep ulceration anywhere in the esophagus or confluent erosion of > 50% of the mucosal surface area on the last 5 cm of esophageal squamous mucosa


Grade V


Stricture that precludes the passage of the endoscope


From Hetzel DJ, Dent J, Reed WD et al. Healing and relapse of severe peptic esophagitis after treatment with omeprazole. Gastroenterology. 1988;95:903–912. With permission from the American Gastroenterological Association.


Comments

Estimation of the damaged surface area is difficult and prone to error. Grade I is based on the so-called minor or equivocal changes without tissue breaks which are poorly reproducible. The system is not validated for intra- and inter-observer variability.


References

Hetzel DJ, Dent J, Reed WD et al. Healing and relapse of severe peptic esophagitis after treatment with omeprazole. Gastroenterology. 1988;95:903–912.


GERD: the Los Angeles Classification of Reflux Esophagitis


Aims

The aim here was to produce a simple clinician-friendly grading system using unequivocal terminology.


 




















The Los Angeles classification

Grade


Description


A


One or more mucosal breaks confined to the folds, each no longer than 5 mm


B


At least one mucosal break more than 5 mm long confined to the mucosal folds but not continuous between the tops of the mucosal folds


C


At least one mucosal break continuous between the tops of two or more mucosal folds, but not circumferential


D


Mucosal break ≥ three-quarters of circumference


From Armstrong D, Bennett JR, Blum AL et al. The endoscopic assessment of esophagitis: a progress report on observer agreement. Gastroenterology. 1996;111:85–92. With permission from the American Gastroenterological Association.


Comments

The Los Angeles system is the only classification which has been validated and which has been shown to be responsive to acid suppressant therapy.


References

Armstrong D, Bennett JR, Blum AL et al. The endoscopic assessment of esophagitis: a progress report on observer agreement. Gastroenterology. 1996;111:85–92.


Lundell L, Dent J, Bennet JR et al. Endoscopic assessment of oesophagitis—clinical and functional correlates and further validation of the Los Angeles Classification. Gut 1999;45: 172–80.


image


Fig. 2.12a–d a Two single erosions, both < 5 cm. Savary–Miller grade II, Hetzel grade II, and Los Angeles grade A. b Multiple long linear erosions, with a 3-cm hiatal hernia. Savary–Miller grade II, Hetzel grade II, and Los Angeles grade B. c Erosions with whitish exudates involving the longitudinal folds and extending into the valley between folds. A stricture is also present. Savary–Miller grade IV, Hetzel grade III, and Los Angeles grade C. d Example of deep circumferential esophageal ulcers, with stenosis of the lumen. Los Angeles grade D.


GERD: The ZAP Classification


Aims

To describe the appearance of the Z-line or squamocolumnar mucosal junction.


 

























Grade


Z-line appearance


Comments


0


Sharp and circular


Z-line may be wave-like because of the mucosal folds; no narrow linear extensions (tongues) or islands of columnar epithelium


I


Irregular with tongue-like protrusions and/or islands of columnar-appearing epithelium


 


II


A distinct, obvious tongue of columnar-appearing epithelium < 3 cm in length


Width of tongue at the base must be less than its length


III


Distinct tongues of columnar-appearing epithelium > 3cm in length or Cephalad displacement of Z-line > 3cm


 


From Wallner B, Sylvan A, Stenling R, Janunger KG. The esophageal Z-line appearance correlates to the prevalence of intestinal meta-plasia. Scand J Gastroenterol. 2000;35:17–22. By permission of Taylor & Francis AS.


Comments

The classification is based on a threshold of 3 cm when describing the extent of intestinal metaplasia.


References

Tewari V, Tewari D, Solanki K et al. Assessment of squamocolumnar junction in distal esophagus using ZAP classification. Gastrointest Endosc. 2003;57:136.


Wallner B, Sylvan A, Stenling R, Janunger KG. The esophageal Z-line appearance correlates to the prevalence of intestinal metaplasia. Scand J Gastroenterol. 2000;35:17–22.


Wallner B, Sylvan A, Janunger KG. Endoscopic assessment of the “Z-line” (squamocolumnar junction) appearance: reproducibility of the ZAP classification among endoscopists. Gastrointest Endosc. 2002;55:65–69.


GERD: AFP Classification According to Bancewicz and Matthews


Aims

To produce a comprehensive grading system including anatomical functional and pathologic features.


 








































AFP score

A = anatomy


0 No hiatal hernia on routine radiology (no provocative maneuvers) or endoscopy


 


1 Sliding hiatal hernia which reduces on screening or spot film radiology


 


2 Sliding hiatal hernia which was not seen to reduce on screening or spot films


 


3 Mixed sliding and paraesophageal hernia or pure paraesophageal hernia


F = function (pH assessment)


0 No pathologic reflux on 24-hour pH testing


 


1 Reflux related to meals


 


2 Upright reflux not related to meals


 


3 Supine reflux with or without upright reflux*


P = pathology**


0 No macroscopic mucosal abnormality


 


1 Macroscopic esophagitis consisting of at least linear streaks in the distal esophagus


 


2 Concentric fibrosis (= stricture)


 


3 Longitudinal fibrosis (= short esophagus) or penetrating ulcer


*The letter L (low), N (normal), or H (high) can be added to the code in this section to denote diminished, normal, or increased pressures in the body of the esophagus on manometry, e.g., F3 L.


**The letter CLO can be added to the code in this section to indicate the presence of columnar-lined esophagus. PS may denote previous surgery. An X is used for missing data.


Comments

The AFP score is easy to use in clinical practice. It enables a detailed description of the preoperative and postoperative condition of the patient undergoing anti-reflux surgery. Full coding might read as follows: A2, F3, N, P1, CLO, PS.


References

Little AG, Ferguson MK, Skinner DB. Diseases of the Esophagus. Vol II: Benign Diseases. Mount Kisco, NY: Futura Publishing Company Inc; 1990:177–180.


GERD: Grading the Gastroesophageal Flap Valve


Aims

To grade the presence and significance of a gastroesophageal flap valve at the gastroesophageal junction by retroflexed endoscopy.


 



















Grade 1


Prominent fold of tissue along the lesser curvature, closely opposed to the endoscope


Grade 2


Fold present, but periods of opening and rapid closing around the endoscope


Grade 3


Fold is not prominent and the endoscope is not tightly gripped by the tissue


Grade 4


No fold recognizable and the lumen of the esophagus gaped open, allowing the squamous epithelium to be viewed from below; hiatal hernia


From Hill LD, Kozarek RA, Kraemer SJ et al. The gastroesophageal flap valve: in vitro and in vivo observations. Gastrointest Endosc. 1996;44: 541–547. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

Grades 1 and 2 are graded as “normal” appearance, and grades III and IV as “reflux” appearance.


References

Hill LD, Kozarek RA, Kraemer SJ et al. The gastroesophageal flap valve: in vitro and in vivo observations. Gastrointest Endosc. 1996;44:541–547.


image


Fig. 2.13a–d Gastroesophageal reflux: grading the gastroesophageal flap valve. a Grade 1; b Grade 2; c Grade 3; d Grade 4.


Neoplastic Lesions


Columnar Metaplasia: CM Classification of Columnar Metaplasia—The Prague System


Aims

To produce an accurate and reproducible assessment of the endoscopic extent of esophageal columnar metaplasia, applicable in clinical practice.


 






CM staging

The length of esophagus lined circumferentially (C) with columnar mucosa is measured in addition to the total length (M) of esophagus involved with columnar mucosa at any point


image


Fig. 2.14 Columnar metaplasia: the Prague Barrett’s C and M endoscopic criteria.


From Armstrong D. Review article: towards consistency in the endoscopic diagnosis of Barrett’s oesophagus and columnar metaplasia. Aliment Pharmacol Ther. 2004;20(5):40–47. With permission of Blackwell Publishing.


Comments

The International Working Group on the Classification of Esophagitis developed the CM classification. A clear definition of endoscopic landmarks for the identification and localization of the gastroesophageal junction as well as the squamocolumnar junction is essential.


References

Armstrong D. Review article: towards consistency in the endoscopic diagnosis of Barrett’s oesophagus and columnar metaplasia. Aliment Pharmacol Ther. 2004;20(5):40–47.


Columnar Metaplasia: Magnifying Endoscopy Classification of Barrett’s Esophagus According to Guelrud


Aims

This classification system was established to train endoscopists to detect mucosal changes and to define a reproducible terminology.


 



















2001 Classification (magnification ×35)

Pattern I


Round pits: characterized by an organized and regularly arranged area of circular dots


Pattern II


Reticular: characterized by circular or oval similarly shaped tubules in regular arrangement


Pattern III


Villous: characterized by a fine villiform appearance with a regular shape and arrangement without any pits


Pattern IV


Ridged: characterized by a cerebriform appearance consisting of a thick villous convoluted shape with a regular arrangement


From Guelrud M, Herrera I, Essenfeld H, Castro J. Enhanced magnification endoscopy: a new technique to identify specialized intestinal metaplasia in Barrett’s esophagus. Gastrointest Endosc. 2001;53: 559–565. With permission from the American Society for Gastrointestinal Endoscopy.


 




























2004 New classification (magnification ×80)

Round pits pattern


Formerly Pattern I, small round pits with a uniform dot-like appearance, signifying fundic columnar epithelium


Tubular pits pattern


Formerly Pattern II, ovoid or short tubular pits with a uniform arrangement, signifying cardia mucosal


Thin linear pattern


New category, thin superficial grooves that may represent cardia mucosa and/or Barrett’s epithelium


Deep linear pattern


New category, deep, coarse grooves signifying Barrett’s epithelium


Villous pattern


Formerly Pattern III, uniform mesh-like villous appearance or, more rarely, fingerlike projections, signifying Barrett’s epithelium


Foveolar pattern


New category, flat surface interspersed with wide circular pits and the absence of villous structures, signifying Barrett’s epithelium


Cerebroid pattern


Formerly Pattern IV, uniform thick, straight tubular or convoluted ridges reminiscent of cerebral gyri, signifying Barrett’s epithelium


From Guelrud M, Ehrlich EE. Endoscopic classification of Barrett’s esophagus. Gastrointest Endosc. 2004;59:58–65. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

In 2004 the original patterns were renamed, and three new categories were added: thin linear, deep linear, and foveolar. In the new classification, detailed mucosal surface descriptions were substituted for mucosal “patterns.”


References

Guelrud M, Herrera I, Essenfeld H, Castro J. Enhanced magnification endoscopy: a new technique to identify specialized intestinal metaplasia in Barrett’s esophagus. Gastrointest Endosc. 2001;53:559–565.


Guelrud M, Ehrlich EE. Endoscopic classification of Barrett’s esophagus. Gastrointest Endosc. 2004;59:58–65.


Columnar Metaplasia: Classification of Barrett’s Epithelium by Magnifying Endoscopy According to Endo


Aims

To classify pit patterns of Barrett’s esophagus by magnifying endoscopy.


 
























Pit-1


Small round


Small round pits of relatively uniform size and shape


Pit-2


Straight


Long straight lines


Pit-3


Long oval


Exhibits long extended pits, larger than those of the dot type


Pit-4


Tubular


Complicated and twisted pattern that is similar to a branch or gyrus like structure


Pit-5


Villous


Flat, finger-like projections


image


Fig. 2.15 Columnar metaplasia: Classification of Barrett’s epithelium by magnifying endoscopy according to Endo.


From Endo T, Awakawa T, Takahashi H et al. Classification of Barrett’s epithelium by magnifying endoscopy. Gastrointest Endosc. 2002;55: 641–647. With permission from the American Society of Gastrointestinal Endoscopy.


Comments

The classification of the superficial mucosal appearance of Barrett’s epithelium by magnifying endoscopy reflects histologic features and mucin phenotypes.


References

Endo T, Awakawa T, Takahashi H et al. Classification of Barrett’s epithelium by magnifying endoscopy. Gastrointest Endosc. 2002;55:641–647.


Columnar Metaplasia: Magnifying Endoscopy Classification of Barrett’s Esophagus According to Toyoda


Aims

To determine the sensitivity and specificity of magnification endoscopy for the detection of intestinal metaplasia by using a revised pit classification. Three types of mucosal pattern are classified:


 













Type 1


Normal pits


Type 2


Slit-reticular pattern


Type 3


Gyrus-villous pattern


See Fig. 2.16 next page.


From Toyoda H, Rubio C, Befrits R, Hamamoto N, Adachi Y, Jaramillo E. Detection of intestinal metaplasia in distal esophagus and esophagogastric junction by enhanced-magnification endoscopy. Gastrointest Endosc. 2004;59:15–21. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

The presence of type 1 and type 2 surface patterns are characteristic of, respectively, fundic epithelium and cardiac epithelium. The type 3 pattern correlated with intestinal metaplasia.


References

Toyoda H, Rubio C, Befrits R, Hamamoto N, Adachi Y, Jaramillo E. Detection of intestinal metaplasia in distal esophagus and esophagogastric junction by enhanced-magnification endoscopy. Gastrointest Endosc. 2004;59:15–21.


image


Fig. 2.16a–e Classification of surface pattern of Barrett’s epithelium and esophagogastric junctional mucosa by enhanced magnifying endoscopy. a Small round pits of uniform size and shape (type 1, corpus). b Slit (arrows) and reticular (arrowhead) pattern (type 2, cardiac). c Gyrus pattern (type 3, intestinal metaplasia). d Villous pattern (type 3, intestinal metaplasia. e Mixed gyrus and villous pattern (type 3, intestinal metaplasia). Findings illustrated in c, d, and e constitutes a single mucosal pattern (type 3).


Esophageal Cancer: Classification of the Intrapapillary Capillary Loop Pattern According to Inoue


Aims

To describe intra-papillary capillary loop patterns and the characteristic changes of carcinoma in situ.


 























Classification of the intra-papillary capillary loop pattern (IPCL)

Typing


IPCL and iodine staining pattern


Type I (normal)


Normal IPCL; iodine stained


Type II (esophagitis)


Dilatation and elongation of IPCL; slightly stained


Type III (mild dysplasia)


Minimal changes on IPCL; unstained


Type IV (severe dysplasia)


Two or three out of four changes; unstained


Type V (carcinoma)


All four changes on IPCL; unstained


 

























Classification of intra-papillary capillary loop pattern (IPCL) destruction according to the infiltration of T1 esophageal cancer

Infiltration


Grade of IPCL destruction


Description


m1


 


Characteristic IPCL changes to intraepithelial carcinoma (Type V changes) only affect the top of the IPCL


m2


 


Type V changes affect the middle part of the IPCL, and are observed as an elongation of the affected IPCL


m3


Severely destroyed


Type V changes affect the total length of the IPCL and the original shape of the IPCL has been destroyed


sm


Disappeared


Abnormal tumor vessels with large diameters have appeared


image


Fig. 2.17 Classification of IPCL according to Inoue.


From Inoue H. Magnification endoscopy in the esophagus and stomach. Dig Endosc. 2001;13:40–41. With permission of Blackwell Publishing.


Comments

The four characteristic changes in IPCL pattern are: dilatation, tortuous running, caliber changes, and shape.


References

Inoue H. Magnification endoscopy in the esophagus and stomach. Dig Endosc. 2001;13:40–41.


Inoue H, Honda T, Yoshida T et al. Ultra-high magnification endoscopy of the normal esophageal mucosa. Dig Endosc. 1996;8:134–138.


Inoue H, Honda T, Nagai K et al. Ultra-high magnification endoscopic observation of carcinoma in situ of the esophagus. Dig Endosc. 1997;9:16–18.


Inoue H, Youichi K, Yoshida T, Kawano T, Endo M, Iwai T. High-magnification endoscopic diagnosis of the superficial esophageal cancer. Dig Endosc. 2000;12:32–35.


Esophageal Cancer: Macroscopic Classification of Superficial and Advanced Esophageal Cancer by the Japanese Society of Esophageal Disease


Aims

To describe a new endoscopic classification of esophageal cancer that includes both early and advanced lesions.


 




























Macroscopic classification of superficial esophageal cancer

Type 0


Superficial type


Type 0–I


Superficial and protruding type


Type 0–II


Superficial and flat type


 


Type 0–IIa


Slightly elevated type


 


Type 0–IIb


Flat type


 


Type 0–IIc


Slightly depressed type


Type 0–III


Superficial and distinctly depressed type


 














Macroscopic classification of advanced esophageal cancer

Type 1: Protruding type


Type 2: Ulcerative and localized type


Type 3: Ulcerative and infiltrating type


Type 4: Diffusely infiltrating type


Type 5: Unclassified type


image


Comments

Commonly used endoscopic grading system for early and advanced cancer.


References

Japanese Society for Esophageal Diseases. Guidelines for Clinical and Pathologic Studies on Carcinoma of the Esophagus. 8th ed. Tokyo: Kanehara; 1992:31–58.


Kumagai Y, Makuuchi H, Mitomi T, Ohmori T. A new classification system for early carcinomas of the esophagus. Dig Endosc. 1993;5:139–50.


Esophageal Cancer: Classification of Depth of Invasion of Superficial Esophageal Cancer by the Japanese Society of Esophageal Disease


Aims

To estimate the depth of infiltration.


 






















Classification of depth of invasion of superficial esophageal cancer

m1:


Epithelial cancer


m2:


Mucosal cancer with invasion between m1 and m3


m3:


Mucosal cancer almost and/or already invading the lamina muscularis mucosae


sm1:


Submucosal cancer with invasion to the first third of the submucosal layer


sm2:


Submucosal cancer with invasion to the middle third of the submucosal layer


sm3:


Massive invasion of the cancer to the submucosal layer


image


Fig. 2.18 Classification of depth of invasion of superficial esophageal cancer: the Japanese Society of Esophageal Disease. Depth of invasion of squamous cell neoplasia in the esophagus. Mucosal carcinoma is divided into three groups: m1 or intraepithelial, m2 or micro-invasive (invasion through the basement membrane), m3 or intramucosal (invasion to the muscularis mucosae). The depth of invasion in the submucosa is divided into three sections of equivalent thickness: superficial (sm1), middle (sm2), and deep (sm3).


Comments

Increasingly used staging; lesions beyond sm1 are not amendable to endoscopic resection because of increased risk of lymph node metastasis.


References

Japanese Society for Esophageal Diseases. Guidelines for the clinical and pathologic studies on carcinoma of the esophagus. 8th ed. Tokyo: Kanehara; 1992:31–58.


Esophageal Cancer: AJCC TNM Staging System of Esophageal Cancer


 

























Primary tumor (T)

TX


Primary tumor cannot be assessed


T0


No evidence of primary tumor


Tis


Carcinoma in situ


T1


Tumor invades lamina propria or submucosa


T2


Tumor invades muscularis propria


T3


Tumor invades adventitia


T4


Tumor invades adjacent structures


 













Regional lymph nodes (N)

NX


Regional lymph nodes cannot be assessed


N0


No regional lymph node metastasis


N1


Metastasis in a single ipsilateral lymph node, 3 cm or less in greatest dimension


 













Distant metastasis (M)

MX


Distant metastasis cannot be assessed


M0


No distant metastasis


M1


Distant metastases


 










Tumors of the lower thoracic esophagus

M1a


Metastasis in celiac lymph nodes


M1b


Other distant metastasis


 










Tumors of the mid thoracic esophagus

M1a


Not applicable


M1b


Nonregional lymph nodes and/or other distant metastasis


 










Tumors of the upper thoracic esophagus

M1a


Metastasis in cervical nodes


M1b


Other distant metastasis


image


 



















Histologic grade (G)

GX


Grade cannot be assessed


G1


Well differentiated


G2


Moderately differentiated


G3


Poorly differentiated


G4


Undifferentiated


 
















Residual tumor (R)

RX


Presence of residual tumor cannot be assessed


R0


No residual tumor


R1


Microscopic residual tumor


R2


Macroscopic residual tumor


From Greene, FL, Page, DL, Fleming ID et al. Used with the permission of the American Joint Committee on Cancer (AJCC), Chicago, Illinois. The original source for this material is the AJCC Cancer Staging Manual, 6th ed (2002) published by Springer-New York, www.springeronline.com.


Comments

The AJCC TNM staging system uses three basic descriptors that are then grouped into stage categories. The first component is “T,” which describes the extent of the primary tumor. The next component is “N,” which describes the absence or presence and extent of regional lymph node metastasis. The third component is “M,” which describes the absence or presence of distant metastasis. The final stage groupings (determined by the different permutations of “T,” “N,” and “M”) range from Stage 0 through Stage IV.


References

Greene, FL, Page, DL, Fleming ID et al. AJCC Cancer Staging Manual. 6th ed. New York: Springer, 2002. http://www.cancerstaging.org/.


Esophageal Cancer: Preoperative Risk Analysis According to Bartels


Aims

To assess the preoperative risk for postoperative death after operation for resectable esophageal cancer.


image


From Bartels H, Stein HJ, Siewert JR. Preoperative risk analysis and postoperative mortality of oesophagectomy for resectable oesophageal cancer. Br J Surg. 1998;85:840–844. With permission granted by John Wiley & Sons, Ltd. on behalf of the BJSS, Ltd.


Comments

The total score ranges from 11 (without risk) to 33 (highest risk). On the basis of the score patients can be divided in three risk groups; “low risk” 11–15 points, “moderate risk” 16–21 points, “high risk” 22–33 points.


References

Bartels H, Stein HJ, Siewert JR. Preoperative risk analysis and postoperative mortality of oesophagectomy for resectable oesophageal cancer. Br J Surg. 1998;85:840–844.


Esophageal Cancer: Bronchoscopy Findings in Carcinoma of the Esophagus


Aims

The aim here was to classify bronchoscopy findings in the pre-operative staging of supracarinal esophageal cancer.


 




















Bronchoscopy findings in carcinoma of the esophagus

Category


Bronchoscopy findings


I


No abnormalities


IIa


Slight compression of the trachea or bronchus; normal mobility of the wall; and parallel and regular longitudinal folds of the pars membranacea


IIb


Impingement or deviation of the trachea or bronchus or widening of the carina, associated with reduction of the mobility during coughing and breathing


III


Frank mucosal invasion


From Choi TK, Siu KF, Lam KH, Wong J. Bronchoscopy and carcinoma of the esophagus I. Findings of bronchoscopy in carcinoma of the esophagus. Am J Surg. 1984;147:757–759. With permission from Excerpta Medica Inc.


Comments

Airway involvement as in category IIb is a predictor of irresectability.


References

Baisi A, Bonavina L, Peracchia A. Bronchoscopic staging of squamous cell carcinoma of the upper thoracic esophagus. Arch Surg. 1999;134:140–143.


Choi TK, Siu KF, Lam KH, Wong J. Bronchoscopy and carcinoma of the esophagus I. Findings of bronchoscopy in carcinoma of the esophagus. Am J Surg. 1984;147:757–759.


Choi TK, Siu KF, Lam KH, Wong J. Bronchoscopy and carcinoma of the esophagus II. Carcinoma of the esophagus with tracheobronchial involvement. Am J Surg. 1984;147:760–762.


Carcinoma of the Esophagogastric Junction: The Siewert Classification


Aims

To classify esophagogastric tumors based on the anatomic location of the tumor center.


 
















The Siewert classification

Type I


Adenocarcinoma of the distal esophagus, which usually arises from an area with specialized intestinal metaplasia of the esophagus (i. e., Barrett’s esophagus) and may infiltrate the esophagogastric junction from above


Type II


True carcinoma of the cardia arising immediately at the esophagogastric junction


Type III


Subcardial gastric carcinoma that infiltrates the esophagogastric junction and distal esophagus from below


From Siewert JR, Stein HJ. Carcinoma of the cardia: carcinoma of the gastroesophageal junction—classification, pathology and extent of re-section. Dis Esophagus. 1996;9:173–182. With permission of Blackwell Publishing.


image


Fig. 2.19 Classification of esophagogastric tumors according to Siewert: Type I, esophageal; type II, cardiac; type III, subcardiac. (Fein M et al. Surgery. 1998;124:707–714. Reproduction approved.)


Comments

This classification was approved at a consensus conference of the International Gastric Cancer Association and the International Society for Diseases of the Esophagus in 1997. The authors treat type I tumors as esophageal cancer and type II and III tumors as gastric cancer.


References

Siewert JR, Stein HJ. Carcinoma of the cardia: carcinoma of the gastroesophageal junction—classification, pathology and extent of resection. Dis Esophagus. 1996;9:173–182.


Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction. Br J Surg. 1998;85:1457–1459.



Stomach


Symptoms


Dyspepsia: The Glasgow Dyspepsia Severity Score


Aims

The aim was to develop a questionnaire for measuring the global and personal impact of dyspeptic symptoms.


 






































































































































The Glasgow dyspepsia severity score

(A) Frequency of dyspeptic symptoms


Score


Over the past six months how frequently have you experienced dyspeptic symptoms?


 


Never


0


On only 1 or 2 days


1


On approximately 1 day per month


2


On approximately 1 day per week


3


On approximately 50 % of days


4


On most days


5


(B) Effect on normal activities


 


Does the dyspepsia interfere with normal activities such as eating, sleeping, or socializing?


 


Never


0


Sometimes


1


Regularly


2


(C) Time off work


 


How many days have you lost off work due to your dyspepsia in the past six months?


 


None


0


1–7 days


1


More than 7 days


2


(D) Consultation with medical profession


 


How often have you attended a doctor due to dyspepsia in the past 6 months?


 


None


0


Once


1


Twice or more


2


(E) GP visits to patient’s home


 


How often have you called your GP to visit you at home because of your dyspepsia in the past six months?


 


None


0


Once


1


Twice or more


2


(F) Tests for dyspepsia


 


How many tests have you had for your dyspepsia in the past 6 months?


 


None


0


One


1


Two or more


2


(G) Treatment for dyspepsia


 


(1) Over the last six months, how frequently have you used drugs that you have obtained by yourself?


 


Never


0


Less than once per week


1


More than once per week


2


(2) Over the last 6 months, for how long have you used drugs prescribed by a doctor?


 


Never


0


For 1 month or less


1


For 1–3 months


2


For more than 3 months


3


From El-Omar EM, Banerjee S, Wirz A, McColl KEL. The Glasgow Dyspepsia Severity Score—a tool for the global measurement of dyspepsia. Eur J GE Hepatol. 1996;8:967–971. With permission of Lippincott Williams & Wilkins (LWW).


Comments

The score was assessed for validity and reproducibility. The mean score in the general population was 1.16 compared with 10.5 in the nonulcer dyspepsia patients and 11.1 in the duodenal ulcer patients.


References

El-Omar EM, Banerjee S, Wirz A, McColl KEL. The Glasgow Dyspepsia Severity Score—a tool for the global measurement of dyspepsia. Eur J GE Hepatol. 1996;8:967–971.


Dyspepsia: Dyspepsia Symptom Severity Index (DSSI)


Aims

To develop a self-report measure to quantify the severity of dyspepsia symptoms in clinical practice and research.


 





















































Dyspepsia symptom severity index (DSSI)

Scale/item


Dysmotility-like


1. Frequent burping or belching


4. Bloating


5. Feeling full after meals


6. Inability to finish normal-sized meals


7. Abdominal (belly) discomfort, without pain, after meals


8. Abdominal (belly) distension (feels as though you need to loosen your clothes)


12. Nausea before meals


13. Nausea after meals


14. Nausea when you wake up in the morning


15. Retching (heaving as if to vomit, with little result)


16. Vomiting


Reflux-like


3. Burping with bitter tasting fluid in throat


17. Regurgitation of bitter fluid into your mouth (reflux) during the day


18. Regurgitation (reflux) at night


19. Burning feeling in your chest (heartburn)


20. Burning feeling in your stomach


Ulcer-like


9. Abdominal (belly) ache or pain right after meals


10. Abdominal (belly) pain before meals or when hungry


11. Abdominal (belly) pain at night


Overall item


From Leidy NK, Farup C, Rentz AM, Ganoczy D, Koch KL. Patient-based assessment in dyspepsia: Development and validation of dyspepsia symptom severity index (DSSI). Dig Dis Sci. 2000;45:1172– 1179. With kind permission of Springer Science and Business Media.


Comments

This is a self-administered questionnaire of 20 items. The numbers in the table state the following order of the items in the questionnaire. The grading of the items by the patient should be on a 0 (absent) to 4 (very severe) Likert scale. One global item is included in order to gather data on the patient’s overall impression of his/her dyspepsia symptom severity.


A total score is represented by the mean across the three subscales (dysmotility-, reflux-, and ulcer-like symptoms).


References

Leidy NK, Farup C, Rentz AM, Ganoczy D, Koch KL. Patient-based assessment in dyspepsia: Development and validation of dyspepsia symptom severity index (DSSI). Dig Dis Sci. 2000;45:1172–1179.


Dyspepsia: Patient Symptom Questionnaire According to Talley


Aims

The aim was to assess changes in symptom severity in a placebo-controlled trial for patients with functional dyspepsia.


image


Comments

This is a patient-completed measure of disease. Severity is scored for each symptom on a 100-mm visual analogue scale. The outcome is defined a priori as the sum of the severity of the eight symptoms, to create the total upper abdominal discomfort severity score (minimum 0, maximum 800 mm). Symptom frequency (from not at all to every day) and impact (from not at all bothersome to extremely bothersome) are scored on five graded Likert scales. Duration (from not at all to continuous or almost continuous) is graded on a seven-graded Likert scale.


References

Talley NJ, Verlinden M, Snape W et al. Failure of a motilin receptor agonist (ABT-229) to relieve the symptoms of functional dyspepsia in patients with and without delayed gastric emptying: a randomized double-blind placebo-controlled trial. Aliment Pharmacol Ther. 2000;14:1653–1661.


Dyspepsia: Symptom Severity Quantitation According to Parkman


Aims

The aim is to assess functional dyspepsia and motor dysfunction by grading six symptoms.


 































Symptom severity quantitation according to Parkman

Symptom


Score


Upper abdominal discomfort


0 none


1 mild


2 moderate


3 severe


Early satiety


0 none


1 mild


2 moderate


3 severe


Postprandial abdominal distension


0 none


1 mild


2 moderate


3 severe


Nausea


0 none


1 mild


2 moderate


3 severe


Vomiting


0 none


1 mild


2 moderate


3 severe


Anorexia


0 none


1 mild


2 moderate


3 severe


Total score


From Parkman HP, Miller MA, Trate D et al. Electrogastrography and gastric emptying scintigraphy are complementary for assessment of dyspepsia. J Clin Gastroenterol. 1997;24(4):214–219. With permission of Lippincott Williams & Wilkins (LWW).


Comments

The patient has to grade six symptoms of dyspepsia and upper gastrointestinal motor dysfunction. The score ranges from 0 points (no symptoms) to a maximum of 18 points.


References

Parkman HP, Miller MA, Trate D et al. Electrogastrography and gastric emptying scintigraphy are complementary for assessment of dyspepsia. J Clin Gastroenterol. 1997;24(4): 214–219.


Dyspepsia: The Leeds Dyspepsia Questionnaire (LDQ)


Comments

The Leeds Dyspepsia Questionnaire (LDQ) contains eight items. Each with two stems, relating to the frequency and severity of dyspepsia symptoms over the previous 6 months and one item on the most troublesome symptom experienced by the subject. The LDQ gives a range of scores from 0–40, and contains questions on epigastric pain, retrosternal pain, regurgitation, nausea, vomiting, belching, early satiety, and dysphagia. The first five questions are used to determine the presence of dyspepsia, whilst all eight questions are required to measure the severity of dyspepsia. The questionnaire is available on request.


References

Moayyedi P, Duffett S, Braunholtz D et al. The Leeds Dyspepsia Questionnaire: a valid tool for measuring the presence and severity of dyspepsia. Aliment Pharmacol Ther. 1998;12: 1257–1262.


Dyspepsia: Rome IIII Criteria for Functional Dyspepsia


 


















































Functional dyspepsia

1


Bothersome postprandial fullness; or


Early satiation; or


Epigastsric pain or


Epigastric burning


2


No evidence of structural disease (including at upper endoscopy) that is likely to explain the symptoms


Postprandial Distress Syndrome (PDS)


1


Bothersome postprandial fullness, occurring after ordinary sized meals, at least several times per week; or


2


Early satiation that prevents finishing a regular meal, at least several times per week Supportive Criteria:


 


1. Upper abdominal bloating or postprandial nausea or excessive belching can be present


 


2. EPS may co-exist


Epigrastric Pain Syndrome (EPS)


1


Pain or burning localized to the epigastrium, or at least moderate severity at least once per week


2


The pain is intermittent


3


Not generalized or localized to other abdominal or chest regions


4


Not relieved by defecation or passage of flatus


5


Not fulfilling criteria for biliary pain


6


The pain may be of a burning quality, but without a retrosternal component


7


The pain is commonly induced or relieved by ingestion of a meal, but may occur while fasting


8


PDS may co-exist


References

Drossman DA. Rome III. The functional gastrointestinal disorders. Lawrence, KW, USA: Allen Press, Inc., 2006.


Gastric Symptom Questionnaires: Severity of Common Symptoms (SCS)


SCS is an eight-question symptom measure developed for use in nonulcer dyspepsia and H. pylori -associated gastritis. The instrument is not specific for dyspepsia and lacks sensitivity.


References

Kuykendall DH, Rabeneck L, Campbell CJ, Wray NP. Dyspepsia: how should we measure it? J Clin Epidemiol. 1998;51:99–106.


Veldhuyzen van Zanten SJ, Tytgat KM, Pollak PT et al. Can severity of symptoms be used as an outcome measure in trials of non-ulcer dyspepsia and Helicobacter pylori associated gastritis? J Clin Epidemiol. 1993;46:273–279.


Gastric Symptom Questionnaires: Aberdeen Dyspepsia Questionnaire (ADQ) by Garratt


References

Garratt AM, Ruta DA, Russell I et al. Developing a condition-specific measure of health for patients with dyspepsia and ulcer-related symptoms. J Clin Epidemiol. 1996;49:565–571.


Gastric Symptom Questionnaires: Dyspepsia Questionnaire by Mansi


References

Mansi C, Borro P, Giacomini M et al. Comparative effects of levosulpiride and cisapride on gastric emptying and symptoms in patients with functional dyspepsia and gastroparesis. Aliment Pharmacol Ther. 2000;14:561–569.


Gastroparesis Patient Symptom Questionnaire


Aims

The aim is to grade upper GI symptoms in patients with idiopathic gastroparesis.


 











































































Patient symptom questionnaire

Symptom


None


 


 


 


Extreme


Postprandial fullness


0


1


2


3


4


Early satiety


0


1


2


3


4


Bloating


0


1


2


3


4


Epigastric discomfort (an ache or discomfort after meals, poorly localized)


0


1


2


3


4


Epigastric pain (a sharp, easy to pinpoint pain after eating)


0


1


2


3


4


Postprandial nausea


0


1


2


3


4


Belching after meals


0


1


2


3


4


Vomiting


0


1


2


3


4


From Miller LS, Szych GA, Kantor SB et al. Treatment of idiopathic gastroparesis with injection of botulinum toxin into the pyloric sphincter muscle. Am J Gastroenterol. 2002;97:1653–1660. With permission of Blackwell Publishing.


Comments

A total score is calculated as the sum of each of the eight individual symptom scores. The maximum score is 32. The score correlates with improved solid phase gastric emptying scintigraphy in patients treated with botulinum toxin injection into the pyloric sphincter muscle.


References

Miller LS, Szych GA, Kantor SB et al. Treatment of idiopathic gastroparesis with injection of botulinum toxin into the pyloric sphincter muscle. Am J Gastroenterol. 2002;97: 1653–1660.


Gastroparesis Cardinal Symptom Index (GCSI)


Aims

To develop a medical treatment measure of gastroparesis-related symptoms.


 








This questionnaire asks you about the severity of symptoms you may have related to your gastrointestinal problem. There are no right or wrong answers. Please answer each question as accurately as possible.


For each symptom, please circle the number that best describes how severe the symptom has been during the past week. If you have not experienced this symptom, circle 0. If the symptom has been very mild, circle 1. If the symptom has been mild, circle 2. If it has been moderate, circle 3. If it has been severe, circle 4. If it has been very severe, circle 5. Please be sure to answer every question. Please rate the severity of the following symptoms during the past week.


image


Comments

The GCSI was developed as part of a larger patient outcomes project for the development of the Patient Assessment of Upper Gastrointestinal Disorders-Symptom Severity Index (PAGISYM).


The GCSI consists of three subscales of the PAGI-SYM instrument, selected to measure important symptoms related to gastroparesis: nausea/vomiting (three items). Postprandial fullness/early satiety (four items) and bloating (two items).


A six-point Likert response scale ranging from 0 (none) to 5 (very severe) with a 2-week recall period is used to rate the severity of each symptom. The GCSI total score is constructed as the average of the three symptom subscales.


References

Rentz AM, Schmier J, De La Loge C et al. Development and preliminary psychometric validation of the patient assessment of upper gastrointestinal disorders-symptom severity index (PAGI-SYM) in GI patients. Presented at the annual meeting of the International Society for Pharmaco-economics and Outcome Research, Antwerp, Belgium, November 2000.


Revicki DA, Rentz AM, Dubois D et al. Development and validation of a patient-assessed gastroparesis symptom severity measure: the Gastroparesis Cardinal Symptom Index. Aliment Pharmacol Ther. 2003;18:141–150.


Anatomical Variants, Injury


Paraesophageal Hernia: Classification of Hiatal Hernias According to Allison


Aims

Allison performed hiatus herniorrhaphy to prevent reflux esophagitis.


 



















Classification of hiatal hernias

Type 1


Sliding hernia. The gastroesophageal junction moves cephalad and may predispose to gastroesophageal reflux


Type 2


Pure paraesophageal hernias. The esophagogastric junction is below the diaphragm. The fundus herniates alongside the esophagus through an anterior weakness of the gastrophrenic ligament


Type 3


A combination of both a sliding and a paraesophageal defect


Type 4


Other viscera including the colon, spleen, small bowel, and omentum herniate through the defect


From Allison P. Reflux, esophagitis, sliding hiatal hernia, and the anatomy of repair. Surg Gynecol Obstet. 1951;92:419–431. With permission of Elsevier.


Comments

Type 2 is rare. Type 3 hernias are usually large and repair is advocated regardless of the symptoms because of the potential incarceration.


References

Allison P. Reflux, esophagitis, sliding hiatal hernia, and the anatomy of repair. Surg Gynecol Obstet. 1951;92:419–431.


Gastric Outlet Obstruction: The Gastric Outlet Obstruction Scoring System (GOOSS)


Aims

A scoring system was created to grade the ability to eat.


 




















GOOSS

Score


 


0


No oral intake


1


Liquids only


2


Soft foods


3


Low residue or full diet


From Adler DG, Baron TH. Endoscopic palliation of malignant gastric outlet obstruction using self-expanding metal stents: experience in 36 patients. Am J Gastroenterol. 2002;97:72–78. With permission of Blackwell Publishing.


Comments

This represents a simple grading system for gastric emptying quality. Used for endoscopic treatment of malignant gastric outlet obstruction. The GOOSS can be applied pre- and poststent placement.


References

Adler DG, Baron TH. Endoscopic palliation of malignant gastric outlet obstruction using self-expanding metal stents: experience in 36 patients. Am J Gastroenterol. 2002;97:72–78.


image


Fig. 2.20 Paraesophageal hernia: classification according to Allison. a Type II. b Type III.


Gastric Volvulus: Types of Gastric Volvulus


Aims

Gastric volvulus is classified on the basis of the orientation of the rotational axis.


 

















Types of gastric volvulus

Type


 


1


Organo-axial, the greater curvature rotates anteriorly and superiorly. The rotational axis is the line between the gastro-esophageal junction and pylorus


2


Mesentero-axial, the greater curvature remains in its caudal position, but the pyloroantral region rotates in more of a right-to-left direction. The rotational axis is a line between the midlines of the greater and lesser curvatures


3


Vertical, the rarest type of volvulus, it is a combination of the first and second types


image


Fig. 2.21 Types of gastric volvulus.


Reproduced from Wastell C, Ellis H. Volvulus of the stomach. A review with a report of 8 cases. Br J Surg. 1971;58:557–562. With permission granted by John Wiley & Sons, Ltd. on behalf of the BJSS, Ltd.


Comments

The gastric organo-axial volvulus is the most common. The volvulus can be further categorized as: primary or secondary depending on accompanying additional pathologies; as acute or chronic according to its onset; and as intra-thoracic or intra-abdominal according to its location. Determination of the type of volvulus usually is of limited clinical significance, as the tightness and rapidity of the twist are more predictive of eventual gangrene.


References

Wastell C, Ellis H. Volvulus of the stomach. A review with a report of 8 cases. Br J Surg. 1971;58:557–562.


Injury to the Stomach: Organ Injury Scaling of the Stomach


Aims

The aim is to grade the degree of injury to the stomach.


 




















Organ injury scaling of the stomach

Grade


Description


I


Superficial hematoma; partial thickness laceration


II


Laceration < 1cm


III


Laceration 1–5 cm


IV


Laceration 5–10 cm


From Moore EE, Jurkovich GJ, Knudson MM et al. Organ injury scaling. VI: Extrahepatic biliary, esophagus, stomach, vulva, vagina, uterus (nonpregnant), uterus (pregnant), fallopian tube, and ovary. J Trauma. 1995;39:1069–1070. With permission of Lippincott Williams & Wilkins (LWW).


Comments

This is a useful grading system in abdominal trauma.


References

Moore EE, Jurkovich GJ, Knudson MM et al. Organ injury scaling. VI: Extrahepatic biliary, esophagus, stomach, vulva, vagina, uterus (nonpregnant), uterus (pregnant), fallopian tube, and ovary. J Trauma. 1995;39:1069–1070.


Vascular Disorders


Gastric Varices: Classification According to Sarin and Kumar


Aims

To classify gastric varices according to their localization.


 
















The Sarin and Kumar classification

GEV1


Gastroesophageal varices extending down the lesser curvature, they are more or less straight


GEV2


Gastroesophageal varices extending into the fundus of the stomach, they are long and tortuous


IGV1


Isolated gastric varices in the fundus (fundal varices)


IGV2


Isolated ectopic varices at other sites in the stomach or duodenum


GEV, gastroesophageal varix; IGV, isolated gastric varices; EGJ, esophagogastric junction.


image


Fig. 2.22a, b Gastric varices: classification according to Sarin and Kumar. a Gastroesophageal varices. b Isolated gastric varices.


Comments

Fundal varices (GEV2, IGV1) are more ominous. Tips are less effective in gastric varices compared to esophageal varices.


References

Sarin SK. Long–term follow-up of gastric variceal sclerotherapy: an eleven-year experience. Gastrointest Endosc. 1997;46:8–14.


Sarin SK, Kumar A. Gastric varices: profile, classification, and management. Am J Gastroenterol. 1989;84:1244–1249.


Sarin SK, Lahoti D, Saxena SP, Murthi NS, Makwane UK. Prevalence, classification and natural history of gastric varices: long term follow-up study in 568 patients with portal hypertension. Hepatology. 1992;16:1343–1349.


Portal Hypertensive Gastropathy: Classification of the Severity of Portal Hypertensive Gastropathy According to McCormack


Aims

The aim was to develop a classification to describe the progression of changes in gastric mucosal lesions during the endoscopic follow-up of portal hypertension.


 


























Classification of the severity of portal hypertensive gastropathy according to McCormack

Classification


Gastric mucosal changes


Nil


Normal appearance


Mild


(i) A fine pink speckling or scarlatina type rash


 


(ii) Superficial reddening, particularly on the surface of the rugae, giving a striped appearance


 


(iii) A fine white reticular pattern separating areas of raised red edematous mucosae resembling a ‘snake skin’


Severe


(i) Discrete red spots analogous to the cherry red spots described in the esophagus. These spots can become confluent giving a local area of severe gastritis that may bleed


 


(ii) A diffuse hemorrhagic gastritis


From McCormack TT, Sims J, Eyre-Brook I et al. Gastric lesions in portal hypertension: inflammatory gastritis or congestive gastropathy. Gut. 1985;26:1226–1232. With permission of BMJ Publishing Group.


Comments

Macroscopic gastritis in portal hypertension is unrelated to the etiology of the portal hypertension and it does not respond to conventional anti-inflammatory drug therapy. There is an increased risk of bleeding in severe gastritis (38–62 %) compared with mild cases (3.5–31 %).


References

McCormack TT, Sims J, Eyre-Brook I et al. Gastric lesions in portal hypertension: inflammatory gastritis or congestive gastropathy. Gut. 1985;26:1226–1232.


image


Fig. 2.23a, b Portal hypertensive gastropathy. a Mild. b Severe.


Portal Hypertensive Gastropathy: Grading According to Tanoue


Aims

The classification was used in a trial that studied the effect of injection sclerotherapy for esophageal varices on portal hypertensive gastropathy.


 
















Grade I


Mild reddening, congestive mucosa, no mosaic-like pattern


Grade II


Severe redness and a fine reticular pattern separating the areas of raised edematous mucosa (mosaic-like pattern) or a fine speckling


Grade III


Point bleeding + grade II


From Tanoue K, Hashizume M, Wada H, Ohta M, Kitano S, Sugimachi K. Effects of endoscopic injection sclerotherapy on portal hypertensive gastropathy: a prospective study. Gastrointest Endosc. 1992;38: 582–585. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

Portal hypertensive gastropathy worsens after endoscopic injection sclerotherapy.


References

Tanoue K, Hashizume M, Wada H, Ohta M, Kitano S, Sugimachi K. Effects of endoscopic injection sclerotherapy on portal hypertensive gastropathy: a prospective study. Gastrointest Endosc. 1992;38:582–585.


Portal Hypertensive Gastropathy: Grading According to the New Italian Endoscopy Club


Aims

To classify elementary endoscopic lesions of portal hypertensive gastropathy, assess their reproducibility, prevalences, sensitivity, and specificity in the diagnosis of cirrhosis of the liver.


 













Grading of the mosaic-like pattern

Mild


Areola uniformly pink


Moderate


Areola has red centre


Severe


Areola uniformly red


 










Grading of portal hypertensive gastropathy

Mild


Presence of mosaic-like pattern of any severity


Severe


Presence of mosaic-like pattern and any of the following: 1. Red marks (red point lesions); or 2. Cherry red spots.


From Primignani M, Carpinelli L, Preatoni P et al. Natural history of portal hypertensive gastropathy in patients with liver cirrhosis. The New Italian Endoscopic Club for the study and treatment of esophageal varices (NIEC). Gastroenterology. 2000;119:181–187. With permission of the American Gastroenterological Association.


Comments

With the New Italian Endoscopic Club classification a sufficient degree of agreement can be achieved in recording portal hypertensive gastropathy.


References

Carpinelli L, Primignani M, Preatoni P et al. Portal hypertensive gastropathy: reproducibility of a classification, prevalence of elementary lesions, sensitivity and specificity in the diagnosis of cirrhosis of the liver. A NIEC multicentre study. New Italian Endoscopic Club. Ital J Gastroenterol Hepatol. 1997;29:533–540.


Primignani M, Carpinelli L, Preatoni P et al. Natural history of portal hypertensive gastropathy in patients with liver cirrhosis. The New Italian Endoscopic Club for the study and treatment of esophageal varices (NIEC). Gastroenterology. 2000;119:181–187.


Portal Hypertensive Gastropathy: Second Baveno Portal Hypertensive Gastropathy Scoring System


Aims

The goals of the Baveno workshops are to develop consensus definitions of key events related to portal hypertension.


 



































Parameter


Score


1. Mucosal mosaic pattern


 


Mild


1


Severe


2


2. Red markings


 


Isolated


1


Confluent


2


3. Gastric antral vascular ectasia


 


Absent


0


Present


2


From de Franchis R. Developing consensus in portal hypertension. J Hepatol. 1996;25:390–394. With permission from The European Association for the Study of the Liver.


Comments

Mild gastropathy = score ≤ 3; severe gastropathy = score ≥ 4. The mosaic pattern consists of small polygonal areas demarcated by a distinct white-to-yellow border, with or without a central bulge. Red marks are flat or slightly bulging red lesions.


References

de Franchis R. Developing consensus in portal hypertension. J Hepatol. 1996;25:390–394.


de Franchls R, ed. Portal Hypertension II. Proceedings of the Second Baveno International Consensus Workshop on Definitions, Methodology and Therapeutic Strategies. Oxford: Blackwell Science; 1996.


Portal Hypertensive Gastropathy: 2-Category and 3-Category Classification System According to Yoo


Aims

To determine the accuracy of a 2- and a 3-category system for the endoscopic classification of portal hypertensive gastropathy.


 










2-Category classification system

Mild


Fine pink speckling (scarlatina-type rash)


Superficial reddening


Mosaic pattern


Severe


Discrete red spots


Diffuse hemorrhagic lesion


 













3-Category classification system

Mild


Mild reddening


Congestive mucosa


Diffuse pink areola


Moderate


Flat red spot in center of a pink areola


Severe redness and a fine reticular pattern separating the areas of raised edematous mucosa


Severe


Diffusely red areola


Pinpoint bleeding


Discrete or confluent red mark lesion


From Yoo HY, Eustace JA, Verma S et al. Accuracy and reliability of the endoscopic classification of portal hypertensive gastropathy. Gastrointest Endosc. 2002;56:675–680. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

There is better inter-observer agreement in the 2-category classification.


References

Yoo HY, Eustace JA, Verma S et al. Accuracy and reliability of the endoscopic classification of portal hypertensive gastropathy. Gastrointest Endosc. 2002;56:675–680.


Portal Hypertensive Gastropathy: Grading According to Sarin


Aims

To grade portal hypertensive gastropathy by upper gastrointestinal endoscopy.


 










Portal hypertensive gastropathy grading according to Sarin

Mild


Presence of discrete cherry red spots with or without mosaic pattern (small elevated erythematous areas outlined by subtle yellowish network) on the gastric mucosa


Severe


Presence of confluent red spots, diffusely distributed in a large portion of the stomach with or without active oozing


From Sarin SK, Sreenivas DV, Lahoti D, Saraya A. Factors influencing development of portal hypertensive gastropathy in patients with portal hypertension. Gastroenterology. 1992;102:994–999. With permission of the American Gastroenterological Association.


Comments

The mucosa of the stomach and the duodenum are inspected carefully especially by bringing the tip of the endoscope close to the mucosa. The precise distribution of portal hypertensive gastropathy in the antrum, corpus, fundus, or whole stomach is recorded.


References

Sarin SK, Sreenivas DV, Lahoti D, Saraya A. Factors influencing development of portal hypertensive gastropathy in patients with portal hypertension. Gastroenterology. 1992;102: 994–999.


Inflammatory Disorders


Drug-Induced (Aspirin, Nonsteroidal Anti-Inflammatory Drug) Mucosal Damage: Lanza Scale (1)


Aims

To define the severity of endoscopically detectable mucosal injury induced by aspirin or NSAID’s.


 



















Endoscopic grading scale of damage

0


Normal


1


One submucosal hemorrhage or superficial ulceration


2


More than one submucosal hemorrhage or superficial ulceration but not numerous or widespread


3


Numerous areas with submucosal hemorrhages or superficial ulcerations


4


Widespread involvement of the stomach with submucosal hemorrhage or superficial ulceration. Invasive ulcer of any size


From Lanza FL, Nelson RS, Rack MF. A controlled endoscopic study comparing the toxic effects of sulindac, naproxen, aspirin, and placebo on the gastric mucosa of healthy volunteers. J Clin Pharmacol. 1984;24:89–95. With permission of Sage Publications.


Comments

An invasive ulcer is defined as a lesion that produces an actual crater, i. e., a depression below the normal plane of the mucosal surface. The Lanza scale is commonly used to evaluate the mucosal injurious potential of drugs. A score of 3 or 4 represents clinically significant injury.


References

Lanza FL, Nelson RS, Rack MF. A controlled endoscopic study comparing the toxic effects of sulindac, naproxen, aspirin, and placebo on the gastric mucosa of healthy volunteers. J Clin Pharmacol. 1984;24:89–95.


Drug-Induced (Aspirin, Nonsteroidal Anti-Inflammatory Drug) Mucosal Damage: Lanza Scale (2)


Aims

To define the severity of endoscopically detectable mucosal injury induced by aspirin or NSAID’s.


 



















Endoscopic grading scale of damage

0


No visible injury (i. e. hemorrhages, erosions, or ulcers)


1


Mucosal hemorrhages only (< 10)


2


10–25 hemorrhages and/or 1 to 5 erosions


3


> 25 hemorrhages and/or 6 to 10 erosions


4


> 10 erosions or an ulcer


From Lanza FL, Codispoti JR, Nelson EB. An endoscopic comparison of gastroduodenal injury with over-the-counter doses of ketoprofen and acetaminophen. Am J Gastroenterol. 1998;93:1051–1054. With permission from the American College of Surgeons.


Comments

An erosion is defined as a definite discontinuation of the mucosa without depth. An ulcer is defined as any lesion of unequivocal depth. The Lanza scale is commonly used to evaluate the mucosal injurious potential of drugs. A score of 3 or 4 represents clinically significant injury.


References

Lanza FL, Codispoti JR, Nelson EB. An endoscopic comparison of gastroduodenal injury with over-the-counter doses of ketoprofen and acetaminophen. Am J Gastroenterol. 1998;93: 1051–1054.


image


Fig. 2.24a–c Drug-induced mucosal damage: Lanza Scale. a Drug-induced erosive gastritis (Lanza Grade 1). b Mild drug-induced hemorrhagic gastritis (Lanza Grade 2). c Extensive drug-induced ulceration along the greater curvature (Lanza Grade 4).


Drug-Induced (Aspirin, NonSteroidal Anti-Inflammatory Drug) Mucosal Damage: Lanza Scale (3)


Aims

To define the severity of endoscopically detectable mucosal injury induced by naproxen, and to determine the effect of different scoring systems.


 



















Rating system emphasizing mucosal erosions

0


Normal stomach


1


Mucosal hemorrhages only


2


One or two erosions


3


Numerous (3–10) areas of erosions


4


Large number of erosions (> 10) or an ulcer


 



















Rating system emphasizing mucosal hemorrhages

0


No visible lesions


1


One area of hemorrhage


2


More than one, but less than 10 areas of hemorrhage


3


10–25 areas of hemorrhage


4


Hemorrhages in most anatomic areas with more than 25 hemorrhages overall


 



















Combined endoscopic scoring system

0


No visible lesions


1


One hemorrhage or erosion (two lesions < 1 cm apart = scored as a single lesion)


2


2–10 hemorrhages or erosions


3


11–25 hemorrhages or erosions


4


More than 25 hemorrhages or erosions, or ulcers of any size


From Lanza FL, Graham DY, Davis RE, Rack MF. Endoscopic comparison of cimetidine and sucralfate for prevention of naproxen-induced acute gastro-duodenal injury: Effect of scoring method. Dig Dis Sci. 1990;35:1494–1499. With kind permission of Springer Science and Business Media.


Comments

Hemorrhages are defined as punctate hemorrhagic lesions that cannot be washed from the mucosa. Erosions and ulcers are defined as white-based mucosal breaks. Erosions are flat and ulcers demonstrate unequivocal depth. The Lanza scale is commonly used to evaluate the mucosal injurious potential of drugs. A score of 3 or 4 represent clinically significant injury.


References

Lanza FL, Graham DY, Davis RE, Rack MF. Endoscopic comparison of cimetidine and sucralfate for prevention of naproxen-induced acute gastro-duodenal injury: Effect of scoring method. Dig Dis Sci. 1990;35:1494–1499.


Drug-Induced (Aspirin, Nonsteroidal Anti-Inflammatory Drug) Mucosal Damage: Upper GI Endoscopy Scoring System According to Simon


Aims

To score upper gastrointestinal mucosal damage in studies comparing COX-2 with nonspecific NSAIDs.


 





























Upper GI endoscopy scoring system

Score


Appearance of gastric and duodenal mucosae


1


Normal


2


1–10 petechiae


3


> 10 petechiae


4


1–5 erosions


5


6–10 erosions


6


11–25 erosions


7


Ulcer


From Simon LS, Lanza FL, Lipsky PE et al. Preliminary study of the safety and efficacy of SC-58635, a novel cyclooxygenase 2 inhibitor: efficacy and safety in two placebo-controlled trials in osteoarthritis and rheumatoid arthritis, and studies of gastrointestinal and platelet effects. Arthritis Rheum. 1998;41:1591–1602. With permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.


Comments

The score is based on the number of petechiae, erosions, and ulcers. An erosion is defined as a lesion producing a definite discontinuance in the mucosa, but without depth. An ulcer is defined as any lesion of any size with unequivocal depth.


References

Simon LS, Lanza FL, Lipsky PE et al. Preliminary study of the safety and efficacy of SC-58635, a novel cyclooxygenase 2 inhibitor: efficacy and safety in two placebo-controlled trials in osteoarthritis and rheumatoid arthritis, and studies of gastrointestinal and platelet effects. Arthritis Rheum. 1998; 41:1591–1602.


Drug-Induced (Aspirin, Nonsteroidal Anti-Inflammatory Drug) Mucosal Damage: Assessing Macroscopic Mucosal Injury According to Cryer


Aims

The aim of this scoring system is to grade macroscopic damage of the gastric mucosa.


 


























Endoscopic scoring to assess macroscopic injury

Score


Definition


0


Normal or mucosal erythema


1


Mucosal hemorrhage or edema, without erosions or ulcers


2


One erosion*, with or without hemorrhage or edema


3


Two to four erosions, with or without hemorrhage or edema


4


Five or more erosions, with or without hemorrhage or edema


5


One or more ulcers#, with or without erosion(s), hemorrhage or edema


*Erosion = mucosal break, not fulfilling definition of ulcer. #Ulcer = ≥ 5 mm mucosal break in largest dimension, with appreciable depth


From Cryer B, Feldman M. Effects of very low dose daily, long-term aspirin therapy on gastric, duodenal, and rectal prostaglandin levels and on mucosal injury in healthy humans. Gastroenterology. 1999;117:17–25. With permission of the American Gastroenterological Association.


Comments

The authors base the scoring on the following premises: (a) hemorrhages and/or edema are abnormal but are less serious than erosions or ulcers; (b) the more erosions, the more injury; and (c) ulcer formation is more serious than even multiple erosions.


References

Cryer B, Feldman M. Effects of very low dose daily, long-term aspirin therapy on gastric, duodenal, and rectal prostaglandin levels and on mucosal injury in healthy humans. Gastroenterology. 1999;117:17–25.


Feldman M, Cryer B, Mallat D, Go MF. Role of Helicobacter pylori infection in gastroduodenal injury and gastric prostaglandin synthesis during long term/low dose aspirin therapy: a prospective placebo-controlled, double-blind randomized trial. Am J Gastroenterology. 2001;96:1751–1757.


Gastritis: The Tarnawski Endoscopic Grading of Alcohol Injury


Aims

The aim is to grade gastric mucosal appearance in patients with alcoholic gastritis.


 


























The Tarnawski endoscopic grading of alcohol injury

Grade


Description


0


Normal mucosa


1


Marked diffuse hyperemia


2


Single hemorrhagic lesion (≤ 2 mm)


3


2–5 hemorrhagic lesions (≤ 2 mm)


4


6–10 hemorrhagic lesions, partially confluent


5


> 10 hemorrhagic lesions, or large area of confluent hemorrhagic lesions


From Tarnawski A, Glick ME, Stachura J, Hollander D, Gergely H. Efficacy of sucralfate and cimetidine in protection of the human gastric mucosa against alcohol injury. Am J Med. 1987;83(Suppl 3B):31–37. With permission from Excerpta Medica Inc.


Comments

The score was used in a study that evaluated the protective effect of cimetidine and sucralfate on alcoholic gastritis.


References

Tarnawski A, Glick ME, Stachura J, Hollander D, Gergely H. Efficacy of sucralfate and cimetidine in protection of the human gastric mucosa against alcohol injury. Am J Med. 1987;83(Suppl 3B):31–37.


Gastritis: Grading and Staging—The Sydney System


Aims

This was the first attempt to grade histologic and endoscopic gastric inflammation.


The system is summarized in Fig. 2.25.


From Tytgat GN. The Sydney System: endoscopic division. Endoscopic appearances in gastritis/duodenitis. J Gastroenterol Hepatol. 1991;6: 223–234. With permission of Blackwell Publishing.


Comments

Commonly used grading system for the evaluation of gastritis.


References

Misiewicz JJ, Tytgat GNJ, Goodwin CS et al. The Sydney system: a new classification of gastritis. In: Working Party Reports, World Congress of Gastroenterology 1990. Sydney: Blackwell Scientific Publications;1990: 1–10.


Tytgat GN. The Sydney System: endoscopic division. Endoscopic appearances in gastritis/duodenitis. J Gastroenterol Hepatol. 1991;6:223–234.


image


Fig. 2.25 Gastritis: grading and staging—the Sydney System. Histological and endoscopic division.


Gastritis: The Updated Sydney System


Aims

To establish an agreed terminology for gastritis, that emphasizes the distinction between the atrophic and nonatrophic stomach.


image


Comments

Including a visual analogue (Fig. 2.26) scale improves inter-observer agreement. Biopsies should be taken from the lesser and the greater curvature of the antrum, within 2–3 cm of the pylorus; from the lesser curvature at approximately 4 cm from the angulus; from the greater curvature at approximately 8 cm from the cardia; and from the incisura angularis.


References

Dixon MF, Genta RM, Yardley JH, Correa P. Classification and grading of gastritis. The updated Sydney system. Am J Surg Pathol. 1996;20:1161–1181.


image


Fig. 2.26 Gastritis: the updated Sydney system. Using the visual analogue scales: The observer should attempt to evaluate one feature at a time. The most prevalent appearance on each side should be matched with the grading panel that resembles it most closely. Observers should keep in mind that these drawings are not intended to represent realistically the histopathologic appearance of the gastric mucosa, rather they provide a schematic representation of the magnitude of each feature and, as such, have certain limitations. Thus, for example, the decreasing thickness of the mucosa usually observed with increasing atrophy is not depicted realistically. Particularly with Helicobacter pylori and neutrophils, there may be a considerable variation of intensity within the same biopsy sample. In such cases, the observer should attempt to average the different areas and score the specimen accordingly.


image


Fig. 2.27 Histologic atrophic gastritis with moderate atrophy.


image


Fig. 2.28 Histologic atrophic gastritis with marked atrophy.


image


Fig. 2.29 Histologic atrophic gastritis with marked intestinal meta-plasia.


Gastritis: Classification of Chronic Gastritis According to RAO


Aims

To minimize inter-observer error by proposing a classification that is purely descriptive.


 














Classification according to RAO

Criteria


 


Chronic inflammatory cell infiltration


Mild


Moderately severe


Severe


Atrophy


Of the glands


Of the mucosa as a whole


From Rao SS, Krasner N, Thomson TJ. Chronic gastritis—a simple classification. J Pathol. 1975;117:93–96. With permission granted by John Wiley & Sons, Ltd on behalf of The Pathological Society.


Comments

This classification is based on two descriptive terms that describe the morphological abnormalities of chronic gastritis namely the degree of infiltration of the whole gastric mucosa by inflammatory cells and the presence or absence of atrophy. To this basic classification metaplasia may be added if present.


References

Rao SS, Krasner N, Thomson TJ. Chronic gastritis—a simple classification. J Pathol. 1975;117:93–96.


Gastritis: Classification of Chronic Gastritis According to Whitehead


Aims

To propose a classification of chronic gastritis, which is applicable to all areas of the gastric mucosa.


image


Comments

The score is based on four biopsy features: (1) the mucosal type; (2) the grade of chronic gastritis; (3) the activity of gastritis; and (4) the presence and type of metaplasia.


References

Whitehead R, Truelove SC, Gear MWL. The histological diagnosis of chronic gastritis in fiberoptic gastroscope biopsy specimens. J Clin Pathol. 1972;25:1–11.


Whitehead R. Simple (non specific) gastritis. In: Mucosal Biopsy of the Gastrointestinal Tract. Saunders, Philadelphia, London, Toronto 1985:33–85.


Gastritis: Scoring of Chronic Gastritis According to Kekki and Siurala


Aims

To define a grading that is based on the loss of glands, regardless of the presence of inflammatory signs or metaplasia.


 























Scoring of chronic gastritis according to Kekki and Siurala

Grade


 


0. Normal mucosa


No round cell infiltration, no loss of glands


1. Superficial gastritis


Round cell infiltration without loss of glands


2. Slight atrophic gastritis


Slight loss of glands


3. Moderate atrophic gastritis


Moderate loss of glands


4. Severe atrophic gastritis


Severe loss of glands


From Kekki M, Siurala M, Varis K, Sipponen P, Sistonen P, Nevanlinna HR. Classification principles and genetics of chronic gastritis. Scand J Gastroenterol. 1987;22(Suppl 141), 1–28. By permission of Taylor & Francis AS.


Comments

The scoring system is simple and equally applicable to both antral and body gastritis.


References

Kekki M, Siurala M, Varis K, Sipponen P, Sistonen P, Nevanlinna HR. Classification principles and genetics of chronic gastritis. Scand J Gastroenterol. 1987;22(Suppl 141):1–28.


Siurala M, Sipponen P, Kekki M. Chronic gastritis: dynamic and clinical aspects. Scan J Gastroenterol. 1985;20(Suppl 109): 69–76.


Gastritis: Morphologic Classification of Chronic Gastritis According to Correa


Aims

The aim of this classification is to define the quality and extent of inflammatory-atrophic changes of the gastric mucosa.


 
























Morphologic classification of chronic gastritis according to Correa

Not atrophic


Superficial (SG)


Band-like infiltrate of lymphocytes and plasma cells occupying the superficial portion of the gastric mucosa, mostly at the gastric pits (foveola) and the necks. Association with spicy food, alcohol, analgesics, and C. pylori.


Diffuse antral (DAG)


A dense infiltrate of lymphocytes and plasma cells occupying full thickness of the antral mucosa. The infiltrate expands the lamina propria and separates the gastric glands. Lymphoid follicles may be prominent. Association with duodenal or pyloric peptic ulcers.


Atrophic


Diffuse corporal (DCG)


A diffuse loss of the oxyntic glands (corpus and fundus). Part of pernicious anemia syndrome.


Multifocal atrophic (MAG)


Correlation with population at risk of stomach cancer. Independent foci of atrophy (gland loss) and mononuclear infiltrate that decreases with intensity as atrophy progresses. Topographic distribution is age-dependant.


From Correa P. Chronic gastritis: a clinico-pathologic classification. Am J Gastroenterol. 1990;2:779–780. With permission of Blackwell Publishing.


Comments

This is a commonly used system to grade gastric inflammation and atrophy. The qualification “acute injury” can be added to any lesion, indicating: polymorphonuclear infiltrate, depletion of cytoplasmic mucus in the foveolar cells, and a mild degree of architectural distortion of the foveolar portion of the mucosa.


References

Correa P. Chronic gastritis: a clinico-pathologic classification. Am J Gastroenterol. 1990;2:779–780.


Gastritis: Endoscopic Classification of Atrophic Gastritis According to Kimura and Takemoto


Aims

To grade endoscopically the extent of atrophy in the stomach.


 































Classification according to Kimura and Takemoto

Type


C–0


No atrophic changes


C–1


Atrophic changes only in the antrum


C–2


Atrophic border on the lesser curvature of the lower portion of the body


C–3


Atrophic border on the lesser curvature of the upper portion of the body


O–1


Atrophic border lies on the lesser curvature and anterior wall of the body


O–2


Atrophic border lies on the anterior wall of the body


O–3


The atrophic region spreads between the anterior wall and the greater curvature of the body


O–p


Pangastritis, atrophic region throughout the entire stomach


image


Fig. 2.30 Atrophic gastritis: classification according to Kimura and Takemoto, closed and open type.


Comments

The atrophic border is the boundary between the pyloric and fundic gland territories. It is recognized endoscopically by the difference in color and height of the mucosa. Biopsies should be taken at the following four sites: (Site 1) the lesser curvature of the mid antrum; (Site 2) the lesser curvature of the angulus; (Site 3) the lesser curvature of the mid body (at a position midway between the angulus and the esophagogastric junction); (Site 4) the greater curvature of the mid body (at a position between the entrance to the antrum and the boundary between the corpus and the fornix).


References

Kimura K, Takemoto T. An endoscopic recognition of the atrophic border and its significance in chronic gastritis. Endoscopy. 1969;3:87–97.


Satoh K, Kimura K, Taniguchi Y et al. Distribution of inflammation and atrophy in the stomach of Helicobacter pylori -positive and -negative patients with chronic gastritis. Am J Gastroenterol. 1996;91:963–969.


Gastritis: Endoscopic Classification of Chronic Gastritis by the Research Society For Gastritis


Aims

To establish a universal, easily applicable grading system.













































































































































Definition of each grade in endoscopic findings

Findings


Grade


Definition


Surface irregularity


1


Fine granular change


 


2


Uniform granular change


 


3


Granular change with various sizes


Rugal hypertrophy


1


Partial


 


2


Entire


 


3


More than 10 mm in width


Visibility of vascular pattern


1


Partial


 


2


Uniform and continuous


 


3


Blood vessels rising to the surface


Intestinal metaplasia


1


Solitary


 


2


Multiple but localized


 


3


Diffuse


Spotty erythema


1


Localized


 


2


Widely scattered


 


3


Widely and densely presented


Patchy erythema


1


Scattered


 


2


Densely presented


 


3


Mixed with fused redness


Linear erythema


1


Intermittent


 


2


Continuous


 


3


Wide with erosion or bleeding


Edema


1


Glossy


 


2


With exudate


 


3


Wide


Flat or depressed erosion


1


Solitary


 


2


Multiple but localized


 


3


Multiple and widely presented


Raised erosion


1


Solitary


 


2


Scattered (< 5 lesions)


 


3


Multiple (> 6 lesions)


Hemorrhage


1


Solitary


 


2


Multiple but localized


 


3


Diffuse


image


Fig. 2.31a–c Surface irregularity. a Grade 1: fine granular change. b Grade 2: uniform granular change. c Grade 3: granular change with various sizes.


image


Fig. 2.32a–c Rugal hyperplasia. a Grade 1: partial. b Grade 2: entire. c Grade 3: > 10 mm wide.


image


Fig. 2.33a–c Visibility of vascular pattern. a Grade 1: partial. b Grade 2: uniform and continuous. c Grade 3: blood vessels rising to the surface.


image


Fig. 2.34a–c Intestinal metaplasia. a Grade 1: solitary. b Grade 2: multiple but localized. c Grade 3: diffuse.


image


Fig. 2.35a–c Spotty erythema. a Grade 1: localized. b Grade 2: widely scattered. c Grade 3: widely and densely presented.


image


Fig. 2.36a–c Patchy erythema. a Grade 1: scattered. b Grade 2: densely presented. c Grade 3: mixed with fused redness.


image


Fig. 2.37a–c Linear erythema. a Grade 1: intermittent. b Grade 2: continuous. c Grade 3: wide with erosion or bleeding.


image


Fig. 2.38a–c Edema. a Grade 1: glossy. b Grade 2: with exudates. c Grade 3: wide.


image


Fig. 2.39a–c Flat or depressed erosion. a Grade 1: solitary. b Grade 2: multiple but localized. c Grade 3: multiple and widely presented.


image


Fig. 2.40a–c Raised erosion. a Grade 1: solitary. b Grade 2: scattered (< 5 lesions). c Grade 3: multiple (> 6 lesions).


image


Fig. 2.41a–c Hemorrhage. a Grade 1: solitary. b Grade 2: multiple but localized. c Grade 3: diffuse.


 













































Fundamental types of endoscopic findings and their diagnostic criteria

Fundamental types


Code number


Definition according to endoscopic findings


Superficial gastritis


1


Findings including edema and redness (spotted, patchy, linear) are observed


Hemorrhagic gastritis


2


Hemorrhage is evidenced


Erosive gastritis


3


Erosive changes including flat or depressed types


Verrucous gastritis


4


Erosive changes including elevated type


Atrophic gastritis


5


Findings such as color change of mucosa, visible vascular pattern, and thinning are observed


Metaplastic gastritis


6


Intestinal metaplasia is noted


Hyperplastic gastritis


7


Remarkable irregularity of mucosa or rugal hypertrophy of greater curvature in corpus


Special gastritis


8


Any types not included in the seven fundamental types, for example, granulomatous gastritis, eosinophilic gastritis, lymphocytic gastritis, Ménétrier disease, and amyloidosis


 




























Histologic classification of gastritis and its diagnostic criteria

Superficial gastritis


Atrophy and inflammation are hardly observed in glands with observation of inflammatory cell infiltration only at the surface of mucosa


Hemorrhagic gastritis


Hemorrhage, hemosiderin sedimentation, hemosiderin phagocytic macrophage are observed


Erosive gastritis


Defect of superficial mucosa is observed, with relevant bioresponse (fibrin precipitation, hemorrhage, edema, neutrophil infitration, and growth of capillary) being evidenced


Verrucous gastritis


This is in the state of hyper-regeneration after erosion, with irregular running of muscle fibers of muscularis mucosae and hyperplasia of pyloric glands surrounded by myofibers in the area of pyloric glands, as well as replacement of pseudopyloric glands and alterations in regeneration of foveolar epithelium


Atrophic gastritis


Atrophy of glands is observed


Metaplastic gastritis


In the biopsy specimens, intestinal metaplastic tubule is observed in more than ⅓ of mucosal tissues


Hypertrophic gastritis


Hypertrophy of glands is observed while foveolar epithelium is almost normal or hypertrophic


From Kaminishi M, Yamaguchi H, Nomura S et al. Endoscopic classification of chronic gastritis based on a pilot study by the research society for gastritis. Digestive Endoscopy. 2002;14:138–151. With permission of Blackwell Publishing.


Comments

The use of five basic types of gastritis is advised: superficial, erosive, verrucous, atrophic, and special types. Metaplastic and hyperplastic types should be excluded from the fundamental types; however, they might be additionally described if they are significant.


References

Kaminishi M, Yamaguchi H, Nomura S et al. Endoscopic classification of chronic gastritis based on a pilot study by the research society for gastritis. Digestive Endoscopy. 2002;14: 138–151.


Intestinal Metaplasia: Classification of Intestinal Metaplasia According to Jass and Filipe


Aims

Intestinal metaplasia is classified into three types based on the histochemical detection of mucins.


 

















Type


Description


I Complete


Straight crypts and regular architecture. The epithelium consists of mature absorptive cells and goblet cells. Goblet cells secrete sialomucin. The absorptive cells are nonsecretory and have well-formed brush borders. Paneth cells are often present.


II Incomplete without sulphomucins


Crypts are elongated and tortuous with mild architectural distortion. The epithelium has few or no absorptive cells, and there are columnar cells in various stages of differentiation. These cells secrete neutral mucin and/or small amounts of sialomucin. Goblet cells secrete sialomucin and occasionally sulphomucins. Paneth cells are rarely present.


III Incomplete with sulphomucins


Distortion of glandular architecture is more pronounced, with cell atypia and loss of differentiation. More marked than in type II. Columnar cells secrete predominantly sulphomucins, and goblet cells contain sialo- and/or sulphomucins. Paneth cells are usually absent.


image


Fig. 2.42a–c Intestinal metaplasia: classification according to Jass and Filipe. a Type I. b Type III.


From Jass JR, Filipe MI. The mucin profiles of normal gastric mucosa, intestinal metaplasia and its variants and gastric carcinoma. Histochem J. 1981;13:931–939. With kind permission of Springer Science and Business Media.


Comments

Small intestinal goblet cells produce sialoglycoproteins that stain with periodic acid–Schiff (PAS) and alcian blue (AB). Colonic goblet cells produce sulphomucin that is detected by AB/high-iron diamine (HID) staining. Progression from type I to type III is proposed during the development of gastric cancer.


References

Filipe IM, Ramachandra S. The histochemistry of intestinal mucins; changes in disease. In: Whitehead R, ed. Gastrointestinal and Oesophageal Pathology, 2nd ed. Edinburgh: Churchill Livingstone; 1995:73–95.


Jass JR, Filipe MI. The mucin profiles of normal gastric mucosa, intestinal metaplasia and its variants and gastric carcinoma. Histochem J. 1981;13:931–939.


Intestinal Metaplasia: Classification of Intestinal Metaplasia According to Matsukura


Aims

The aim of this classification is to characterize intestinal meta-plasia.


 










Complete type


Intestinal metaplasia associated with the intestinal marker enzymes sucrose alpha-D-glucohydrolase, alpha, alpha-trehalase, aminopeptidase (microsomal) (APM), and alkaline phosphatase (ALP). Tissue of this type contains goblet cells and Paneth’s cells, but does not show high-iron diamine (HID)-positive mucin staining with HID-Alcian blue.


Incomplete type


Intestinal metaplasia associated with sucrose alpha-D-glucohydrolase, APM, goblet cells, and HID-positive mucin but not with alpha, alpha-trehalase, ALP, or Paneth’s cells.


From Matsukura N, Suzuki K, Kawachi T et al. Distribution of marker enzymes and mucin in intestinal metaplasia in human stomach and relation to complete and incomplete types of intestinal metaplasia to minute gastric carcinomas. J Natl Cancer Inst. 1980;65:231–240. With permission of Oxford University Press.


Comments

Gastrointestinal metaplasia is divided into complete (small intestinal) and incomplete (colonic) varieties using enzyme expression. Metaplastic tissue that secretes sulphomucins is known to accompany intestinal type gastric cancer.


References

Matsukura N, Suzuki K, Kawachi T et al. Distribution of marker enzymes and mucin in intestinal metaplasia in human stomach and relation to complete and incomplete types of intestinal metaplasia to minute gastric carcinomas. J Natl Cancer Inst. 1980;65:231–240.


Gastric Ulcer: Classification of Benign Gastric Ulcers


Aims

To classify gastric ulcers depending on the distribution in the stomach.


 

















Classification of benign gastric ulcers

Type


 


1


Ulcers are located at the level of the angular notch, or above. This is usually in the area of the antrocorporeal transitional zone, which may rise along the lesser curve as a function of age.


2


Ulcers are in the same location, but occur in association with evidence of actual or prior duodenal ulcer disease.


3


Ulcers are in the prepyloric region, usually within 2.5 cm of the pylorus.


From Johnson HD. Gastric ulcer: classification, blood group characteristics, secretion pattern and pathogenesis. Ann Surg. 1965;162: 996–1004. With permission of Lippincott Williams & Wilkins (LWW).


Comments

Type I ulcers are associated with low or normal acid secretion, whereas types II and III have high acid secretion.


References

Johnson HD. Gastric ulcer: classification, blood group characteristics, secretion pattern and pathogenesis. Ann Surg. 1965;162:996–1004.


Gastric Ulcer: Phases of Gastric Ulcer Healing According to Sakita and Fukutomi


Aims

The aim was to describe the various stages of healing of gastric ulcers.


 

































Phases of gastric ulcer healing according to Sakita and Fukutomi

Phase


 


 


A1


Active stages


Ulcer that contains mucus coating, with marginal elevation because of edema


A2


 


Mucus-coated ulcer with discrete margin and less edema than active stage 1


H1


Healing stages


Unhealed ulcer covered by regenerating epithelium less than 50%, with or without converging folds


H2


 


Ulcer with a mucosal break but almost covered with regenerating epithelium


S1


Scar stages, red scar


Red scar with rough epithelialization without mucosal break


S2


Scar stages, white scar


White scar with complete reepithelialization


image


Fig. 2.43 The various phases of healing of a gastric ulcer (adapted from Sakita and Fukutomi. A1, A2: active stages; H1, H2: healing stages; S1, S2: scar stages (red scar, white scar).


From Lee SY, Kim JJ, Lee JH et al. Healing rate of EMR-induced ulcer in relation to the duration of treatment with omeprazole. Gastrointest Endosc. 2004;60:213–217. With permission from the American Society for Gastrointestinal Endoscopy.


Comments

This system is often used in Japan.


References

Lee SY, Kim JJ, Lee JH et al. Healing rate of EMR-induced ulcer in relation to the duration of treatment with omeprazole. Gastrointest Endosc. 2004;60:213–217.


Sakita T, Fukutomi H. Endoscopy of gastric ulcer. In: Yoshitoshi Y, ed. Peptic Ulcer. Tokyo: Nonkodo; 1971:198–208.


Peptic Ulcer: Postoperative Evaluation According to Visick


Aims

To evaluate patient symptoms after peptic ulcer surgery.


 



















Visick classification

Grade I


No symptoms


Grade II


Mild symptoms relieved by care


Grade III s


Mild symptoms not relieved by care, but satisfactory


Grade III u


Mild symptoms not relieved by care, unsatisfactory


Grade IV


Not improved


From Visick AH. A study of failures after gastrectomy. Ann R Coll Surg Engl. 1948;3:266–284. With permission of The Royal College of Surgeons of England.


Comments

Grades I and II are usually considered a satisfactory outcome. “Relieved by care” is poorly specified.


References

Visick AH. A study of failures after gastrectomy. Ann R Coll Surg Engl. 1948;3:266–284.


Goligher JC, Hill GL, Kenny TE, Nutter E. Proximal gastric vagotomy without drainage for duodenal ulcer—results after 5–8 years. Br J Surg. 1978;3:145–151.


Intussusception: Classification of Jejunogastric Intussusception According to Michael


Aims

 





























Classification of jejunogastric intussusception according to Michael

Type


 


% of cases


1


Afferent limb intussusception


6


2a


Efferent limb intussusception


70–75 combined


2b


Afferent-efferent limb intussusception


 


3


A combination of type 1 and 2


10


4


Intussusception through a Braun side-to-side jejunojejunal anastomosis


 


image


Fig. 2.44 Classification of jejunogastric intussusception according to Michael.


From Michael JW. Jejunogastric intussusception diagnosis and management. J Clin Gastroenterol. 1989;11:452–453. With permission of Lippincott Williams & Wilkins (LWW).


Comments

The jejunogastric intussusception is a rare complication of gastric surgery.


References

Michael JW. Jejunogastric intussusception diagnosis and management. J Clin Gastroenterol. 1989;11:452–453.


Intussusception: Classification of Jejunogastric Intussusception According to Shackmann


Aims

Three anatomic types of jejunogastric intussusception are described.


 





















Classification of jejunogastric intussusception according to Shackman

Type


 


% of cases


1


Antegrade or afferent limb intussusception


10


2


Retrograde or efferent limb intussusception


80


3


A combination of type 1 and 2


10


From Shackman R. Jejunogastric intussusception. Br. J. Surg. 1940;27: 475–480. With permission granted by John Wiley & Sons, Ltd. on behalf of the BJSS, Ltd.


Comments

The jejunogastric intussusception is a rare complication of gastric surgery. Retrograde peristalsis seems to be the cause of type II intussusception.


References

Shackman R. Jejunogastric intussusception. Br. J. Surg. 1940;27:475–480.


Neoplastic Lesions


Gastric Polyps: Morphologic Classification According to the WHO


Aims

To categorize gastric polypoid lesions according to the World Health Organization’s (WHO) histologic classification of gastrointestinal tumors was revised in 1990.


 













Morphologic classification according to the WHO

Neoplasia


a. Epithelial


Intestinal-type adenoma


– Tubular


– Tubulovillous


– Villous


Pyloric gland adenoma


Adenocarcinoma


b. Endocrine


Carcinoid tumor


c. Mesenchymal


Leiomyoma


Neurogenic tumors


– Neurinoma, neurofibroma


Granular cell tumor


Lipoma


Sarcoma


– Neurosarcoma, fibrosarcoma,


– Leiomyosarcoma


d. Lymphatic


Mucosa-associated lymphoid tissue lymphoma


Tumorlike lesion


Fundic gland polyp


Hyperplastic polyp


Inflammatory fibroid polyp


Brunner’s gland heterotopia


Pancreatic heterotopia


Peutz–Jeghers polyp


Cronkhite–Canada polyp


Juvenile polyp


Differential diagnosis


Focal foveolar hyperplasia


Lymphatic follicles


Giant folds


Varioliform gastritis


From Watanabe H, Jass JR, Sobin LH. Histological Typing of Esophageal and Gastric Tumors. Berlin: Springer; 1990. With kind permission of Springer Science and Business Media.


Comments

According to the WHO classification of gastric tumors and polyps the frequency of malignant transformation depends on histologic type.


References

Jass JR, Sobin LH, Watanabe H. The World Health Organization’s histologic classification of gastrointestinal tumors. A commentary on the second edition. Cancer. 1990;66: 2162–2167.


Watanabe H, Jass JR, Sobin LH. Histological Typing of Esophageal and Gastric Tumors. Berlin: Springer; 1990.


Gastric Polyps: Morphologic Classification According to Petras


Aims

To propose a useful classification of gastric polyps.


 










Morphologic classification

Nonneoplastic polyps


A. Possibly inflammatory


1. Hyperplastic polyp (synonyms: regenerative polyp, reparative polyp, polyp type I and II of Nakamura)


2. Inflammatory fibroid polyp


3. Gastric xanthelasma


B. Possibly hamartomatous


1. Peutz–Jeghers polyp


2. Juvenile polyp


3. Cronkhite–Canada polyp


4. Fundic gland polyp (synonyms: retention polyp, cystic polyp, Elster’s glandular cysts)


a. Associated with familial adenomatous polyposis


b. Not associated with familial adenomatous polyposis


C. Heterotopia-pancreatic rest (synonyms: adenomyoma, adenomyomatous hamartoma)


Neoplastic polyps


A. Adenoma with or without associated carcinoma


1. Tubular (usually flat)


2. Tubulovillous


3. Villous


B. Lymphoma


C. Carcinoid


Comments

The most common polyps are fundic gland polyps, hyperplastic polyps, and adenomas.


References

Petras RE. Comments on the Proceedings of the Endoscopy Masters Forum: endoscopy in the precancerous and early-stage cancerous conditions of the gastrointestinal tract. Endoscopy. 1995;27:58–63.


Gastric Dysplasia: The Padova Classification


Aims

To achieve consensus in the classification of gastric dysplasia between the Japanese and the Western system.


 























The Padova classification

Category


Microscopic image


1


Negative for neoplasia/dysplasia


1.0 Normal


1.1 Reactive foveolar hyperplasia


1.2 Intestinal metaplasia (IM)


1.2.1 IM complete type


1.2.2 IM incomplete type


2


Indefinite for dysplasia


2.1 Foveolar proliferation


2.2 Hyperproliferative IM


3


Noninvasive neoplasia [flat or elevated (synonymous with adenoma)]


3.1 Low grade


3.2 High grade


3.2.1 Including suspicious for carcinoma without invasion (intra-glandular)


3.2.2 Including carcinoma without invasion (intra-glandular)


4


Suspicious for invasive carcinoma


5


Invasive adenocarcinoma


From Rugge M, Correa P, Dixon MF et al. The Padova classification. Am J Surg Pathol. 2000;24:167–176. With permission of Lippincott Williams & Wilkins (LWW).


Comments

Categories 1 and 2 are considered benign alterations.


References

Rugge M, Correa P, Dixon MF et al. The Padova classification. Am J Surg Pathol. 2000;24:167–176.


For the grading of gastric neoplasia please also see the Vienna Classification, the modified Vienna classification, and the WHO classification schemes discussed in Chapter 1, Neoplastic Disorders, pages 57–59.


Gastric Cancer: Group Classification of the Japanese Research Society for Gastric Cancer (JRSGC)


Aims

This is a recommendation on reporting gastric biopsies proposed by the Japanese Research Society for Gastric Cancer.


 



















Group I


Normal mucosa and benign lesions with no atypia


Group II


Lesions showing atypia but not diagnosed as benign (nonneoplastic)


Group III


Borderline lesions between benign (nonneoplastic) and malignant lesions


Group IV


Lesions strongly suspected of being carcinoma


Group V


Carcinoma


From Japanese Research Society for Gastric Cancer. Group classification of gastric biopsy specimens. In: Nishi M, Omori Y, Miwa K, eds. Japanese Classification of Gastric Carcinoma. Tokyo: Kanehara; 1995: 73–76. With kind permission of Springer Science and Business Media.


Comments

Influenced by the early histologic studies on gastric neoplasia by Nakamura et al., the JRSGC guidelines propose five groups, ranging from normal mucosa to carcinoma that is not necessarily infiltrating.


References

Japanese Research Society for Gastric Cancer. Group classification of gastric biopsy specimens. In: Nishi M, Omori Y, Miwa K, eds. Japanese Classification of Gastric Carcinoma. Tokyo: Kanehara; 1995:73–76.


Nakamura K, Sugano H, Takagi K, Fuchigami A. Histopathological study on early carcinoma of the stomach: criteria for diagnosis of atypical epithelium. Gann. 1966;57:613–620.


Gastric Cancer: Macroscopic Classification of Early and Advanced Gastric Cancer by the Japanese Research Society for Gastric Cancer


Aims

To classify early and advanced gastric cancer based on endoscopic appearance.


 



















Classification of early gastric cancer

Type 0-I


Protruding type


Type 0-IIa


Superficial elevated type


Type 0-IIb


Flat type


Type 0-IIc


Superficial depressed type


Type 0-III


Excavated type


 



















Classification of advanced gastric cancer

Type 1:


Polypoid tumors, sharply demarcated from the surrounding mucosa, usually attached on a wide base


Type 2:


Ulcerated carcinomas with sharply demarcated and raised margins


Type 3:


Ulcerated carcinomas without definite limits, infiltrating into the surrounding wall


Type 4:


Diffusely infiltrating carcinomas in which ulceration is usually not a marked feature


Type 5:


Nonclassifiable carcinomas that cannot be classified into any of the above types


From Japanese Gastric Cancer Association. Japanese Classification of Gastric Carcinoma. 2nd English ed. Gastric Cancer. 1998;1:10–24. With kind permission of Springer Science and Business Media.


Comments

When there is a combination of types of early gastric cancer, the type that occupies the largest area should be described first. In the case where the lesion has twice the thickness of normal mucosa, it should be typed 0-I. If it has less than twice the thickness of normal mucosa, it should be typed 0-IIa.


References

Japanese Research Society for Gastric Cancer. Japanese Classification of Gastric Carcinoma. 1st English ed. Tokyo: Kanehara; 1995:73–88.


Japanese Gastric Cancer Association. Japanese Classification of Gastric Carcinoma. 2nd English ed. Gastric Cancer. 1998;1: 10–24.


image


Fig. 2.45 Endoscopic classification of early gastric carcinoma. Type I: protruded; Type II: superficial (IIa: elevated; IIb: flat; IIc: depressed); type III: excavated (Japanese Research Society for Gastric Cancer).


Gastric Cancer: Classification of Advanced Gastric Cancer According to Borrmann


Aims

To stage various presentations of advanced gastric cancer.


 




















Borrmann classification

Type


 


I


A polypoid cauliflower-like mass projecting into the gastric lumen, without substantial necrosis or ulceration


II


Ulcerated mass or fungating lesion with sharp margin. Cancerous tissue is present in the periphery and in the center of the ulceration


III


Diffusely infiltrating type, with superimposed ulcerations, lesions often have indistinct margins


IV


Diffuse infiltrating linitis plastica-type malignancy


From Borrmann R. Geschwulste des Magens und Duodenum. In: Henke F, Lubarch O, eds. Handbuch der speziellen pathologischen Anatomie und Histologie. Berlin: Springer Verlag; 1926. With kind permission of Springer Science and Business Media.


Comments

This grading system is still occasionally used.


References

Borrmann R. Geschwulste des Magens und Duodenum. In: Henke F, Lubarch O, eds. Handbuch der speziellen pathologischen Anatomie und Histologie. Berlin: Springer Verlag; 1926.


image


Fig. 2.46 The classification of advanced gastric cancer with four categories, according to Borrmann.


image


Fig. 2.47a–d a Borrmann type I gastric adenocarcinoma. b Borrmann type II lesion. c Borrmann type III lesion, with superimposed ulceration. d Borrmann type IV lesion, with diffusely infiltrating carcinoma.


Staging Gastric Cancer: AJCC TNM Staging System of Gastric Cancer


 































Primary tumor (T)

TX


Primary tumor cannot be assessed


T0


No evidence of primary tumor


Tis


Carcinoma in situ: intra-epithelial tumor without invasion of the lamina propria


T1


Tumor invades lamina propria or submucosa


T2


Tumor invades muscularis propria or submucosa


T2a


Tumor invades muscularis propria


T2b


Tumor invades subserosa


T3


Tumor penetrates serosa (visceral peritoneum) without invasion of adjacent structures


T4


Tumor invades adjacent structures


 



















Regional lymph nodes (N)

NX


Regional lymph nodes cannot be assessed


N0


No regional lymph node metastasis


N1


Metastasis in 1 to 6 regional lymph nodes


N2


Metastasis in 7 to 15 regional lymph nodes


N3


Metastasis in more than 15 regional lymph nodes


 













Distant metastasis (M)

MX


Presence of distant metastasis cannot be assessed


M0


No distant metastasis


M1


Distant metastases


image


 



















Histologic grade (G)

GX


Grade cannot be assessed


G1


Well differentiated


G2


Moderately differentiated


G3


Poorly differentiated


G4


Undifferentiated


 
















Residual tumor (R)

RX


Presence of residual tumor cannot be assessed


R0


No residual tumor


R1


Microscopic residual tumor


R2


Macroscopic residual tumor


From Greene FL, Page DL, Fleming ID et al. AJCC Cancer Staging Manual, 6th ed. New York: Springer, 2002. Used with the permission of the American Joint Committee on Cancer (AJCC), Chicago, Illinois. The original source for this material is the AJCC Cancer Staging Manual, 6th ed (2002) published by Springer-New York, www.springeronline.com.


Comments

The AJCC TNM staging system uses three basic descriptors that are then grouped into stage categories. The first component is “T,” which describes the extent of the primary tumor. The next component is “N,” which describes the absence or presence and extent of regional lymph node metastasis. The third component is “M,” which describes the absence or presence of distant metastasis. The final stage groupings (determined by the different permutations of “T,” “N,” and “M”) range from Stage 0 through Stage IV.


References

Greene FL, Page DL, Fleming ID et al. AJCC Cancer Staging Manual, 6th ed. New York: Springer, 2002. http://www.cancerstaging.org/.


Gastric Cancer: Japanese Staging of Gastric Carcinoma


Aims

To provide a common language for the clinical and pathologic description of gastric carcinoma.


 



















Depth of invasion (T)

T1:


Tumor invasion of mucosa and/or muscularis mucosa (M) or submucosa (SM)


T2:


Tumor invasion of muscularis propria (MP) or subserosa (SS)


T3:


Tumor penetration of serosa (SE)


T4:


Tumor invasion of adjacent structures (SI)


TX:


Unknown


 










































































































Lymph nodes (N)

No. 1


Right paracardial LN


No. 2


Left paracardial LN


No. 3


LN along the lesser curvature


No. 4sa


LN along the short gastric vessels


No. 4sb


LN along the left gastroepiploic vessels


No. 4d


LN along the right gastroepiploic vessels


No. 5


Suprapyloric LN


No. 6


Infrapyloric LN


No. 7


LN along the left gastric artery


No. 8a


LN along the common hepatic artery (anterosuperior group)


No. 8p


LN along the common hepatic artery (posterior group)


No. 9


LN around the celiac artery


No. 10


LN at the splenic hilum


No. 11p


LN along the proximal splenic artery


No. 11d


LN along the distal splenic artery


No. 12a


LN in the hepatoduodenal ligament (along the hepatic artery)


No. 12b


LN in the hepatoduodenal ligament (along the bile duct)


No. 12p


LN in the hepatoduodenal ligament (behind the portal vein)


No. 13


LN on the posterior surface of the pancreatic head


No. 14v


LN along the superior mesenteric vein


No. 14a


LN along the superior mesenteric artery


No. 15


LN along the middle colic vessels


No. 16a1


LN in the aortic hiatus


No. 16a2


LN around the abdominal aorta (from the upper margin of the celiac trunk to the lower margin of the left renal vein)


No. 16b1


LN around the abdominal aorta (from the lower margin of the left renal vein to the upper margin of the inferior mesenteric artery)


No. 16b2


LN around the abdominal aorta (from the upper margin of the inferior mesenteric artery to the aortic bifurcation)


No. 17


LN on the anterior surface of the pancreatic head


No. 18


LN along the inferior margin of the pancreas


No. 19


Infradiaphragmatic LN


No. 20


LN in the esophageal hiatus of the diaphragm


No. 110


Paraesophageal LN in the lower thorax


No. 111


Supradiaphragmatic LN


No. 112


Posterior mediastinal LN


image


Fig. 2.48a–d Japanese staging of gastric carcinoma: lymph node groups.


image


 



















Extent of lymph node metastasis (N)

N0


No evidence of lymph node metastasis


N1


Metastasis to Group 1 lymph nodes, but no metastasis to Groups 2 or 3 lymph nodes


N2


Metastasis to Group 2 lymph nodes, but no metastasis to Group 3 lymph nodes


N3


Metastasis to Group 3 lymph nodes


NX


Unknown


image


 
















Lymph node dissection (D)

D0


No dissection or incomplete dissection of the Group 1 nodes


D1


Dissection of all the Group 1 nodes


D2


Dissection ofall the Group 1 and Group 2 nodes


D3


Dissection of all the Group 1, Group 2, and Group 3 nodes


References

From Japanese Gastric Cancer Association. Japanese Classification of Gastric Carcinoma. 2nd English ed. Gastric Cancer. 1998;1:10-24.With kind permission of Springer Science and Business Media. http://www.jgca.jp/PDFfiles/JCGC-2E.PDF.


Gastric Cancer: The Lauren Classification of Gastric Cancer


Aims

This is a classification of gastric cancer proposed in 1965 by Lauren that divides gastric cancer into either intestinal or diffuse forms.


 

















The Laurén classification of gastric cancer

Type


Description


Intestinal


Differentiated cancer with a tendency to form glands. More likely to involve the distal stomach and to occur in patients with atrophic gastritis. Strong environmental association.


Diffuse


There is little cell cohesion and there is a predilection for extensive submucosal spread and early metastasis. May involve any part of the stomach and has worse prognosis. Has similar frequency in all geographic areas.


Others


 


Comments

The Laurén classification is a useful classification for epidemiological studies. It only roughly stratifies gastric carcinoma into three histopathologic types, and has little prognostic relevance.


References

Laurén P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal type carcinoma. Acta Pathol Microbiol Scand. 1965;64:31-49.


Gastric Cancer: Pathologic Classification According to Ming


Aims

To propose a simple biological classification, which divides gastric carcinoma into two types: the expanding and the infiltrative type.


 









































































































Gastric carcinoma classification according to Ming

Features


Expanding carcinoma


Infiltrative carcinoma


Pathologic features


 


 


1. Cell


 


 


Glandular pattern


Common


Rare


Cell differentiation


Variable


Variable


Cell aggregation


Common


 


Border of aggregate


Distinct


Indistinct


Cell density


Dense


Loose


Goblet cell


Common


Common


Striated border


Common


Rare


2. Mucus


 


 


Acidic mucus


Common


Common


Extra-cellular pool


Occasional


Frequent


3. In situ carcinoma


Common


Rare


4. Stroma


 


 


Collagenous tissue


Moderate


Marked


Lymphocytes, plasmocytes


Common


Rare


5. Surrounding tissue


Compressed


Not compressed


6. Adjacent mucosa


 


 


Intestinal metaplasia


Severe


Mild


Dysplasia


Common


Absent


Clinical features


 


 


1. Sex (male:female)


2:1


1:1


2. Age, under 50 years


6%


14%


3. 5-year survival


27.4%


9.9%


Reproduced from Ming SC. Gastric carcinoma, a pathological classification. Cancer. 1977;39:2475-2485. With permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.


Comments

There is a close correspondence between the classification of Laurén and Ming, where intestinal-type cancer has features of expanding carcinoma and diffuses type cancer resembles infiltrative carcinoma.


References

Ming SC. Gastric carcinoma, a pathological classification. Cancer. 1977;39:2475-2485.


Stemmermann GN, Brown C. A survival study of intestinal and diffuse types of gastric carcinoma. Cancer. 1974;33: 1190-1195.


Gastric Cancer: Goseki Classification of Gastric Carcinoma


Aims

To create a histologic classification of gastric cancer based on the degree of differentiation of the glandular tubules and the amount of mucus in the cytoplasm.


 

























Goseki classification of gastric carcinoma

Type


Tubular differentiation


Mucus in cytoplasm


I


Well


Poor


II


Well


Rich


III


Poor


Poor


IV


Poor


Rich


image


Fig. 2.49 Gastric carcinoma: Classification according to Goseki.


From Goseki N, Takizawa T, Koike M. Differences in the mode of the extension of gastric cancer classified by histological type: new histological classification of gastric carcinoma. Gut. 1992;33:606-612. With permission of BMJ Publishing Group.


Comments

The frequency of hematogenous metastasis is high in group I. In group IV the frequency of lymph node metastasis, direct invasion into surrounding organs, and peritoneal dissemination is higher.


References

Dixon MF, Martin IG, Sue-Ling HM, Wyatt JI, Quirke P, Johnston D. Goseki grading in gastric cancer: comparison with existing systems of grading and its reproducibility. Histopathology. 1994;25:309-316.


Goseki N, Takizawa T, Koike M. Differences in the mode of the extension of gastric cancer classified by histological type: new histological classification of gastric carcinoma. Gut. 1992;33:606-612.


Gastric Cancer: Carneiro Classification


Aims

The aim of this classification is to classify gastric carcinoma based upon histologic type.


Comments

The Carneiro classification recognizes four histologic types: glandular, isolated cells, solid glandular, and mixed glandular type. There are differences in the biological behavior of gland forming and isolated cell gastric carcinomas. Mixed carcinoma has an isolated cell component and another component. Diagnosis may be difficult as the presence of a second component in the tumor occupying 5% of its volume may change the classification. Mixed carcinoma has the worst prognosis.


References

Carneiro F, Seixas M, Sobrinho-Simoes M. New elements for an updated classification of the carcinomas of the stomach. Pathology, Research and Practice. 1995;191:571-584.


Gastric Cancer: WHO Classification of Gastric Carcinoma


Aims

The aim of the classification is to stage gastric cancer according to the dominant histologic abnormality. The WHO classification of gastric carcinoma defines six types: papillary, tubular, mucinous, signet-ring cell, undifferentiated, and others.


Comments

The WHO classification presents histologically well-described tumor categories. However, it is a purely descriptive classification that has very little relationship to prognosis and biological behavior.


References

Fenoglio-Preiser C, Carneiro F, Correa P et al. Gastric carcinoma. In: Hamilton SR, Aaltonen LA, eds. Pathology and Genetics of Tumours of the Digestive System. WHO Classification of Tumours. Lyon: IARC; 2000:39-52.


Carcinoma of the Esophagogastric Junction: The Siewert Classification


Aims

To classify esophagogastric tumors based on the anatomic location of the tumor center.


 
















The Siewert classification

Type I


Adenocarcinoma of the distal esophagus, which usually arises from an area with specialized intestinal metaplasia of the esophagus (i. e., Barrett esophagus) and may infiltrate the esophagogastric junction from above


Type II


True carcinoma of the cardia arising immediately at the esophagogastric junction


Type III


Subcardial gastric carcinoma that infiltrates the esophagogastric junction and distal esophagus from below


From Siewert JR, Stein HJ. Carcinoma of the cardia: carcinoma of the gastroesophageal junction—classification, pathology and extent of resection. Dis Esophagus. 1996;9:173-182. With permission of Blackwell Publishing.


Comments

This classification was approved at a consensus conference of the International Gastric Cancer Association and the International Society for Diseases of the Esophagus in 1997. The authors treat type I tumors as esophageal cancer and type II and III tumors as gastric cancer.


References

Siewert JR, Stein HJ. Carcinoma of the cardia: carcinoma of the gastroesophageal junction—classification, pathology and extent of resection. Dis Esophagus. 1996;9:173-182.


Siewert JR, Stein HJ. Classification of adenocarcinoma of the oesophagogastric junction. Br JSurg. 1998;85:1457-1459.


Gastric MALT Lymphoma: Histologic Grading According to Wotherspoon


Aims

To stage gastric MALT lymphoma.


 

































Histologic scoring of MALT lymphoma

Grade


Description


Histologic features


0


Normal


Scattered plasma cells in lamina propria. No lymphoid follicles.


1


Chronic active gastritis


Small clusters of lymphocytes in lamina propria. No lymphoid follicles. No lymphoepithelial lesions.


2


Chronic active gastritis with florid lymphoid follicle formation


Prominent lymphoid follicles with surrounding mantle zone and plasma cells. No lymphoepithelial lesions.


3


Suspicious lymphoid infiltrate in lamina propria, probably reactive


Lymphoid follicles surrounded by small lymphocytes that infiltrate diffusely in lamina propria and occasionally into epithelium.


4


Suspicious lymphoid infiltrate in lamina propria, probably lymphoma


Lymphoid follicles surrounded by centrocyte-like cells that infiltrate diffusely in lamina propria and into epithelium in small groups.


5


Low-grade B-cell lymphoma of MALT


Presence of dense diffuse infiltrate of centrocyte-like cells in lamina propria with prominent lymphoepithelial lesions.


From Wotherspoon AC, Doglioni C, Diss TC et al. Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet. 1993;342:575–577. With permission of Elsevier.


Comments

Most H. pylori associated low-grade gastric MALT lymphomas, limited to the gastric wall in the absence of regional lymph node involvement, do respond to antimicrobial therapy.


References

Wotherspoon AC, Doglioni C, Diss TC et al. Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori. Lancet. 1993;342:575–577.


Gastric MALT Lymphoma: GELA (Groupe D’Etude des Lymphomes de L’Adulte) Histologic Grading System for Post-Treatment Evaluation


Aims

A post-treatment histologic grading system for gastric MALT lymphoma was established as part of a multicenter clinical study.


image


Comments

The GELA grading is a post-treatment histologic grading based on H&E stained sections of three essential diagnostic features: the lymphoid infiltrate, the presence of lymphoepithelial lesions, and stromal changes.


References

Copie-Bergman C, Gaulard P, Lavergne-Slove A et al. Proposal for a new histological grading system for post-treatment evaluation of gastric MALT lymphoma. Gut. 2003;52:1656.


Gastric MALT Lymphoma: Criteria for Regression According to Neubauer


Aims

The aim is to grade the regression of gastric MALT lymphoma.


 










Criteria for regression according to Neubauer

Complete histologic regression


No remnant lymphoma cells can be detected in the post-treatment biopsy specimens and an “empty” tunica propria with small basal clusters of lymphocytes and scattered plasma cells are found instead


Partial histologic regression


Defined by the presence of post-treatment biopsy samples exhibiting only partial depletion of atypical lymphoid cells from the tunica propria or focal lymphoepithelial destruction


From Neubauer A, Thiede C, Morgner A et al. Cure of Helicobacter pylori infection and duration of remission of low-grade gastric mucosa-associated lymphoid tissue lymphoma. J Natl Cancer Inst. 1997;89:1350–1355. With permission of Oxford University Press.


Comments

The regression of gastric MALT lymphoma after H. pylori eradication is assessed by endoscopic biopsies.


References

Isaacson PG. Gastrointestinal lymphoma. Hum Pathol. 1994;25: 1020–1029.


Neubauer A, Thiede C, Morgner A et al. Cure of Helicobacter pylori infection and duration of remission of low-grade gastric mucosa-associated lymphoid tissue lymphoma. J Natl Cancer Inst. 1997;89:1350–1355.


Gastric Endocrine Cell Classification According to Solcia


Aims

This classification is an attempt to classify hyperplastic and neoplastic endocrine cell changes in the stomach by arranging them in a sequence of morphological lesions with increasing oncological potential.


 

















Classification according to Solcia

Normal pattern


Normal number and distribution of endocrine cells


Hyperplasia


Simple (diffuse) hyperplasia


Linear or chain-forming hyperplasia


Micronodular hyperplasia


Adenomatoid hyperplasia


Dysplasia


Dysplastic (precarcinoid) growth


– Enlarging micronodule


– Fusing micronodules


– Microinvasive lesion


– Nodule with newly formed stroma


Neoplasia


Intra-mucosal neoplasia (intra-mucosal carcinoid)


Invasive neoplasm (invasive carcinoid)


image


Fig. 2.50a–f Gastroendocrine cell classification according to Solcia. a Simple (diffuse) hyperplasia. b Linear or chain-forming hyperplasia. c Micronodular hyperplasia. d Enlarging micronodule. e Intramucosal neoplasm. f Invasive neoplasm.


Comments

As a cut-off point between dysplasia and neoplasia a lesion size of 0.5 mm is chosen.


References

Solcia E, Bordi C, Creutzfeldt W et al. Histopathological classification of nonantral gastric endocrine growth in man. Digestion. 1988;41:185–200.



Small Intestine


Anatomical Variants, Injury


Malrotation of the Duodenum: Classification According to Gravgaard


Aims

The aim of this classification is to type duodenal malrotation according to the anatomical site of the malrotation.


 



































Classification according to Gravgaard

Type


 


A


Normal duodenum


B


Malrotation of the duodenal bulb


C


Malrotation of the superior part of the duodenum


D


Malrotation of the superior duodenal flexure


E


Malrotation of the descending part of the duodenum


F


Malrotation of the inferior duodenal flexure


G


Malrotation of the transverse part of the duodenum


H


Malrotation of the ascending part of the duodenum


I


Malrotation of the transverse and ascending parts of the duodenum


image


Fig. 2.51 Malrotation of the duodenum: classification according to Gravgaard.


From Gravgaard E, Holm Möller S, Andersen D. Malrotation of the duodenum. Frequency in a radiographic control group. Scan J Gastroent. 1977;12:585–588. By permission of Taylor & Francis AS.


Comments

The classification shows various patterns of duodenal malrotation causing luminal narrowing.


References

Gravgaard E, Holm Möller S, Andersen D. Malrotation of the duodenum. Frequency in a radiographic control group. Scan J Gastroent. 1977;12:585–588.


Small Intestine: Classification of Rotatory Anomalies













1


Nonrotation: with a right-sided small intestine and left-sided colon


2


Reverse rotation: when a clockwise 90° rotation leaves the small bowel superficial to the transverse colon, which often passes through the small intestinal mesentery


3


Malrotation: failure to complete the normal rotatory process


image


Fig. 2.52 Small intestine: classification of rotatory anomalies according to Bouchier: nonrotation type 1. Different types of nonfixation and nonrotation: (a) nonrotation; (b) nonrotation of right colon. Both situations allow volvulus.


References

Bouchier IAD, Allan RN, Hodgson HJS, Keighley MRB, eds. In: Gastroenterology. Vol 1. 2nd ed. London: WB Saunders Company; 1993:360.


Classification of Rotatory Anomalies According to Stinger































Type Ia


Nonrotation of the colon and duodenum: the duodenum and jejunum remain to the right of the spine and the colon to the left.


Type IIa


Nonrotation of the duodenum only


Type IIb


The duodenum and colon show reversed rotation


Type IIc


Only reversed rotation of the duodenum. The duodenum passes anterior to the SMA and the large bowel passes in front of both of them


Type IIIa


Both the duodenum and colon are not rotated


Type IIIb


Incomplete fixation of the hepatic flexure


Type IIIc


Incomplete attachment of the cecum


Type IIId


Internal hernia near the ligament of Treitz


Comments

Several types of malrotation are classified according to the embryologic state of development. Type Ia (nonrotation) is estimated to be an incidental finding in 0.2% of adults.


References

Stinger DA. Pediatric Gastrointestinal Imaging. Philadelphia: BC Decker; 1989: 235–239.


Classification of Rotatory Anomalies According to Rescorla and Grosfeld













Nonrotation


Early arrest of rotation of the duodenojejunal loop leaving it in the right side of the abdomen


Incomplete rotation


Arrest of the duodenojejunal loop after it has partially rotated around the superior mesenteric artery but has not ascended to a normal position


From Rescorla FJ, Grosfeld JL. Anomalies of rotation and fixation. Surgery. 1990;108:710–715. With permission of Elsevier.


References

Rescorla FJ, Grosfeld JL. Anomalies of rotation and fixation. Surgery. 1990;108:710–715.


Classification of Rotatory Anomalies According to Gohl and Demeester













Stage I


Omphaloceles caused by failure of the gut to return to the abdomen


Stage II


Nonrotation, malrotation, and reversed rotation


Stage III


Unattached duodenum, mobile cecum, and an unattached small bowel mesentery


From Gohl ML, DeMeester TR. Midgut nonrotation in adults. Am J Surg. 1975;129:319–323. With permission from Excerpta Medica Inc.


Comments

Intestinal anomalies are categorized by the stage of their occurrence.


References

Gohl ML, DeMeester TR. Midgut nonrotation in adults. Am J Surg. 1975;129:319–323.


Intestinal Atresia: Classification According to Bland-Sutton
















Type I


Atresia with a complete occlusion of the intestinal lumen by a diaphragm


Type II


Atresia with complete occlusion of the intestinal lumen terminating into a blind end and joined by a cord-like structure


Type III


Atresia with complete occlusion as in Type II but without any connection between the proximal and distal segments and, in addition, a V-shaped defect in the mesentery


From Bland-Sutton JD. Imperforate ileum. Am J Med Sci. 1889;98:457–462. With permission of Lippincott Williams & Wilkins (LWW).


Comments

Intestinal atresia is typically classified using the 1889 descriptions by Bland-Sutton and the 1964 descriptions by Louw.


References

Bland-Sutton JD. Imperforate ileum. Am J Med Sci. 1889;98: 457–462.


 


Intestinal Atresia: Classification According to Louw






















Type I


Intra-luminal diaphragm with seromuscular continuity


Type II


Cord-like segment between the bowel blind ends


Type IIIa


Atresia with complete separation of blind ends and V-shaped mesenteric defect


Type IIIb


Jejunal atresia with extensive mesenteric defect and distal ileum acquiring its blood supply entirely from a single ileocolic artery. The distal bowel coils itself around the vessel, giving the appearance of an apple peel deformity


Type IV


Multiple atresias of the small intestine


image


Fig. 2.53 Intestinal atresia: classification according to Louw.


From Louw JH, Barnard CN. Congenital intestinal atresia. Observation on its origin The Lancet. 1955;2:1065–1067. With permission of Elsevier.


Comments

Type IIIa is most commonly found.


References

Grosfeld JL, Ballantine TV, Shoemaker R. Operative management of intestinal atresia and stenosis based on pathologic findings. J Pediatr Surg. 1979;14:368–375.


Louw JH, Barnard CN. Congenital intestinal atresia. Observation on its origin. Lancet. 1955;2:1065–1067.


Louw JH. Investigations into the etiology of congenital atresia of the colon. Dis Colon Rectum. 1964;7:471–478.


Mid-Gut Atresia: Types of Intestinal Atresia














(a) Septal


(b) Fibrous cord


(c) Gap with defect in the mesentery


(d) Multiple atresias


(e) Apple peel intestine syndrome


image


Fig. 2.54 Mid-gut atresia: types of intestinal atresia.


From De Lorimier AA, Fonkalsrud EW, Hays DM. Congenital atresia and stenosis of the jejunum and ileum. Surgery. 1969;65:819–827. With permission of Elsevier.


Comments

Anatomic variants of intestinal atresia are described.


References

Blyth H, Dickson JAS. Apple peel syndrome (congenital intestinal atresia). A family study of seven index patients. J Med Genet. 1969;6:275–277.


De Lorimier AA, Fonkalsrud EW, Hays DM. Congenital atresia and stenosis of the jejunum and ileum. Surgery. 1969;65: 819–827.


Intestinal Atresia: Classification According to Martin
















Type I


Membranous obstruction


Type II


Interrupted bowel


Type III


Apple-peel deformity


Type IV


Multiple atresias


From Martin LW, Zerella JT. Jejunoileal atresia: a proposed classification. J Pediatr Surg. 1976;11:399–403. With permission of Elsevier.


Comments

Proposed classification based on a combination of morphology and clinical characteristics.


References

Martin LW, Zerella JT. Jejunoileal atresia: a proposed classification. J Pediatr Surg. 1976;11:399–403.


Injury to the Duodenum: Organ Injury Scaling of the Duodenum


Aims

The aim was to develop a grading system for severity of duodenal injury.


 









































Organ injury scaling of the duodenum

Grade


Injury


Description


I


Hematoma


Involving single portion of the duodenum


 


Laceration


Partial thickness, no perforation


II


Hematoma


Involving more than one portion


 


Laceration


Disruption < 50% of circumference


III


Laceration


Disruption 50%–75% circumference of D2; disruption 50%–100% circumference of D1, D3, D4


IV


Laceration


Disruption > 75% circumference of D2; involving ampulla or distal common bile duct


V


Laceration


Massive disruption of duodenopancreatic complex


 


Vascular


Devascularization of duodenum


From Asensio JA, Feliciano DV, Britt LD, Kerstein MD. Management of duodenal injuries. Curr Probl Surg. 1993;30:1023–1093. With permission of Elsevier.


Comments

The grading should advance one grade for multiple injuries to the same organ.


References

Asensio JA, Feliciano DV, Britt LD, Kerstein MD. Management of duodenal injuries. Curr Probl Surg. 1993;30:1023–1093.


Inflammatory Disorders


Duodenitis: Grading of Duodenitis According to Joffe


Aims

To grade the endoscopic appearance of bulbo duodenal inflammation.


 























Grading of duodenitis according to Joffe

Grade


 


0


Normal mucosa


1


Edematous mucosa


2


Hyperemia of the mucosa


3


Petechiae of the mucosa


4


Erosions of the mucosa


From Joffe SN, Lee FD, Blumgart LH. Duodenitis. Clin Gastroenterol. 1978;7:635–650 (continued as Gastroenterology Clinics of North America). With permission of Elsevier Inc.


Comments

Duodenitis is described as a clinical entity that can give rise to dyspepsia and, on rare occasions, gastrointestinal hemorrhage.


References

Joffe SN, Lee FD, Blumgart LH. Duodenitis. Clin Gastroenterol. 1978;7:635–650.


Perforated Peptic Ulcer Disease: The Boey Classification of Bad Outcome


Aims

To examine operative risk factors for patients with perforated duodenal ulcers.


 










Boey risk factors

Major medical illness


Preoperative shock, systolic blood pressure < 100 mmHg


Longstanding perforation (more than 24 hours)


From Boey J, Wong J, Ong GB. A prospective study of operative risk factors in perforated duodenal ulcers. Ann Surg. 1982;195:265–269. With permission of Lippincott Williams & Wilkins (LWW).


Comments

The scoring system can predict mortality after open and laparoscopic surgery for perforated peptic ulcers. Major medical illness includes severe cardiac disease, severe pulmonary disease, renal failure, diabetes mellitus, and hepatic precoma. The mortality rate increases progressively with increasing numbers of risk factors: 0, 10, 45.5, and 100% in patients with none, one, two, and all three risk factors, respectively.


References

Boey J, Wong J, Ong GB. A prospective study of operative risk factors in perforated duodenal ulcers. Ann Surg. 1982;195: 265–269.


Boey J, Choi SK, Poon A, Alagaratnam TT. Risk stratification in perforated duodenal ulcers. A prospective validation of predictive factors. Ann Surg. 1987;205:22–26.


Coeliac Disease (Gluten Enteropathy): Grading Morphologic Severity According to Marsh


Aims

To grade the histologic severity of the mucosal injury in the proximal small bowel.


 






















Marsh grading

Pre-infiltrative (type 0)


Normal intestinal lesion


Infiltrative (type 1)


Normal mucosal architecture in which the villous epithelium is markedly infiltrated by intra-epithelial lymphocytes


Hyperplastic (type 2)


Similar to type 1, and crypt enlargement


Destructive (type 3)


Flat mucosa


Grade IIIa Partial villous atrophy


Grade IIIb Subtotal villous atrophy


Grade IIIc Total villous atrophy


Hypoplastic (type 4)


Irreversible hypoplastic or atrophic


 






















Grade I


Intra-epithelial lymphocytosis (> 30/100 enterocytes)


Grade II


Intra-epithelial lymphocytosis and crypt hyperplasia


Grade IIIa


Partial villous atrophy


Grade IIIb


Subtotal villous atrophy


Grade IIIc


Total villous atrophy


Grade IV


Irreversible hypoplastic/atrophic changes


From Marsh MN. Gluten major histocompatibility complex and the small intestine: A molecular and immunobiologic approach to the spectrum of gluten-sensitivity (celiac sprue). Gastroenterology. 1992;102:330–354. With permission of the American Gastroenterological Association.


Comments

The Marsh grading system is increasingly used to quantify the morphologic alterations in coeliac disease.


References

Marsh MN. Gluten major histocompatibility complex and the small intestine: A molecular and immunobiologic approach to the spectrum of gluten-sensitivity (celiac sprue). Gastroenterology. 1992;102:330–354.


Wahab PJ, Crusius BA, Meijer JWR, Mulder CJJ. Gluten challenge in borderline gluten-sensitive enteropathy. Am J Gastroenterol. 2001;96:1464–1469.


image


Fig. 2.55a–e Coeliac disease: grading according to Marsh. a Marsh I: Lymphocytic enteritis. b Marsh II: lymphocytic enteritis with crypt hyperplasia. c Marsh IIIA: partial villous atrophy.


image


Fig. 2.55d, e d Marsh IIIB: subtotal villous atrophy. e Marsh IIIC: total villous atrophy.


Coeliac Disease (Gluten Enteropathy): Grading According to Marsh Modified by Oberhuber


Aims

To modify the stages according to Marsh, so that they may be used for diagnostic purposes.


image


 













Grading of villous atrophy according to Oberhuber

Mild villous atrophy


Minor or moderate degrees of shortening and blunting of the villi


Marked villous atrophy


Only short tent-like remainders of the villi are seen


Flat mucosa/total villous atrophy


No more villi are to be recognized and the surface is flat


From Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11:1185–1194. With permission of Lippincott Williams & Wilkins (LWW).


Comments

Grading based on the number of intra-epithelial lymphocytes, crypt hypertrophia, and villous atrophy.


References

Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11:1185–1194.


Coeliac Disease: European Society for Pediatric Gastroenterology and Nutrition (ESPGAN)—Revised Criteria for Diagnosing Coeliac Disease


Aims

The Working Group of the European Society of Paediatric Gastroenterology and Nutrition in 1990 revised the criteria for gluten enteropathy.


 












Characteristic morphologic abnormality of small bowel mucosa while the patient is on a diet containing gluten


While on a gluten-containing diet the presence of serum IgA antibodies to one or more of the following:


– Gliadin (AGA)


– Endomysium (IgA-EMA)


– Tissue transglutaminase (IgA—tTG)


Clinical remission within a few weeks of starting a gluten-free diet


Disappearance of antibodies within a few weeks of starting a gluten-free diet


From Walker-Smith JA, Schmitz J, Shmerling DH, Visakorpi JK. Revised criteria for diagnosis of coeliac disease: report of the Working Group of European Society of Paediatric Gastroenterology and Nutrition. Arch Dis Child. 1990;65:909. With permission of BMJ Publishing Group.


Comments

The criteria include histopathologic alterations in duodenal biopsies, clinical response to gluten withdrawal, and presence of antiendomysial antibodies.


References

Walker-Smith JA, Schmitz J, Shmerling DH, Visakorpi JK. Revised criteria for diagnosis of coeliac disease: report of the Working Group of European Society of Paediatric Gastroenterology and Nutrition. Arch Dis Child. 1990;65:909.


Enterocutaneous Fistulae: Classification of Fistulae According to Schein and Decker


Aims

Postoperative enterocutaneous fistulas are classified based on the origin of the fistula and the daily output.


 


























Type


 


I


Esophageal, gastric, and duodenal fistulae


II


Small bowel


III


Large bowel


IV


All the aforementioned drain through a large abdominal wall defect


High output


> 500 ml/day


Low output


< 500 ml/day


From Schein M, Decker GA. Postoperative external alimentary tract fistulas. Am J Surg. 1991;161:435–438. With permission from Excerpta Medica Inc.


Comments

This classification is based on the Sitges-Serra classification. Type IV has the highest mortality rate (60%), followed by type II (33%), type III (20%) and type I (17%). High output fistulas usually require surgical intervention.


References

Schein M, Decker GA. Postoperative external alimentary tract fistulas. Am J Surg. 1991;161:435–438.


Sitges-Serra A, Jaurrieta E, Sitges-Creus A. Management of postoperative enterocutaneous fistulas: The role of parenteral nutrition and surgery. Br J Surg. 1989;69:147–150.


Enterocutaneous Fistulae: Classification of Fistulae According to Rolstad and Bryant


Aims

Fistulas are classified according to complexity, anatomic location, and physiology.


 













Complexity

Simple fistulas


No organ involvement or associated abscess


Type I complex


Associated with an abscess or multiple organs


Type II complex


Enterostoma within the base of a disrupted wound


image


Fig. 2.56a, b Enterocutaneous fistulae: classification according to Rolstad and Bryant. a Simple enterocutaneous fistula. b Complex type I fistula with associated abscess.


 
















Anatomic location

Type I


Esophageal, gastric, and duodenal


Type II


Small bowel


Type III


Large bowel


Type IV


From large abdominal wall defects greater than 20 cm2


 













Physiology (fistula output)

Low volume


< 200 ml/24 hours


Moderate volume


Between 200 and 500 ml/24 hours


High volume


> 500 ml/24 hours


Comments

High volume fistulas are generally associated with high morbidity, high mortality, and less chance of spontaneous closure.


References

Rolstad BS, Bryant R. Management of drain sites and fistulas. In: Bryant R, ed. Acute and Chronic Wounds. St. Louis: Mosby; 2000:317–341.


Neoplasia


Duodenal Polyposis: Classification of Duodenal Polyposis According to Spiegelman


Aims

To develop a classification system for duodenal polyps based on the number and size of polyps, histology, and degree of dysplasia.


image


 























Stage


Points


0


0


I


1–4


II


5–6


III


7–8


IV


9–12


From Spigelman AD, Williams CB, Talbot IC, Domizio P, Phillips RK. Upper gastrointestinal cancer in patients with familial adenomatous polyposis. Lancet. 1989;2:783–785. With permission of Elsevier.


Comments

This is a commonly used grading of duodenal adenomatosis in FAP.


References

Offerhaus GJ, Entius MM, Giardiello FM. Upper gastrointestinal polyps in familial adenomatous polyposis. Hepatogastroenterology. 1999;46:667–669.


Spigelman AD, Williams CB, Talbot IC, Domizio P, Phillips RK. Upper gastrointestinal cancer in patients with familial adenomatous polyposis. Lancet. 1989;2:783–785.


Small Intestinal Cancer: AJCC TNM Staging System of Small Intestinal Cancer


 

























Primary tumor (T)

TX


Primary tumor cannot be assessed


T0


No evidence of primary tumor


Tis


Carcinoma in situ


T1


Tumor invades lamina propria or submucosa


T2


Tumor invades muscularis propria


T3


Tumor invades through the muscularis propria into the subserosa or into the nonperitonealized perimuscular tissue (mesentery or retroperitoneum) with extension 2 cm or less


T4


Tumor perforates the visceral peritoneum or directly invades other organs or structures (includes other loops of small intestine, mesentery, or retroperitoneum more than 2 cm, and abdominal wall by way of serosa; for duodenum only, invasion of pancreas


 













Regional lymph nodes (N)

NX


Regional lymph nodes cannot be assessed


N0


No regional lymph node metastasis


N1


Regional lymph node metastasis


 













Distant metastasis (M)

MX


Distant metastasis cannot be assessed


M0


No distant metastasis


M1


Distant metastases


image


 



















Histologic grade (G)

GX


Grade cannot be assessed


G1


Well differentiated


G2


Moderately differentiated


G3


Poorly differentiated


G4


Undifferentiated


 
















Residual tumor (R)

RX


Presence of residual tumor cannot be assessed


R0


No residual tumor


R1


Microscopic residual tumor


R2


Macroscopic residual tumor


From Greene FL, Page DL, Fleming ID et al. AJCC Cancer Staging Manual, 6th ed. New York: Springer, 2002. Used with the permission of the American Joint Committee on Cancer (AJCC), Chicago, Illinois. The original source for this material is the AJCC Cancer Staging Manual, 6th ed (2002) published by Springer-New York, www.springeronline.com.


Comments

The AJCC TNM staging system uses three basic descriptors that are then grouped into stage categories. The first component is “T,” which describes the extent of the primary tumor. The next component is “N,” which describes the absence or presence and extent of regional lymph node metastasis. The third component is “M,” which describes the absence or presence of distant metastasis. The final stage groupings (determined by the different permutations of “T,” “N,” and “M”) range from Stage 0 through Stage IV.


References

Greene FL, Page DL, Fleming ID et al. AJCC Cancer Staging Manual, 6th ed. New York: Springer, 2002. http://www.cancerstaging.org/.


Immunoproliferative Small Intestinal Disease: The WHO Memorandum


Aims

To classify the histopathology of immunoproliferative small-intestinal disease.


 













From the WHO memorandum

(a)


Diffuse, dense, compact, and apparently benign lymphoproliferative mucosal infiltration:


– (i) Pure plasmacytic


– (ii) Mixed lymphoplasmacytic


(b)


As in (a), plus circumscribed “immunoblastic” sarcoma (s), in either the intestine and/or mesenteric lymph nodes


(c)


Diffuse “immunoblastic” or plasmocytic sarcoma with or without demonstrable, apparently benign lymphoplasmacytic infiltration


Comments

(a) Atypical plasma cells or large lymphoid cells (immuno-blasts) may be scattered within the lymphoplasmacytic infiltration; (b) and (c) cells resembling Sternberg–Reed cells may be part of the tumor infiltrate.


References

WHO. Alpha-chain disease and related small-intestinal lymphoma: a memorandum. WHO Bull. 1976;54:615–624.


Immunoproliferative Small Intestinal Disease: Staging According to Salem


Aims

To propose a pathologic staging classification for biopsies taken during staging laparotomy.


 






















Staging according to Salem

0


Benign-appearing lymphoplasmacytic mucosal infiltrate (LPI), no evidence of malignancy


I


LPI and malignant lymphoma in either intestine (Ii) or mesenteric lymph nodes (In), but not both


II


LPI and malignant lymphoma in both intestine and mesenteric lymph nodes


III


Involvement of retroperitoneal and/or extra-abdominal lymph nodes


IV


Involvement of noncontiguous nonlymphatic tissues


Unknown


Inadequate staging


Reproduced from Salem PA, Nassar VH, Shahid MJ et al. “Mediterranean abdominal lymphoma” or immunoproliferative small intestinal disease. Part I: clinical aspects. Cancer. 1977;40:2941–2947. With permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.


Comments

Staging laparotomy consists of gross examination of abdominal organs, resection of grossly involved segments of small intestine and multiple resections of uninvolved adjacent segments, splenectomy in the presence of alpha heavy-chain disease or lymphomatous changes on preoperative intestinal biopsies or frozen section, wedge and needle liver biopsies, lymph node resection from splenic pedicle, porta hepatis, mesentery and paraortic region, iliac crest bone marrow biopsy.


Mesenteric lymph nodes may harbor malignant lymphoma even when mucosa reveals a benign-appearing infiltrate only. Stage 0 can be totally reversible when treated with antibiotics.


References

Salem PA, Nassar VH, Shahid MJ et al. “Mediterranean abdominal lymphoma” or immunoproliferative small intestinal disease. Part I: clinical aspects. Cancer. 1977;40: 2941–2947.


Immunoproliferative Small Intestinal Disease (IPSID)


Aims

To achieve increased knowledge of alpha-chain disease and to create a practical staging system on which therapeutic schemes can be based.


 

















Stage


 


A


A mature plasmacytic or lymphocytoplastic infiltrate invades the mucosal lamina propria and is sometimes associated with a variable amount of villous atrophy


B


Intermediate. The infiltrate invades the submucosa


C


Immunoblastic lymphoma either forming discrete ulcerated tumors or extensively infiltrating long segments of the intestinal wall


Reproduced from Galian A, Lecestre MJ, Scotto Y, Bognel C, Matuchansky C, Rambaud JC. Pathological study of alpha-chain disease with special emphasis on evolution. Cancer. 1977;39:2081–101. With permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc.


Comments

Stage A is characterized by a diffuse, dense, and apparently benign plasmacytic infiltration of mucosa and/or lymph nodes, and Stage C by the presence of low or high grade lymphomas with or without benign lymphoplasmacytic infiltration. Stage B is intermediate between Stages A and C and is characterized by mixed mature and dystrophic plasma cells infiltrating the mucosa and villous atrophy.


Complete and prolonged remission has been seen in stage A only, sometimes with oral antibiotics alone.


References

Fermand JP, Brouet JC. Heavy-chain diseases. Hematol Oncol Clin North Am. 1999;13:1281–1294.


Galian A, Lecestre MJ, Scotto Y, Bognel C, Matuchansky C, Rambaud JC. Pathological study of alpha-chain disease with special emphasis on evolution. Cancer. 1977;39:2081–101.


May 30, 2016 | Posted by in GASTROENTEROLOGY | Comments Off on Organ-Related Staging and Grading

Full access? Get Clinical Tree

Get Clinical Tree app for offline access