Your activity: 6 p.v.

Pathology and prognostic determinants of colorectal cancer

Pathology and prognostic determinants of colorectal cancer
Author:
Carolyn C Compton, MD, PhD
Section Editor:
Kenneth K Tanabe, MD
Deputy Editor:
Diane MF Savarese, MD
Literature review current through: Dec 2022. | This topic last updated: Nov 23, 2022.

INTRODUCTION — Carcinoma of the colon or rectum (colorectal cancer [CRC]) is a common and lethal disease. Approximately 151,030 new cases are diagnosed each year in the United States, of which 106,180 are colon cancers and the remainder are rectal cancers [1]. Annually, approximately 52,980 Americans die of CRC, accounting for approximately 9 percent of all cancer deaths. Global, country-specific data on incidence and mortality are available from the World Health Organization (WHO) GLOBOCAN database.

Surgical resection is the primary treatment modality for early stage CRC (stage I through III) (table 1), and the most powerful tool for assessing prognosis following potentially curative surgery is pathologic analysis of the resected specimen. Although the parameters that determine pathologic stage are the strongest predictors of postoperative outcome, other clinical, molecular, and histologic features may influence prognosis independent of stage. Among patients with stage IV disease, prognosis is more closely tied to the location and extent of distant metastatic disease.

Here we will discuss the pathology of CRC and the major determinants of prognosis following surgical resection, with particular attention to the strength of the evidence supporting each factor. The molecular pathogenesis of CRC and the clinical presentation and staging evaluation for colon and rectum cancer are discussed elsewhere. (See "Molecular genetics of colorectal cancer" and "Clinical presentation, diagnosis, and staging of colorectal cancer" and "Pretreatment local staging evaluation for rectal cancer".)

PATHOLOGY

Gross appearance — Although all colorectal cancers (CRCs) originate from adenomas or flat dysplasia, they evolve into different morphologic patterns with invasion and expansion. Tumors in the proximal or right colon usually appear grossly as polypoid or fungating exophytic masses. Occult bleeding may result in the clinical presentation of an unexplained iron deficiency anemia.

By contrast, tumors involving the distal or left colon are more commonly annular or encircling lesions that produce an "apple-core" or "napkin-ring" appearance (image 1A-B). The bowel lumen becomes constricted and narrowed, producing symptoms of bowel dysfunction (eg, constipation, diarrhea, or bowel obstruction). The presence of clinical bowel obstruction or perforation of the bowel wall worsens the prognosis overall [2,3].

Despite differences in their gross appearance, right- and left-sided colon cancers are microscopically similar, and they seem to have a similar prognosis when they present with locoregional disease [4]. However, in the setting of metastatic disease, at least some data suggest a worse prognosis for those with a right-sided primary tumor [5,6].

Synchronous colon cancers, defined as two or more distinct primary tumors separated by normal bowel and not due to direct extension or metastasis, occur in 3 to 5 percent of patients with CRC [7,8]. The incidence is closer to 2.5 percent when patients with Lynch syndrome (hereditary nonpolyposis CRC [HNPCC]) are excluded [9]. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis".)

Synchronous primaries have the same prognosis as a solitary cancer when the highest stage of disease at presentation is compared [8].

Histology — The vast majority of malignant tumors of the colon and rectum are carcinomas. Other histologic types of tumors (neuroendocrine neoplasms, hamartomas, mesenchymal tumors, lymphomas) are relatively unusual. Of the carcinomas, more than 90 percent are adenocarcinomas. The most recent World Health Organization (WHO) classification of tumors of the colon and rectum is provided in the table (table 2).

Some of these morphologic variants carry prognostic significance. As an example, signet ring cell carcinomas are an aggressive adenocarcinoma subtype with a poor prognosis overall, while the medullary adenocarcinoma subtype, which is often associated with deficient mismatch repair (MMR) proteins, has a relatively favorable prognosis [10]. (See 'Mismatch repair deficiency' below.)

Histologic grade of differentiation takes into account the degree to which there are well-formed glands, an indication of the degree of cell-cell cooperation, maintenance of polarization, and coordinated secretion of cell product into a central lumen. The greater the presence of these features, the higher the degree of differentiation and the lower the grade. The inclusion of cytologic or other features in the estimation of grade is variable. Gland formation is always present to greater or lesser degrees in well-differentiated and moderately differentiated tumors (low-grade tumors when using a two-tiered grading system), respectively. By contrast, poorly differentiated or undifferentiated adenocarcinomas (high-grade tumors) do not form well-defined glandular structures, consisting predominantly of solid sheets or cords of infiltrating cells, often with marked cellular atypia, pleomorphism, and a high mitotic rate. In the majority of studies, the prognostic significance of grade falls out on statistical analysis as a two-tiered stratification variable: low grade (well and moderately differentiated) versus high grade (poorly differentiated or undifferentiated).

The College of American Pathologists (CAP) and the American Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) both specify the use of a four-tiered grading system for CRC [11,12]. This is primarily to ensure correct mapping of cancer grade in United States cancer registries, which have long used three- or four-tiered systems. The CAP recommends grade assignment based solely on the degree of gland formation, as follows [12]:

Grade 1 – Well differentiated (>95 percent gland formation)

Grade 2 – Moderately differentiated (50 to 95 percent gland formation)

Grade 3 – Poorly differentiated (<50 percent gland formation)

Grade 4 – Undifferentiated (no gland or mucin formation; no squamous or neuroendocrine differentiation)

In contrast, in the most recent (fifth) edition of the WHO Classification of Digestive System Tumours, the WHO recommends use of a two-tiered system, collapsing well and moderately differentiated into low grade, and poorly differentiated into high grade [13]. This simplifies grading but preserves the prognostic power of grade as demonstrated in most publications on the topic, which typically collapsed three- and four-tiered data into a two-tiered grade classification for prognostication analysis.

Many tumors produce mucin, which may remain within the cells (eg, signet ring cells) or be secreted. Extracellular mucin not confined within tumor glands may facilitate the dissection and penetration of a tumor through the colonic wall [14]. Tumors producing copious amounts of extracellular mucin (ie, mucin comprising ≥50 percent of the tumor mass) are classified as mucinous carcinomas (table 3). This histologic type accounts for approximately 11 to 17 percent of all CRCs [14-17]. Mucinous carcinomas have a predilection for the right side of the colon [18,19], and they may have a poor responsiveness to upfront (neoadjuvant) chemoradiotherapy [20] and adjuvant chemotherapy [21], although this is a somewhat controversial area, particularly with appendiceal mucinous neoplasms [22]. (See 'Histologic type, grade of differentiation, and presence of mucin' below.)

In some non-gland-forming carcinomas, intracellular mucin may be a dominant feature that displaces tumor cell nuclei to the side. When ≥50 percent of the tumor is made up of cells of this type, it is classified as a signet ring cell carcinoma (picture 1). It accounts for only 1 to 2 percent of all CRCs, but signet ring cell carcinoma is an aggressive variant with a propensity for extensive intramural spread and peritoneal carcinomatosis [14,23,24]. As an example, in one report of 1600 consecutive patients with CRC, 13 of the 14 patients with signet ring cell carcinomas had stage III or IV disease at diagnosis, and nine had peritoneal spread [24]. Signet ring cell carcinoma has a high incidence of microsatellite instability and a strong association with Lynch syndrome. (See 'Mismatch repair deficiency' below.)

Some cancers, particularly those arising in the distal colon, contain areas of squamous differentiation and are termed adenosquamous carcinomas [25]. These rare tumors account for between 0.05 and 0.2 percent of all colorectal malignancies and are associated with higher overall and colorectal-specific mortality as compared with adenocarcinoma [26,27].

Approximately 10 percent of CRCs, particularly poorly differentiated tumors, contain foci of neuroendocrine differentiation. Non-gland forming tumors with a predominance of neuroendocrine differentiation are classified as well-differentiated neuroendocrine (carcinoid) tumors, which have a more favorable prognosis than adenocarcinomas, and poorly differentiated neuroendocrine carcinomas, which have a poor prognosis overall. However, the prognostic importance of focal neuroendocrine differentiation within other histologic types such as an adenocarcinoma is unclear. Assessment of neuroendocrine differentiation should be made on routine hematoxylin and eosin (H&E) stained sections; the data are insufficient to recommend the use of special stains (ie, chromogranin, neuron-specific enolase, or synaptophysin). (See 'Focal neuroendocrine differentiation' below and "Pathology, classification, and grading of neuroendocrine neoplasms arising in the digestive system".)

The medullary carcinoma subtype (table 3) is a distinctive type of non-gland forming cancer composed of large eosinophilic, polygonal cells that grow in solid sheets and are heavily infiltrated by small lymphocytes (ie, tumor-infiltrating lymphocytes) [28]. The importance of recognizing this tumor type is its association with tumors that are deficient in one or more MMR proteins, including those that arise in the setting of Lynch syndrome [29]. These tumors characteristically have a high degree of microsatellite instability, frequently in combination with a BRAF mutation, and are associated with a good prognosis. Large bowel tumors with microsatellite instability are more common in the right colon; are more often of the mucinous, signet ring, or medullary histologic type; and have abundant tumor-infiltrating lymphocytes or are rimmed by a Crohn-like, germinal center-producing lymphoid reaction. (See 'Mismatch repair deficiency' below.)

Appendix cancer — Cancer of the vermiform appendix is rare. Primary malignant tumors of the appendix are found in 0.1 percent of all appendectomy specimens; the majority are carcinoids, but approximately one-third are adenocarcinomas. Significantly, metachronous or synchronous primary neoplasms, particularly involving the gastrointestinal (GI) tract, are common in patients with appendiceal tumors. (See "Epithelial tumors of the appendix" and "Well-differentiated neuroendocrine tumors of the appendix".)

Appendiceal adenocarcinomas commonly spread intraperitoneally. When these carcinomas are mucin secreting and present with peritoneal dissemination, they produce the clinical picture of a pseudomyxoma peritonei. The histologic spectrum of tumors that are associated with this clinical pattern ranges from low-grade appendiceal mucinous neoplasms (LAMNs) to invasive high-grade signet ring cell adenocarcinomas. (See "Epithelial tumors of the appendix", section on 'Peritoneal disease spread and pseudomyxoma peritonei'.)

Primary mucinous epithelial ovarian carcinoma only rarely presents similarly, but may be considered in the differential diagnosis [30]. Immunohistochemistry (IHC) can help with the distinction. (See 'Immunohistochemistry' below and "Epithelial carcinoma of the ovary, fallopian tube, and peritoneum: Clinical features and diagnosis".)

The natural history and prognosis of appendiceal adenocarcinomas differ from that of adenocarcinomas arising in other large bowel sites. As a result, the current AJCC Tumor, Node, Metastasis (TNM) cancer staging manual contains a separate staging classification for appendiceal cancers (table 4). (See "Epithelial tumors of the appendix".)

Immunohistochemistry — Cytokeratin 20 (CK20) and caudal-type homeobox 2 (CDX2) are two of the most sensitive and specific markers of intestinal differentiation and are extremely useful IHC markers in correctly identifying adenocarcinomas of colorectal origin [31-33]. IHC for CK20 and CK7 can be particularly helpful in the differential diagnosis of a primary appendiceal malignancy versus mucinous ovarian cancer (table 5) [34,35]. (See 'Appendix cancer' above.)

However, an important caveat is that unlike conventional colorectal adenocarcinomas, medullary carcinomas of the colon with high levels of microsatellite instability frequently lack CDX2 and CK20 expression, and instead, they may express markers not commonly associated with CRC, such as calretinin, CK7, SATB2, and CDH17 [36-39].

IHC is also helpful to identify deficient expression of DNA MMR. This is clinically useful in three ways:

With sporadic somatic MMR gene mutation, tumor tissue will show loss of staining for one or more of the MMR proteins, but the surrounding normal mucosa will be positive, which also serves as an internal control for the stain. Tumors with deficient MMR proteins have a relatively favorable prognosis, and this may be used in the decision-making process for adjuvant chemotherapy, particularly for patients with stage II (node-negative) colon cancer. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Molecular factors, including circulating tumor DNA'.)

Germline mutations in the MMR genes that cause Lynch syndrome typically result in a truncated or lost MMR protein that can be detected as loss of staining of the protein on tumor IHC testing [40,41]. Typically testing is carried out for MLH1, PMS2, MSH2 and MSH6. The likelihood of finding a germline mutation in one of the MMR genes based on IHC results varies depending on the protein that is absent (table 6) [42]. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis", section on 'Immunohistochemistry'.)

Deficient MMR status may identify those patients who might benefit from immunotherapy targeting the immune checkpoint programmed cell death receptor 1 (PD-1).

PATTERNS OF SPREAD AND STAGING — Colorectal cancers (CRCs) can spread by lymphatic and hematogenous dissemination, as well as contiguous and transperitoneal spread. The most common metastatic sites are the regional lymph nodes, liver, and lungs. Because the venous drainage of the intestinal tract is via the portal system, the first site of hematogenous dissemination is usually liver, followed by lungs, bone, and many other sites, including (rarely) brain. However, tumors arising in the distal rectum may metastasize initially to the lungs because the inferior rectal vein drains into the inferior vena cava rather than into the portal venous system.

TNM staging — The Tumor, Node, Metastasis (TNM) staging system of the American Joint Committee on Cancer (AJCC)/Union for International Cancer Control (UICC) is the preferred staging system for CRC [43]. The older Duke's classification, including the Astler-Coller modification, is no longer used.

As in all of the TNM staging classifications, radiographic, endoscopic (including biopsy), and intraoperative findings are used to assign a clinical stage (cT, cN, cM), while assessment of pathologic stage (pT, pN, pM) requires gross pathologic or histopathologic examination. (See "Clinical presentation, diagnosis, and staging of colorectal cancer", section on 'Staging' and "Pretreatment local staging evaluation for rectal cancer", section on 'Principles of rectal cancer staging by imaging'.)

If distant metastases are confirmed by pathologic assessment, assignment of pM1 is appropriate. Otherwise, no pM category is assigned. The designation pM0 does not exist since it would denote pathologic confirmation of the absence of distant metastases anywhere in the body (a determination possible only at autopsy).

The most recent (eighth edition, 2017) revision of the TNM staging classification is outlined in the table (table 1) [44]. Compared with the earlier 2010 version, the M1c stage has been introduced to reflect peritoneal carcinomatosis as a poor prognostic factor, and nodal micrometastases (tumor clusters >0.2 mm in diameter) are now scored as positive given the results of a meta-analysis demonstrating a poor prognosis in these patients [45]. (See 'Nodal micrometastases' below.)

This version also acknowledges the following factors, which are important to consider when making decisions about treatment, given their prognostic and predictive impact, but are not yet incorporated into the formal staging criteria:

Preoperative serum carcinoembryonic antigen (CEA) levels. (See "Clinical presentation, diagnosis, and staging of colorectal cancer", section on 'Tumor markers'.)

Tumor regression score, which reflects the pathologic response to preoperative radiation therapy, chemoradiotherapy, or chemotherapy (table 7), and status of the circumferential resection margin for rectal cancers. (See 'Tumor regression after neoadjuvant therapy' below and 'Circumferential (radial) margin' below and "Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma", section on 'Prognosis and extent of tumor regression'.)

Lymphovascular and perineural invasion. (See 'Lymphovascular invasion' below and 'Perineural invasion' below.)

Microsatellite instability, which reflects deficiency of mismatch repair enzymes and is both a prognostic factor and predictive of a lack of response to fluoropyrimidine therapy. (See 'Mismatch repair deficiency' below.)

Mutation status of KRAS, NRAS, and BRAF, because mutations in these genes are associated with treatment resistance to agents targeting the epidermal growth factor receptor (EGFR). (See 'RAS and BRAF' below.)

Regional nodes — The regional lymph nodes for each segment of the large bowel are designated in the figure (figure 1) [44]. The prognostic impact of the total number of resected and involved nodes is discussed in detail below. (See 'Regional nodes' below.)

In situ cancer — Traditionally, the term "carcinoma in situ" refers to cytologically malignant epithelial cells that do not penetrate the basement membrane or invade the underlying stroma. However, in the staging of CRC, the pTis category includes stromal invasion of malignant cells through the lamina propria into but not through the muscularis mucosa. This is unique to the large bowel and justified because the colorectal mucosa, unlike the mucosa elsewhere in the gastrointestinal (GI) tract and in other organs, lacks stromal lymphatics. As a result, tumors invading up to the muscularis mucosa lack access to the regional lymphatic vasculature and cannot metastasize via this route. It has been recommended that the terms "intraepithelial carcinoma" (without stromal invasion) and "intramucosal carcinoma" (with mucosal stromal invasion) be used to differentiate the two lesions included in the pTis category [46], but this distinction has not been made in either the current 2010 or newest 2017 TNM classifications [43,44].

Posttherapy staging for rectal cancer — The TNM staging system was initially devised for tumors that had not been previously treated. Preoperative or neoadjuvant therapy is increasingly employed for rectal tumors. Compared with initial surgery followed by adjuvant therapy, neoadjuvant therapy is associated with less long-term treatment-related toxicity, and in some cases, preservation of sphincter function. (See "Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma".)

For rectal cancers, the pathologic TNM stage of disease in the resected specimen following neoadjuvant treatment is classified differently (the ypTNM classification) (table 1) as compared with tumors that have not received neoadjuvant treatment [43]. The prognostic implication of the posttreatment pathologic stage is discussed below. (See 'Tumor regression after neoadjuvant therapy' below and "Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma".)

PROGNOSTIC DETERMINANTS — The most important indicator of outcome after resection of colorectal cancer (CRC) is the pathologic stage at presentation [47]. Five-year survival stratified by tumor stage at diagnosis for rectal cancer using the 2010 American Joint Committee on Cancer (AJCC; seventh edition of the AJCC Cancer Staging Manual) staging criteria is illustrated in the figure (figure 2) [43]. Five-year survival stratified by tumor stage at diagnosis for colon cancer using the 2010 staging criteria is illustrated in the figure (figure 3) [43]. (See 'TNM staging' above.)

Among patients receiving neoadjuvant (presurgical) treatment for rectal cancer, the posttreatment stage (given a yp designation in the AJCC staging system, see above) is a more accurate predictor of outcome than the pretreatment clinical stage. Five-year survival according to pathologic stage after chemoradiotherapy for rectal cancer is outlined in the table (table 8). (See 'Posttherapy staging for rectal cancer' above and 'Tumor regression after neoadjuvant therapy' below.)

Beyond pathologic stage at presentation, the most important prognostic determinants for CRC are the presence of extramural tumor deposits, lymphovascular and perineural invasion, histologic grade of differentiation, the preoperative level of serum carcinoembryonic antigen (CEA), microsatellite instability (MSI), and RAS and BRAF mutations.

The below sections will summarize the most recent information on prognostic determinants for CRC, according to the most recent (2017) Tumor, Node, Metastasis (TNM) staging classification from the AJCC [44].

Pathologic features

Local tumor extent — The local extent of disease (ie, depth of tumor penetration) independently influences survival [48-51]. However, evaluation and reporting of features that determine the T category is variable, particularly the presence or absence of serosal involvement. (See 'Histology' above.)

There exists significant confusion as to the definition of serosal "involvement." Histologic determination of serosal penetration is difficult, and conservative interpretation may lead to understaging of disease. As an example, cytologic examination of serosal scrapings reveals malignant cells in up to 26 percent of histologically defined pT3 specimens [51,52]. However, when there is uncertainty as to the greatest degree of tumor extension, assignment of the lesser value is justified by the general rules of AJCC staging. The AJCC Cancer Staging Manual suggests multiple-level sectioning and/or submission of additional blocks of tissue to assess serosal involvement by direct extension of tumor or through inflammation, either of which are categorized as T4a [44]. The prognostic significance of tumor that closely (<1 mm) approaches the serosal surface but does not penetrate the serosa is unclear but has been suggested to portend a higher risk of peritoneal relapse [53].

The histopathologic findings associated with peritoneal tumor involvement are heterogeneous, and standard guidelines for their diagnostic interpretation are lacking. Local peritoneal involvement may be reflected by any of the following [51]:

A mesothelial inflammatory and/or hyperplastic reaction with tumor close to the serosal surface

Tumor present at the serosal surface with an inflammatory reaction, mesothelial hyperplasia, and/or erosion or ulceration

Free tumor cells on the serosal surface within the peritoneum with underlying ulceration of the visceral peritoneum

All three types of local peritoneal involvement can be used to define serosal involvement (ie, T4a tumors [43]), and all adversely impact prognosis [29,51]. The presence of free cells on the serosal surface is more likely than the other two forms of peritoneal involvement to predict intraperitoneal recurrence and/or persistence [51]. In cases in which peritoneal involvement is uncertain, the lesser category (ie, T3) should be assigned [54,55]. (See 'TNM staging' above.)

For portions of the colorectum that are not peritonealized (eg, posterior aspects of the ascending and descending colon, lower portion of the rectum), the T4a category is not applicable (figure 4).

Among the factors that have been studied and determined not to exert a significant impact on prognosis are tumor size [49,56,57] and gross tumor configuration [56,58]. However, newer data suggest that tumor size may be an adverse prognostic factor for colon but not rectal cancer [59]. Overall size >4.5 cm was an independent predictor of poor outcome, but the optimal cut-point for size indicative of an adverse prognosis varied with anatomic location in the colon, decreasing from right to left.

Residual tumor — Residual tumor after definitive therapy is an adverse prognostic factor [29,46,60,61]. As an example, in a report of 152 patients with T4 colon cancers (table 1), 10-year recurrence-free survival was significantly less for the 42 patients with incompletely resected cancers compared with those with fully resected T4N0 or T4 node-positive disease (19 versus 88 and 58 percent, respectively) [60].

The completeness of resection is largely dependent on the status of the circumferential (radial) resection margin, although the designation is global, and includes the transverse margins and other disease observed but not removed at surgery [43]. (See 'Circumferential (radial) margin' below.)

The R designation indicates local residual disease after therapy is complete and is appropriate only in the setting of M0 disease. The residual disease codes in the TNM staging system (R0, R1, and R2) are defined as follows [62]:

R0 – Indicates complete tumor resection with all margins histologically uninvolved

R1 – Incomplete tumor resection with microscopic surgical resection margin involvement (margins grossly uninvolved)

R2 – Incomplete tumor resection with gross residual tumor that was not resected (primary tumor, regional nodes, or macroscopic margin involvement)

An expanded R classification has been proposed that takes into account the minimal distance between tumor and resection margin [63]. However, these modifications to the R classification have not yet been adopted by the AJCC, and they are not a part of either the 2010 or the 2017 edition of the TNM staging manual [43,44].

Circumferential (radial) margin — The circumferential resection margin (CRM) corresponds to the surgically dissected nonperitonealized surface of the specimen. This term applies to any aspect of the colorectum that is not covered (or partially covered, as with the ascending and descending colon and rectosigmoid) by a serosal layer of mesothelial cells and that must be dissected from the retroperitoneum (or subperitoneum in order to remove the viscus). For mid and distal rectal cancers that are entirely subperitoneal in location, the entire external surface of the specimen is considered a CRM. (See "Radical resection of rectal cancer", section on 'Radial margins'.)

By contrast, for colonic segments that are completely encased by a peritonealized (serosal) surface (eg, cecum, transverse, and sigmoid colon), the only surgical margin that is surgically dissected is the mesenteric margin, unless the cancer is adherent to or invading an adjacent organ or structure. In such cases, the CRM is a relevant radial margin only when the point of deepest tumor penetration is on the mesenteric aspect of the bowel and extends to the surface of this margin with or without serosal penetration. These concepts are illustrated in the figure (figure 5). (See "Surgical resection of primary colon cancer", section on 'Resection margins'.)

For rectal cancer, the quality of the surgical technique and the status of the CRM is one of most important predictive factors for both local and distant recurrence as well as survival [64-69]. Total mesorectal excision (TME) with adequate surgical clearance around the penetrating edge of the tumor decreases the rate of local relapse. With this approach, all mesorectal soft tissues encasing the rectum (which includes the mesentery and all regional nodes) are removed intact, and the circumferential surface is the mesorectal fascia. Guidelines for grading the quality and completeness of the mesorectum in a TME are outlined in the table (table 9) [70]. (See "Radical resection of rectal cancer", section on 'Total mesorectal excision'.)

In the analysis of the surgical specimen, the distance between the closest leading edge of the tumor and the CRM should be measured and recorded, in mm. Whether or not the CRM is designated as "positive" [71,72], there is level 1 evidence that the risk of local recurrence and death is increased if the distance between the deepest point of penetration by the tumor and the CRM is ≤1 mm [64,73-76]. In the rectum, a positive CRM denotes T3 disease with an R1 or R2 resection (ie, microscopic or macroscopic involvement of the margin by tumor) and not T4a disease (figure 4). Therefore, the College of American Pathologists (CAP) recommends recording a CRM as positive if tumor is identified 1 mm or less from the nonperitonealized surface of a resection specimen [12].

Although studies defining the relationship between CRM status and outcome are lacking for colonic cancers, it is reasonable to expect a greater risk for local tumor recurrence in any nonperitonealized segment, as with rectal cancer. For patients who have not received radiation therapy preoperatively, CRM positivity represents an indication for postoperative radiation therapy, regardless of the local tumor extent, particularly for rectal cancers. (See "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy", section on 'Indications and rationale for adjuvant therapy'.)

Regional nodes — Regional lymph node involvement is one of the strongest predictors of outcome following surgical resection of CRC, second only to the presence of distant metastasis. Nodal spread is an indication for adjuvant therapy for both colon cancer and rectal cancer to reduce the risk of lethal cancer metastases recurring in distant organs. (See "Adjuvant therapy for resected stage III (node-positive) colon cancer" and "Adjuvant therapy for resected rectal adenocarcinoma in patients not receiving neoadjuvant therapy".)

Whether fatal distant metastases are seeded by lymph node metastases or whether regional lymph nodes act as a barrier for trapping primary CRC cells that have intrinsic potential for metastatic competence is debated. Distant metastatic spread typically follows venous and not lymphatic drainage, and at least some data suggest that lymphatic and distant metastases arise from independent tumor subclones in a substantial number of cases [77,78]. These findings support the view that lymph node involvement reflects intrinsic metastatic potential.

For both colon and rectal cancers, the incidence of regional node involvement is related both to the depth of transmural invasion of the primary tumor and to histologic grade. The number of involved lymph nodes is a strong predictor of outcome [43,79-82]. As a result, the TNM classification stratifies nodal involvement according to the number of involved lymph nodes (table 1). The number of involved nodes influences outcome in both the N1 and N2 categories, regardless of the T category, both for rectal and colon cancer. In addition to the number of involved nodes, the total number of lymph nodes in the surgical specimen directly influences prognosis for both stage II (node-negative) and stage III (node-positive) disease [83-88]. These relationships are depicted in the figures (figure 6A-B) [44].

The use of a lymph node ratio (LNR; the ratio of metastatic to examined lymph nodes) has been suggested as a means of incorporating both the number of involved nodes and the total number examined into prognostic stratification [89-91]. As an example, five-year survival rates according to LNR from the INT-0089 trial are illustrated in the table (table 10) [89].

A systematic review of 16 studies including 33,984 patients with stage III colon or rectal cancer concluded that the LNR was an independent predictor of overall, disease-free, and cancer-specific survival, and that the prognostic separation obtained by the LNR was superior to that of the number of positive nodes (ie, the N stage) [91]. However, there is no consensus as to the optimal cutoff values defining the LNR category with the best outcomes. There are no prospective trials addressing this issue, and the values used in the available retrospective analyses vary widely [91].

The reason for the relationship between total number of nodes in the specimen and outcomes is unclear. The most obvious explanation is that removal of more nodes increases the accuracy of staging. However, the strong association between total lymph node count and survival is not entirely explained by improvements in staging [88,92,93]. The larger number of nodes may reflect the quality of the surgery and a more complete resection of the mesenteric pedicle. Alternatively, the differences between individual patients with colon cancer in terms of the number of lymph nodes present may reflect underlying biologic variability. As an example, tumor factors may stimulate lymph nodes to enlarge, reflecting immune system recognition of the tumor and more favorable survival outcomes with a higher number of lymph nodes that is independent of the number of metastatic lymph nodes. Such biological differences could contribute to the lack of association between average node counts, the frequency of nodal positivity, and survival in several hospital-based series [94-97] and in population-based analyses from the Surveillance, Epidemiology, and End Results (SEER) database [88,93].

Despite the disparate findings, guidelines from expert groups recommend at least 12 nodes be examined histologically to accurately determine nodal status; however, this number was derived empirically based on older observational data that was not adjusted for variables such as T-stage and tumor grade and the use of preoperative chemoradiotherapy for rectal cancer [29,83,84,98,99]. (See 'Tumor regression after neoadjuvant therapy' below.)

At least in the United States, this "benchmark" is not being met in a sizable minority of patients who undergo initial surgery, particularly those treated in community hospitals [100,101]. Rates of inadequate node retrieval seem to be improving over time in academic institutions (dropping from 38 to 5 percent between 1998 and 2005 in National Comprehensive Cancer Network [NCCN] member institutions in one report [102]).

It has long been recognized that the frequency of detecting nodal disease is influenced by the methodology of node handling and assessment, which is quite variable [103]. Surgical technique may contribute to variation in the number of nodes contained in a resection specimen [104], while diligence in the search for nodes, the use of fat clearing or other techniques to increase macroscopic visualization of nodes, and the pathologist's threshold for an acceptable number of evaluated nodes are variables in pathologic technique. If fewer than 12 nodes are present in the surgical specimen, additional techniques such as fat clearing should be used to try to increase the nodal yield [29,105].

The relevance of the 12-node threshold has been questioned for patients with rectal cancer who have received neoadjuvant therapy prior to resection. Removal of 12 or more nodes is often not achievable after neoadjuvant therapy, and lower nodal counts in this setting have not been associated with understaging or inferior survival [106]. (See "Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma".)

Extranodal extension of tumor is associated with an increased risk of recurrent disease and mortality [107]; it should be noted in the pathologic assessment of resection specimens [54,55].

Nodal micrometastases — Controversy exists as to the prognostic significance of isolated tumor cells (ITCs) or micrometastases in regional nodes.

The influence of ITCs and nodal micrometastases has been brought to the forefront by the development of sentinel node mapping and techniques such as immunohistochemistry (IHC) and reverse-transcriptase polymerase chain reaction (RT-PCR) to detect tumor-specific RNA, which detect small clusters or single tumor cells in lymph nodes [108,109]. The use of these supplemental assays may result in upstaging of up to 50 percent of patients with CRC who have histologically negative nodes, depending on the pT category [110-113]. (See "Surgical resection of primary colon cancer", section on 'Role of sentinel node mapping'.)

However, the biological significance of these micrometastases is unclear; their detection has been shown to alter prognosis in some [114] but not all studies. One reason is that some authors define ITCs as not only single-tumor cells in the subcapsular or marginal sinus, but also clumps of up to 20 tumor cells or small clusters of tumor cells measuring ≤0.2 mm in greatest dimension [115]. Micrometastases, on the other hand, are defined as clusters of tumor cells measuring ≥0.2 mm in greatest dimension. A meta-analysis demonstrated a poor prognosis for patients with tumor clusters ≥0.2 mm in diameter, but not for ITCs [45].

In the current 2010 TNM staging classification, ITCs and micrometastases do not count as positive nodal disease, although the presence of ITCs, whether detected by standard histologic techniques or IHC, are coded as pN0 (i+), and a tumor that is detected only by special molecular techniques such as RT-PCR is coded as pN0 (mol+) [43]. (See 'TNM staging' above.)

However, largely based on the meta-analysis, in the most recent (2017) eighth edition revision, which is scheduled to take effect in the United States on January 1, 2018, nodal micrometastases (tumor clusters >0.2 mm in diameter) are specifically designated as positive, while isolated tumor cells are not scored at all (table 1). Outside of the United States, the Union for International Cancer Control (UICC) has implemented the eighth edition changes as of January 1, 2017.

Extramural non-nodal tumor deposits — The seventh edition AJCC Cancer Staging Manual classification was the first to use the term "tumor deposits" to designate discrete nodules of tumor of any size or shape within the pericolic or perirectal fat or in adjacent mesentery in the lymphatic drainage area of the tumor that are not associated with identifiable lymph node or vascular structures [43]. In the eighth edition, tumor deposits are defined as "discrete tumor nodules within the lymph drainage area of the primary carcinoma without identifiable lymph node tissue or identifiable vascular or neural structure" and are codified as N1c [11]. They are considered the equivalent of nodal metastases (a lymph node replaced by tumor), even if they lack residual nodal architecture, and even in the absence of metastases in any of the identified regional lymph nodes elevate the cancer to stage III. Each tumor deposit should be counted separately, and the number recorded in the pathology report. However, the number of tumor deposits is not added to the number of positive nodes in the N category if one or more of the identified regional nodes contains metastatic tumor (see 'TNM staging' above). The most recent 2017 revision further clarifies the definition of tumor deposits (table 1) [44].

The presence of these tumor deposits is a strong adverse prognostic feature [116-122]. The adverse influence on prognosis is seen in patients both with [117,121] and without [116,118] nodal metastases. The presence of extramural extranodal tumor deposits is strongly linked to the presence of extramural venous invasion [116,120].

Tumor regression after neoadjuvant therapy — In appropriately selected patients with rectal cancer, neoadjuvant chemoradiotherapy is associated with significant tumor response and downstaging [123]. For these patients, prognosis is best determined by the posttreatment pathologic stage (ie, the ycTNM or ypTNM classification). (See 'TNM staging' above.)

Among patients receiving initial chemoradiotherapy for rectal cancer, tumor eradication, as detected by pathologic examination of the resected specimen, is associated with a significantly better prognosis as compared with patients with residual tumor, particularly residual nodal disease [124]. Similarly, minimal residual disease is associated with a better prognosis than is gross residual disease [125]. Outcomes stratified by posttreatment pathologic stage after chemoradiotherapy for rectal cancer are depicted in the table (table 8). Although several grading systems for tumor response have been advocated (table 11), a four-point tumor regression score is recommended by the AJCC (table 7) [43,44]. (See "Neoadjuvant chemoradiotherapy, radiotherapy, and chemotherapy for rectal adenocarcinoma", section on 'Prognosis and extent of tumor regression'.)

Lymphovascular invasion — Tumor invasion into veins or small nonmuscularized vessels that may represent either postcapillary lymphatics or venules is an important prognostic determinant [58,126-129]. Both venous invasion, particularly of extramural veins, and lymphatic invasion represent independent adverse prognostic factors [29,57,126,130-134].

Venous and angiolymphatic invasion should be reported as present or absent in all tumors, including malignant polyps, and the anatomic location specified as intramural or extramural. The diagnosis of lymphovascular invasion (LVI) requires identification of tumor cells either within an endothelial-lined channel or surrounded by an elastic lamina. Small vessels not definitively interpreted as lymphatics or venules should be identified as angiolymphatic vessels.

Given its prognostic importance, LVI is one of the clinicopathologic factors that is included in the definition of "high-risk" stage II colon cancer from the American Society of Clinical Oncology (ASCO) [135], National Comprehensive Cancer Network (NCCN) [136], and European Society for Medical Oncology (ESMO) (table 12) [137]. The presence of these high-risk features might influence the use of adjuvant chemotherapy in this setting. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Clinicopathologic features'.)

Perineural invasion — There is level 1 evidence that perineural invasion (PNI) is associated with a poor prognosis in multivariate analysis [128,138-144]. Given its prognostic importance, PNI is another of the clinicopathologic factors included in the definition of "high-risk" stage II colon cancer from ASCO [135], the NCCN [136], and ESMO (table 12) [137] that might influence the use of adjuvant chemotherapy in this setting. The finding of PNI should be recorded in the pathology report and designated on the TNM staging form even though it does not influence stage. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Clinicopathologic features'.)

Histologic type, grade of differentiation, and presence of mucin — As a general rule, histologic type has not proven to be an independent prognostic factor for colorectal adenocarcinomas with the exception of a few high-grade subtypes (eg, signet ring, poorly differentiated, or undifferentiated tumors) [15,56,58,145-148].

Histologic grade reflects the degree of tumor differentiation and is a feature that has consistently been demonstrated to be a stage-independent prognostic factor [48,49,56,149-152]. However, histologic grading is subjective, with significant interobserver variability and no single widely accepted, uniformly used system [153,154]. The interpretation of grade can be based on the entire tumor or by one single worst area, by the amount of gland formation alone, or by a combination of glandular formation and other structural or cytologic features. This issue is discussed in detail above. (See 'Histology' above.)

Many tumors produce mucin, which may remain within the cells (signet ring cells) or be secreted. Extracellular mucin dissects through the colonic wall, aiding the local extension of the tumor [14]. Tumors producing copious amounts of extracellular mucin (ie, mucin comprising ≥50 percent of the tumor mass) are classified as mucinous carcinomas. (See 'Histology' above.)

While signet ring cancers are clearly associated with an inferior prognosis, the influence of an extracellular mucinous component on prognosis is unclear; the available data are conflicting [148]. Tumor site, mismatch repair (MMR) status, and the effect of preoperative treatment (for rectal cancer) may influence the prognostic impact of mucinous histology:

At least some data suggest that the presence of mucin is independently associated with worse outcomes in rectal but not colonic tumors [148], but there are conflicting data about the independent prognostic impact of mucinous subtype in rectal cancer [155]. Others report that modern treatment using preoperative short-course radiation therapy or long-course chemoradiotherapy for rectal cancer may have improved outcomes, narrowing the gap between common adenocarcinomas and mucinous cancers [156].

Furthermore, tumors, particularly right-sided tumors, are associated with MMR deficiency (dMMR; the biologic footprint of which is the presence of high levels of MSI [MSI-H]), which have a relatively more favorable prognosis. This was shown in a series of 394 consecutive patients with stage III CRC who received adjuvant chemotherapy; three-year survival was significantly better for those with nonmucinous histology (79 versus 57 percent) [157]. Although the numbers were small, for the 26 patients with tumors that were deficient in one or more MMR proteins, there was no difference in outcomes for mucinous versus nonmucinous tumors. Similarly, among the 123 patients with proximal (right-sided) tumors, mucinous histology was not associated with a significantly worse outcome. (See 'Mismatch repair deficiency' below.)

Tumor border and tumor budding — The configuration of the tumor at the advancing edge (ie, the tumor border) has prognostic significance that is independent of stage. Specifically, an irregular, infiltrating pattern of growth as opposed to a smooth pushing (expansile) border has been demonstrated to be an independent adverse prognostic factor by several multivariate analyses [146,158,159]. The following characteristics are indicative of an infiltrating border [146]:

Inability to define the limits of tumor invasion and/or distinguish host tissue from malignant tissue by naked eye examination of a microscopic slide of the border

On microscopic examination of the tumor border, dissection of tumor through the full thickness of the muscularis propria without a stromal response and/or dissection of mesenteric adipose tissue by small glands or irregular clusters or cords of cells and/or perineural invasion

Tumor "budding" is another specific tumor border feature that is defined as microscopic clusters of undifferentiated cancer cells just ahead of the invasive front of the tumor [160]; this has also been referred to as "focal dedifferentiation" [154,161]. It is hypothesized that tumor budding reflects a detachment of cells and invasion of stroma at the invasive front of a carcinoma, and it is presumed to be an early step in the metastatic process. Some data suggest that tumor budding, especially when it is extensive, may be of greater prognostic value than grade and that its prognostic value is independent of the overall tumor border configuration [154,160,162]. A three-tiered grading system for tumor budding (BD1 through 3) was endorsed by the International Tumor Budding Consensus Conference [163,164], with specimens having BD3 budding (≥ 10 buds) being associated with a greater risk of recurrence in stage II colorectal cancer. A systematic review concluded that resected colorectal cancers with high levels of tumor budding are more likely to develop disease recurrence (odds ratio 5.50, 95% CI 3.64-8.29) and a cancer-related death at five years (odds ratio 4.51, 95% CI 2.55-7.99) than those without high levels of tumor budding.

High levels of tumor budding are included as one of the clinicopathologic features of high-risk stage II colon cancer that are used in the decision-making process for adjuvant chemotherapy, and according to an updated ASCO guideline on treatment of stage II cancer, patients whose tumors display this feature may be offered systemic therapy [135]. (See "Adjuvant therapy for resected stage II colon cancer".)

Host immune response — As in many other types of cancer, the presence of tumor-infiltrating lymphocytes is a favorable prognostic factor in most studies [165-174]. In particular, a high density of CD8+ T cells and CD45RO+ cells (both CD4+ and CD8+ lymphocytes that have been exposed to antigen, ie memory, cells) within these lymphoid populations has been associated with the absence of pathologic evidence of early metastatic invasion, earlier stage, and improved patient survival [166,168,169,171,173]. These data have led some to suggest that this response indicates that defensive host mechanisms were operative [166]. However, there are no direct data to support such a hypothesis.

More recently, a high density of a different type of T cell, regulatory T cells (which are characterized by the CD4+CD25+ phenotype and thought to modulate the antitumor immune response [175]), was found to have a stronger prognostic significance in CRC than either infiltrating CD8+ or CD45RO+ cells [176]. (See "Normal B and T lymphocyte development", section on 'Regulatory T cells (Tregs)'.)

Further, a Crohn disease-like lymphoid reaction characterized by peritumoral lymphoid aggregates is a common feature of MSI-H tumors [177,178] and has also been associated with an improved prognosis [174,179-181].

Several large studies of CRC have shown that tumor lymphocytic reaction and T-cell subpopulations are significant prognostic biomarkers, even after adjusting for stage, lymph node count, and well-established prognostic biomarkers, including BRAF mutation [179,182]. Lymphoid infiltration may represent a favorable prognostic marker because of its association with deficiency of MMR, the biologic footprint of which is MSI. Tumors containing numerous lymphoid cells frequently have mutations in DNA MMR genes and high levels of MSI-H [183-186]. Medullary carcinoma, a unique histologic type of CRC that is almost always MSI-H and has a strong association with Lynch syndrome, is characterized by intratumoral lymphocytic infiltrates. However, in at least one report, MSI-H and intratumoral lymphocytic infiltration were each independent prognostic factors [187]. (See 'Mismatch repair deficiency' below.)

The contribution of tumor infiltrating lymphocytes to the favorable prognosis of MSI-H tumors is further supported by studies showing that subpopulations of these cells are associated with a distinct molecular phenotype [188] and that the good prognosis of molecularly defined subgroups of CRC that are enriched for MSI-H tumors is characterized by overexpression of genes specific to cytotoxic lymphocytes. (See 'Molecular classification' below.)

Despite these convincing data, at present, the host immune response is not yet considered to represent a standard prognostic indicator for clinical use. Nevertheless, multinational efforts are underway combining tumor and immune factors to develop and validate an "immunoscore" to quantify the in situ immune infiltrate as a novel instrument for classification and prognostication of CRC [189-192].

Peritumoral fibrosis — A desmoplastic stromal response is common in invasive CRCs and is responsible for their typical hard consistency. In some [51,146,193,194], but not all, studies [195-197], fibrosis is an independent adverse prognostic factor.

Microvessel density — Intratumoral microvessel density (MVD) is a reflection of tumor-induced angiogenesis. MVD has been independently associated with shorter survival in some but not all studies. A meta-analysis of all studies relating MVD expression to prognosis concluded that at least some of the variability could be explained by the different methods of MVD assessment [198]. There was a significant inverse correlation between IHC expression and survival when MVD was assessed using antibodies against CD31 or CD34, but not factor VIII.

There is a need for validation of MVD in large studies of prognostic factors using multivariate analysis; however, standard guidelines for staining, evaluation, and interpretation of MVD are lacking [29].

Focal neuroendocrine differentiation — Although extensive neuroendocrine differentiation is an adverse prognostic factor in CRC, the significance of scattered or focal neuroendocrine differentiation is unclear; the available data are conflicting [199-204]. (See 'Histology' above.)

Tumor location — In many (but not all [205]) studies, primary tumor location is a prognostic factor in CRC [206-211]. In a meta-analysis of 66 studies including 1,427,846 patients with all stages of disease, left-sided primary tumor location (tumor location at or beyond the splenic flexure) was associated with a significantly reduced risk of death (hazard ratio [HR] 0.82, 95% CI 0.79-0.84), and this was independent of stage (although the effect size was greater for metastatic disease), race, use of adjuvant chemotherapy, year of study, and quality of the included studies [208].

Tumor location might simply be a proxy for molecular biology. As an example, in one study, mutations in BRAF or KRAS (which are associated with an inferior prognosis) were more common in proximal (right-sided) cancers, while distal (left-sided) tumors were more likely to be nonmutated [212]. Data from The Cancer Genome Atlas have demonstrated a difference in the distribution of the consensus molecular subtypes between right- and left-sided tumors [213]. (See 'Molecular classification' below.)

However, the data are inconsistent:

A better prognosis for left-sided as compared with right-sided tumors has been shown in populations with RAS wild-type tumors [210,214]. (See 'RAS and BRAF' below.)

Others have shown a better outcome for right-sided tumors that are RAS but not BRAF mutated [214].

Hypermutated tumors (ie, those with higher mutation burdens, often described as >10 mutations per megabase [215,216]) that are MSI-H (and are associated with a better outcome) are much more likely to be right sided.

Accumulating data suggest that tumor sidedness might also be a predictive factor for response to therapies that target the epidermal growth factor receptor (EGFR) in patients with RAS wild-type metastatic CRC. This subject is discussed in detail elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Anti-EGFR agent versus bevacizumab and the influence of tumor sidedness'.)

Clinical features — Certain clinical features are associated with prognosis, including high preoperative levels of the serum tumor marker carcinoembryonic antigen (CEA) and the presence of obstruction or perforation.

Preoperative serum CEA — In most (but not all [217]) studies, high levels of preoperative serum of the tumor marker CEA are of prognostic significance, although the optimal cutoff value is debated:

In several studies, CEA levels ≥5.0 ng/mL have an adverse impact on survival that is independent of tumor stage [48,128,218-225]. As an example, in a series of 17,910 patients diagnosed with colon cancer of any stage and reported to the SEER database in 2004, at a short median follow-up (27 months), an elevated preoperative CEA level was associated with a significantly increased risk of overall mortality (HR for death 1.60, 95% CI 1.46-1.76) [224]. Elevated CEA was an independent prognostic factor across all stages, and within each stage grouping, the prognosis of the subset of patients with elevated CEA was similar to or worse than a subset of patients identified as having a normal preoperative CEA level and who belonged to a higher AJCC stage grouping. An intriguing finding was that patients with node-negative disease and an elevated preoperative CEA level had a worse prognosis (HR for death 1.75, 95% CI 1.48-2.09) than did those with node-positive disease and a normal preoperative CEA level (HR for death 1.58, 95% CI 1.30-1.91).

Others suggest that a cutoff level of 3.0 ng/mL yields maximal sensitivity and specificity for recurrence [226].

Regardless of the cutoff value used, these data have led some to recommend that preoperative CEA elevation be incorporated into the conventional TNM staging system for colon cancer [29]. The current 2017 revision (table 1) does not include serum CEA in stage assignment, but recommends that the information be collected for prognostic value (and for postoperative monitoring for recurrence) [43,44]. However, while patients with node-negative cancer and a high preoperative CEA could be considered at higher than average risk for recurrence after surgery alone, and this might influence the decision to administer adjuvant chemotherapy, particularly if other risk factors (eg, obstruction, perforation) are present, there are no data that directly support benefit for adjuvant chemotherapy in this particular setting. Furthermore, at least one report refutes the association of high preoperative serum CEA levels with poor prognosis, as long as levels normalize following resection [217].

Elevated preoperative CEA levels are not considered one of the clinicopathologic factors that define "high-risk" stage II colon cancer by ASCO [135], or the NCCN [136], but they are considered a minor factor in the ESMO guidelines (table 12) [137]. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Higher-risk stage II disease'.)

Bowel obstruction and/or perforation — Many (but not all [128,227,228]) series report an adverse prognostic impact of clinical obstruction at the time of diagnosis for colon and rectal cancers [229-236]. Similarly, gross perforation is also noted to be an adverse prognostic feature in most [230,234-240] but not all [128,228,241] reports. Although many of these reports indicate that obstruction and/or perforation were independently predictive of poorer survival on multivariate analysis, some have concluded that CRCs that need emergency surgery because of issues like obstruction or perforation generally show a more aggressive histopathologic profile (more advanced stage, unfavorable histologic features) than do elective cases [242].

Both obstruction and perforation are considered to represent clinicopathologic factors that define "high-risk" stage II colon cancer from the NCCN [136], ESMO [137], and ASCO [135] (table 12). The presence of these high-risk features might influence the use of adjuvant chemotherapy in this setting. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Clinicopathologic features'.)

Molecular factors — The prognostic value of a wide variety of promising and potentially clinically applicable molecular markers has been studied in CRC. To date, the only factor that is routinely used for decision-making in clinical care is MMR deficiency. Another marker is used for its predictive capacity (RAS mutations predict a lack of efficacy for agents targeting the EGFR), and a third marker (BRAF mutations) has potential utility as a prognostic and predictive factor. An updated guideline on molecular biomarkers for the evaluation of CRC from the combined American Society for Clinical Pathology (ASCP)/CAP/Association for Molecular Pathology (AMP)/ASCO recommends the following [12,243]:

Clinicians should order MMR status testing in patients with CRC for prognostic stratification and/or for the identification of patients at high risk for Lynch syndrome. (See 'Mismatch repair deficiency' below.)

Colorectal carcinoma patients being considered for anti-EGFR therapy must receive RAS mutational testing, including KRAS and NRAS codons 12 and 13 of exon 2; 59 and 61 of exon 3; and 117 and 146 of exon 4 (expanded or extended RAS testing). (See 'RAS and BRAF' below.)

BRAF V600 mutational analysis should be performed in dMMR tumors with loss of MLH1 to evaluate for Lynch syndrome risk. The presence of a BRAF mutation strongly favors a sporadic tumor, but the absence of a BRAF mutation does not exclude the risk of Lynch syndrome. (See 'RAS and BRAF' below.)

There is insufficient evidence to recommend use of BRAF c.1799 p.V600 mutational status as a predictive molecular biomarker for response to anti-EGFR inhibitors. In support of the lack of a recommendation, the committee cited conflicting results from two meta-analyses addressing the benefit of anti-EGFR therapy in patients with RAS wild-type but BRAF-mutant CRC [244,245].

However, this is a controversial area. The most recent TNM staging classification (eighth edition, 2017) considers BRAF mutations to have both prognostic (level 1 evidence) and predictive (level 1 evidence) significance [44]. (See 'TNM staging' above.)

This subject is discussed in detail elsewhere. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'RAS wild-type, BRAF mutated tumors'.)

Mismatch repair deficiency — Mutations in one of several DNA MMR genes are found in Lynch syndrome (hereditary nonpolyposis CRC [HNPCC]) and in 15 to 20 percent of sporadic colon cancers [246]. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis".)

The characteristic genetic signature of dMMR tumors is a high number of DNA replication errors (RER+) and high levels of DNA MSI, defined as instability in ≥30 percent of microsatellite loci [247,248]. The term MSI refers to the expansion or contraction of short repeated DNA sequences that are caused by the insertion or deletion of repeated units.

Among patients with localized CRCs, tumors that are dMMR (MSI-H) are associated with longer survival than proficient MMR (pMMR) tumors that either have low microsatellite instability (MSI-L; ie, showing instability in <30 to 40 percent of microsatellite loci) or are microsatellite stable (MSS), both in Lynch-related and sporadic cases, despite often being poorly differentiated [187,247-256]. (See "Molecular genetics of colorectal cancer", section on 'Microsatellite instability high versus low'.)

The biologic basis for this finding is unknown. MSI is one of the clinically significant prognostic factors that are recommended for collection in both the current 2010 and the newest 2017 TNM staging criteria for CRC [43,44]. (See 'TNM staging' above.)

The better prognosis of dMMR tumors in early stage disease may be limited to individuals who lack a BRAF V600E mutation [257].

The prognostic influence of MSI is less clear in patients with metastatic CRC, a population in which the prevalence of MSI-H disease is low (approximately 3.5 percent) [258]. At least one report suggests an adverse influence of MSI on prognosis in this setting, a finding attributed, at least in part, to the higher frequency of BRAF mutations in this population [259,260].

In addition to the better prognosis afforded by the presence of MMR deficiency in an individual tumor, the bulk of available data support that adjuvant fluorouracil-based chemotherapy is less beneficial for patients with MSI-H (dMMR) tumors. This topic is discussed in detail elsewhere. (See "Adjuvant therapy for resected stage II colon cancer", section on 'Patients with deficient MMR'.)

MMR deficiency also identifies patients who might benefit from immunotherapy using immune checkpoint inhibitors in the setting of advanced disease.

As noted above, MMR deficiency is found in tumors from patients with Lynch syndrome (HNPCC, which accounts for one-third of all dMMR CRCs) as well as in 15 to 20 percent of sporadic colon cancers (which account for two-thirds of all dMMR CRCs). Emerging data suggest that there might be differences in the biologic behavior of these two groups. In one study, Lynch-associated dMMR patients presented with more somatic mutations and neoantigens compared with sporadic dMMR tumors, which may be associated with a stronger immune reaction and better survival overall [261]. Whether this heterogeneity will translate into a better response to immune checkpoint inhibitors in patients with advanced disease is not established. Furthermore, whether the utility of dMMR status as a predictive factor for adjuvant fluoropyrimidine therapy in earlier stage disease might differ according to the molecular mechanism underlying the loss of MMR awaits further study [262].

Methods for testing for dMMR/MSI are discussed in detail elsewhere. (See "Lynch syndrome (hereditary nonpolyposis colorectal cancer): Clinical manifestations and diagnosis", section on 'Identification of individuals at risk for Lynch syndrome'.)

RAS and BRAF — The pathogenesis of CRC involves the accumulation of genetic and epigenetic modifications within pathways that regulate proliferation, apoptosis, and angiogenesis. (See "Molecular genetics of colorectal cancer".)

RAS and BRAF mutations are of prognostic and predictive value in metastatic CRC:

KRAS mutations involving either codon 12 or 13 can be identified in 12 to 75 percent of tumors; they have been independently associated with a worse prognosis in most [263-269] (albeit not all [270-272]) studies. One potential explanation is that different mutations within the gene may exert disparate prognostic influences. As an example, the multicenter RASCAL study evaluated the prognostic significance of mutations in codon 12 or 13 in 2721 patients from 13 countries [264]. In multivariate analysis, only codon 12 mutations were independently associated with an increased risk of recurrence and death. However, in a later analysis that was expanded to include 4268 patients and 12 possible mutations within codons 12 and 13, only one specific mutation on codon 12, found in 9 percent of the cohort, was significantly associated with an adverse outcome and only in patients with node-positive disease [273].

Although less information is available, NRAS mutations are also associated with an inferior prognosis [274].

The activation of the EGFR signaling cascade is a well-described pathway leading to colon tumorigenesis. Mutations within the RAS and BRAF oncogenes located downstream of EGFR within this pathway lead to its constitutive activation, even if the EGFR is blocked. Consequently, tumors with mutated KRAS and NRAS are unresponsive to anti-EGFR therapy. (See "Systemic chemotherapy for metastatic colorectal cancer: General principles", section on 'RAS mutations'.)

BRAF activating mutations, most of which occur in codon 600 (V600E), occur in less than 10 percent of sporadic CRCs (but are especially associated with cigarette smoking) and are a strong negative prognostic marker for both early stage and advanced/recurrent tumors in non-MSI-H tumors [255,260,267,268,275-279]. For MSI-H tumors, in which most of the BRAF mutations occur, the presence of a mutation does not have the same adverse prognostic significance [269,280].

The available data also support the view that BRAF V600E mutations confer resistance to anti-EGFR therapy. (See "Molecular genetics of colorectal cancer", section on 'Hypermethylation phenotype (CIMP+) pathway'.)

Less is known about BRAF mutations that occur outside of codon 600. In one report in which 9643 patients with metastatic CRC underwent next-generation sequencing, there were 208 with non-V600 BRAF mutations (2.2 percent of the total and accounting for 22 percent [one-fifth] of all of the BRAF mutations) [281]. Compared with BRAF V600E mutations, the cancers that harbored a non-V600 BRAF mutation arose in younger individuals (58 versus 68 years), were less often found in women (46 versus 65 percent), and were less often high grade (13 versus 64 percent) or right sided (36 versus 81 percent). Furthermore, patients whose tumors had a non-V600 BRAF mutation also had significantly better median overall survival as compared with both BRAF V600E mutations and wild-type BRAF metastatic CRC (61 versus 11 versus 43 months, respectively). There are no data addressing the predictive value of non-V600 BRAF mutations for response to anti-EGFR agents.

In view of these data, the most recent TNM staging classification considers RAS and BRAF V600E mutations to have both prognostic (level 1 evidence) and predictive (level 1 evidence) significance. Therefore, testing for RAS and BRAF mutational status has become part of routine pathological evaluation for CRC greater than stage I. (See "Systemic chemotherapy for metastatic colorectal cancer: General principles", section on 'BRAF' and "Systemic chemotherapy for metastatic colorectal cancer: General principles", section on 'RAS mutations'.)

All other molecular markers — The prognostic value of a wide variety of promising and potentially clinically applicable molecular markers has been studied in CRC, including:

DNA content (aneuploidy)

Tumor suppressor genes (LOH 1p, LOH 8p; LOH 5q; SMAD4, TP53 gene)

Oncogenes other than RAS and BRAF (eg, c-myc)

Apoptosis/cell suicide-related genes (Bcl-2; BAX; Apoptosis protease activating factor-1 [APAF1])

DNA synthesis-related genes (thymidylate synthetase; thymidine phosphorylase)

Growth factors and growth factor receptor genes (transforming growth factors a and b, human epidermal growth factor receptor-2 [HER2])

Cyclins and cyclin dependent kinase inhibitor genes (p27; p21)

Angiogenesis-related genes (vascular endothelial growth factor [VEGF])

Adhesion molecule and glycoprotein genes (CD44; E-cadherin [CDH1]; sialo-Tn antigen)

Matrix metalloproteinases and their inhibitors (MMPs; urokinase-type plasminogen activator)

Metastasis suppressor genes (eg, NM23H1B)

Overexpression of, and high circulating levels of microRNA

Epigenetic aberrations such as methylation levels

Peroxisome proliferator-activated receptor-gamma (PPARG)

Circulating tumor DNA

Soluble type IV collagen

Deletions in 18q

The independent influence of these markers on prognosis remains unproven. Variability in assay methodology, conflicting results from various studies examining the same factor, and the prevalence of multiple small studies that lack statistically robust, multivariate analyses all contribute to the lack of conclusive data.

As an example, TP53, which probably represents the most widely studied gene in CRC, has been analyzed according to loss of heterozygosity, p53 protein overexpression by IHC, and detection of mutations by either direct sequencing or single strand polymorphism analysis. Despite a large number of publications and a systematic review, the impact of TP53 abnormalities on prognosis in CRC remains uncertain [282]. Even among reports that use a single-assay system, the reagents used vary considerably, confounding interpretation of the results.

Before any of these markers can be incorporated into clinically meaningful prognostic stratification systems, more studies are required using multivariate analysis, well-characterized patient populations, reproducible and current methodology, and standardized reagents. Guidelines to standardize reporting of tumor marker studies have been proposed by an international group [283].

Prognostic molecular profiles — Gene expression profiling has identified molecular signatures, such as the 12-gene recurrence score (Oncotype DX Colon Cancer Assay), that may augment conventional prognostic indicators in their ability to predict colon cancer outcomes among patients with node-negative (stage II) and node-positive (stage III) disease [284]. However, before these tools can be adopted for widespread clinical use, external validation in prospective clinical trials is needed to assess their reliability [285] and prognostic and predictive utility (ie, their ability to predict which patients would be predicted to benefit from specific chemotherapy approaches) [286].

Molecular classification — Efforts are underway to develop gene expression-based classifications of CRC. There appear to be at least three molecular pathways leading to colorectal tumorigenesis (table 13): the chromosomal instability (CIN) pathway, which is typified by the inherited condition familial adenomatous polyposis (FAP); the mutator-phenotype/DNA mismatch repair pathway, which is implicated in the inherited condition Lynch syndrome as well as in a proportion of sporadic CRCs in which there is loss of DNA MMR function; and the hypermethylation phenotype hyperplastic/serrated polyp pathway, which is characterized by a high frequency of methylation of some CpG islands (CpG island hypermethylation phenotype [CIMP]-positive) [287]. (See "Molecular genetics of colorectal cancer", section on 'Molecular pathways to colorectal tumorigenesis'.)

Several independent groups have proposed CRC subtypes based on distinct global gene expression profiles [288-291]. One proposed molecular classification system suggests the presence of four unique clinically relevant molecular subtypes with distinguishing features [288]:

CMS1 (MSI-like) – Contains most MSI-H tumors with mutations in genes encoding DNA MMR proteins, resulting in high mutational burden. The MSI-like subtype is also enriched for tumors with a CIMP and mutations in the BRAF oncogene.

CMS2 (canonical) – Subtype with high CIN as well as activation of the Wnt and MYC pathways. (See "Molecular genetics of colorectal cancer", section on 'APC gene'.)

CMS3 (metabolic) – Enriched in tumors with KRAS mutations and shows disruption of metabolic pathways.

CMS4 (mesenchymal) – Mesenchymal phenotype and frequently, CIMP phenotype.

Increasingly, molecular classification schemes such as these have been associated with prognosis and response to therapy both in the adjuvant and metastatic disease settings [286,288,292]. The CMS1 tumors have a good prognosis, the CMS4 tumors have a poor prognosis, and the CMS2 and CMS3 types have an intermediate prognosis.

Furthermore, in keeping with the prognostic importance of peritumoral inflammatory response, fibrosis, and microvessel density, the available evidence suggests that these molecular subtypes are correlated with microenvironmental (ie, immune, fibroblastic, and angiogenic) signatures [293-295]:

The good-prognosis MSI-enriched subgroup (CMS1) is characterized by overexpression of genes specific to cytotoxic lymphocytes [293]. (See 'Host immune response' above.)

The poor prognosis mesenchymal subgroup expresses markers of lymphocytes and of cells of monocytic origin, and they display an angiogenic, inflammatory, and immunosuppressive signature that is also found in other tumors, including breast and kidney cancers. This initially led to speculation that epithelial-to-mesenchymal transition of the tumor cells might be responsible for the tumor aggressiveness [289-291].

However, pathologically, these tumors have a high density of fibroblasts [293], and they share a gene program induced by transforming growth factor-beta (TGF-b) in tumor stromal cells [294], suggesting that the elevated expression of mesenchymal genes is mainly contributed by tumor-associated stromal cells and not epithelial tumor cells [294]. In fact, transcriptional signatures built to specifically report the abundance of cancer-associated fibroblasts, leucocytes, or endothelial cells all have significantly higher expression in samples of the mesenchymal subtype. Stromal cells in these tumors likely produce the chemokines and cytokines that favor tumor-associated inflammation and support angiogenesis, resulting in a poor prognosis. (See 'Peritumoral fibrosis' above.)

By contrast, the canonical and metabolic subtypes with intermediate prognosis exhibit low immune and inflammatory signatures.

While these results are intriguing and may pave the way for future molecularly based prognostic stratification systems that aid in the selection of specific therapy [296-298], molecular classification is not yet ready for incorporation into available staging systems or prognostic models, and it is difficult to reproduce at point of care since it requires analysis of a large number of genes.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Colorectal cancer".)

SUMMARY

Histology and immunohistochemistry

The vast majority of tumors of the colon and rectum are carcinomas. Other histologic types (neuroendocrine neoplasms, hamartomas, mesenchymal tumors, lymphomas) are relatively unusual. Of the carcinomas, more than 90 percent are adenocarcinomas. (See 'Histology' above.)

Cytokeratin (CK) 20 and caudal-type homeobox 2 (CDX2) are two of the most sensitive and specific markers of intestinal differentiation and are extremely useful immunohistochemistry (IHC) markers in correctly identifying adenocarcinomas of colorectal origin. IHC for CK20 and CK7 can be particularly helpful in distinguishing a primary appendiceal malignancy versus mucinous ovarian cancer (table 5). (See 'Immunohistochemistry' above.)

Prognostic determinants

Tumor stage

-The most powerful tool for assessing prognosis following potentially curative surgery for colorectal cancer (CRC) is pathologic analysis of the resected specimen. The most important pathologic characteristics are the presence of distant metastases, local tumor extent, nodal positivity (particularly the number of involved lymph nodes), residual disease, extramural tumor deposits, lymphovascular and perineural invasion, histologic grade of differentiation, and for patients treated with neoadjuvant therapy prior to resection of a rectal cancer, the tumor regression grade. (See 'Pathologic features' above.)

-The depth of tumor invasion, the status of the regional nodes, and the presence or absence of metastases are used to define the tumor stage, which is then used to derive the prognostic stage groupings. Five-year survival stratified by tumor stage at diagnosis for colon cancer using the American Joint Committee on Cancer (AJCC) 2010 staging criteria is illustrated in the figure (figure 3). Five-year survival stratified by tumor stage at diagnosis for rectal cancer using the 2010 staging criteria is illustrated in the figure (figure 2). (See 'TNM staging' above.)

Among patients receiving neoadjuvant (presurgical) treatment for rectal cancer, the posttreatment stage (given a yp designation in the AJCC staging system) is a more accurate predictor of outcome than the pretreatment clinical stage. Five-year survival according to pathologic stage after chemoradiotherapy for rectal cancer is outlined in the table (table 8). (See 'Posttherapy staging for rectal cancer' above and 'Tumor regression after neoadjuvant therapy' above.)

Other clinicopathologic features – Further stratification of outcomes can be achieved by the identification of patients with a high preoperative serum level of the tumor marker carcinoembryonic antigen (CEA), clinical obstruction at the time of diagnosis, and macroscopic perforation of the tumor. (See 'Clinical features' above.)

Molecular factors

-Molecular features may also influence outcome, independent of stage at presentation. However, the only factors that are used for clinical decision making at present are the status of the DNA mismatch repair (MMR) proteins (the biologic footprint of deficient MMR [dMMR] status is microsatellite instability), and BRAF and RAS mutations. These factors are prognostic, but dMMR status is also predictive of efficacy of fluoropyrimidine-based chemotherapy. (See 'Mismatch repair deficiency' above and 'RAS and BRAF' above.)

-Until large, statistically robust studies are completed with multivariate analysis of an array of potential prognostic factors and their interaction with each other, no other molecular or genetic markers should be used routinely to develop treatment recommendations or to estimate prognosis in patients with resected CRC. (See 'All other molecular markers' above.)

-Gene expression-based tumor classification may help clarify the importance of the tumor microenvironment to biologic behavior, and molecularly based prognostic stratification systems may someday be used to select specific therapy. However, molecular classification is not yet ready for incorporation into available staging systems or other prognostic models. (See 'Molecular classification' above.)

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022; 72:7.
  2. Carraro PG, Segala M, Cesana BM, Tiberio G. Obstructing colonic cancer: failure and survival patterns over a ten-year follow-up after one-stage curative surgery. Dis Colon Rectum 2001; 44:243.
  3. Katoh H, Yamashita K, Wang G, et al. Prognostic significance of preoperative bowel obstruction in stage III colorectal cancer. Ann Surg Oncol 2011; 18:2432.
  4. Weiss JM, Pfau PR, O'Connor ES, et al. Mortality by stage for right- versus left-sided colon cancer: analysis of surveillance, epidemiology, and end results--Medicare data. J Clin Oncol 2011; 29:4401.
  5. Price TJ, Beeke C, Ullah S, et al. Does the primary site of colorectal cancer impact outcomes for patients with metastatic disease? Cancer 2015; 121:830.
  6. Loupakis F, Yang D, Yau L, et al. Primary tumor location as a prognostic factor in metastatic colorectal cancer. J Natl Cancer Inst 2015; 107.
  7. Langevin JM, Nivatvongs S. The true incidence of synchronous cancer of the large bowel. A prospective study. Am J Surg 1984; 147:330.
  8. Passman MA, Pommier RF, Vetto JT. Synchronous colon primaries have the same prognosis as solitary colon cancers. Dis Colon Rectum 1996; 39:329.
  9. Fante R, Roncucci L, Di GregorioC, et al. Frequency and clinical features of multiple tumors of the large bowel in the general population and in patients with hereditary colorectal carcinoma. Cancer 1996; 77:2013.
  10. Knox RD, Luey N, Sioson L, et al. Medullary colorectal carcinoma revisited: a clinical and pathological study of 102 cases. Ann Surg Oncol 2015; 22:2988.
  11. AJCC Cancer Staging Manual, 8th, Amin MB (Ed), Springer, New York 2017. p.269.
  12. Updated recommendations fro the College of American Pathologists for examination of colon and rectum cancer specimens. https://documents.cap.org/protocols/cp-gilower-colonrectum-17protocol-4010.pdf (Accessed on October 08, 2019).
  13. Nagtegaal ID, Arends MJ, Salto-Tellez M. Colorectal adenocarcinoma. In: WHO Classification of Tumours: Digestive System Tumours, 5th, WHO Classification of Tumours Editorial Board (Ed), International Agency for Research on Cancer, Lyon 2019. p.177.
  14. Green JB, Timmcke AE, Mitchell WT, et al. Mucinous carcinoma--just another colon cancer? Dis Colon Rectum 1993; 36:49.
  15. Secco GB, Fardelli R, Campora E, et al. Primary mucinous adenocarcinomas and signet-ring cell carcinomas of colon and rectum. Oncology 1994; 51:30.
  16. Minsky BD, Mies C, Rich TA, et al. Colloid carcinoma of the colon and rectum. Cancer 1987; 60:3103.
  17. Minsky BD. Clinicopathologic impact of colloid in colorectal carcinoma. Dis Colon Rectum 1990; 33:714.
  18. Consorti F, Lorenzotti A, Midiri G, Di Paola M. Prognostic significance of mucinous carcinoma of colon and rectum: a prospective case-control study. J Surg Oncol 2000; 73:70.
  19. Nitsche U, Zimmermann A, Späth C, et al. Mucinous and signet-ring cell colorectal cancers differ from classical adenocarcinomas in tumor biology and prognosis. Ann Surg 2013; 258:775.
  20. Shin US, Yu CS, Kim JH, et al. Mucinous rectal cancer: effectiveness of preoperative chemoradiotherapy and prognosis. Ann Surg Oncol 2011; 18:2232.
  21. Lee DW, Han SW, Lee HJ, et al. Prognostic implication of mucinous histology in colorectal cancer patients treated with adjuvant FOLFOX chemotherapy. Br J Cancer 2013; 108:1978.
  22. Hugen N, Verhoeven RH, Radema SA, et al. Prognosis and value of adjuvant chemotherapy in stage III mucinous colorectal carcinoma. Ann Oncol 2013; 24:2819.
  23. Nissan A, Guillem JG, Paty PB, et al. Signet-ring cell carcinoma of the colon and rectum: a matched control study. Dis Colon Rectum 1999; 42:1176.
  24. Psathakis D, Schiedeck TH, Krug F, et al. Ordinary colorectal adenocarcinoma vs. primary colorectal signet-ring cell carcinoma: study matched for age, gender, grade, and stage. Dis Colon Rectum 1999; 42:1618.
  25. Frizelle FA, Hobday KS, Batts KP, Nelson H. Adenosquamous and squamous carcinoma of the colon and upper rectum: a clinical and histopathologic study. Dis Colon Rectum 2001; 44:341.
  26. Masoomi H, Ziogas A, Lin BS, et al. Population-based evaluation of adenosquamous carcinoma of the colon and rectum. Dis Colon Rectum 2012; 55:509.
  27. Petrelli NJ, Valle AA, Weber TK, Rodriguez-Bigas M. Adenosquamous carcinoma of the colon and rectum. Dis Colon Rectum 1996; 39:1265.
  28. Jass JR, Sobin LH. Histological typing of intestinal tumours. In: WHO International Histological Classification of Tumours, 2nd, Springer-Verlag, Berlin-New York 1989.
  29. Compton CC, Fielding LP, Burgart LJ, et al. Prognostic factors in colorectal cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 2000; 124:979.
  30. Ronnett BM, Kurman RJ, Shmookler BM, et al. The morphologic spectrum of ovarian metastases of appendiceal adenocarcinomas: a clinicopathologic and immunohistochemical analysis of tumors often misinterpreted as primary ovarian tumors or metastatic tumors from other gastrointestinal sites. Am J Surg Pathol 1997; 21:1144.
  31. Werling RW, Yaziji H, Bacchi CE, Gown AM. CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas. Am J Surg Pathol 2003; 27:303.
  32. Moll R, Zimbelmann R, Goldschmidt MD, et al. The human gene encoding cytokeratin 20 and its expression during fetal development and in gastrointestinal carcinomas. Differentiation 1993; 53:75.
  33. Landau MS, Kuan SF, Chiosea S, Pai RK. BRAF-mutated microsatellite stable colorectal carcinoma: an aggressive adenocarcinoma with reduced CDX2 and increased cytokeratin 7 immunohistochemical expression. Hum Pathol 2014; 45:1704.
  34. Guerrieri C, Frånlund B, Fristedt S, et al. Mucinous tumors of the vermiform appendix and ovary, and pseudomyxoma peritonei: histogenetic implications of cytokeratin 7 expression. Hum Pathol 1997; 28:1039.
  35. Chu P, Wu E, Weiss LM. Cytokeratin 7 and cytokeratin 20 expression in epithelial neoplasms: a survey of 435 cases. Mod Pathol 2000; 13:962.
  36. Hinoi T, Tani M, Lucas PC, et al. Loss of CDX2 expression and microsatellite instability are prominent features of large cell minimally differentiated carcinomas of the colon. Am J Pathol 2001; 159:2239.
  37. McGregor DK, Wu TT, Rashid A, et al. Reduced expression of cytokeratin 20 in colorectal carcinomas with high levels of microsatellite instability. Am J Surg Pathol 2004; 28:712.
  38. Winn B, Tavares R, Fanion J, et al. Differentiating the undifferentiated: immunohistochemical profile of medullary carcinoma of the colon with an emphasis on intestinal differentiation. Hum Pathol 2009; 40:398.
  39. Lin F, Shi J, Zhu S, et al. Cadherin-17 and SATB2 are sensitive and specific immunomarkers for medullary carcinoma of the large intestine. Arch Pathol Lab Med 2014; 138:1015.
  40. Weissman SM, Bellcross C, Bittner CC, et al. Genetic counseling considerations in the evaluation of families for Lynch syndrome--a review. J Genet Couns 2011; 20:5.
  41. Shia J. Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn 2008; 10:293.
  42. Hampel H, Frankel WL, Martin E, et al. Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol 2008; 26:5783.
  43. AJCC Cancer Staging Manual, 7th ed, Edge SB, Byrd DR, Compton CC, et al (Eds), Springer, New York 2010. p.143.
  44. Jessup JM, Goldberg RM, Asare EA, et al. Colon and Rectum. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), AJCC, Chicago 2017. p.251.
  45. Sloothaak DA, Sahami S, van der Zaag-Loonen HJ, et al. The prognostic value of micrometastases and isolated tumour cells in histologically negative lymph nodes of patients with colorectal cancer: a systematic review and meta-analysis. Eur J Surg Oncol 2014; 40:263.
  46. Compton C, Fenoglio-Preiser CM, Pettigrew N, Fielding LP. American Joint Committee on Cancer Prognostic Factors Consensus Conference: Colorectal Working Group. Cancer 2000; 88:1739.
  47. AJCC (American Joint Committee on Cancer) Cancer Staging Manual, 7th ed, Edge, SB, Byrd, DR, Compton, CC, et al (Eds), Springer, New York 2010. p.133.
  48. Wiggers T, Arends JW, Volovics A. Regression analysis of prognostic factors in colorectal cancer after curative resections. Dis Colon Rectum 1988; 31:33.
  49. Chapuis PH, Dent OF, Fisher R, et al. A multivariate analysis of clinical and pathological variables in prognosis after resection of large bowel cancer. Br J Surg 1985; 72:698.
  50. Tominaga T, Sakabe T, Koyama Y, et al. Prognostic factors for patients with colon or rectal carcinoma treated with resection only. Five-year follow-up report. Cancer 1996; 78:403.
  51. Shepherd NA, Baxter KJ, Love SB. The prognostic importance of peritoneal involvement in colonic cancer: a prospective evaluation. Gastroenterology 1997; 112:1096.
  52. Zeng Z, Cohen AM, Hajdu S, et al. Serosal cytologic study to determine free mesothelial penetration of intraperitoneal colon cancer. Cancer 1992; 70:737.
  53. Panarelli NC, Schreiner AM, Brandt SM, et al. Histologic features and cytologic techniques that aid pathologic stage assessment of colonic adenocarcinoma. Am J Surg Pathol 2013; 37:1252.
  54. Washington MK, Berlin J, Branton PA, et al. Protocol for the examination of specimens from patients with primary carcinomas of the colon and rectum. Arch Pathol Lab Med 2008; 132:1182.
  55. http://www.cap.org/ShowProperty?nodePath=/UCMCon/Contribution Folders/WebContent/pdf/cp-colon-16protocol-3400.pdf (Accessed on January 12, 2017).
  56. Newland RC, Dent OF, Lyttle MN, et al. Pathologic determinants of survival associated with colorectal cancer with lymph node metastases. A multivariate analysis of 579 patients. Cancer 1994; 73:2076.
  57. Takahashi Y, Tucker SL, Kitadai Y, et al. Vessel counts and expression of vascular endothelial growth factor as prognostic factors in node-negative colon cancer. Arch Surg 1997; 132:541.
  58. Michelassi F, Ayala JJ, Balestracci T, et al. Verification of a new clinicopathologic staging system for colorectal adenocarcinoma. Ann Surg 1991; 214:11.
  59. Kornprat P, Pollheimer MJ, Lindtner RA, et al. Value of tumor size as a prognostic variable in colorectal cancer: a critical reappraisal. Am J Clin Oncol 2011; 34:43.
  60. Willett CG, Goldberg S, Shellito PC, et al. Does postoperative irradiation play a role in the adjuvant therapy of stage T4 colon cancer? Cancer J Sci Am 1999; 5:242.
  61. Wittekind C, Compton CC, Greene FL, Sobin LH. TNM residual tumor classification revisited. Cancer 2002; 94:2511.
  62. Gress DM, Edge SB, Greene FL, et al.. Principles of Cancer Staging. In: AJCC Cancer Staging Manual, 8th, Amin MB (Ed), AJCC, Chicago 2017. p.29.
  63. Wittekind C, Compton C, Quirke P, et al. A uniform residual tumor (R) classification: integration of the R classification and the circumferential margin status. Cancer 2009; 115:3483.
  64. Nagtegaal ID, Quirke P. What is the role for the circumferential margin in the modern treatment of rectal cancer? J Clin Oncol 2008; 26:303.
  65. Adam IJ, Mohamdee MO, Martin IG, et al. Role of circumferential margin involvement in the local recurrence of rectal cancer. Lancet 1994; 344:707.
  66. Stocchi L, Nelson H, Sargent DJ, et al. Impact of surgical and pathologic variables in rectal cancer: a United States community and cooperative group report. J Clin Oncol 2001; 19:3895.
  67. Quirke P, Durdey P, Dixon MF, Williams NS. Local recurrence of rectal adenocarcinoma due to inadequate surgical resection. Histopathological study of lateral tumour spread and surgical excision. Lancet 1986; 2:996.
  68. de Haas-Kock DF, Baeten CG, Jager JJ, et al. Prognostic significance of radial margins of clearance in rectal cancer. Br J Surg 1996; 83:781.
  69. Gosens MJ, Klaassen RA, Tan-Go I, et al. Circumferential margin involvement is the crucial prognostic factor after multimodality treatment in patients with locally advanced rectal carcinoma. Clin Cancer Res 2007; 13:6617.
  70. Parfitt JR, Driman DK. The total mesorectal excision specimen for rectal cancer: a review of its pathological assessment. J Clin Pathol 2007; 60:849.
  71. Nagtegaal ID, Marijnen CA, Kranenbarg EK, et al. Circumferential margin involvement is still an important predictor of local recurrence in rectal carcinoma: not one millimeter but two millimeters is the limit. Am J Surg Pathol 2002; 26:350.
  72. Bernstein TE, Endreseth BH, Romundstad P, et al. Circumferential resection margin as a prognostic factor in rectal cancer. Br J Surg 2009; 96:1348.
  73. Jessup JM, Goldberg RM, Aware EA, et al. Colon and Rectum. In: AJCC Cancer Staging Manual, 8th ed, Amin MB (Ed), AJCC, Chicago 2017. p.251. Corrected at 4th printing, 2018.
  74. Yasuda K, Adachi Y, Shiraishi N, et al. Pattern of lymph node micrometastasis and prognosis of patients with colorectal cancer. Ann Surg Oncol 2001; 8:300.
  75. Clarke G, Ryan E, O'Keane JC, et al. The detection of cytokeratins in lymph nodes of Duke's B colorectal cancer subjects predicts a poor outcome. Eur J Gastroenterol Hepatol 2000; 12:549.
  76. Tschmelitsch J, Klimstra DS, Cohen AM. Lymph node micrometastases do not predict relapse in stage II colon cancer. Ann Surg Oncol 2000; 7:601.
  77. Markowitz SD. Cancer bypasses the lymph nodes. Science 2017; 357:35.
  78. Naxerova K, Reiter JG, Brachtel E, et al. Origins of lymphatic and distant metastases in human colorectal cancer. Science 2017; 357:55.
  79. Wolmark N, Fisher B, Wieand HS. The prognostic value of the modifications of the Dukes' C class of colorectal cancer. An analysis of the NSABP clinical trials. Ann Surg 1986; 203:115.
  80. Greene FL, Stewart AK, Norton HJ. A new TNM staging strategy for node-positive (stage III) colon cancer: an analysis of 50,042 patients. Ann Surg 2002; 236:416.
  81. Swanson RS, Compton CC, Stewart AK, Bland KI. The prognosis of T3N0 colon cancer is dependent on the number of lymph nodes examined. Ann Surg Oncol 2003; 10:65.
  82. Chen SL, Bilchik AJ. More extensive nodal dissection improves survival for stages I to III of colon cancer: a population-based study. Ann Surg 2006; 244:602.
  83. Tepper JE, O'Connell MJ, Niedzwiecki D, et al. Impact of number of nodes retrieved on outcome in patients with rectal cancer. J Clin Oncol 2001; 19:157.
  84. Chang GJ, Rodriguez-Bigas MA, Skibber JM, Moyer VA. Lymph node evaluation and survival after curative resection of colon cancer: systematic review. J Natl Cancer Inst 2007; 99:433.
  85. Johnson PM, Porter GA, Ricciardi R, Baxter NN. Increasing negative lymph node count is independently associated with improved long-term survival in stage IIIB and IIIC colon cancer. J Clin Oncol 2006; 24:3570.
  86. Le Voyer TE, Sigurdson ER, Hanlon AL, et al. Colon cancer survival is associated with increasing number of lymph nodes analyzed: a secondary survey of intergroup trial INT-0089. J Clin Oncol 2003; 21:2912.
  87. Compton CC. Optimal pathologic staging: defining stage II disease. Clin Cancer Res 2007; 13:6862s.
  88. Baxter NN, Ricciardi R, Simunovic M, et al. An evaluation of the relationship between lymph node number and staging in pT3 colon cancer using population-based data. Dis Colon Rectum 2010; 53:65.
  89. Berger AC, Sigurdson ER, LeVoyer T, et al. Colon cancer survival is associated with decreasing ratio of metastatic to examined lymph nodes. J Clin Oncol 2005; 23:8706.
  90. Meyers MO, Hollis DR, Mayer RJ, et al. Ratio of metastatic to examined lymph nodes is a powerful predictor of overall survival in rectal cancer: An analysis of Intergroup 0114 (abstract). J Clin Oncol 2007; 25:165s.
  91. Ceelen W, Van Nieuwenhove Y, Pattyn P. Prognostic value of the lymph node ratio in stage III colorectal cancer: a systematic review. Ann Surg Oncol 2010; 17:2847.
  92. Moore J, Hyman N, Callas P, Littenberg B. Staging error does not explain the relationship between the number of lymph nodes in a colon cancer specimen and survival. Surgery 2010; 147:358.
  93. Parsons HM, Tuttle TM, Kuntz KM, et al. Association between lymph node evaluation for colon cancer and node positivity over the past 20 years. JAMA 2011; 306:1089.
  94. Wong SL, Ji H, Hollenbeck BK, et al. Hospital lymph node examination rates and survival after resection for colon cancer. JAMA 2007; 298:2149.
  95. Tsikitis VL, Larson DL, Wolff BG, et al. Survival in stage III colon cancer is independent of the total number of lymph nodes retrieved. J Am Coll Surg 2009; 208:42.
  96. Prandi M, Lionetto R, Bini A, et al. Prognostic evaluation of stage B colon cancer patients is improved by an adequate lymphadenectomy: results of a secondary analysis of a large scale adjuvant trial. Ann Surg 2002; 235:458.
  97. Bui L, Rempel E, Reeson D, Simunovic M. Lymph node counts, rates of positive lymph nodes, and patient survival for colon cancer surgery in Ontario, Canada: a population-based study. J Surg Oncol 2006; 93:439.
  98. National voluntary consensus standards for quality of cancer care from the National Quality Forum available online at www.qualityforum.org/pdf/cancer/txAppA-Specifications_web.pdf (Accessed on May 02, 2011).
  99. de Campos-Lobato LF, Stocchi L, de Sousa JB, et al. Less than 12 nodes in the surgical specimen after total mesorectal excision following neoadjuvant chemoradiation: it means more than you think! Ann Surg Oncol 2013; 20:3398.
  100. Baxter NN, Virnig DJ, Rothenberger DA, et al. Lymph node evaluation in colorectal cancer patients: a population-based study. J Natl Cancer Inst 2005; 97:219.
  101. Bilimoria KY, Bentrem DJ, Stewart AK, et al. Lymph node evaluation as a colon cancer quality measure: a national hospital report card. J Natl Cancer Inst 2008; 100:1310.
  102. Chang GJ, Taylor SH, Rodriguez-Bigas MA, Skibber JM, et al. The twelve lymph node standard in colon cancer: Can it be achieved? (abstract 448). Data presented at the 2008 ASCO GI Cancers Symposium, Orlando, FL, January 24, 2008.
  103. Morris EJ, Maughan NJ, Forman D, Quirke P. Identifying stage III colorectal cancer patients: the influence of the patient, surgeon, and pathologist. J Clin Oncol 2007; 25:2573.
  104. West NP, Hohenberger W, Weber K, et al. Complete mesocolic excision with central vascular ligation produces an oncologically superior specimen compared with standard surgery for carcinoma of the colon. J Clin Oncol 2010; 28:272.
  105. Abbassi-Ghadi N, Boshier PR, Goldin R, Hanna GB. Techniques to increase lymph node harvest from gastrointestinal cancer specimens: a systematic review and meta-analysis. Histopathology 2012; 61:531.
  106. Govindarajan A, Gönen M, Weiser MR, et al. Challenging the feasibility and clinical significance of current guidelines on lymph node examination in rectal cancer in the era of neoadjuvant therapy. J Clin Oncol 2011; 29:4568.
  107. Veronese N, Nottegar A, Pea A, et al. Prognostic impact and implications of extracapsular lymph node involvement in colorectal cancer: a systematic review with meta-analysis. Ann Oncol 2016; 27:42.
  108. Waldman S, Hyslop T, Schulz S, et al. A prospective multicenter study of guanyl cyclase C (GCC), quantified by the reverse transcriptase-polymerase chain reaction (qRT-PCR), as a prognostic marker of occult metastases in lymph nodes of pN0 colorectal cancer patients (abstract). J Clin Oncol 2008; 26:580s.
  109. Sargent DJ, Shi Q, Gill S, et al. Molecular testing for lymph node metastases as a determinant of colon cancer recurrence: results from a retrospective multicenter study. Clin Cancer Res 2014; 20:4361.
  110. Paramo JC, Summerall J, Wilson C, et al. Intraoperative sentinel lymph node mapping in patients with colon cancer. Am J Surg 2001; 182:40.
  111. Bernini A, Spencer M, Frizelle S, et al. Evidence for colorectal cancer micrometastases using reverse transcriptase-polymerase chain reaction analysis of MUC2 in lymph nodes. Cancer Detect Prev 2000; 24:72.
  112. Bilchik AJ, Nora DT, Sobin LH, et al. Effect of lymphatic mapping on the new tumor-node-metastasis classification for colorectal cancer. J Clin Oncol 2003; 21:668.
  113. Gill S, Haince JF, Shi Q, et al. Prognostic Value of Molecular Detection of Lymph Node Metastases After Curative Resection of Stage II Colon Cancer: A Systematic Pooled Data Analysis. Clin Colorectal Cancer 2015; 14:99.
  114. Rahbari NN, Bork U, Motschall E, et al. Molecular detection of tumor cells in regional lymph nodes is associated with disease recurrence and poor survival in node-negative colorectal cancer: a systematic review and meta-analysis. J Clin Oncol 2012; 30:60.
  115. Mescoli C, Albertoni L, Pucciarelli S, et al. Isolated tumor cells in regional lymph nodes as relapse predictors in stage I and II colorectal cancer. J Clin Oncol 2012; 30:965.
  116. Goldstein NS, Turner JR. Pericolonic tumor deposits in patients with T3N+MO colon adenocarcinomas: markers of reduced disease free survival and intra-abdominal metastases and their implications for TNM classification. Cancer 2000; 88:2228.
  117. Lo DS, Pollett A, Siu LL, et al. Prognostic significance of mesenteric tumor nodules in patients with stage III colorectal cancer. Cancer 2008; 112:50.
  118. Belt EJ, van Stijn MF, Bril H, et al. Lymph node negative colorectal cancers with isolated tumor deposits should be classified and treated as stage III. Ann Surg Oncol 2010; 17:3203.
  119. Nagtegaal ID, Knijn N, Hugen N, et al. Tumor Deposits in Colorectal Cancer: Improving the Value of Modern Staging-A Systematic Review and Meta-Analysis. J Clin Oncol 2017; 35:1119.
  120. Lord AC, D'Souza N, Pucher PH, et al. Significance of extranodal tumour deposits in colorectal cancer: A systematic review and meta-analysis. Eur J Cancer 2017; 82:92.
  121. Delattre JF, Cohen R, Henriques J, et al. Prognostic Value of Tumor Deposits for Disease-Free Survival in Patients With Stage III Colon Cancer: A Post Hoc Analysis of the IDEA France Phase III Trial (PRODIGE-GERCOR). J Clin Oncol 2020; 38:1702.
  122. Peacock O, Limvorapitak T, Hu C-Y, et al. Improving the AJCC/TNM staging classification for colorectal cancer: The prognostic impact of tumor deposits (abstract). J Clin Oncol 38:2020 (suppl; abstr 4012. Abstract available online at https://meetinglibrary.asco.org/record/185534/abstract (Accessed on June 15, 2020).
  123. Ruo L, Tickoo S, Klimstra DS, et al. Long-term prognostic significance of extent of rectal cancer response to preoperative radiation and chemotherapy. Ann Surg 2002; 236:75.
  124. Gavioli M, Luppi G, Losi L, et al. Incidence and clinical impact of sterilized disease and minimal residual disease after preoperative radiochemotherapy for rectal cancer. Dis Colon Rectum 2005; 48:1851.
  125. Ryan R, Gibbons D, Hyland JM, et al. Pathological response following long-course neoadjuvant chemoradiotherapy for locally advanced rectal cancer. Histopathology 2005; 47:141.
  126. Betge J, Pollheimer MJ, Lindtner RA, et al. Intramural and extramural vascular invasion in colorectal cancer: prognostic significance and quality of pathology reporting. Cancer 2012; 118:628.
  127. Hogan J, Chang KH, Duff G, et al. Lymphovascular invasion: a comprehensive appraisal in colon and rectal adenocarcinoma. Dis Colon Rectum 2015; 58:547.
  128. Quah HM, Chou JF, Gonen M, et al. Identification of patients with high-risk stage II colon cancer for adjuvant therapy. Dis Colon Rectum 2008; 51:503.
  129. Sternberg A, Sibirsky O, Cohen D, et al. Validation of a new classification system for curatively resected colorectal adenocarcinoma. Cancer 1999; 86:782.
  130. Takebayashi Y, Aklyama S, Yamada K, et al. Angiogenesis as an unfavorable prognostic factor in human colorectal carcinoma. Cancer 1996; 78:226.
  131. Santos C, López-Doriga A, Navarro M, et al. Clinicopathological risk factors of Stage II colon cancer: results of a prospective study. Colorectal Dis 2013; 15:414.
  132. Lim SB, Yu CS, Jang SJ, et al. Prognostic significance of lymphovascular invasion in sporadic colorectal cancer. Dis Colon Rectum 2010; 53:377.
  133. Amri R, England J, Bordeianou LG, Berger DL. Risk Stratification in Patients with Stage II Colon Cancer. Ann Surg Oncol 2016; 23:3907.
  134. Siddiqui MRS, Simillis C, Hunter C, et al. A meta-analysis comparing the risk of metastases in patients with rectal cancer and MRI-detected extramural vascular invasion (mrEMVI) vs mrEMVI-negative cases. Br J Cancer 2017; 116:1513.
  135. Baxter NN, Kennedy EB, Bergsland E, et al. Adjuvant Therapy for Stage II Colon Cancer: ASCO Guideline Update. J Clin Oncol 2022; 40:892.
  136. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology. Available at: https://www.nccn.org/professionals/physician_gls (Accessed on May 18, 2022).
  137. Argilés G, Tabernero J, Labianca R, et al. Localised colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2020; 31:1291.
  138. Liebig C, Ayala G, Wilks J, et al. Perineural invasion is an independent predictor of outcome in colorectal cancer. J Clin Oncol 2009; 27:5131.
  139. Peng J, Sheng W, Huang D, et al. Perineural invasion in pT3N0 rectal cancer: the incidence and its prognostic effect. Cancer 2011; 117:1415.
  140. Huh JW, Kim HR, Kim YJ. Prognostic value of perineural invasion in patients with stage II colorectal cancer. Ann Surg Oncol 2010; 17:2066.
  141. Fujita S, Shimoda T, Yoshimura K, et al. Prospective evaluation of prognostic factors in patients with colorectal cancer undergoing curative resection. J Surg Oncol 2003; 84:127.
  142. Jessup JM, Goldberg RM, Asare EA, et al. Colon and Rectum. In: AJCC Cancer Staging Manual, 8th, Amin MB (Ed), AJCC, Chicago 2017. p.265.
  143. Cienfuegos JA, Martínez P, Baixauli J, et al. Perineural Invasion is a Major Prognostic and Predictive Factor of Response to Adjuvant Chemotherapy in Stage I-II Colon Cancer. Ann Surg Oncol 2017; 24:1077.
  144. Alotaibi AM, Lee JL, Kim J, et al. Prognostic and Oncologic Significance of Perineural Invasion in Sporadic Colorectal Cancer. Ann Surg Oncol 2017; 24:1626.
  145. Harrison JC, Dean PJ, el-Zeky F, Vander Zwaag R. From Dukes through Jass: pathological prognostic indicators in rectal cancer. Hum Pathol 1994; 25:498.
  146. Halvorsen TB, Seim E. Association between invasiveness, inflammatory reaction, desmoplasia and survival in colorectal cancer. J Clin Pathol 1989; 42:162.
  147. Ofner D, Riedmann B, Maier H, 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.
  148. Hyngstrom JR, Hu CY, Xing Y, et al. Clinicopathology and outcomes for mucinous and signet ring colorectal adenocarcinoma: analysis from the National Cancer Data Base. Ann Surg Oncol 2012; 19:2814.
  149. Hermanek P, Guggenmoos-Holzmann I, Gall FP. Prognostic factors in rectal carcinoma. A contribution to the further development of tumor classification. Dis Colon Rectum 1989; 32:593.
  150. Jessup JM, Stewart AK, Menck HR. The National Cancer Data Base report on patterns of care for adenocarcinoma of the rectum, 1985-95. Cancer 1998; 83:2408.
  151. Scott NA, Wieand HS, Moertel CG, et al. Colorectal cancer. Dukes' stage, tumor site, preoperative plasma CEA level, and patient prognosis related to tumor DNA ploidy pattern. Arch Surg 1987; 122:1375.
  152. Fisher ER, Sass R, Palekar A, et al. Dukes' classification revisited. Findings from the National Surgical Adjuvant Breast and Bowel Projects (Protocol R-01). Cancer 1989; 64:2354.
  153. Blenkinsopp WK, Stewart-Brown S, Blesovsky L, et al. Histopathology reporting in large bowel cancer. J Clin Pathol 1981; 34:509.
  154. Goldstein NS, Hart J. Histologic features associated with lymph node metastasis in stage T1 and superficial T2 rectal adenocarcinomas in abdominoperineal resection specimens. Identifying a subset of patients for whom treatment with adjuvant therapy or completion abdominoperineal resection should be considered after local excision. Am J Clin Pathol 1999; 111:51.
  155. Tarantino I, Hüttner FJ, Warschkow R, et al. Prognostic Relevance of Mucinous Subtype in a Population-based Propensity Score Analysis of 40,083 Rectal Cancer Patients. Ann Surg Oncol 2016; 23:1576.
  156. Hugen N, van de Velde CJ, Bosch SL, et al. Modern Treatment of Rectal Cancer Closes the Gap Between Common Adenocarcinoma and Mucinous Carcinoma. Ann Surg Oncol 2015; 22:2669.
  157. Kim SH, Shin SJ, Lee KY, et al. Prognostic value of mucinous histology depends on microsatellite instability status in patients with stage III colon cancer treated with adjuvant FOLFOX chemotherapy: a retrospective cohort study. Ann Surg Oncol 2013; 20:3407.
  158. Shepherd NA, Saraga EP, Love SB, Jass JR. Prognostic factors in colonic cancer. Histopathology 1989; 14:613.
  159. Morikawa T, Kuchiba A, Qian ZR, et al. Prognostic significance and molecular associations of tumor growth pattern in colorectal cancer. Ann Surg Oncol 2012; 19:1944.
  160. Hase K, Shatney C, Johnson D, et al. Prognostic value of tumor "budding" in patients with colorectal cancer. Dis Colon Rectum 1993; 36:627.
  161. Minsky BD, Mies C, Rich TA, Recht A. Lymphatic vessel invasion is an independent prognostic factor for survival in colorectal cancer. Int J Radiat Oncol Biol Phys 1989; 17:311.
  162. Oh BY, Park YA, Huh JW, et al. Prognostic Impact of Tumor-Budding Grade in Stages 1-3 Colon Cancer: A Retrospective Cohort Study. Ann Surg Oncol 2018; 25:204.
  163. Lugli A, Zlobec I, Berger MD, et al. Tumour budding in solid cancers. Nat Rev Clin Oncol 2021; 18:101.
  164. Lugli A, Kirsch R, Ajioka Y, et al. Recommendations for reporting tumor budding in colorectal cancer based on the International Tumor Budding Consensus Conference (ITBCC) 2016. Mod Pathol 2017; 30:1299.
  165. Canna K, McArdle PA, McMillan DC, et al. The relationship between tumour T-lymphocyte infiltration, the systemic inflammatory response and survival in patients undergoing curative resection for colorectal cancer. Br J Cancer 2005; 92:651.
  166. Pagès F, Berger A, Camus M, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med 2005; 353:2654.
  167. Naito Y, Saito K, Shiiba K, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 1998; 58:3491.
  168. Prall F, Dührkop T, Weirich V, et al. Prognostic role of CD8+ tumor-infiltrating lymphocytes in stage III colorectal cancer with and without microsatellite instability. Hum Pathol 2004; 35:808.
  169. Pagès F, Kirilovsky A, Mlecnik B, et al. In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer. J Clin Oncol 2009; 27:5944.
  170. Lee WS, Park S, Lee WY, et al. Clinical impact of tumor-infiltrating lymphocytes for survival in stage II colon cancer. Cancer 2010; 116:5188.
  171. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction. J Clin Oncol 2011; 29:610.
  172. Huh JW, Lee JH, Kim HR. Prognostic significance of tumor-infiltrating lymphocytes for patients with colorectal cancer. Arch Surg 2012; 147:366.
  173. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006; 313:1960.
  174. Rozek LS, Schmit SL, Greenson JK, et al. Tumor-Infiltrating Lymphocytes, Crohn's-Like Lymphoid Reaction, and Survival From Colorectal Cancer. J Natl Cancer Inst 2016; 108.
  175. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol 2006; 6:295.
  176. Salama P, Phillips M, Grieu F, et al. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J Clin Oncol 2009; 27:186.
  177. Jenkins MA, Hayashi S, O'Shea AM, et al. Pathology features in Bethesda guidelines predict colorectal cancer microsatellite instability: a population-based study. Gastroenterology 2007; 133:48.
  178. Greenson JK, Huang SC, Herron C, et al. Pathologic predictors of microsatellite instability in colorectal cancer. Am J Surg Pathol 2009; 33:126.
  179. Ogino S, Nosho K, Irahara N, et al. Lymphocytic reaction to colorectal cancer is associated with longer survival, independent of lymph node count, microsatellite instability, and CpG island methylator phenotype. Clin Cancer Res 2009; 15:6412.
  180. Väyrynen JP, Sajanti SA, Klintrup K, et al. Characteristics and significance of colorectal cancer associated lymphoid reaction. Int J Cancer 2014; 134:2126.
  181. Harrison JC, Dean PJ, el-Zeky F, Vander Zwaag R. Impact of the Crohn's-like lymphoid reaction on staging of right-sided colon cancer: results of multivariate analysis. Hum Pathol 1995; 26:31.
  182. Nosho K, Baba Y, Tanaka N, et al. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol 2010; 222:350.
  183. Jass JR, Do KA, Simms LA, et al. Morphology of sporadic colorectal cancer with DNA replication errors. Gut 1998; 42:673.
  184. Messerini L, Vitelli F, De Vitis LR, et al. Microsatellite instability in sporadic mucinous colorectal carcinomas: relationship to clinico-pathological variables. J Pathol 1997; 182:380.
  185. Rüschoff J, Dietmaier W, Lüttges J, et al. Poorly differentiated colonic adenocarcinoma, medullary type: clinical, phenotypic, and molecular characteristics. Am J Pathol 1997; 150:1815.
  186. Kim H, Jen J, Vogelstein B, Hamilton SR. Clinical and pathological characteristics of sporadic colorectal carcinomas with DNA replication errors in microsatellite sequences. Am J Pathol 1994; 145:148.
  187. Lanza G, Gafà R, Santini A, et al. Immunohistochemical test for MLH1 and MSH2 expression predicts clinical outcome in stage II and III colorectal cancer patients. J Clin Oncol 2006; 24:2359.
  188. Angelova M, Charoentong P, Hackl H, et al. Characterization of the immunophenotypes and antigenomes of colorectal cancers reveals distinct tumor escape mechanisms and novel targets for immunotherapy. Genome Biol 2015; 16:64.
  189. Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 2014; 232:199.
  190. Pagès F, Mlecnik B, Marliot F, et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018; 391:2128.
  191. Yomoda T, Sudo T, Kawahara A, et al. The Immunoscore is a Superior Prognostic Tool in Stages II and III Colorectal Cancer and is Significantly Correlated with Programmed Death-Ligand 1 (PD-L1) Expression on Tumor-Infiltrating Mononuclear Cells. Ann Surg Oncol 2019; 26:415.
  192. Sinicrope FA, Shi Q, Catteau A, et al. Immunoscore Is Prognostic in Low-Risk and High-Risk Stage III Colon Carcinomas Treated With Adjuvant Infusional Fluorouracil, Leucovorin, and Oxaliplatin in a Phase III Trial. JCO Precis Oncol 2022; 6:e2200010.
  193. Sis B, Sarioglu S, Sokmen S, et al. Desmoplasia measured by computer assisted image analysis: an independent prognostic marker in colorectal carcinoma. J Clin Pathol 2005; 58:32.
  194. Crispino P, De Toma G, Ciardi A, et al. Role of desmoplasia in recurrence of stage II colorectal cancer within five years after surgery and therapeutic implication. Cancer Invest 2008; 26:419.
  195. Roncucci L, Fante R, Losi L, et al. Survival for colon and rectal cancer in a population-based cancer registry. Eur J Cancer 1996; 32A:295.
  196. Deans GT, Heatley M, Anderson N, et al. Jass' classification revisited. J Am Coll Surg 1994; 179:11.
  197. Caporale A, Amore Bonapasta S, Scarpini M, et al. Quantitative investigation of desmoplasia as a prognostic indicator in colorectal cancer. J Invest Surg 2010; 23:105.
  198. Des Guetz G, Uzzan B, Nicolas P, et al. Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer 2006; 94:1823.
  199. Foley EF, Gaffey MJ, Frierson HF Jr. The frequency and clinical significance of neuroendocrine cells within stage III adenocarcinomas of the colon. Arch Pathol Lab Med 1998; 122:912.
  200. Liu Y, Xu J, Jiao Y, et al. Neuroendocrine differentiation is a prognostic factor for stage II poorly differentiated colorectal cancer. Biomed Res Int 2014; 2014:789575.
  201. Cho YB, Yang SS, Lee WY, et al. The clinical significance of neuroendocrine differentiation in T3-T4 node-negative colorectal cancer. Int J Surg Pathol 2010; 18:201.
  202. Gaffey MJ, Mills SE, Lack EE. Neuroendocrine carcinoma of the colon and rectum. A clinicopathologic, ultrastructural, and immunohistochemical study of 24 cases. Am J Surg Pathol 1990; 14:1010.
  203. de Bruïne AP, Wiggers T, Beek C, et al. Endocrine cells in colorectal adenocarcinomas: incidence, hormone profile and prognostic relevance. Int J Cancer 1993; 54:765.
  204. Grabowski P, Schindler I, Anagnostopoulos I, et al. Neuroendocrine differentiation is a relevant prognostic factor in stage III-IV colorectal cancer. Eur J Gastroenterol Hepatol 2001; 13:405.
  205. Karim S, Brennan K, Nanji S, et al. Association Between Prognosis and Tumor Laterality in Early-Stage Colon Cancer. JAMA Oncol 2017; 3:1386.
  206. Zhang Y, Ma J, Zhang S, et al. A prognostic analysis of 895 cases of stage III colon cancer in different colon subsites. Int J Colorectal Dis 2015; 30:1173.
  207. Holch JW, Ricard I, Stintzing S, et al. The relevance of primary tumour location in patients with metastatic colorectal cancer: A meta-analysis of first-line clinical trials. Eur J Cancer 2017; 70:87.
  208. Petrelli F, Tomasello G, Borgonovo K, et al. Prognostic Survival Associated With Left-Sided vs Right-Sided Colon Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2016; 3:211.
  209. O'Dwyer PJ, Manola J, Valone FH, et al. Fluorouracil modulation in colorectal cancer: lack of improvement with N -phosphonoacetyl- l -aspartic acid or oral leucovorin or interferon, but enhanced therapeutic index with weekly 24-hour infusion schedule--an Eastern Cooperative Oncology Group/Cancer and Leukemia Group B Study. J Clin Oncol 2001; 19:2413.
  210. Tejpar S, Stintzing S, Ciardiello F, et al. Prognostic and Predictive Relevance of Primary Tumor Location in Patients With RAS Wild-Type Metastatic Colorectal Cancer: Retrospective Analyses of the CRYSTAL and FIRE-3 Trials. JAMA Oncol 2016.
  211. Loree JM, Pereira AAL, Lam M, et al. Classifying Colorectal Cancer by Tumor Location Rather than Sidedness Highlights a Continuum in Mutation Profiles and Consensus Molecular Subtypes. Clin Cancer Res 2018; 24:1062.
  212. Sinicrope FA, Mahoney MR, Yoon HH, et al. Analysis of Molecular Markers by Anatomic Tumor Site in Stage III Colon Carcinomas from Adjuvant Chemotherapy Trial NCCTG N0147 (Alliance). Clin Cancer Res 2015; 21:5294.
  213. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012; 487:330.
  214. Taleb J, Kourie HR, Emile J-F, et al.. Association of prognostic value. JAMA Oncol 2018.
  215. Roberts SA, Gordenin DA. Hypermutation in human cancer genomes: footprints and mechanisms. Nat Rev Cancer 2014; 14:786.
  216. Campbell BB, Light N, Fabrizio D, et al. Comprehensive Analysis of Hypermutation in Human Cancer. Cell 2017; 171:1042.
  217. Konishi T, Shimada Y, Hsu M, et al. Association of Preoperative and Postoperative Serum Carcinoembryonic Antigen and Colon Cancer Outcome. JAMA Oncol 2018; 4:309.
  218. Wolmark N, Fisher B, Wieand HS, et al. The prognostic significance of preoperative carcinoembryonic antigen levels in colorectal cancer. Results from NSABP (National Surgical Adjuvant Breast and Bowel Project) clinical trials. Ann Surg 1984; 199:375.
  219. Park YJ, Park KJ, Park JG, et al. Prognostic factors in 2230 Korean colorectal cancer patients: analysis of consecutively operated cases. World J Surg 1999; 23:721.
  220. Meling GI, Rognum TO, Clausen OP, 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.
  221. Lindmark G, Bergström R, Påhlman L, Glimelius B. The association of preoperative serum tumour markers with Dukes' stage and survival in colorectal cancer. Br J Cancer 1995; 71:1090.
  222. Harrison LE, Guillem JG, Paty P, Cohen AM. Preoperative carcinoembryonic antigen predicts outcomes in node-negative colon cancer patients: a multivariate analysis of 572 patients. J Am Coll Surg 1997; 185:55.
  223. Park IJ, Choi GS, Lim KH, et al. Serum carcinoembryonic antigen monitoring after curative resection for colorectal cancer: clinical significance of the preoperative level. Ann Surg Oncol 2009; 16:3087.
  224. Thirunavukarasu P, Sukumar S, Sathaiah M, et al. C-stage in colon cancer: implications of carcinoembryonic antigen biomarker in staging, prognosis, and management. J Natl Cancer Inst 2011; 103:689.
  225. Thirunavukarasu P, Talati C, Munjal S, et al. Effect of Incorporation of Pretreatment Serum Carcinoembryonic Antigen Levels Into AJCC Staging for Colon Cancer on 5-Year Survival. JAMA Surg 2015; 150:747.
  226. Kim CG, Ahn JB, Jung M, et al. Preoperative Serum Carcinoembryonic Antigen Level as a Prognostic Factor for Recurrence and Survival After Curative Resection Followed by Adjuvant Chemotherapy in Stage III Colon Cancer. Ann Surg Oncol 2017; 24:227.
  227. Liu ZH, Li C, Huang NQ, et al. No difference of complete or incomplete left-sided malignant colonic obstruction on both short- and long-term outcomes. Genet Mol Res 2014; 13:7965.
  228. Niedzwiecki D, Bertagnolli MM, Warren RS, et al. Documenting the natural history of patients with resected stage II adenocarcinoma of the colon after random assignment to adjuvant treatment with edrecolomab or observation: results from CALGB 9581. J Clin Oncol 2011; 29:3146.
  229. Faivre-Finn C, Bouvier-Benhamiche AM, Phelip JM, et al. Colon cancer in France: evidence for improvement in management and survival. Gut 2002; 51:60.
  230. Chen HS, Sheen-Chen SM. Obstruction and perforation in colorectal adenocarcinoma: an analysis of prognosis and current trends. Surgery 2000; 127:370.
  231. Mohd Suan MA, Tan WL, Soelar SA, et al. Intestinal obstruction: predictor of poor prognosis in colorectal carcinoma? Epidemiol Health 2015; 37:e2015017.
  232. Tougeron D, Sickersen G, Mouillet G, et al. Predictors of disease-free survival in colorectal cancer with microsatellite instability: An AGEO multicentre study. Eur J Cancer 2015; 51:925.
  233. Hong KD, Um JW, Ji WB, et al. Endoscopic obstruction in rectal cancers: survival and recurrence patterns following curative surgery. J Laparoendosc Adv Surg Tech A 2015; 25:278.
  234. Hatano S, Ishida H, Ishibashi K, et al. Identification of risk factors for recurrence in high-risk stage II colon cancer. Int Surg 2013; 98:114.
  235. Koebrugge B, Vogelaar FJ, Lips DJ, et al. The number of high-risk factors is related to outcome in stage II colonic cancer patients. Eur J Surg Oncol 2011; 37:964.
  236. Ho YH, Siu SK, Buttner P, et al. The effect of obstruction and perforation on colorectal cancer disease-free survival. World J Surg 2010; 34:1091.
  237. Petersen VC, Baxter KJ, Love SB, Shepherd NA. Identification of objective pathological prognostic determinants and models of prognosis in Dukes' B colon cancer. Gut 2002; 51:65.
  238. Belt EJ, Stockmann HB, Abis GS, et al. Peri-operative bowel perforation in early stage colon cancer is associated with an adverse oncological outcome. J Gastrointest Surg 2012; 16:2260.
  239. Honoré C, Goéré D, Souadka A, et al. Definition of patients presenting a high risk of developing peritoneal carcinomatosis after curative surgery for colorectal cancer: a systematic review. Ann Surg Oncol 2013; 20:183.
  240. Saha AK, Smith KJ, Sue-Ling H, et al. Prognostic factors for survival after curative resection of Dukes' B colonic cancer. Colorectal Dis 2011; 13:1390.
  241. Banaszkiewicz Z, Woda Ł, Tojek K, et al. Colorectal cancer with intestinal perforation - a retrospective analysis of treatment outcomes. Contemp Oncol (Pozn) 2014; 18:414.
  242. Ghazi S, Berg E, Lindblom A, et al. Clinicopathological analysis of colorectal cancer: a comparison between emergency and elective surgical cases. World J Surg Oncol 2013; 11:133.
  243. Sepulveda AR, Hamilton SR, Allegra CJ, et al. Molecular Biomarkers for the Evaluation of Colorectal Cancer: Guideline From the American Society for Clinical Pathology, College of American Pathologists, Association for Molecular Pathology, and the American Society of Clinical Oncology. J Clin Oncol 2017; 35:1453.
  244. Rowland A, Dias MM, Wiese MD, et al. Meta-analysis of BRAF mutation as a predictive biomarker of benefit from anti-EGFR monoclonal antibody therapy for RAS wild-type metastatic colorectal cancer. Br J Cancer 2015; 112:1888.
  245. Pietrantonio F, Petrelli F, Coinu A, et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: a meta-analysis. Eur J Cancer 2015; 51:587.
  246. Shibata D, Peinado MA, Ionov Y, et al. Genomic instability in repeated sequences is an early somatic event in colorectal tumorigenesis that persists after transformation. Nat Genet 1994; 6:273.
  247. Thibodeau SN, Bren G, Schaid D. Microsatellite instability in cancer of the proximal colon. Science 1993; 260:816.
  248. Lothe RA, Peltomäki P, Meling GI, et al. Genomic instability in colorectal cancer: relationship to clinicopathological variables and family history. Cancer Res 1993; 53:5849.
  249. Gryfe R, Kim H, Hsieh ET, et al. Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer. N Engl J Med 2000; 342:69.
  250. Sankila R, Aaltonen LA, Järvinen HJ, Mecklin JP. Better survival rates in patients with MLH1-associated hereditary colorectal cancer. Gastroenterology 1996; 110:682.
  251. Watanabe T, Wu TT, Catalano PJ, et al. Molecular predictors of survival after adjuvant chemotherapy for colon cancer. N Engl J Med 2001; 344:1196.
  252. Kohonen-Corish MR, Daniel JJ, Chan C, et al. Low microsatellite instability is associated with poor prognosis in stage C colon cancer. J Clin Oncol 2005; 23:2318.
  253. Malesci A, Laghi L, Bianchi P, et al. Reduced likelihood of metastases in patients with microsatellite-unstable colorectal cancer. Clin Cancer Res 2007; 13:3831.
  254. Hutchins G, Southward K, Handley K, et al. Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol 2011; 29:1261.
  255. Sinicrope FA, Mahoney MR, Smyrk TC, et al. Prognostic impact of deficient DNA mismatch repair in patients with stage III colon cancer from a randomized trial of FOLFOX-based adjuvant chemotherapy. J Clin Oncol 2013; 31:3664.
  256. Zaanan A, Shi Q, Taieb J, et al. Role of Deficient DNA Mismatch Repair Status in Patients With Stage III Colon Cancer Treated With FOLFOX Adjuvant Chemotherapy: A Pooled Analysis From 2 Randomized Clinical Trials. JAMA Oncol 2018; 4:379.
  257. Taieb J, Shi Q, Pederson L, et al. Prognosis of microsatellite instability and/or mismatch repair deficiency stage III colon cancer patients after disease recurrence following adjuvant treatment: results of an ACCENT pooled analysis of seven studies. Ann Oncol 2019; 30:1466.
  258. Smith CG, Fisher D, Claes B, et al. Somatic profiling of the epidermal growth factor receptor pathway in tumors from patients with advanced colorectal cancer treated with chemotherapy ± cetuximab. Clin Cancer Res 2013; 19:4104.
  259. Venderbosch S, Nagtegaal ID, Maughan TS, et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: a pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014; 20:5322.
  260. Gonsalves WI, Mahoney MR, Sargent DJ, et al. Patient and tumor characteristics and BRAF and KRAS mutations in colon cancer, NCCTG/Alliance N0147. J Natl Cancer Inst 2014; 106.
  261. Liu GC, Liu RY, Yan JP, et al. The Heterogeneity Between Lynch-Associated and Sporadic MMR Deficiency in Colorectal Cancers. J Natl Cancer Inst 2018; 110:975.
  262. Sinicrope FA, Foster NR, Thibodeau SN, et al. DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst 2011; 103:863.
  263. Cerottini JP, Caplin S, Saraga E, et al. The type of K-ras mutation determines prognosis in colorectal cancer. Am J Surg 1998; 175:198.
  264. Andreyev HJ, Norman AR, Cunningham D, et al. Kirsten ras mutations in patients with colorectal cancer: the multicenter "RASCAL" study. J Natl Cancer Inst 1998; 90:675.
  265. Samowitz WS, Curtin K, Schaffer D, et al. Relationship of Ki-ras mutations in colon cancers to tumor location, stage, and survival: a population-based study. Cancer Epidemiol Biomarkers Prev 2000; 9:1193.
  266. Yoon HH, Tougeron D, Shi Q, et al. KRAS codon 12 and 13 mutations in relation to disease-free survival in BRAF-wild-type stage III colon cancers from an adjuvant chemotherapy trial (N0147 alliance). Clin Cancer Res 2014; 20:3033.
  267. Modest DP, Ricard I, Heinemann V, et al. Outcome according to KRAS-, NRAS- and BRAF-mutation as well as KRAS mutation variants: pooled analysis of five randomized trials in metastatic colorectal cancer by the AIO colorectal cancer study group. Ann Oncol 2016; 27:1746.
  268. Taieb J, Zaanan A, Le Malicot K, et al. Prognostic Effect of BRAF and KRAS Mutations in Patients With Stage III Colon Cancer Treated With Leucovorin, Fluorouracil, and Oxaliplatin With or Without Cetuximab: A Post Hoc Analysis of the PETACC-8 Trial. JAMA Oncol 2016; :1.
  269. Taieb J, Le Malicot K, Shi Q, et al. Prognostic Value of BRAF and KRAS Mutations in MSI and MSS Stage III Colon Cancer. J Natl Cancer Inst 2017; 109.
  270. Clarke GA, Ryan E, Crowe JP, et al. Tumour-derived mutated K-ras codon 12 expression in regional lymph nodes of stage II colorectal cancer patients is not associated with increased risk of cancer-related death. Int J Colorectal Dis 2001; 16:108.
  271. Troungos C, Valavanis C, Kapranos N, Kittas C. K-ras mutation in Greek patients with poorly and moderately differenciated tumours of the lower intestinal tract. Anticancer Res 1997; 17:1399.
  272. Esteller M, González S, Risques RA, et al. K-ras and p16 aberrations confer poor prognosis in human colorectal cancer. J Clin Oncol 2001; 19:299.
  273. Andreyev HJ, Norman AR, Cunningham D, et al. Kirsten ras mutations in patients with colorectal cancer: the 'RASCAL II' study. Br J Cancer 2001; 85:692.
  274. Cercek A, Braghiroli MI, Chou JF, et al. Clinical Features and Outcomes of Patients with Colorectal Cancers Harboring NRAS Mutations. Clin Cancer Res 2017; 23:4753.
  275. Roth AD, Tejpar S, Delorenzi M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol 2010; 28:466.
  276. Gavin PG, Colangelo LH, Fumagalli D, et al. Mutation profiling and microsatellite instability in stage II and III colon cancer: an assessment of their prognostic and oxaliplatin predictive value. Clin Cancer Res 2012; 18:6531.
  277. Popovici VC, Budinska E, Roth A, et al. BRAF and KRAS mutations as additional risk factors in the context of clinical parameters of patients with colorectal cancer (abstract). J Clin Oncol 31, 2013 (suppl; abstr 3522). BRAF and KRhttp://meetinglibrary.asco.org/content/111853-132 (Accessed on June 18, 2013).
  278. Samowitz WS, Sweeney C, Herrick J, et al. Poor survival associated with the BRAF V600E mutation in microsatellite-stable colon cancers. Cancer Res 2005; 65:6063.
  279. Yokota T, Ura T, Shibata N, et al. BRAF mutation is a powerful prognostic factor in advanced and recurrent colorectal cancer. Br J Cancer 2011; 104:856.
  280. Seppälä TT, Böhm JP, Friman M, et al. Combination of microsatellite instability and BRAF mutation status for subtyping colorectal cancer. Br J Cancer 2015; 112:1966.
  281. Jones JC, Renfro LA, Al-Shamsi HO, et al. Non-V600 BRAF Mutations Define a Clinically Distinct Molecular Subtype of Metastatic Colorectal Cancer. J Clin Oncol 2017; :JCO2016714394.
  282. Munro AJ, Lain S, Lane DP. P53 abnormalities and outcomes in colorectal cancer: a systematic review. Br J Cancer 2005; 92:434.
  283. McShane LM, Altman DG, Sauerbrei W, et al. Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst 2005; 97:1180.
  284. Yamanaka T, Oki E, Yamazaki K, et al. 12-Gene Recurrence Score Assay Stratifies the Recurrence Risk in Stage II/III Colon Cancer With Surgery Alone: The SUNRISE Study. J Clin Oncol 2016; 34:2906.
  285. Mahar AL, Compton C, Halabi S, et al. Personalizing prognosis in colorectal cancer: A systematic review of the quality and nature of clinical prognostic tools for survival outcomes. J Surg Oncol 2017; 116:969.
  286. Ten Hoorn S, de Back TR, Sommeijer DW, Vermeulen L. Clinical Value of Consensus Molecular Subtypes in Colorectal Cancer: A Systematic Review and Meta-Analysis. J Natl Cancer Inst 2022; 114:503.
  287. Noffsinger AE. Serrated polyps and colorectal cancer: new pathway to malignancy. Annu Rev Pathol 2009; 4:343.
  288. Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer. Nat Med 2015; 21:1350.
  289. Sadanandam A, Lyssiotis CA, Homicsko K, et al. A colorectal cancer classification system that associates cellular phenotype and responses to therapy. Nat Med 2013; 19:619.
  290. De Sousa E Melo F, Wang X, Jansen M, et al. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat Med 2013; 19:614.
  291. Marisa L, de Reyniès A, Duval A, et al. Gene expression classification of colon cancer into molecular subtypes: characterization, validation, and prognostic value. PLoS Med 2013; 10:e1001453.
  292. Phipps AI, Limburg PJ, Baron JA, et al. Association between molecular subtypes of colorectal cancer and patient survival. Gastroenterology 2015; 148:77.
  293. Becht E, de Reyniès A, Giraldo NA, et al. Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy. Clin Cancer Res 2016; 22:4057.
  294. Calon A, Lonardo E, Berenguer-Llergo A, et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet 2015; 47:320.
  295. Isella C, Terrasi A, Bellomo SE, et al. Stromal contribution to the colorectal cancer transcriptome. Nat Genet 2015; 47:312.
  296. Dienstmann R, Vermeulen L, Guinney J, et al. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer 2017; 17:79.
  297. Ubink I, Elias SG, Moelans CB, et al. A Novel Diagnostic Tool for Selecting Patients With Mesenchymal-Type Colon Cancer Reveals Intratumor Subtype Heterogeneity. J Natl Cancer Inst 2017; 109.
  298. Sveen A, Bruun J, Eide PW, et al. Colorectal Cancer Consensus Molecular Subtypes Translated to Preclinical Models Uncover Potentially Targetable Cancer Cell Dependencies. Clin Cancer Res 2018; 24:794.
Topic 2484 Version 99.0

References