Background.—Under the auspices of the College of American Pathologists, the current state of knowledge regarding pathologic prognostic factors (factors linked to outcome) and predictive factors (factors predicting response to therapy) in colorectal carcinoma was evaluated. A multidisciplinary group of clinical (including the disciplines of medical oncology, surgical oncology, and radiation oncology), pathologic, and statistical experts in colorectal cancer reviewed all relevant medical literature and stratified the reported prognostic factors into categories that reflected the strength of the published evidence demonstrating their prognostic value. Accordingly, the following categories of prognostic factors were defined. Category I includes factors definitively proven to be of prognostic import based on evidence from multiple statistically robust published trials and generally used in patient management. Category IIA includes factors extensively studied biologically and/or clinically and repeatedly shown to have prognostic value for outcome and/or predictive value for therapy that is of sufficient import to be included in the pathology report but that remains to be validated in statistically robust studies. Category IIB includes factors shown to be promising in multiple studies but lacking sufficient data for inclusion in category I or IIA. Category III includes factors not yet sufficiently studied to determine their prognostic value. Category IV includes factors well studied and shown to have no prognostic significance.

Materials and Methods.—The medical literature was critically reviewed, and the analysis revealed specific points of variability in approach that prevented direct comparisons among published studies and compromised the quality of the collective data. Categories of variability recognized included the following: (1) methods of analysis, (2) interpretation of findings, (3) reporting of data, and (4) statistical evaluation. Additional points of variability within these categories were defined from the collective experience of the group. Reasons for the assignment of an individual prognostic factor to category I, II, III, or IV (categories defined by the level of scientific validation) were outlined with reference to the specific types of variability associated with the supportive data. For each factor and category of variability related to that factor, detailed recommendations for improvement were made. The recommendations were based on the following aims: (1) to increase the uniformity and completeness of pathologic evaluation of tumor specimens, (2) to enhance the quality of the data needed for definitive evaluation of the prognostic value of individual prognostic factors, and (3) ultimately, to improve patient care.

Results and Conclusions.—Factors that were determined to merit inclusion in category I were as follows: the local extent of tumor assessed pathologically (the pT category of the TNM staging system of the American Joint Committee on Cancer and the Union Internationale Contre le Cancer [AJCC/UICC]); regional lymph node metastasis (the pN category of the TNM staging system); blood or lymphatic vessel invasion; residual tumor following surgery with curative intent (the R classification of the AJCC/UICC staging system), especially as it relates to positive surgical margins; and preoperative elevation of carcinoembryonic antigen elevation (a factor established by laboratory medicine methods rather than anatomic pathology). Factors in category IIA included the following: tumor grade, radial margin status (for resection specimens with nonperitonealized surfaces), and residual tumor in the resection specimen following neoadjuvant therapy (the ypTNM category of the TNM staging system of the AJCC/UICC). Factors in category IIB included the following: histologic type, histologic features associated with microsatellite instability (MSI) (ie, host lymphoid response to tumor and medullary or mucinous histologic type), high degree of MSI (MSI-H), loss of heterozygosity at 18q (DCC gene allelic loss), and tumor border configuration (infiltrating vs pushing border). Factors grouped in category III included the following: DNA content, all other molecular markers except loss of heterozygosity 18q/DCC and MSI-H, perineural invasion, microvessel density, tumor cell–associated proteins or carbohydrates, peritumoral fibrosis, peritumoral inflammatory response, focal neuroendocrine differentiation, nuclear organizing regions, and proliferation indices. Category IV factors included tumor size and gross tumor configuration. This report records findings and recommendations of the consensus conference group, organized according to structural guidelines defined herein.

Local Extent of Tumor Assessed Pathologically (pT Category)

Overview.—Despite universal recognition of the prognostic importance of the local extent of disease as determined by pathologic assessment,1 variations in approach to the acquisition, interpretation, reporting, and analysis of this vital information still exist on a fundamental level. Lack of uniformity in methodologic approach and variations in the interpretation and reporting of pathologic findings are currently the most problematic issues associated with this factor.1–6 

Method Issues

  • Specimen processing variation

    Processing fresh versus fixed tissue from specimen

    Fixing specimen closed or opened

    Opening of specimen along long axis versus across short axis

    Fixation time before sampling for microscopic evaluation

    Pinning techniques versus no pinning

    Inking versus no inking (if inked, which surfaces marked)

    Type of fixative used

    Number of blocks submitted

  • Variability in handling of cases with nonperitonealized surfaces (radial) margins (discussed separately in category IIA variables)

Recommendation.—For standardization purposes, inking of radial margins should be carried out. Tissue should be fixed in 10% buffered formalin before processing. The duration of fixation, open versus closed fixation, and pinned versus unpinned should be at the pathologists discretion. Overfixation should be avoided, however (see p 985). At least 3 blocks of tumor should be submitted (5 blocks may be submitted to optimize identification of extramural venous invasion; see below), or the entire tumor should be submitted if it is less than 3 blocks, taken perpendicular to the bowel wall and cut transversely to demonstrate deepest extent of tumor and tumor border configuration.

Interpretation Issues

  • pTis: variability in use of the term carcinoma in situ. Used descriptively (and traditionally), the term connotes malignant epithelial cells that do not penetrate their basement membrane and do not invade the underlying stroma. Used as a staging term in colorectal cancer, the term also includes malignant cells that invade the lamina propria up to and including the muscularis mucosae.

  • pT4: confusion regarding the definition of serosal perforation and the microscopic features by which it is recognized. Breach of the serosal surface (serosal involvement by tumor) may have variable histopathologic manifestations, many of which are not interpreted by pathologists as serosal perforation, leading to an underestimation of pT4b disease.

  • pT3 with positive radial margin versus pT4b: confusion about evaluation of peritonealized versus nonperitonealized surfaces of the specimen.

Recommendation.—For pTis (carcinoma in situ): specify as either (1) high-grade dysplasia/intraepithelial carcinoma (pTie) or (2) intramucosal carcinoma (pTim). Clarification of the definition for pT3 is required to indicate that the serosal surface is to be uninvolved by tumor.5 Clarification of category pT4b (serosal perforation) is needed, and its definition should include disruption of the serosal (mesothelial) cells on the bowel surface. This disruption may include the following: (1) mesothelial inflammatory and/or hyperplastic reactions with tumor close to but not at the serosal surface; (2) tumor present at the serosal surface with inflammatory reaction, mesothelial hyperplasia, and/or erosion or ulceration; and (3) free tumor cells on the serosal surface (in the peritoneum) with underlying ulceration of the visceral peritoneum.5 

Reporting Issues

  • Variability in staging system and terms used (eg, Dukes' or Astler-Coller staging systems used instead of TNM system)

  • Variable reporting of pT4 tumors as pT4a versus pT4b

Recommendation.—Use of terms and definitions of the T categories set forth by the American Joint Committee on Cancer and the Union Internationale Contre le Cancer (AJCC/UICC) in the 1997 AJCC/UICC Cancer Staging Manual6 and the 1993 TNM Supplement.7 

Statistical Issues

  • Considerable variation in staging system used and in the use of pathologic stage data in prognostic marker studies

Recommendation.—Report pT, pN, and pM categories in all cases. Each surgical margin (proximal, distal, and radial) should be reported separately.

Other

  • Obtaining fresh tissue or additional fixed tissue for research while maintaining integrity of surgical pathology evaluation

Recommendation.—There are currently insufficient data to make recommendations, but archiving of additional tumor for molecular studies may be advisable.

Regional Lymph Node Metastasis Assessed Pathologically (pN Category)

Overview.—Metastasis to regional lymph nodes as determined by pathologic assessment is, among the factors that most strongly predict outcome following surgical resection, second only to distant metastatic disease in importance. Nevertheless, significant methodologic variation still exists in routine pathology practice with regard to both lymph node harvesting and processing of lymph nodes for microscopic examination. Lack of uniformity in approach is currently the most problematic issue associated with this factor. Newer (nontraditional) methods of lymph node examination for micrometastatic disease and the biologic significance of metastasis identified by these methods currently lack validation.8–20 

Method Issues

  • Variations in surgical technique contributing to variation in number of nodes contained in resection specimens

  • Variations in handling of specimen (using conventional techniques)Diligence of search for nodes

    Use of clearing or other solutions to increase macroscopic visualization of nodes

    Threshold for acceptable number of nodes

    Submission of whole versus half of each node found for microscopic examination

    Acquisition of tissue levels (and in number of levels, if acquired) for microscopic examination

    Separation of lymph nodes by anatomic site in large specimens (ie, regional vs nonregional as pertains to the anatomic site of the tumor)

  • Use of special techniques as adjuncts to or replacement for light microscopy (nonhistologic or nonconventional techniques)

    Immunohistochemical staining: cytokeratin, carcinoembryonic antigen (CEA), epithelial membrane antigen

    Polymerase chain reaction amplification of tumor RNA and DNA—considerable variation in method and control comparisons in investigational studies

Recommendation.—All identified lymph nodes should be sectioned. It has been shown that 12 to 15 negative lymph nodes predict for regional node negativity.9, 19 If fewer than 12 nodes are found, additional visual enhancement techniques should be considered. All grossly negative or equivocal lymph nodes are to be submitted entirely. For grossly positive lymph nodes, a representative sample should be submitted for microscopic confirmation. Data are insufficient to recommend routine use of tissue levels or ancillary special techniques.

Interpretation Issues

  • Variation in lower limit of acceptable nodal harvest

  • Failure to interpret tumor directly invading node as metastatic disease

  • Failure to recognize nonregional lymph node metastasis as pM1 disease

  • Variable interpretation of micrometastasis by light microscopy

  • Variable interpretation of minute foci of tumor (including single cells) or tumor detected by nonhistologic or nonconventional methods as biologically significant

Recommendation.—Use guidelines for definitions of nodal metastasis given in the 1997 TNM Cancer Staging Manual.6 Any histologically confirmed focus of tumor that measures 2 mm or less in greatest dimension is to be regarded as a micrometastasis and classified as N1. Tumor detected by nonhistologic methods is classified as pN0.20 

Reporting Issues

  • Variability in reporting of regional lymph node status (pN missing from many reports)

  • Variability in assignment of a pN category by the pathologist

Recommendation.—Regional lymph node status (both numbers of nodes examined and number of nodes positive) always should be reported and always assigned a pN category by the pathologist.

Statistical Issues

  • Various methods for analyzing nodal data: categorical, continuous, percentage positive

Recommendation.—There are currently insufficient data to make recommendations.

Blood or Lymphatic Vessel Invasion

Overview.—The prognostic importance of involvement of small (thin-walled, presumably lymphatic) vessels in the submucosa has been well documented with respect to polypectomies of malignant polyps and shown to be associated with an increased risk of regional lymph node metastasis. The prognostic importance of involvement of extramural veins by tumor and its association with increased risk of liver metastasis has also been demonstrated. Despite recognition of the importance of blood or lymphatic vessel involvement by tumor, considerable heterogeneity exists in the methodologic approach to, assessment of, and reporting of this feature.1,3,8,21–57 

Method Issues

  1. Malignant polyps or local excisions (pT1 tumors)

    • Variable number of tissue levels examined

    • Variable use of special stains or immunohistochemical staining to visualize vessels

  2. pT2, pT3, and pT4 tumors

    • Variable sectioning of specimens and number of samples submitted

      Data demonstrating increased likelihood of finding venous invasion with submission of additional sections suggest that 5 blocks of tumor may be optimal8

      Variable estimates of cost-effectiveness of more extensive examination

      Practical feasibility of more extensive examination

    • Variable number of tissue levels examined

    • Variability in use of special stains or immunohistochemical staining to visualize vessels

Recommendation.—At least 3 blocks of tumor (optimally 5 or more blocks) should be submitted. A single hematoxylin-eosin–stained section from each block should be examined for blood or lymphatic vessel invasion; data are insufficient to recommend that additional tissue levels be examined. No special stains or immunohistochemical stains are recommended.

Interpretation Issues

  • Differentiation of postcapillary venules from lymphatic vessels (both thin-walled, small-caliber vessels) often not possible, but these vessels variably definitively diagnosed

  • Malignant polyps: interobserver variability in diagnosis of small vessel invasion (impact of retraction or cautery artifact on interpretation)

  • The importance of suspicion of small vessel invasion in a malignant polyp variably recognized (outcome resembles that of diagnostic small vessel invasion)

  • Mural penetration of tumor variably interpreted as large vessel invasion

Recommendation.—Identification of tumor within an endothelial-lined channel or surrounded by an elastic lamina is required for diagnosis of vessel invasion. Small vessels not definitively interpreted as lymphatics or venules should be identified as angiolymphatic vessels.

Reporting Issues

  • Variability in reporting of small vessel invasion

  • Variability in reporting of large vessel invasion

  • Variability in reporting anatomic location of small vessel invasion (eg, submucosal, mural, extramural)

  • Variability in reporting anatomic location of large vessel invasion (eg, submucosal, mural, extramural)

Recommendation.—For all tumors, including malignant polyps and rectal tumors removed by transanal disk excision, venous and angiolymphatic invasion should always be reported as present or absent and its anatomic location specified as intramural or extramural.

Statistical Issues

  • Considerable variation in types of vascular invasion data in prognostic marker studies (eg, lymphatic only, venous only, both lumped together, separation by anatomic location, or any vessel invasion without specification of type or site)—unclear whether vessel type, vessel location, or both are prognostically significant

Recommendation.—In prognostic marker studies, large vessel and small vessel invasion should be designated separately. Anatomic site within the bowel wall should be considered a separate variable.

Residual Tumor Classification (R Classification)

Overview.—The residual tumor (R) classification has been shown to have prognostic significance. The following discussion is included for anatomic pathologists for educational purposes and for its relationship, in some circumstances, to a positive surgical margin. The presumption underlying the finding of tumor at a surgical resection margin is that tumor remains in the patient at the surgical interface, and based on this premise, classification of a positive margin as residual tumor (R) is appropriate.1,6 

Method Issues

  • Lack of understanding of appropriate usage of R classification for tumor remaining in patient following therapy of any type

  • Variable inappropriate use of the R category to refer to residual tumor in the resection specimen after neoadjuvant therapy

  • Variable use of R category to refer to residual tumor in the patient after incomplete resection (eg, a positive radial margin)

  • Variable inappropriate use of ypTNM to modify the R category

  • Lack of guidelines for the appropriate use of ypTNM

Recommendation.—Tumor at a surgical resection margin should be considered the counterpart of residual tumor in the patient and classified according to the R classification as defined in the 1997 AJCC/UICC Cancer Staging Manual.6 Further definition of the residual tumor category is needed to distinguish residual tumor in the patient following treatment versus residual tumor in a resection specimen following neoadjuvant treatment (see “Tumor Classification After Neoadjuvant Therapy (ypTNM)” in the “Category IIA” section).

Interpretation Issues

  • Positive margins (including radial) may or may not be interpreted as evidence of residual disease in the patient—role of pathologist versus surgeon in defining residual disease in incomplete resections

Recommendation.—Positive margins should be interpreted as the counterpart of residual tumor in the patient unless proven otherwise.

Reporting Issues

  • Reporting of surgical margin status alone versus surgical margin status plus corresponding R classification

Recommendation.—Surgical margin status should always be reported. If positive, the appropriate R category (R1 indicates microscopic residual disease; R2 indicates macroscopic residual disease) should be assigned.

Statistical Issues

  • Few data on the relevance of the R category as it relates to radial margins due primarily to the lack of recognition, reporting, and studies on prognostic significance of radial margins, except in rectal cancer

Recommendation.—All studies on prognostic importance of residual disease as it relates to margin status (including the radial margin) should also include the R classification.

Preoperative CEA Elevation

Overview.—Preoperative CEA has been shown to have prognostic significance. The following discussion is included for anatomic pathologists for educational purposes only. Anatomic pathologists rarely know if testing is performed or, if performed, what the results are.58–66 

Method Issues

  • Variation in laboratory measurement methods

  • Variation in preoperative testing for CEA according to treating physician

Recommendation.—Standard laboratory testing of a preoperative serum sample should be performed for all patients.

Interpretation Issues

  • Variation in level of elevation that is regarded as significant

Recommendation.—Significant level of elevation is greater than 5 ng/mL. (This should not be taken to preclude the individual laboratory's right to determine local normal ranges for this analyte.)

Reporting Issues

  • Ordering physician may or may not report results as part of tumor staging

Recommendation.—Preoperative CEA levels, if known, should be reported as a clinical or (clinical) pathologic parameter as follows: CX, CEA level not assessed; C0, CEA level not elevated (<5 ng/mL); or C1, CEA level elevated (≥5 ng/mL).

Statistical Issues

  • Use of different cutoffs for elevation (eg, anything greater than hospital norm, 5 ng/mL, 7.5 ng/mL, 10 ng/mL) in analyses demonstrating the significance of CEA

  • Variable adjustment for preoperative treatment

Recommendation.—In prognostic marker studies, preoperative CEA levels should be reported as elevated if 5 ng/mL or greater and handled as a separate element in multivariate analyses.

Histologic Grade

Method Issues

  • Multiple grading systems suggested during the past several decades but none widely accepted

  • Variation in number of strata in different grading systems

  • Assessment of grade largely subjective overall

  • Semiquantitative grading suggested by the College of American Pathologists despite lack of data to justify use

  • Prognostic significance of grade demonstrated in most studies by collapsing 4 into 2 grades as follows: well and moderately (grades 1 and 2) defined as low grade, and poorly and undifferentiated (grades 3 and 4) defined as high grade

Recommendation.—A 2-tiered grading system (high grade and low grade) would both reduce interobserver variation and retain or improve prognostic significance.1–3,8,9,62,65,68–80 

Interpretation Issues

  • Determination of grade largely a subjective exercise with few or no defined criteria

  • Substantial interobserver variability demonstrated8 

  • Fundamental basis of grade controversial:

    Overall impression

    Worst area

    Amount of gland formation alone

    Combination of gland formation and other structural or cytologic features (eg, nuclear grade)

  • Relationship between grade, DNA replication error, and high degree of microsatellite instability (MSI-H) status may be the most important issue in high-grade tumors

    For medullary tumors, problem may be obviated if these are defined by the World Health Organization (WHO) as a separate histologic type (WHO classification now under revision) and not assigned a grade by convention

    For mucinous tumors, grade assignment may be inappropriate

Recommendations.—Gland formation should be the only feature used to assign grade (<50% gland formation defines high grade).

Reporting Issues

  • Variation in wording used in surgical pathology reports

  • Variable use of numerical versus descriptive grade

  • Variable reporting of single grade for entire tumor versus range of grades within a single tumor

  • Number of categories from 2 to 4 often goes unstated

  • Variable or inappropriate assignment of grade to histologic types that should not be graded or are always assigned a specific grade by convention

Recommendation.—Report grade in a 2-tiered descriptive system as either high grade or low grade.

Statistical Issues

  • Variable practice of grouping of grades to reduce the number of categories

Recommendation.—Use 2 categories to include well- and moderately differentiated tumors in a low-grade category and poorly and undifferentiated tumors in a high-grade category for all statistical analyses.

Radial Margin (Specimens With Nonperitonealized Surfaces)

Method Issues81–84 

  • Lack of understanding of definition, importance, and need for separate analyses of radial margins by both pathologists and surgeons

  • Open or closed fixation at the discretion of the pathologist, but closed fixation and gross serial sectioning through the bowel used in most studies demonstrating the importance of radial margins

  • Variable use of ink to mark radial margin

Recommendation.—Radial margin status (positive or negative) and, if negative, surgical clearance (the distance between the tumor and the radial margin at its closest approach) should be a standard part of assessment for all specimens with nonperitonealized surfaces. Grossly identified radial margins should be inked. Open or closed fixation is at the discretion of the pathologist.

Interpretation Issues

  • Peritonealized surface misinterpreted as a radial margin when mesothelial cells are missing

  • Variability in the consideration of specimen-specific anatomical issues, with all external surfaces variably treated as peritonealized surfaces and involvement variably misinterpreted as pT4b

Recommendation.—As recommended by the Report from the National Cancer Institute Colorectal Cancer Surgery Guidelines Conference (April 1–2, 1999), the surgeon should be aware of the radial margins at the time of operation. Labeling the specimen and marking areas of concern so the specimen can be properly oriented and areas of specific concern can be correctly identified by the pathologist should be considered in every case and performed when appropriate. For the pathologist, careful gross assessment to identify the location of the peritoneal reflection should be performed in the fresh state, if possible, on specimens with both peritonealized and nonperitonealized surfaces. The nonperitonealized surface should be marked with ink and reported separately. For rectal specimens that lack a peritonealized surface, the entire external surface of the specimen is a radial margin.

Reporting Issues

  • Variable lack of reporting of positive radial margins by pathologists

  • If positive radial margin reported, variable reporting of anatomic location of involvement

  • If radial margin assessed and reported to be negative, variable reporting of the surgical clearance (the distance between the tumor and the radial margin at its closest approach)

Recommendation.—Radial margin status (positive or negative) and surgical clearance in all cases with negative radial margins should be a standard part of reporting for all specimens with nonperitonealized surfaces. Whenever orientation of the specimen is possible, the anatomic location of the positive radial margin should be reported. Positive radial margins should be classified by the R classification, which connotes residual disease in the patient (see above).

Statistical Issues

  • Current limitation of studies on the importance of radial margins are limited to rectal cancers; no data at all on radial margins in resection specimens with partially peritonealized surfaces

  • Among current studies, numbers of patients relatively small; no statistically robust studies with multivariate analysis

Recommendation.—All studies on prognostic factors should include assessment of radial margin and surgical clearance (see below).

Tumor Classification After Neoadjuvant Therapy (ypTNM)

Method Issues1,6,7 

  • Lack of understanding of restriction of use of cTNM or pTNM to previously untreated tumors

  • Lack of understanding that the prognostic significance of a ypTNM stage grouping cannot be equated with the prognostic significance of a p/cTNM stage grouping (based on previously untreated tumor)

  • Variable use of the ypTNM category to refer to residual tumor in resection specimen after neoadjuvant therapy

Recommendation.—Tumor remaining in a resection specimen following neoadjuvant therapy should always be classified by ypTNM to distinguish it from untreated tumor.

Interpretation Issues

  • Viable versus necrotic tumor in determining residual disease

  • Acellular mucin pools or other probable “footprints” of tumor interpreted as evidence of residual tumor

Recommendations.—Only histologically viable tumor should be interpreted as residual disease and classified by ypTNM.

Reporting Issues

  • Variable reporting of uncertainty about viability of residual tumor

  • ypTNM variably assigned and reported

Recommendation.—Histologically viable tumor remaining in a resection specimen following neoadjuvant therapy should always be classified by ypTNM to distinguish it from untreated tumor are reported using this classification.

Statistical Issues

  • Limited number and size of studies on the importance of residual disease in the resection specimen after neoadjuvant therapy

  • Importance of level of precision in assessment of residual disease (ie, assessment by ypTNM) unknown

Recommendation.—All studies on the prognostic importance of residual disease in resection specimens following neoadjuvant therapy should include ypTNM.

Histologic Type

Method Issues3,38,39,46,47,51,70,72,78,85–96 

  • Variable use of routine histology alone versus use of special studies (eg, assessment of neuroendocrine differentiation in small cell carcinomas by immunostaining)

Recommendation.—Perform assessment of tumor type by routine histologic method alone. Special studies may be used at the discretion of the pathologist.

Interpretation Issues

  • Mixed patterns variably interpreted (eg, signet ring cells in a mucinous carcinoma)

Recommendations.—Mixed patterns should be classified by predominant type.

Reporting Issues

  • WHO classification variably used

  • Medullary carcinoma variably reported as a distinct entity

Recommendation.—Report histologic type according to the WHO classification. Medullary carcinoma should be reported separately from undifferentiated carcinoma (see below).

Statistical Issues

  • Histologic type not statistically significant in most studies of prognostic variables except for tumor types that are, by definition, high grade (poorly or undifferentiated), specifically, signet-ring cell carcinoma and small cell carcinoma

  • Limited number of studies on the prognostic significance of histologic type after stratification by MSI status

Recommendation.—Tumor type should be correlated with outcome after adjustment for MSI status in statistically robust studies with multivariate analysis in order to definitively determine its prognostic significance.

Histologic Features Associated With MSI-H: Host Lymphoid Response to Tumor and Medullary or Mucinous Histologic TypeHost Lymphoid Response to Tumor

Method Issues38,89,97–106 

  • Variable use of histologic appearance alone versus use of immunohistochemical staining for lymphocytes

  • Variation in sampling location and amount—unclear affect on assessment of this parameter

Recommendation.—Perform assessment by routine histology only. Both the perimeter and center of tumor should be examined in this assessment.

Interpretation Issues

  • Variable interpretation of pattern of lymphoid response

    Intratumoral lymphocytes

    Peritumoral lymphoid reaction

    Transmural Crohn's-like lymphoid reaction

  • Intensity of lymphoid response variably interpreted (eg, all or none versus graded intensity)

  • One or more than one pattern variably recognized

  • Variable differentiation between an inflammation versus immune response

  • Intratumoral lymphocytes variably interpreted as a separate feature versus integral feature of a medullary carcinoma

  • Variable recognition of the relationship between replication error status and intratumoral lymphocytes

Recommendations.—Intratumoral lymphocytic infiltrates should be distinguished from peritumoral inflammatory infiltrates. The former are closely associated with MSI-H and medullary architecture (see below). Only moderate- and high-density intratumoral infiltrates (4 or more per high-power field) should be considered significant.

Reporting Issues

  • Variable reporting of host lymphoid response

  • Variation in description of type and grading of host lymphoid response

Recommendation.—Separate reporting of host lymphoid response is optional. If reported, distinction should be made between peritumoral and intratumoral lymphoid infiltrates.

Statistical Issues

  • Statistically robust studies are needed to confirm the relationship between host lymphoid response and prognosis and among intratumoral lymphocytes, MSI-H, and prognosis.

Recommendation.—Lymphoid response as outlined above (especially intratumoral lymphocytes) should be included in multivariate analyses to correlate with outcome and MSI-H status in statistically robust studies.

Histologic Type: Medullary Carcinoma, Mucinous Carcinoma98,107–113 

Method Issues

  • Variable use of routine histology alone versus use of special studies

Recommendation.—Perform assessment by routine histologic method alone. Special studies should be performed at the discretion of the pathologist.

Interpretation Issues

  • Variable interpretation of mixed patterns common in MSI-H tumors (eg, signet ring cells in a mucinous carcinoma)

  • Medullary type not included in the WHO classification prior to 2000

  • Medullary types commonly confused with neuroendocrine carcinomas

Recommendations.—Medullary carcinoma should be distinguished and separately classified. Mixed patterns should be classified by predominant type.

Reporting Issues

  • WHO classification variably used

Recommendation.—Report histologic type according to WHO classification as revised in 2000. Medullary carcinoma should be reported separately as a specific type according to the newly revised classification.

Statistical Issues

  • Histologic type not proven statistically significant independent of tumor grade

  • Relationship among tumor type, MSI-H, and outcome lacking

Recommendation.—Correlate tumor type with outcome and MSI status in statistically robust studies with multivariate analysis.

High Degree of MSI

Method Issues114–118 

  1. Molecular techniques

    • Tissue source may be variably contaminated with nonneoplastic cells

    • Variable probes may be used

    • Variable duration of fixation and type of fixative used (eg, containing heavy metals)

    • Variability in quality control

    • Variable use of requisite paired tumor and normal specimens for comparison

    • Innate variability in technique using polymerase chain reaction assays

  2. Immunostaining techniques

    • Various antibodies (different clones) used

    • Tissue source fresh or fixed

    • Variable use of antigen retrieval methods leading to variable staining quality with individual antibodies to the major DNA repair enzymes (hMLH1 and hMSH2) (eg, hMLH1 is articularly dependent on appropriate antigen retrieval techniques)

Recommendations.—For molecular methods, good general polymerase chain reaction quality control is required. Overfixation (>72 hours) should be avoided. Manual microdissection is usually required to obtain ≥70% tumor DNA. Consensus probe panels developed by the National Cancer Institute for MSI are recommended. Paired tumor and normal samples must be used. For immunostaining methods, immunostaining procedures for hMLH1 and hMSH2 should include antigen retrieval based on steam heating with EDTA or citrate buffer.

Interpretation Issues

  1. Molecular techniques

    • Variable recognition of cutoff of 30% instability for diagnosis of MSI-H

    • Misinterpretation of dilution by normal cells as a negative assay

  2. Immunostaining techniques

    • Low sensitivity of staining and/or high background leading to misinterpretation

    • Focal chromogen deposition occurring within truly negative nuclei with antigen retrieval techniques misinterpreted as positivity

Recommendations.—For molecular methods, interpretation of MSI-H should be strictly based on 30% or more of microsatellites assayed showing instability. Paired tumor and normal samples must be used. Dilution of tumor sample by normal cells (>30%) should be avoided. For immunostaining methods, internal and external staining controls should be used. Only diffuse nuclear staining should be interpreted as positive.

Reporting Issues

  1. Molecular techniques

    • Variable use of multiple synonyms and related terms: microsatellite stable, low-level instability, MSI-H, replication error positive, mutator phenotype, ubiquitous somatic mutation

  2. Immunostaining techniques

    • Uniform reporting format lacking

Recommendations.—For molecular methods, report should include specific probes used. The terms proposed by the National Cancer Institute Workshop on Microsatellite Instability (microsatellite stable, low-level instability, MSI-H) should be used.118 The MSI-H status should be defined as more than 30% of markers analyzed demonstrating instability. For immunostaining methods, hMLH1 and hMSH2 expression should be reported as intact or absent.

Statistical Issues

  1. Molecular techniques

    • Incomplete data on MSI-H as a therapeutic predictive factor

    • The importance of low-level instability uncertain

  2. Immunostaining techniques

    • Familial MSI-H cases may have intact expression of both hMLH1 and hMSH2 due to either (1) missense mutation or (2) mutation in another of the DNA mismatch repair genes

Recommendations.—For molecular methods, prospective therapeutic trials are required to test predictive value. For immunostaining methods, when being used for heredity nonpolyposis colorectal cancer proband identification, MSI molecular testing should also be included in the testing algorithm.

Loss of Heterozygosity at 18q and Allelic Loss of Deleted in Colon Cancer Gene

Method Issues119–128 

  1. Molecular techniques

    • Variable contamination of tissue source with nonneoplastic cells

    • Variable probes used (eg, different clones)

    • Tissue source fresh or fixed

  2. Immunostaining techniques

    • Various different monoclonal and polyclonal antibodies used

    • Variations in tissue antigenicity

Recommendations.—For molecular methods, good general polymerase chain reaction quality control is required. Overfixation (more than 72 hours) should be avoided. Manual microdissection is usually required to obtain ≥70% tumor DNA. Paired tumor and normal samples must be used. For immunostaining methods, internal and external positive and negative staining controls should be closely monitored. Antigen retrieval techniques are recommended by most authors.

Interpretation Issues

  1. Molecular techniques

    • Variable interpretation of differential band intensity for diagnosis of allele loss if less than twofold.

    • Misinterpretation of noninformative assays as loss of heterozygosity (LOH) or as normal

    • Potential misinterpretation of dilution by normal cells as a negative assay

  2. Immunostaining techniques

    • Weak positive staining variably interpreted as loss of expression

Recommendations.—For molecular methods, interpretation of allelic loss should be based on a twofold difference in band intensity. Paired tumor and normal samples must be used. Dilution of tumor sample by normal cells (>30%) should be avoided. For immunostaining methods, internal and external staining controls should be carefully monitored. Only completely negative nuclei should be interpreted as showing loss of deleted in colon cancer (DCC) gene expression. Grading of intensity of nuclear staining is inappropriate.

Reporting Issues

  1. Molecular techniques

    • Multiple terms used as if synonymous: LOH, allelic loss, allelic imbalance, DCC (eg, not the only gene on 18q)

  2. Immunostaining techniques

    • Uniform reporting format not yet established

Recommendations.—For molecular methods, report should include specific probes used and quantitative threshold for LOH (allelic imbalance). For immunostaining methods, DCC expression should be reported as intact or absent.

Statistical Issues

  1. Molecular techniques

    • Variations in strength and utility of DCC as a prognostic marker vary among studies

  2. Immunostaining techniques

    • Variation in relationship between loss of DCC expression and 18q LOH

    • Incomplete data on DCC loss as a therapeutic predictive factor

Recommendation.—For molecular methods, statistically robust prospective studies are needed to confirm prognostic value. For immunostaining methods, statistically robust studies are needed to establish the relationship among loss of DCC expression, LOH with various 18q probes, and patient outcome.

Tumor Border Configuration

Method Issues38,51,85,89,96–98,129–132 

  • Variation in criteria for assessment according to author

  • Gross versus microscopic versus combination approaches to assessment

  • Assessment variably subjective

Recommendation.—The 2-tiered evaluation system (pushing border vs infiltrating border) that has been defined by Jass et al and tested for interobserver variability should be used.

Interpretation Issues

  • Substantial interobserver variability unless pathologists educated to definition

  • Variation in opinions as to what features should be included in the definition

Recommendation.—Definitions of features of pushing versus infiltrating border published by Jass et al should be followed for interpretation and to reduce interobserver variability.

Reporting Issues

  • Tumor border configuration rarely reported

  • Recognition of significance of tumor border configuration not widely recognized

Recommendation.—If reported, report tumor border configuration described as pushing or infiltrating.

Statistical Issues

  • Need for statistically robust studies with multivariate analysis

Recommendation.—Evaluation of tumor border configuration as a 2-tiered variable should be carried out in large studies on prognostic factors using multivariate analysis.

DNA Content

Method Issues73,133–143 

  1. Flow cytometry

    • Methodology not standardized

    • Difficult to quality control

    • Variation with fresh versus archived tissue

    • Variation in quality of histograms with preparatory techniques

    • Variation with ratio of stromal to neoplastic cells

    • Channel setting (for linearity) variably machine dependent

  2. Image analysis

    • Methods not standardized

    • Methods not widely available

Recommendation.—Data are insufficient to recommend specific methods. Comparative evaluation of methods for DNA content is needed.

Interpretation Issues

  • Variation in determination of aneuploidy

    Variation in setting of cutoffs

    Variation in number of repeat analyses (discretion of investigator)

  • Variation in basic definitions and terms (eg, diploid, diploid low, diploid high, nondiploid, aneuploid, tetraploid)

Recommendation.—Data are insufficient to recommend specific interpretation guidelines.

Reporting Issues

  • Variation in terms (eg, diploid, diploid low, diploid high, nondiploid, aneuploid, tetraploid)

Recommendation.—Although standardized terms are needed, data are insufficient to recommend specific terminology. Definition should be reported for specific terms used.

Statistical Issues

  • Univariate versus multivariate analyses

  • Lumping of DNA ploidy and cell proliferation analysis into a single variable

  • Adjustment for treatment variations

Recommendation.—DNA content should be evaluated using consistent and reproducible methods in large studies using multivariate analysis.

Other Molecular Markers

Overview.—A wide variety of molecular markers has been defined in colorectal cancer, but aside from LOH 18q/DCC loss and MSI-H (see category IIB above), the prognostic significance of these factors remains unproven.1,119,144–183 A critical analysis of the variables related to each of these molecular markers is beyond the scope of this review, but the general limitations of the existing data defining the prognostic significance of these markers is outlined below. The categories of molecular markers linked with colorectal cancer include the following:

  • Tumor suppressor genes (LOH 1p/p53, LOH 8p, LOH 1p, LOH 5q)

  • Oncogenes (K-ras, c-myc)

  • Apoptosis and cell suicide-related genes (bcl-2; BAX)

  • DNA synthesis-related genes (thymidylate synthase; thymidine phosphatase)

  • Transforming growth factors (TGF) and epidermal growth factor receptor (EGF-R) genes (TGF-α, TGF-β, c-erb-b/her2/neu, EGF-R)

  • Cyclin-dependent kinase inhibitor genes (p27, p21)

  • Angiogenesis-related genes (vascular endothelial growth factor)

  • Adhesion molecule and glycoprotein genes (CD44, E-cadherin, sialo-Tn antigen)

  • Matrix metalloproteases and inhibitors (urokinase-type plasminogen activator)

  • Metastasis suppressor genes (nm23-H1)

Method Issues

  • Variation of method according to investigator and factor

  • Various methods applied to investigation of a single genetic factor producing different results (various types of aberrant genetic events may ultimately produce the same effect in the cell)

  • No standard guidelines for clinical testing

Recommendation.—Data are not sufficient for specific recommendations.

Interpretation Issues

  • Investigator-dependent interpretation

  • Method-dependent interpretation

Recommendation.—Data are not sufficient for specific recommendations.

Reporting Issues

  • Investigator-dependent reporting and terminology

Recommendation.—Data are not sufficient for specific recommendations.

Statistical Issues

  • Large number of single studies on single factors

  • Small number of studies on a large number of individual molecular factors

  • Conflicting results from various studies of same factor (eg, p53)

  • Almost no statistically robust studies on most factors

  • Almost no multivariate analyses of most factors

Recommendation.—Individual factors should be evaluated as single variables in large studies on prognostic factors using multivariate analysis.

Perineural Invasion

Method Issues1,2,44,87,184,185 

  • Variable use of routine histology alone versus immunostaining to highlight nerves

Recommendation.—Use routine histology alone.

Interpretation Issues

  • None

Recommendation.—None.

Reporting Issues

  • Variably reported

Recommendation.—Report perineural invasion as present or absent in all cases.

Statistical Issues

  • Univariate versus multivariate analyses

  • Study size variation

Recommendation.—Perineural invasion should be evaluated as an individual variable in large studies on prognostic factors using multivariate analysis.

Microvessel Density

Method Issues42,185–191 

  • Variation in dilution of factor VIII (Dako Corporation, Carpinteria, Calif) used for immunostaining of endothelium

    1:250

    1:2400

  • Variation in definition of a high-power field

    ×40 field

    ×20 field

  • Variation in number of fields

Recommendation.—Standard guidelines for staining and evaluation should be established.

Interpretation Issues

  • Granulation tissue due to ulceration versus tumor-induced neovascularization variably interpreted

  • Variation in which vessels are counted

  • Variation in interpretation of a vessel (eg, stained cells in clusters without lumens variably assessed as vessels)

Recommendation.—Interpretation guidelines published by Weidner should be followed.

Reporting Issues

  • Variably reported as a density measurement: mean number of microvessels per high-power field

  • Variably reported as a total number (microscopic area examined fixed) (eg, <25 or ≥25)

Recommendation.—Tumor angiogenesis should be reported as a maximum density measurement.

Statistical Issues

  • Univariate versus multivariate analyses

  • Small numbers of cases

Recommendation.—Microvessel density should be evaluated in large studies on prognostic factors using multivariate analysis.

Cell Proteins and Carbohydrates

Overview.—Among the numerous cell proteins and carbohydrate markers that have been reported in colorectal cancer, none have been extensively studied in clinical trials.1 This class of tumor markers includes all the following substances:

  • Class I HLA molecules

  • Class II HLA molecules

  • CA 19–9

  • CA 72–4

  • Sialyl Lex

  • Sialosyl-Tn

  • Urokinase-type plasminogen activator

  • Plasminogen activator inhibitor 2

  • Glycoprotein 72

  • P-glycoprotein (multidrug resistance gene product)

  • MUC-1 mucin

  • E-cadherin

  • α-Catenin

  • Integrins

  • Type IV collagen

  • Gelatinase B (metalloproteinase-9)

  • Laminin

  • Tenascin

  • Autocrine mobility factor receptor (gp78)

  • Phospholipase C

  • Secretory component of immunoglobulin A

  • Metallothionein

  • EGF-R

  • Gastrin receptor

  • Somatostatin receptors

  • Sucrase-isomaltase

  • Cathepsin B, L, and D (cysteine/aspartyl proteases)

  • Ferritin

  • CD44

  • Vitamin D receptor protein

  • Cytokeratin 20

Method Issues

  • Variation of method according to investigator and factor

  • No standard guidelines

Recommendation.—Data are not sufficient for specific recommendations.

Interpretation Issues

  • Investigator-dependent interpretation of results

Recommendation.—Data are not sufficient for specific recommendations.

Reporting Issues

  • Investigator-dependent reporting of results and use of terminology

Recommendation.—Data are not sufficient for specific recommendations.

Statistical Issues

  • Large number of single studies on single factors

  • Small number of studies on a large number of individual cell elements

  • Almost no statistically robust studies

  • Almost no multivariate analyses

Recommendation.—Individual factors should be evaluated as single variables in large studies on prognostic factors using multivariate analysis.

Peritumoral Fibrosis (Desmoplasia)

Method Issues5,97–99 

  • Histopathologic examination alone versus special stains

  • Sampling variation

Recommendation.—Assessment of tumor-associated stromal response should be performed by routine histopathologic examination of the tumor periphery (no special stains recommended).

Interpretation Issues

  • Interobserver variation and intraobserver variation99 

  • Variation in judgment threshold for how much fibrosis constitutes desmoplasia

  • Peritumoral fibrosis sometimes graded: little, moderate, extensive

  • Variably considered part of tumor border configuration instead of a separate variable

Recommendation.—Explicit guidelines for analysis and interpretation of peritumoral fibrosis should be established.

Reporting Issues

  • Usually not evaluated

  • Usually not reported

Recommendation.—Guidelines for reporting of peritumoral fibrosis should be established.

Statistical Issues

  • Significance of desmoplasia independent of tumor border configuration unclear

  • Small studies insufficient to determine significance

Recommendation.—Peritumoral fibrosis should be evaluated by uniform method as an individual variable in large studies on prognostic factors using multivariate analysis.

Purulent Peritumoral Inflammatory Reaction

Method Issues71,98,107,192–194 

  • Variable sampling with avoidance of necrotic areas likely to be inflamed

Recommendation.—Data are insufficient to recommend specific methodologic guidelines.

Interpretation Issues

  • Variation in interpretation of immune cells as inflammatory cells

  • Variation in interpretation of abscesses as a primary versus secondary phenomenon in relationship to tumor perforation (itself an adverse prognostic factor)

Recommendation.—Inflammatory reactions should be evaluated as a feature separate and distinct from host lymphoid responses (see “Host Lymphoid Response to Tumor”).

Reporting Issues

  • Variably reported at all

  • Variable reporting of inflammatory reaction in association with tumor perforation

Recommendation.—Tumor perforation should be reported in all cases. Inflammatory reaction should be reported as purulent in type to distinguish it from a host lymphoid (immune) response.

Statistical Issues

  • Few studies with little data

  • Variation in recognition and analysis as a unique factor distinguished from lymphoid response

Recommendation.—Peritumoral inflammation should be analyzed as a unique variable in large studies using multivariate analysis.

Foci of Neuroendocrine Differentiation Within Any Histologic Type

Method Variation Issues195–197 

  • Light microscopy—variable identification of neuroendocrine cells in routine hematoxylin-eosin–stained sections

  • Histochemical stains—variable use of argentaffin and argyrophil reactions

  • Immunohistochemical methods—variable use

    • Chromogranin

    • Neuron-specific enolase

    • Synaptophysin

    • Leu-7

    • Specific peptides

Recommendation.—Assessment should be performed by hematoxylin-eosin staining alone; data are insufficient to recommend special stains or immunohistochemical stains.

Interpretation Variation Issues

  • Definition of significant neuroendocrine differentiation (eg, any positive cells detected by immunohistochemical staining or some specific number of cells)

Recommendation.—Data are insufficient to recommend specific interpretation guidelines.

Reporting Issues

  • Dependent on interpretation and relationship to histologic type

Recommendation.—Documentation of neuroendocrine differentiation may be reported as a confirmation of small cell histologic type or rare composite or amphicrine tumors.

Statistical Issues

  • Cut point determination varies

Recommendation.—Data are insufficient to recommend specific statistical guidelines.

Nucleolar Organizing Regions

Method Issues71,192–194,197–199 

  • Variation in thickness of sections (2–5 μm) used in different studies

  • Variation in staining techniques

  • Nucleolar organizing region analysis by automated image analyses versus counting under oil immersion

  • Variation in number of nuclei counted

Recommendation.—Data are not sufficient to recommend specific method.

Interpretation Issues

  • Variation in number of nucleolar organizing regions with plane of section (focusing up and down)

  • Counting alone versus separation into patterns (eg, clusters vs individual) versus area of nucleolar organizing regions per nucleus

  • Interobserver and intraobserver variability193 

Recommendation.—Data are not sufficient to recommend specific method of interpretation.

Reporting Issues

  • Median numbers versus ranges

Recommendation.—Data are not sufficient to recommend specific method of reporting.

Statistical Issues

  • Small patient numbers

  • Univariate versus multivariate analyses

Recommendation.—Data are insufficient to recommend specific statistical guidelines.

Proliferation Indices

Method Issues200–216 

  • Immunohistochemistry (Ki-67, proliferating cell nuclear antigen) method variation

  • Flow cytometric method variation

  • Mitotic counts rarely used for carcinomas—variation in approach

  • Variation in number of counts performed using any method

Recommendation.—Data are insufficient to recommend specific method.

Interpretation Issues

  • Interobserver variability in interpretation of a mitotic figure

  • Immunostaining: variable interpretation of strong versus weak staining

  • Overall interpretation variation—average or region of most intense activity only

  • Specificity: Ki-67 variably expressed in noncycling cells

Recommendation.—Data are insufficient to recommend specific interpretation guidelines.

Reporting Issues

  • Morphologic methods: variable expression of rate as number of cycling cells per high-power field or per fixed number of cells

  • Proliferation index rarely reported at all

Recommendation.—Data are insufficient to recommend specific reporting guidelines.

Statistical Issues

  • Univariate versus multivariate analyses

  • Conflicting data

Recommendation.—Data are insufficient to recommend specific statistical guidelines.

Tumor Size

Method Issues2,3,42,68,76,78,216,217 

  • Variable number of dimensions recorded

Recommendation.—One dimension (largest diameter) is sufficient.

Interpretation Issues

  • None

Recommendation.—None.

Reporting Issues

  • Variably recorded as part of the gross description

Recommendation.—Tumor size should be report as part of permanent record of tumor description. Although the size of the tumor is of no prognostic significance, it may be important for quality control of tumor size determined by nonpathologic means (eg, imaging modalities).

Statistical Issues

  • None.

Recommendation.—Stay in category IV.

Gross Tumor Configuration

Method Issues3,67,74,86–88 

  • None

Recommendation.—None.

Interpretation Issues

  • Variable interpretation of complex configurations

  • Variable numbers of individual configurations considered in evaluation

Recommendation.—Data are insufficient to recommend specific interpretation guidelines.

Reporting Issues

  • Variable reporting of configuration

  • When reported, variable interpretation (see above) produces variable reporting

Recommendation.—Data are insufficient to recommend specific reporting guidelines. Reporting is optional as a point of description and documentation.

Statistical Issues

  • Multivariate versus univariate analyses

  • Variable study size

Recommendation.—Stay in category IV.

Hermanek
,
P.
and
L. H.
Sobin
.
Colorectal carcinoma.
In: Hermanek P, Gospodarowicz MK, Henson DE, Hutter RVP, Sobin LH, eds. Prognostic Factors in Cancer. New York, NY: Springer-Verlag NY Inc; 1995
.
Chapuis
,
P. H.
,
O. F.
Dent
, and
R.
Fisher
.
et al
.
A multivariate analysis of clinical and pathological variables in prognosis after resection of large bowel cancer.
Br J Surg
1985
.
72
:
698
702
.
Newland
,
R. C.
,
O. F.
Dent
, and
M. N.
Lyttle
.
et al
.
Pathologic determinants of survival associated with colorectal cancer with lymph node metastases: a multivariate analysis of 579 patients.
Cancer
1994
.
73
:
2076
2082
.
Tominaga
,
T.
,
T.
Sakabe
, and
Y.
Koyama
.
et al
.
Prognostic factors for patients with colon or rectal carcinoma treated with resection only: five-year follow-up report.
Cancer
1996
.
78
:
403
408
.
Shepherd
,
N.
,
K.
Baxter
, and
S.
Love
.
The prognostic importance of peritoneal involvement in colonic cancer: a prospective evaluation.
Gastroenterology
1997
.
112
:
1096
1102
.
Fleming
,
I. D.
,
J. S.
Cooper
, and
Henson
.
et al (American Joint Committee on Cancer), eds
.
Cancer Staging Manual.
5th ed. Philadelphia, Pa: Lippincott-Raven; 1997
.
Hermanek
,
P.
,
D. E.
Henson
,
R. V. P.
Hutter
, and
L. H.
Sobin
.
TNM Supplement.
New York, NY: Springer-Verlag NY Inc; 1993
.
Blenkinsopp
,
W. K.
,
S.
Stewart-Brown
,
L.
Blesovsky
,
G.
Kearney
, and
L. P.
Fielding
.
Histopathology reporting in large bowel cancer.
J Clin Pathol
1981
.
34
:
509
513
.
Scott
,
K. W. M.
and
R. H.
Grace
.
Detection of lymph node metastases in colorectal carcinoma before and after fat clearance.
Br J Surg
1989
.
76
:
1165
1167
.
Cawthorn
,
S. J.
,
N. M.
Gibbs
, and
C. G.
Marks
.
Clearance technique for the detection of lymph nodes in colorectal cancer.
Br J Surg
1986
.
73
:
58
60
.
Herrera-Ornelas
,
L.
,
J.
Justiniano
,
N.
Castillo
,
N. J.
Petrelli
,
J. P.
Stulc
, and
A.
Mittelman
.
Metastases in small lymph nodes from colon cancer.
Arch Surg
1987
.
122
:
1253
1256
.
Herrera
,
L.
and
J. R.
Villareal
.
Incidence of metastases from rectal adenocarcinoma in small lymph nodes detected by a clearing technique.
Dis Colon Rectum
1992
.
35
:
783
788
.
Jeffers
,
M. D.
,
G. M.
O'Dowd
,
H.
Mulcahy
,
M.
Staff
,
D. P.
O'Donoghue
, and
M.
Toner
.
The prognostic significance of immunohistochemically detected lymph node micrometastases in colorectal carcinoma.
J Pathol
1994
.
172
:
183
187
.
Cutait
,
R.
,
V. A.
Alves
, and
L. C.
Lopes
.
et al
.
Restaging of colorectal cancer based on the identification of lymph node micrometastases through immunoperoxidase staining of CEA and cytokeratins.
Dis Colon Rectum
1991
.
34
:
917
920
.
Adell
,
G.
,
B.
Boeryd
,
B.
Franlung
,
R.
Sjodahl
, and
L.
Hanansson
.
Occurrence and prognostic importance of micrometastases in regional nodes in Duke's B colorectal carcinoma: an immunohistochemical study.
Eur J Surg
1996
.
162
:
637
642
.
Liefers
,
G-J.
,
A-M.
Cleton-Jansen
, and
C. J. H.
van de Velde
.
et al
.
Micrometastases and survival in stage II colorectal cancer.
N Engl J Med
1997
.
337
:
1188
1194
.
Yamamoto
,
N.
,
Y.
Kato
,
A.
Yanagisawa
,
H.
Ohta
,
T.
Takahashi
, and
T.
Kitagawa
.
Predictive value of genetic diagnosis for cancer micrometastases: histologic and experimental appraisal.
Cancer
1997
.
80
:
1393
1398
.
Greenson
,
J. K.
,
C. E.
Isenhart
,
R.
Rice
,
C.
Mojzisik
,
D.
Houchens
, and
E. W. Jr
Martin
.
Identification of occult micrometastases in pericolic lymph nodes of Dukes’B colorectal cancer patients using monoclonal antibodies against cytokeratin and CC49: correlation with long-term survival.
Cancer
1994
.
73
:
563
569
.
Ratto
,
C.
,
L.
Sofo
, and
M.
Ippoliti
.
et al
.
Accurate lymph-node detection in colorectal specimens resected for cancer is of prognostic significance.
Dis Colon Rectum
1999
.
42
:
143
158
.
Hermanek
,
P.
,
R. V. P.
Hutter
,
L. H.
Sobin
, and
C.
Wittekind
.
Communication UICC International Union Against Cancer: classification of isolated tumor cells and micrometastasis.
Cancer
1999
.
86
:
2668
2673
.
Netzer
,
P.
,
C.
Forster
, and
R.
Biral
.
et al
.
Risk factor assessment of endoscopically removed malignant colorectal polyps.
Gut
1988
.
43
:
669
674
.
Geraghty
,
J. M.
,
C. D.
Williams
, and
I. C.
Talbot
.
Malignant colorectal polyp: venous invasion and successful treatment by endoscopic polypectomy.
Gut
1991
.
32
:
774
778
.
Nivatvongs
,
S.
,
A.
Rojanasakul
, and
H. M.
Reiman
.
et al
.
The risk of lymph node metastasis and colorectal polyps with invasive carcinoma.
Dis Colon Rectum
1991
.
34
:
323
328
.
Cunningham
,
K. N.
,
L. R.
Mills
,
B. N.
Schuman
, and
D. H.
Mwakyusa
.
Longterm prognosis of well differentiated adenocarcinoma endoscopically removed colorectal adenomas.
Dig Dis Sci
1994
.
39
:
2034
3037
.
Haggitt
,
R. C.
,
R. E.
Glotzbach
,
E. E.
Soffer
, and
L. D.
Wruble
.
Prognostic factors in colorectal carcinomas arising in adenomas: implications for lesions removed by endoscopic polypectomy.
Gastroenterology
1989
.
89
:
328
336
.
Morson
,
B. C.
,
J. E.
Whiteway
,
E. A.
Jones
,
F. A.
Macrie
, and
C. D.
Williams
.
Histopathological and prognosis of malignant colorectal polyp treated by endoscopic polypectomy.
Gut
1989
.
25
:
437
444
.
Volk
,
E.
,
J. R.
Goldblum
,
R. E.
Petras
,
W. D.
Carey
, and
O.
Fazi
.
Management and outcome of patients with invasive carcinoma arising in colorectal polyps.
Gastroenterology
1995
.
109
:
1801
1807
.
Cooper
,
H. S.
,
L. M.
Deppisch
, and
E. I.
Kahn
.
et al
.
Pathology of the malignant colorectal polyp.
Hum Pathol
1998
.
29
:
15
26
.
Kikuchi
,
R.
,
M.
Takano
, and
K.
Takagi
.
et al
.
Management of early invasive colorectal cancer.
Dis Colon Rectum
1995
.
38
:
289
1295
.
Hase
,
K.
,
C. H.
Shatney
, and
H.
Mochizuki
.
et al
.
Longterm results of curative resection of minimally invasive cancer.
Dis Colon Rectum
1995
.
38
:
19
26
.
Willett
,
C. G.
,
C. C.
Compton
,
P. C.
Schellito
, and
J. T.
Effird
.
Selection factors for local excision for abdominal perineal resection of early stage rectal cancer.
Cancer
1994
.
73
:
2716
2720
.
Minsky
,
B. D.
,
T.
Rich
,
A.
Recht
,
W.
Harvey
, and
Z.
Meis
.
Selection criteria for local excision with or without adjuvant radiation therapy for rectal cancer.
Cancer
1989
.
63
:
1421
1429
.
Brodsky
,
J. T.
,
G. K.
Richard
,
A. M.
Cohen
, and
B. D.
Minsky
.
Variables correlated with the risk of lymph node metastasis in early rectal cancer.
Cancer
1992
.
69
:
322
326
.
Goldstein
,
N. S.
and
J.
Hart
.
Histological features associated with lymph node metastasis in stage I and superficial T2 rectal adenocarcinomas in abdominal perineal resection specimen: identifying a subset of patients for whom treatment with adjuvant therapy or complete abdominal perineal resection should be considered after local excision.
Am J Clin Pathol
1999
.
111
:
51
58
.
Blumberg
,
D.
,
P. P.
Paty
, and
A.
Picon
.
et al
.
Stage I rectal cancer identification of high risk patient.
J Am Coll Surg
1998
.
86
:
574
580
.
Ouchi
,
K.
,
T.
Sugawara
, and
H.
Ono
.
et al
.
Histological features and clinical significance of venous invasion in colorectal carcinoma with hepatic metastasis.
Cancer
1996
.
78
:
2313
2317
.
Talbot
,
I. C.
,
S.
Ritchie
,
M.
Leighton
,
A. O.
Hughes
,
H. J. R.
Bussey
, and
B. C.
Morson
.
Invasion of veins by carcinoma of rectum: method of detection, histological features and significance.
Histopathology
1981
.
5
:
141
163
.
Harrison
,
J. C.
,
P. J.
Dean
,
F.
El-Zeky
, and
R.
Vander Zwaag
.
From Dukes' through Jass: pathological prognostic indicators in rectal cancer.
Hum Pathol
1994
.
25
:
498
505
.
Jass
,
J. R.
,
W. S.
Atkin
, and
J.
Cuzick
.
et al
.
The grading of rectal cancer: historical perspectives in a multivariate analysis of 447 cases.
Histopathology
1986
.
10
:
437
439
.
Takebayashi
,
Y.
,
S. I.
Akiyama
,
K.
Yamada
,
S.
Akiba
, and
T.
Akiu
.
Angiogenesis is an unfavorable prognostic factor in human colorectal carcinoma.
Cancer
1986
.
78
:
226
231
.
Talbot
,
I. C.
,
S.
Ritchie
,
M. H.
Leighton
,
A. O.
Hugh
,
H. J. R.
Bussey
, and
B. C.
Morson
.
Spread of rectal cancer within veins: histological features and clinical significance.
Am J Surg
1981
.
141
:
15
17
.
Takahasi
,
Y.
,
S. L.
Tucker
, and
Y.
Kitadai
.
et al
.
Vessel counts and expression of vascular endothelial growth factor as prognostic factors in known negative colon cancer.
Arch Surg
1997
.
132
:
541
546
.
Freedman
,
L. S.
,
P.
Macaskill
, and
A. N.
Smith
.
Multivariate analysis of prognostic factors for operable rectal cancer.
Lancet
1984
.
1
:
733
736
.
Knudsen
,
J. B.
,
T.
Nilson
,
M.
Sprechler
,
A.
Johannsen
, and
N.
Christensen
.
Venous and nerve invasion as prognostic factors in post-operative survival of patients with resectable cancer of the rectum.
Dis Colon Rectum
1983
.
26
:
613
617
.
Harrison
,
J. C.
,
P. J.
Dean
,
F.
el-Zeky
, and
R.
Vander Zwaag
.
Impact of the Crohn's like lymphoid reaction on staging of right sided colon cancer: results of a multivariate analysis.
Hum Pathol
1995
.
26
:
31
38
.
Minsky
,
B. D.
,
C.
Mies
,
A.
Recht
,
T. A.
Rich
, and
J. T.
Chaffey
.
Resectable adenocarcinoma of the rectosigmoid of rectum, II—the influence of blood vessel invasion.
Cancer
1988
.
61
:
1417
1424
.
Heys
,
S. D.
,
A.
Scherif
,
J. S.
Bagley
,
J.
Brittenden
,
C.
Smart
, and
N.
Eremi
.
Prognostic factors in survival of patients aged less than forty-five years with colorectal cancer.
Br J Surg
1994
.
81
:
685
688
.
Horn
,
A.
,
O.
Dahl
, and
I.
Morild
.
Venous and neural invasion as predictors of recurrence in rectal adenocarcinoma.
Dis Colon Rectum
1991
.
34
:
798
804
.
Krasnam
,
J.
,
L.
Flancbaum
,
R. P.
Cody
,
S.
Sheinbaum
, and
G.
Benari
.
Vascular neural invasion in colorectal carcinoma: incidence and prognostic significance.
Cancer
1988
.
61
:
1018
1023
.
Khankhanian
,
N.
,
G. M.
Mavligit
,
W. O.
Russell
, and
N.
Schimek
.
Prognostic significance of vascular invasion in colorectal cancer of Dukes' B class.
Cancer
1977
.
39
:
1195
1200
.
Jass
,
J. R.
,
S. B.
Love
, and
J. M. A.
Northover
.
A new prognostic classification of rectal cancer.
Lancet
1987
.
1
:
1303
1306
.
Cranley
,
J. P.
,
R. E.
Petras
,
W. D.
Carey
,
K.
Paradis
, and
M. V.
Sivak
.
When is endoscopic polypectomy adequate therapy for colonic polyps containing invasive cancer?
Gastroenterology
1986
.
91
:
419
427
.
Richards
,
W. O.
,
W. A.
Webb
, and
S. J.
Morris
.
et al
.
Patient management after endoscopic removal of the cancerous colon adenoma.
Ann Surg
1987
.
205
:
665
670
.
Coverlizza
,
S.
,
M.
Risio
,
A.
Ferrari
,
C. M.
Fenoglio-Preiser
, and
F. P.
Rossini
.
Colorectal adenomas containing invasive carcinoma: pathologic assessment of lymph node metastatic potential.
Cancer
1989
.
64
:
1937
1947
.
Muller
,
S.
,
I. M.
Chesner
, and
M. J.
Egan
.
et al
.
Significance of venous and lymphatic invasion in malignant polyps of the colon and rectum.
Gut
1989
.
30
:
1385
1391
.
Cooper
,
H. S.
,
L. M.
Deppisch
, and
W. K.
Gourley
.
et al
.
Endoscopically removed malignant colorectal polyps: clinicopathologic correlations.
Gastroenterology
1995
.
108
:
1657
1665
.
Zeng
,
Z.
,
A. M.
Cohen
,
S.
Hajdu
,
S. S.
Sternberg
,
E. R.
Sigurdson
, and
W.
Enker
.
Serosal cytologic study to determine free mesothelial penetration of intraperitoneal colon cancer.
Cancer
1992
.
70
:
737
740
.
Wanebo
,
H. J.
,
B.
Rao
, and
C. M.
Pinsky
.
et al
.
Preoperative carcinoembryonic antigen level as a prognostic indicator in colorectal cancer.
N Engl J Med
1978
.
299
:
448
451
.
Wolmark
,
N.
,
B.
Fisher
, and
H. S.
Wieand
.
et al
.
The significance of preoperative carcinoembryonic antigen levels in colorectal cancer.
Ann Surg
1984
.
199
:
375
382
.
Onetto
,
M.
,
M.
Paganuzzi
, and
G. B.
Secco
.
et al
.
Preoperative carcinoembryonic antigen and prognosis in patients with colorectal cancer.
Biomed Pharmacother
1985
.
39
:
392
395
.
Scott
,
N. A.
,
N. S.
Wieand
,
C. G.
Moertel
,
S. S.
Cha
,
R. W.
Beart
, and
M. M.
Lieber
.
Colorectal cancer: Dukes' stage, tumor site, preoperative plasma CEA level, and patient prognosis related to tumor DNA ploidy pattern.
Arch Surg
1987
.
122
:
1375
1379
.
Wiggers
,
T.
,
J.
Arends
, and
A.
Volovics
.
Regression analysis of prognostic factors in colorectal cancer after curative resections.
Dis Colon Rectum
1988
.
31
:
33
41
.
Meling
,
G. I.
,
T. O.
Rognum
, and
O. P. F.
Clausen
.
et al
.
Serum carcinoembryonic antigen in relation to survival, DNA ploidy pattern, and recurrent disease in 406 colorectal carcinoma patients.
Scand J Gastroenterol
1992
.
27
:
1061
1068
.
Slenz
,
K.
,
A.
Senagore
,
J.
Hibbert
,
W. P.
Mazier
, and
T. M.
Talbott
.
Can preoperative and postoperative CEA predict survival after colon cancer resection?
Am Surg
1994
.
60
:
528
532
.
Lindmark
,
G.
,
R.
Bergström
,
L.
Påhlman
, and
B.
Glimelius
.
The association of preoperative serum tumour markers with Dukes'stage and survival in colorectal cancer.
Br J Cancer
1995
.
71
:
1090
1094
.
Harrison
,
L. E.
,
J. G.
Guillem
,
P.
Paty
, and
A. M.
Cohen
.
Preoperative carcinoembryonic antigen predicts outcomes in node-negative colon cancer patients: a multivariate analysis of 572 patients.
J Am Coll Surg
1997
.
185
:
55
59
.
Freedman
,
L.
,
P.
Macaskill
, and
A.
Smith
.
Multivariate analysis of prognostic factors for operable rectal cancer.
Lancet
1984
.
2
:
733
736
.
Griffin
,
M. R.
,
E. J.
Bergstralh
,
R. J.
Coffey
,
R. W. Jr
Beart
, and
L. J.
Melton
.
Predictors of survival after curative resection of carcinoma of the colon and rectum.
Cancer
1987
.
60
:
2318
2324
.
Fisher
,
E. R.
,
R.
Sass
,
A.
Palekar
,
B.
Fisher
, and
N.
Wolmark
.
Dukes’classification revisited: findings from the National Surgical Adjuvant Breast and Bowel Projects (protocol R-01).
Cancer
1989
.
64
:
2354
2360
.
Hermanek
,
P.
,
I.
Guggenmoos-Holzmann
, and
F. P.
Gall
.
Prognostic factors in rectal carcinoma: a contribution to the further development of tumor classification.
Dis Colon Rectum
1989
.
32
:
593
599
.
Rüschoff
,
J.
,
A.
Bittinger
,
K.
Neumann
, and
P.
Schmitz-Moormann
.
Prognostic significance of nucleolar organizing regions (NORs) in carcinomas of the sigmoid colon and rectum.
Pathol Res Pract
1990
.
186
:
85
91
.
Robey-Cafferty
,
S. S.
,
A. K.
el-Naggar
,
D. J.
Grignon
,
K. R.
Cleary
, and
J. Y.
Ro
.
Histologic parameters and DNA ploidy as predictors of survival in stage B adenocarcinoma of colon and rectum.
Mod Pathol
1990
.
3
:
261
266
.
Böttger
,
T. C.
,
D.
Potratz
,
M.
Stockle
,
S.
Wellek
,
J.
Klupp
, and
T.
Junginger
.
Prognostic value of DNA analysis in colorectal carcinoma.
Cancer
1993
.
72
:
3579
3587
.
Deans
,
G. T.
,
C. C.
Patterson
, and
G. T.
Parks
.
et al
.
Colorectal carcinoma: importance of clinical and pathological factors in survival.
Ann R Coll Surg Engl
1994
.
76
:
59
64
.
Jessup
,
J. M.
,
P. T.
Lavin
, and
C. W. Jr
Andrews
.
et al
.
Sucrase-isomaltase is an independent prognostic marker for colorectal carcinoma.
Dis Colon Rectum
1995
.
38
:
1257
1264
.
D'Eredita
,
G.
,
G.
Serio
,
V.
Neri
,
R. A.
Polizzi
,
G.
Barberior
, and
T.
Losacco
.
A survival regression analysis of prognostic factors in colorectal cancer.
Aust N Z J Surg
1996
.
66
:
445
451
.
Jessup
,
J. M.
,
L. S.
McGinnis
,
G. D. Jr
Steele
,
H. R.
Menck
, and
D. P.
Winchester
.
The National Cancer Data Base report on colon cancer.
Cancer
1996
.
78
:
918
926
.
Mulcahy
,
H. E.
,
M. M.
Skelly
,
A.
Husain
, and
D. P.
O'Donoghue
.
Long-term outcome following curative surgery for malignant large bowel obstruction.
Br J Surg
1996
.
83
:
46
50
.
Ropponen
,
K.
,
M.
Eskelinen
,
V. M.
Kosma
,
P.
Lipponen
,
P.
Paakkinen
, and
E.
Alhava
.
Comparison of classic and quantitative prognostic factors in colorectal cancer.
Anticancer Res
1996
.
16
:
3875
3882
.
Thomas
,
G. D. H.
,
M. F.
Dixon
,
N. C.
Smeeton
, and
N. S.
Williams
.
Observer variation in the histological grading of rectal carcinoma.
J Clin Pathol
1983
.
36
:
385
391
.
Adam
,
I. J.
,
M. O.
Mohamdee
, and
I. G.
Martin
.
et al
.
Role of the circumferential margin involvement in the local recurrence of rectal cancer.
Lancet
1994
.
344
:
707
711
.
Chan
,
K.
,
J.
Boey
, and
S.
Wong
.
A method of reporting radial invasion and surgical clearance of rectal carcinoma.
Histopathology
1985
.
9
:
1319
1327
.
Quirke
,
P.
,
P.
Durdy
,
M. F.
Dixon
, and
N. S.
Williams
.
Local recurrence of rectal adenocarcinoma due to inadequate surgical resection.
Lancet
1986
.
2
:
996
999
.
Quirke
,
P.
and
N.
Scott
.
The pathologists role in the assessment of local recurrence in rectal carcinoma.
Surg Oncol Clin N Am
1992
.
3
:
1
17
.
Roncucci
,
L.
,
R.
Fante
, and
L.
Losi
.
et al
.
Survival for colon and rectal cancer in a population-based cancer registry.
Eur J Cancer
1996
.
32
:
295
302
.
Michelassi
,
F.
,
J.
Ayala
,
T.
Balestracci
,
R.
Goldberg
,
R.
Chappell
, and
G. E.
Block
.
Verification of a new clinicopathologic staging system for colorectal adenocarcinoma.
Ann Surg
1991
.
214
:
11
18
.
Michelassi
,
F.
,
G. E.
Block
,
L.
Vannucci
,
A.
Montag
, and
R.
Chappell
.
A 5- to 21-year follow-up and analysis of 250 patients with rectal adenocarcinoma.
Ann Surg
1988
.
208
:
379
387
.
Crucitti
,
F.
,
L.
Sofo
, and
G.
Doglietto
.
et al
.
Prognostic factors in colorectal cancer: current status and new trends.
J Surg Oncol
1991
.
2
:
76
82
.
Carlon
,
C.
,
G.
Fabris
,
C.
Arslan-Pagnini
,
A. M.
Pluchinotta
,
E.
Chinelli
, and
S.
Carniato
.
Prognostic correlations of operable carcinoma of the rectum.
Dis Colon Rectum
1985
.
28
:
47
50
.
Green
,
J.
,
A.
Timmcke
,
W.
Mitchell
,
T. C.
Hicks
,
J. B.
Gathright
, and
J. E.
Ray
.
Mucinous carcinoma—Just another colon cancer?
Dis Colon Rectum
1993
.
36
:
49
54
.
Spratt
,
J.
and
H.
Spjut
.
Prevalence and prognosis of individual clinical and pathologic variables associated with colorectal carcinoma.
Cancer
1967
.
20
:
1976
1985
.
Umpleby
,
H. C.
and
R. C.
Williamson
.
Carcinoma of the large bowel in the first four decades.
Br J Surg
1984
.
71
:
272
277
.
Secco
,
G.
,
R.
Fardelli
, and
E.
Campora
.
et al
.
Primary mucinous adenocarcinomas and signet-ring cell carcinomas of colon and rectum.
Oncology
1994
.
51
:
30
34
.
Symonds
,
D.
and
A.
Vickery
.
Mucinous carcinoma of the colon and rectum.
Cancer
1976
.
37
:
1891
1900
.
Sasaki
,
O.
,
W. S.
Atkin
, and
J. R.
Jass
.
Mucinous carcinoma of the rectum.
Histopathology
1987
.
11
:
259
272
.
Shepherd
,
N.
,
E.
Saraga
,
S.
Love
, and
J. R.
Jass
.
Prognostic factors in colonic cancer.
Histopathology
1989
.
14
:
613
620
.
Jass
,
J. R.
Lymphocytic infiltration and survival in rectal cancer.
J Clin Pathol
1986
.
39
:
585
589
.
Halvorsen
,
T. B.
and
E.
Seim
.
Association between invasiveness, inflammatory reaction, desmoplasia and survival in colorectal cancer.
J Clin Pathol
1989
.
42
:
162
166
.
Deans
,
G. T.
,
M.
Heatley
,
N.
Anderson
,
C. C.
Patterson
,
B. J.
Rowlands
, and
T. G.
Parks
.
Jass' classification revisited.
J Am Coll Surg
1994
.
179
:
11
17
.
Pihl
,
E.
,
M. A.
Malahy
,
N.
Khankhanian
,
E. M.
Hersh
, and
G. M.
Mavligit
.
Immunomorphological features of prognostic significance in Dukes' class B colorectal carcinoma.
Cancer Res
1977
.
37
:
4145
4149
.
Zhou
,
X. G.
,
B. U.
Yu
, and
Y. X.
Shen
.
Surgical treatment and late results in 1226 cases of colorectal cancer.
Dis Colon Rectum
1983
.
26
:
250
256
.
Svennevig
,
J. L.
,
O. C.
Lunde
,
J.
Holter
, and
D.
Bjfrgsvik
.
Lymphoid infiltration and prognosis in colorectal carcinoma.
Br J Cancer
1984
.
49
:
375
377
.
Jass
,
J. R.
,
K-A.
Do
, and
L. A.
Simms
.
et al
.
Morphology of sporadic colorectal cancer with DNA replication errors.
Gut
1998
.
42
:
673
679
.
Messerini
,
L.
,
F.
Vitelli
, and
L. R.
DeVitis
.
et al
.
Microsatellite instability in sporadic colorectal carcinomas: relationship to clinico-pathological variables.
J Pathol
1997
.
182
:
380
384
.
Bocker
,
T.
,
J.
Schlegel
, and
F.
Kullman
.
et al
.
Genomic instability in colorectal carcinomas: comparison of different evaluation methods and their biologic significance.
J Pathol
1996
.
179
:
15
19
.
Jesserun
,
J.
,
M.
Romero-Guadarrama
, and
J. C.
Manivel
.
Medullary adenocarcinoma of the colon: clinicopathologic study of 11 cases.
Hum Pathol
1999
.
30
:
843
848
.
Öfner
,
D.
,
B.
Riedmann
, and
H.
Maier
.
et al
.
Standardized staining and analysis of argyrophilic nucleolar organizer region associated proteins (AgNORs) in radically resected colorectal adenocarcinoma-correlation with tumour stage and long-term survival.
J Pathol
1995
.
175
:
441
448
.
Rüschoff
,
J.
,
W.
Dietmaier
, and
J.
Lüttges
.
et al
.
Poorly differentiated colonic adenocarcinoma, medullary type: clinical, phenotypic and molecular characteristics.
Am J Pathol
1997
.
150
:
1815
1825
.
Jass
,
J.
,
T. C.
Smyrk
,
S. M.
Stewart
,
M. R.
Lane
,
S. J.
Lanspa
, and
H. T.
Lynch
.
Pathology of hereditary nonpolyposis colon cancer.
Anticancer Res
1994
.
14
:
1631
1634
.
Kim
,
H.
,
J.
Jen
,
B.
Vogelstein
, and
S. R.
Hamilton
.
Clinical and pathological characteristics of sporadic colon carcinomas with DNA replication errors in microsatellite sequences.
Am J Pathol
1994
.
145
:
148
156
.
Lynch
,
H. T.
,
T. C.
Smyrk
, and
P.
Watson
.
et al
.
Genetics, natural history, tumor spectrum, and pathology of hereditary non-polyposis colon cancer: an updated review.
Gastroenterology
1993
.
104
:
1535
1549
.
Mecklin
,
J. P.
,
P.
Sipponen
, and
H. J.
Jarvinen
.
Histopathology of colorectal carcinomas and adenomas in cancer family syndrome.
Dis Colon Rectum
1986
.
29
:
849
853
.
Graham
,
D. M.
and
H. D.
Appelman
.
Crohn's-like lymphoid reaction and colorectal carcinoma: a potential histological prognosticator.
Mod Pathol
1990
.
3
:
332
335
.
Shibata
,
D.
,
M. A.
Peinado
,
Y.
Ionov
,
S.
Malkhosyan
, and
M.
Perucho
.
Genomic instability in repeated sequences is an early somatic event in colorectal tumorigenesis that persists after transformation.
Nat Genet
1994
.
6
:
263
281
.
Chen
,
W. S.
,
J. Y.
Chen
,
J. M.
Liu
,
W. C.
Lin
, and
K. L.
King
.
Microsatellite instability in sporadic-colon-cancer patients with or without liver metastases.
Int J Cancer
1997
.
74
:
470
474
.
Sankila
,
R.
,
L. A.
Aaltonen
,
H. J.
Jarvinen
, and
J-P.
Mecklin
.
Better survival rates in patients with MLH1-associated hereditary colorectal cancer.
Gastroenterology
1997
.
110
:
682
687
.
Watson
,
P.
,
K. M.
Lin
, and
M. A.
Rodriguez-Bigas
.
et al
.
Colorectal carcinoma survival among hereditary nonpolyposis colorectal carcinoma family members.
Cancer
1998
.
83
:
259
266
.
Boland
,
C. R.
,
S. N.
Thibodeau
, and
S. R.
Hamilton
.
et al
.
A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer.
Cancer Res
1998
.
58
:
5248
5257
.
McLoed
,
H. L.
and
G. I.
Murray
.
Tumour markers of prognosis in colorectal cancer.
Br J Cancer
1999
.
79
:
191
203
.
O'Connell
,
M. J.
,
D. J.
Schaid
,
V.
Ganju
,
J.
Cunningham
,
J. S.
Kovach
, and
S. N.
Thibodeau
.
Current status of adjuvant chemotherapy for colorectal cancer: can molecular markers play a role in predicting prognosis?
Cancer
1992
.
70
:
1732
1739
.
Jen
,
J.
,
H.
Kim
, and
S.
Piantadosi
.
et al
.
Allelic loss of chromosome 18q and prognosis in colorectal cancer.
N Engl J Med
1994
.
331
:
213
221
.
Kato
,
M.
,
Y.
Ito
,
S.
Kobayashi
, and
K.
Isono
.
Detection of DCC and Ki-ras gene alterations in colorectal carcinoma tissue as prognostic markers for liver metastatic recurrence.
Cancer
1996
.
77
:
1729
1735
.
Itoh
,
F.
,
Y.
Hinoda
, and
M.
Ohe
.
et al
.
Decreased expression of DCC mRNA in human colorectal cancers.
Int J Cancer
1993
.
53
:
260
263
.
Shibata
,
D.
,
M. A.
Reale
, and
P.
Siverman
.
et al
.
The DCC protein and prognosis in colorectal cancer.
N Engl J Med
1996
.
335
:
1727
3172
.
Laurent-Puig
,
P.
,
S.
Olschwang
, and
O.
Delattre
.
et al
.
Survival and acquired genetic alterations in colorectal cancer.
Gastroenterology
1992
.
102
:
1136
1141
.
Carethers
,
J. M.
,
M. T.
Hawn
,
J. K.
Greenson
,
C. L.
Hitchcock
, and
C. R.
Boland
.
Prognostic significance of allelic loss at chromosome 18q21 for stage II colorectal cancer.
Gastroenterology
1998
.
114
:
1188
1195
.
Martínez-López
,
E.
,
A.
Abad
, and
A.
Font
.
et al
.
Allelic loss on chromosome 18q as a prognostic marker in stage II colorectal cancer.
Gastroenterology
1998
.
114
:
1180
1187
.
Iino
,
H.
,
M.
Fukayama
, and
Y.
Maeda
.
et al
.
Molecular genetics for clinical management of colorectal carcinoma. 17p, 18q, and 22q loss of heterozygosity and decreased DCC expression are correlated with the metastatic potential.
Cancer
1994
.
73
:
1324
1331
.
Thynne
,
G. S.
,
L. H.
Weiland
,
C. G.
Moertel
, and
A.
Silvers
.
Correlation of histopathologic characteristics of primary tumor and uninvolved regional lymph nodes in Dukes' C colonic carcinoma with prognosis.
Mayo Clin Proc
1980
.
55
:
243
245
.
Sinicrope
,
F. A.
,
J.
Hart
,
T. A.
Brasitus
,
F.
Michelassi
,
J. J.
Lee
, and
A. R.
Safa
.
Relationship of P-glycoprotein and carcinoembryonic antigen expression in human colon carcinoma to local invasion, DNA ploidy, and disease relapse.
Cancer
1994
.
74
:
2908
2917
.
Ono
,
M.
,
M.
Sakamoto
, and
Y.
Ino
.
et al
.
Cancer cell morphology at the invasive front and expression of cell adhesion-related carbohydrate in the primary lesion of patients with colorectal carcinoma with liver metastasis.
Cancer
1996
.
78
:
1179
1986
.
Hase
,
K.
,
C.
Shatney
,
D.
Johnson
,
M.
Trollope
, and
M.
Vierra
.
Prognostic value of tumor “budding”in patients with colorectal cancer.
Dis Colon Rectum
1993
.
36
:
627
635
.
Shankey
,
T. V.
,
P. S.
Rabinovitch
, and
C.
Bagwell
.
et al
.
Guidelines for implementation of clinical cytometry.
Cytometry
1993
.
14
:
472
477
.
Nori
,
D.
,
O.
Merimsky
,
D.
Saw
,
E.
Cortes
,
E.
Chen
, and
J.
Chassin
.
Tumor ploidy as a risk factor for disease recurrence and short survival in surgically treated Dukes B2 colon cancer patients.
Tumour Biol
1996
.
17
:
75
80
.
Caruso
,
M. L.
,
M.
Pirrelli
, and
R.
Armentano
.
DNA content in colorectal carcinomas: application of image analysis to archival material.
Pathologica
1994
.
86
:
376
383
.
Witzig
,
T. E.
,
C. L.
Loprinzi
, and
N. J.
Gonchoroff
.
et al
.
DNA ploidy and cell kinetic measurements as predictors or recurrence and survival in stages B2 and C colorectal adenocarcinoma.
Cancer
1991
.
68
:
879
888
.
Enker
,
W. E.
,
M. K.
Kimmel
,
E. S.
Cibas
,
M. L.
Cranor
, and
M. R.
Melamed
.
DNA/RNA content and proliferation fractions of colorectal carcinomas: a five-year prospective study relating flow cytometry to survival.
J Natl Cancer Inst
1991
.
83
:
701
707
.
Dean
,
P. A.
and
A. M.
Vernava
.
Flow cytometric analysis of DNA content in colorectal carcinoma.
Dis Colon Rectum
1992
.
35
:
95
102
.
Bauer
,
K. D.
,
C. B.
Bagwell
, and
W.
Giaretti
.
et al
.
Concensus review of utility of DNA flow cytometry in colorectal cancer.
Cytometry
1993
.
14
:
486
491
.
Fisher
,
E. R.
,
R. H.
Sideritis
,
R.
Sass
, and
B.
Fisher
.
Value of assessment of DNA ploidy in rectal cancers.
Arch Pathol Lab Med
1989
.
113
:
525
528
.
Crissman
,
J. D.
,
R. J.
Zarbo
,
C. D.
Niebylski
,
T.
Corbett
, and
D.
Weaver
.
Flow cytometric analysis of colon adenocarcinomas: a comparative study of preparatory techniques.
Mod Pathol
1988
.
1
:
198
204
.
Zarbo
,
R. J.
,
R. E.
Nakhleh
,
R. D.
Brown
,
J. J.
Kubus
,
C. K.
Ma
, and
P.
Mackowiak
.
Prognostic significance of DNA ploidy and proliferation in 309 colorectal carcinomas as determined by two-color multiparametric DNA flow cytometry.
Cancer
1997
.
79
:
2073
2086
.
Lanza
,
G.
,
R.
Gafa
, and
A.
Santini
.
et al
.
Prognostic significance of DNA ploidy in patients with stage II and stage III colon carcinoma: a prospective flow cytometric study.
Cancer
1998
.
82
:
49
59
.
Troungos
,
C.
,
C.
Valvanis
,
N.
Kapranos
, and
C.
Kittas
.
K-ras mutation in Greek patients with poorly and moderately differentiated tumours of the lower intestinal tract.
Anticancer Res
1997
.
17
:
1399
1404
.
Cerottini
,
J. P.
,
S.
Caplin
,
E.
Sarage
,
J. C.
Givel
, and
J.
Benhattar
.
The type of K-ras mutation determines prognosis in colorectal cancer.
Am J Surg
1998
.
175
:
198
202
.
Finkelstein
,
S. D.
,
R.
Sayegh
,
A.
Bakker
, and
P.
Swalsky
.
Determination of tumor aggressiveness in colorectal cancer by K-ras-2 analysis.
Arch Surg
1993
.
128
:
526
532
.
Ahnen
,
D. J.
,
P.
Feigl
, and
C.
Quan
.
et al
.
K-ras mutation and p53 overexpression predict the clinical behavior of colorectal cancer: a Southwest Oncology Group Study.
Cancer Res
1998
.
58
:
1149
1158
.
Moerkerk
,
P.
,
J. W.
Arends
,
M.
van Driel
,
A.
de Bruine
,
A.
de Goeij
, and
J.
ten Kate
.
Type and number of Ki-ras point mutations relate to stage of human colorectal cancer.
Cancer Res
1994
.
54
:
3376
3378
.
Span
,
M.
,
P. T.
Moerkerk
,
A. F.
de Goeij
, and
J. W.
Arends
.
A detailed analysis of K-ras point mutations in relation to tumor progression and survival in colorectal cancer patients.
Int J Cancer
1996
.
69
:
241
245
.
Andreyev
,
H. J.
,
A. R.
Norman
,
D.
Cunningham
,
J. R.
Oates
, and
P. A.
Clarke
.
Kirsten ras mutations in patients with colorectal cancer: the multicenter “RASCAL” study.
J Natl Cancer Inst
1998
.
90
:
675
684
.
Nemunaitis
,
J.
,
J.
Cox
,
W.
Meyer
,
A.
Courtney
, and
G.
Mues
.
Irinotecan hydrochloride (CPT-11) resistance identified by K-ras mutation in patients with progressive colon cancer after treatment with 5-fluorouracil (5-FU).
Am J Clin Oncol
1997
.
20
:
527
529
.
Johnston
,
P. G.
,
C. M.
Liang
,
S.
Henry
,
B. A.
Chapner
, and
C. J.
Allegra
.
Production and characterization of monoclonal antibodies that localize human thymidylate synthase in the cytoplasm of human cells and tissue.
Cancer Res
1991
.
51
:
6668
6676
.
Johnston
,
P. G.
,
E. R.
Fisher
, and
H. E.
Rockette
.
et al
.
The role of thymidylate synthase expression in prognosis and outcome of adjuvant chemotherapy in patients with rectal cancer.
J Clin Oncol
1994
.
12
:
2640
2647
.
Leichman
,
L.
,
H. J.
Lenz
, and
C. G.
Leichman
.
et al
.
Quantitation of intratumoral thymidylate synthase expression predicts for resistance protracted infusion of 5-fluorouracil and weekly leucovorin in disseminated colorectal cancers: preliminary report from an ongoing trial.
Eur J Cancer
1995
.
31
:
1306
1310
.
Yamachika
,
T.
,
H.
Nakanishi
, and
K.
Inada
.
et al
.
A new prognostic factor for colorectal carcinoma, thymidylate synthase, and its therapeutic significance.
Cancer
1998
.
82
:
70
77
.
Loda
,
M.
,
B.
Cukor
, and
S. W.
Tam
.
et al
.
Increased proteasome-dependent degradation of the cyclin-dependent kinase inhibitor p27 in aggressive colorectal carcinomas.
Nat Med
1997
.
3
:
231
234
.
Ayhan
,
A.
,
W.
Yasui
,
H.
Yokozaki
,
M.
Seto
,
R.
Ueda
, and
E.
Tahara
.
Loss of heterozygosity at the bcl-2 gene locus and expression of bcl-2 in human gastric and colorectal carcinomas.
Jpn J Cancer Res
1994
.
85
:
584
591
.
Bhatavdekar
,
J. M.
,
D. D.
Patel
, and
N.
Ghosh
.
et al
.
Coexpression of bcl-2, c-myc, and p53 oncoproteins as prognostic discriminants in patients with colorectal carcinoma.
Dis Colon Rectum
1997
.
40
:
785
790
.
Levine
,
A. J.
,
J.
Momand
, and
C. A.
Finlay
.
The p53 tumour suppressor gene.
Nature
1991
.
315
:
453
455
.
Lane
,
D. P.
P53, guardian of the genome.
Nature
1992
.
358
:
15
16
.
Yin
,
Y.
,
M. A.
Tainsky
,
F. Z.
Bischoff
,
L. C.
Strong
, and
G. M.
Wahl
.
Wild type p53 restores cell cycle control and inhibits gene amplification cells with mutant p53 alleles.
Cell
1992
.
70
:
937
948
.
Hartwell
,
L.
Defects in a cell cycle checkpoint may be responsible for the genomic instability of cancer cells.
Cell
1992
.
71
:
543
546
.
Kern
,
S. E.
,
E. R.
Fearon
, and
K. W. F.
Tersmette
.
Clinical and pathological associations with allelic loss in colorectal carcinoma.
JAMA
1989
.
261
:
3099
3103
.
Takanishi
,
D. M. A.
,
L.
Yaremko
, and
I.
Angriman
.
et al
.
Chromosome 17p allelic loss in colorectal cancer.
Arch Surg
1995
.
130
:
585
588
.
Swalsky
,
P. A.
and
K. I.
Bland
.
Prognostic value of TP53 and K-ras-2 mutational analysis in stage III carcinoma of the colon.
Am J Surg
1996
.
171
:
41
46
.
Hamelin
,
R.
,
P.
Laurent-Puig
, and
S.
Olschwang
.
et al
.
Association of p53 mutations with short survival in colorectal cancer.
Gastroenterology
1994
.
106
:
42
48
.
Dix
,
B. R.
,
P.
Robbins
,
R.
Soong
,
D.
Jenner
,
A. K.
House
, and
B. J.
Iacopetta
.
The common molecular genetic alterations in Dukes' B and C colorectal carcinomas are not short-term prognostic indicators of survival.
Int J Cancer
1994
.
59
:
747
751
.
Khine
,
K.
,
D.
Smith
, and
H.
Goh
.
High frequency of allelic deletion on chromosome 17 p in advanced colorectal cancer.
Cancer
1994
.
73
:
28
35
.
Diez
,
M.
,
J. M.
Enriquez
, and
J.
Camunas
.
et al
.
Prediction of recurrence in B-C stages of colorectal cancer by p53 nuclear overexpression in comparison with standard pathological features.
Eur J Surg Oncol
1995
.
21
:
635
639
.
Zeng
,
Z. S.
,
A. S.
Sarkis
, and
Z. F.
Zhang
.
et al
.
P53 nuclear overexpression: an independent predictor of survival in lymph node-positive colorectal cancer patients.
J Clin Oncol
1994
.
12
:
2043
2050
.
Flamini
,
G.
,
G.
Curigliano
, and
C.
Ratto
.
et al
.
Prognostic significance of cytoplasmic p53 overexpression in colorectal cancer: an immunohistochemical analysis.
Eur J Cancer
1996
.
32
:
802
806
.
Poller
,
D. N.
,
K. J.
Baxter
, and
N. A.
Shepherd
.
P53 and Rb1 protein expression: are they prognostically useful in colorectal cancer?
Br J Cancer
1997
.
75
:
87
93
.
Starzynska
,
T.
,
M.
Bromley
,
K.
Marlicz
,
S. A.
Roberts
,
M.
Ucinski
, and
P. L.
Stern
.
Accumulation of p53 in relation to long-term prognosis in colorectal carcinoma.
Eur J Gastroenterol Hepatol
1997
.
9
:
183
186
.
Smith
,
Dr
,
C. Y.
Ji
, and
H. S.
Goh
.
Prognostic significance of p53 overexpression and mutation in colorectal adenocarcinomas.
Br J Cancer
1996
.
74
:
216
223
.
Wiggenraad
,
R.
,
R.
Tamminga
,
P.
Blok
,
R.
Rouse
, and
J.
Hermans
.
The prognostic significance of p53 expression for survival and local control in rectal carcinoma treated with surgery and postoperative radiotherapy.
Int J Radiat Oncol Biol Phys
1998
.
41
:
29
35
.
Fante
,
R.
,
C.
Di Gregorio
,
L.
Losi
,
L.
Roncucci
, and
M.
Ponz de Leon
.
Clinico-pathological correlation and prognostic significance of nuclear p53 protein in colorectal cancer: Colorectal Cancer Study Group of the University and Health Care District of Modena.
Ital J Gastroenterol
1996
.
28
:
205
210
.
Ogunbiyi
,
O. A.
,
P. J.
Goodfellow
, and
G.
Gagliardi
.
et al
.
Prognostic value of 1p allelic loss in colon cancer.
Gastroenterology
1997
.
113
:
761
766
.
Nihei
,
Z.
,
W.
Ichikawa
, and
K.
Kojima
.
et al
.
The positive relationship between the expression of CD44 variant 6 and prognosis in colorectal cancer.
Jpn J Surg
1996
.
26
:
760
761
.
Tannapfel
,
A.
,
F.
Kockerling
,
A.
Katalinic
, and
C.
Wittekind
.
Expression of nm23-H1 predicts lymph node involvement in colorectal carcinoma.
Dis Colon Rectum
1995
.
38
:
651
654
.
Wang
,
L.
,
U.
Patel
,
L.
Ghosh
,
H-C.
Chen
, and
S.
Banjerjee
.
Mutation in the nm23 gene is associated with metastasis in colorectal cancer.
Cancer Res
1993
.
53
:
717
720
.
Campo
,
E.
,
R.
Miquel
, and
P.
Jares
.
et al
.
Prognostic significance of the loss of heterozygosity of nm23-H1 and p53 genes in human colorectal carcinomas.
Cancer
1994
.
73
:
2913
2921
.
Bell
,
S. M.
,
N.
Scott
, and
D.
Cross
.
et al
.
Prognostic value of p53 overexpression and c-Ki-ras gene mutations in colorectal cancer.
Gastroenterology
1993
.
104
:
57
64
.
Benhattar
,
J.
,
L.
Losi
,
P.
Chaubert
,
J-C.
Gival
, and
J.
Costa
.
Prognostic significance of K-ras mutations in colorectal carcinoma.
Gastroenterology
1993
.
104
:
1044
1048
.
Cohn
,
K. H.
,
D. L.
Ornstein
, and
F.
Wang
.
et al
.
The significance of allelic deletions and aneuploidy in colorectal carcinoma: results of a 5-year follow-up study.
Cancer
1997
.
79
:
233
244
.
Takebayashi
,
Y.
,
S.
Aklyama
,
K.
Yamada
,
S.
Akiba
, and
T.
Aikou
.
Angiogenesis as an unfavorable prognostic factor in human colorectal carcinoma.
Cancer
1996
.
78
:
226
231
.
Banner
,
B. F.
,
R.
Whitehouse
,
S. P.
Baker
, and
R. S.
Swanson
.
Tumor angiogenesis in stage II colorectal carcinoma: association with survival.
Am J Clin Pathol
1998
.
109
:
733
737
.
Weidner
,
N.
Tumor angiogenesis: review of current applications in tumor prognostication.
Semin Diagn Pathol
1993
.
10
:
302
313
.
Frank
,
R.
,
T. J.
Saclarides
, and
S.
Leurgans
.
et al
.
Tumor angiogenesis as a predictor of recurrence and survival in patients with node-negative colon cancer.
Ann Surg
1995
.
222
:
695
699
.
Saclarides
,
T. J.
,
N. J.
Speziale
, and
E.
Drab
.
et al
.
Tumor angiogenesis and rectal cancer.
Dis Colon Rectum
1994
.
37
:
921
926
.
Engel
,
C. J.
,
S. T.
Bennett
, and
A. F.
Chambers
.
et al
.
Tumor angiogenesis predicts recurrence in invasive colorectal cancer when controlled for Dukes' staging.
Am J Surg Pathol
1996
.
20
:
1260
1265
.
Lindmark
,
G.
,
B.
Gerdin
, and
C.
Sundberg
.
et al
.
Prognostic significance of microvascular count in colorectal cancer.
J Clin Oncol
1996
.
14
:
461
466
.
Adachi
,
Y.
,
A.
Kido
, and
M.
Mori
.
et al
.
Nuclear DNA content and nucleolar organizer regions in colorectal cancer.
J Surg Oncol
1995
.
59
:
177
180
.
Moran
,
K.
,
T.
Cooke
, and
G.
Forster
.
et al
.
Prognostic value of nucleolar organizer regions and ploidy values in advanced colorectal cancer.
Br J Surg
1989
.
76
:
1152
1155
.
Öfner
,
D.
,
M.
Totsch
, and
P.
Sandbichler
.
et al
.
Silver stained nucleolar organizer region proteins (Ag-NORs) as a predictor of prognosis in colonic cancer.
J Pathol
1990
.
162
:
43
49
.
Gaffey
,
M.
,
S.
Mills
, and
E.
Lack
.
Neuroendocrine carcinoma of the colon and rectum: a clinicopathologic, ultrastructural, and immunohistochemical study of 24 cases.
Am J Surg Pathol
1990
.
14
:
1010
1023
.
DeBruine
,
A.
,
T.
Wiggers
, and
C.
Beek
.
et al
.
Endocrine cells in colorectal adenocarcinomas: incidence, hormone profile and prognostic relevance.
Int J Cancer
1993
.
54
:
765
771
.
Griffiths
,
A. P.
,
C. W.
Butler
,
P.
Roberts
,
M. F.
Dixon
, and
P.
Quirke
.
Silver-stained structures (AgNORs), their dependence on tissue fixation and absence of prognostic relevance in rectal adenocarcinoma.
J Pathol
1989
.
159
:
121
127
.
Kram
,
N.
,
S.
Nessim
, and
S. A.
Geller
.
A study of colonic adenocarcinoma with comparison of histopathology, DNA flow cytometric data, and number of nucleolar organizer regions (NORs).
Mod Pathol
1989
.
2
:
468
472
.
Rayter
,
Z.
and
C.
Corbishley
.
The prognostic value of nucleolar organizer regions in colorectal cancer: a 5 year follow-up study.
Ann R Coll Surg Engl
1992
.
74
:
374
.
Choi
,
H. J.
,
I. K.
Jung
,
S. S.
Kim
, and
S. H.
Hong
.
Proliferating cell nuclear antigen expression and its relationship to malignancy potential in invasive colorectal carcinomas.
Dis Colon Rectum
1997
.
40
:
51
59
.
Holt
,
P. R.
,
S. F.
Moss
,
A. M.
Kapetanakis
,
A.
Petrotos
, and
S.
Wang
.
Is Ki-67 a better proliferative marker in the colon than proliferating cell nuclear antigen?
Cancer Epidemiol Biomarkers Prev
1997
.
6
:
131
135
.
Bromley
,
M.
,
D.
Rew
, and
A.
Becciolini
.
et al
.
A comparison of proliferation markers (BrdUrd, KI-67, PCNA) determined at each cell position in the crypts of normal human colonic mucosa.
Eur J Histochem
1996
.
40
:
89
100
.
Kullmann
,
F.
,
M.
Fadaie
, and
V.
Gross
.
et al
.
Expression of proliferating cell nuclear antigen (PCNA) and Ki-67 in dysplasia in inflammatory bowel disease.
Eur J Gastroenterol Hepatol
1996
.
8
:
371
379
.
Noffsinger
,
A. E.
,
M. A.
Miller
,
M. V.
Cusi
, and
C. M.
Fenoglio-Preiser
.
The pattern of cell proliferation in neoplastic and nonneoplastic lesions of ulcerative colitis.
Cancer
1996
.
78
:
2307
2312
.
Mayer
,
A.
,
M.
Takimoto
, and
E.
Fritz
.
et al
.
The prognostic significance of proliferating cell nuclear antigen, epidermal growth factor receptor, and mdr gene expression in colorectal cancer.
Cancer
1993
.
71
:
2454
2460
.
Al-Sheneber
,
I. F.
,
H. R.
Shibata
,
J.
Sampalis
, and
S.
Jothy
.
Prognostic significance of proliferating cell nuclear antigen expression in colorectal cancer.
Cancer
1993
.
71
:
1954
1959
.
Kubota
,
Y.
,
R. E.
Petras
, and
K. A.
Easley
.
et al
.
Ki-67-determined growth fraction versus standard staging and grading parameters in colorectal carcinoma.
Cancer
1992
.
70
:
2602
2609
.
Sahin
,
A. A.
,
J. Y.
Ro
, and
R. W.
Brown
.
et al
.
Assessment of Ki-67-derived tumor proliferation activity in colorectal adenocarcinomas.
Mod Pathol
1994
.
7
:
17
22
.
Neoptolemos
,
J. P.
,
K. M.
Oates
, and
K. M.
Newbold
.
et al
.
Cyclin/proliferation cell nuclear antigen immunohistochemistry does not improve the power of Dukes' or Jass' classifications for colorectal cancer.
Br J Surg
1995
.
82
:
184
187
.
Rew
,
D. A.
,
G. D.
Wilson
,
I.
Taylor
, and
P. C.
Weaver
.
Proliferation characteristics of human colorectal carcinomas measured in vitro.
Br J Surg
1991
.
78
:
60
66
.
Bleiberg
,
H.
,
M.
Buyes
,
B.
Van den Heule
, and
P.
Galand
.
Cell cycle parameters and prognosis of colorectal cancer.
Eur J Cancer Clin Oncol
1984
.
20
:
391
396
.
Wahlstrom
,
B.
,
I.
Branehog
, and
U.
Stierner
.
et al
.
Association of ploidy and cell proliferation, Dukes' classification, and histopathological differentiation in adenocarcinomas of colon and rectum.
Eur J Surg
1992
.
158
:
237
243
.
Witzig
,
T. E.
,
C. L.
Loprinzi
, and
N. J.
Gonchoroff
.
et al
.
DNA ploidy and cell measurements as predictors of recurrence and survival in stages B2 and C adenocarcinoma.
Cancer
1991
.
68
:
879
888
.
Van Oijen
,
M. G. C. T.
,
R. H.
Medema
,
P. J.
Slootweg
, and
G.
Rijken
.
Positivity of the proliferation marker Ki-67 in noncycling cells.
Am J Clin Pathol
1998
.
110
:
24
31
.
Willett
,
C. G.
,
G.
Warland
, and
R.
Cheek
.
et al
.
Proliferating cell nuclear antigen (PCNA) and mitotic activity in rectal cancer: predictor of response to preoperative irradiation.
J Clin Oncol
1994
.
12
:
679
682
.
Frank
,
R. E.
,
T. J.
Saclarides
, and
S.
Leurgans
.
et al
.
Tumor angiogenesis as a predictor of recurrence and survival in patients with node-negative colon cancer.
Ann Surg
1995
.
222
:
695
699
.
Jass
,
J.
,
Y.
Ajioka
, and
J. P.
Allen
.
et al
.
Assessment of invasive growth pattern and lymphocytic infiltration in colorectal cancer.
Histopathology
1996
.
28
:
543
548
.

Presented at the College of American Pathologists Conference XXXV: Solid Tumor Prognostic Factors: Which, How and So What?, Chicago, Ill, June 10–13, 1999.