Skip to main content
Free access
ORIGINAL REPORTS
November 28, 2011

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

Publication: Journal of Clinical Oncology
PDF

Abstract

Purpose

Up to 25% of patients with node-negative colorectal cancer (CRC) on conventional histopathologic analysis ultimately die of recurrent disease. We performed a systematic review with meta-analyses to clarify whether molecular detection of isolated tumor cells or micrometastases in regional lymph nodes indicates high risk of disease recurrence and poor survival in node-negative CRC.

Methods

The following databases were searched in August 2011 to identify studies on the prognostic significance of molecular tumor-cell detection in regional lymph nodes of node-negative CRC: MEDLINE, BIOSIS, Science Citation Index, EMBASE, CCMed, and publisher databases. We extracted hazard ratios (HRs) and associated 95% CIs from the identified studies and performed random-effects model meta-analyses on overall survival, disease-specific survival, and disease-free survival.

Results

A total of 39 studies with a cumulative sample size of 4,087 patients were included. Immunohistochemistry, reverse transcriptase polymerase chain reaction, and both techniques were applied in 30, seven, and two studies, respectively. Thirteen studies were graded with low risk of bias. Meta-analyses revealed that molecular tumor-cell detection in regional lymph nodes was associated with poor overall survival (HR, 2.20; 95% CI, 1.43 to 3.40), disease-specific survival (HR, 3.37; 95% CI, 2.31 to 4.93), and disease-free survival (HR, 2.24; 95% CI, 1.57-3.20). Subgroup analyses showed the prognostic significance of molecular tumor-cell detection of being independent of the applied detection method, molecular target, and number of retrieved lymph nodes.

Conclusion

Molecular detection of occult disease in regional lymph nodes is associated with an increased risk of disease recurrence and poor survival in patients with node-negative CRC.

Introduction

Colorectal cancer (CRC) represents the third most common malignancy among both sexes.1 In theory, patients with localized CRC who present without lymph node or distant metastases (ie, stages I and II) may be cured by surgical resection alone. Therefore, except for patients with high-risk tumors (eg, T4 tumor and tumor perforation), current guidelines do not recommend adjuvant therapy for these patients.2 However, up to 25% of patients with stages I and II CRC ultimately die as a result of recurrent disease.3 Because of the strong prognostic relevance of lymph node metastases in CRC, the presence of isolated tumor cells (ITCs) or micrometastases (MMs) within regional lymph nodes that are not detected on conventional histopathologic examination by using hematoxylin and eosin staining have been suspected to be markers of systemic tumor spread in these patients.4 Thus, the detection of occult disease may help to identify those patients with node-negative CRC who are at high-risk for tumor recurrence and who might benefit from adjuvant therapy. By using molecular detection techniques such as immunohistochemistry or reverse transcriptase polymerase chain reaction (RT-PCR), various studies have demonstrated occult tumor cells in regional lymph nodes in 25% to 50% of patients with node-negative CRC on routine histopathologic analysis.57 However, the prognostic value of molecular tumor-cell detection in patients with node-negative CRC has remained uncertain as a result of inconsistent results of available, individual studies.79
To clarify this issue, and because of the need for more accurate prognostic markers in patients with node-negative CRC, we conducted a systematic review with meta-analyses of studies that evaluated the prognostic significance of molecular tumor-cell detection in regional lymph nodes.

Methods

This systematic review and meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.10

Search Strategy and Selection Criteria

The following databases were searched in August 2011: MEDLINE, BIOSIS, Science Citation Index, EMBASE, CCMed, publisher databases, American Society of Clinical Oncology Abstracts, and the clinical trial registries WHO International Clinical Trials Registry Platform and ClinicalTrials.gov. We also checked reference lists of relevant articles and review articles. No language restrictions and time limits were applied to the initial search. Search strategies, databases, and date ranges are provided in supplemental material (Data Supplement).
Two reviewers (N.N.R. and U.B.) independently screened the identified abstracts for eligibility and retrieved full articles for detailed assessment. We included studies that evaluated the association of molecular tumor-cell detection (ie, ITCs and/or MMs) with overall survival (OS; date of surgery to date of death as a result of any cause), disease-specific survival (DSS; date of surgery to date of death as a result of CRC), disease-free survival (DFS; date of surgery to date of first recurrence or death; ie, events were death or recurrence, whichever came first), or recurrence-free survival (date of surgery to date of first recurrence; ie, events were locoregional and/or systemic recurrence) of patients with node-negative CRC. Molecular detection techniques may have included any form of RT-PCR or immunohistochemistry. Occult disease was defined according to the definition of the International Union Against Cancer (UICC; ie, as a single or small number of tumor cells ≤ 0.2 mm in dimension [ITCs] and as tumor deposits ≤ 2.0 mm but greater than 0.2 mm in dimension [MMs]).11 If the UICC definition was not applied or mentioned explicitly, single or clustered cells detected on molecular analyses were also considered as occult disease. We excluded studies that enrolled less than 20 patients with node-negative CRC who underwent molecular analysis of regional lymph nodes as well as studies for which no hazard ratio (HR) could be calculated for any of the outcomes. Furthermore, we excluded studies that were published in a language other than English, German, or French, studies that were not published in peer-reviewed journals, and studies that did not apply the definitions of ITCs and MMs used in this study. In case of multiple publications from the same institution with identical or overlapping patient cohorts, the most informative report was included. Studies that were excluded for these reasons are listed in the Data Supplement as are identified review articles and relevant meeting abstracts.

Data Extraction

Two authors (N.N.R. and U.B.) independently extracted the following data from eligible articles: first author, year of publication, study period, sample size, patient age, site of disease (colon and/or rectum), stage of disease (stage I and/or stage II), adjuvant chemotherapy, study type, rate of patients with positive lymph nodes on molecular analysis, definition of occult disease, histopathologic analysis, number of retrieved lymph nodes, methods of tumor-cell detection, target genes and antigens, duration of follow-up, prognostic outcomes, and use of multivariate models. Disagreements were resolved by discussion.

Assessment of Study Quality

The study quality of included studies was assessed by using the modified risk of bias tool recommended by the Cochrane Collaboration as described previously.12,13

Statistical Analyses

Synchronized extraction results were pooled statistically as effect estimates in meta-analyses. HRs and their corresponding SEs were extracted for individual time-to-event outcome parameters of each primary study. If an HR and the associated SE or CIs were not reported, we approximated the HR by using the statistical data provided in the article (eg, individual patient data or survival plots).14,15
Extracted HRs were pooled by using the generic inverse-variance method available in the Review Manager software (Version 5.0, The Nordic Cochrane Centre; The Cochrane Collaboration, Copenhagen, Denmark). An HR greater than 1 indicated a worse prognosis in the tumor cell–positive group. A minimum number of three studies was required to perform meta-analyses. A random-effect analysis model was applied because we expected interstudy heterogeneity with respect to, for example, study populations, detection assays, definitions of positive lymph nodes, and duration of follow-up16 Statistical heterogeneity was assessed with I2 statistics. Reasons for statistical heterogeneity were explored by using sensitivity analyses (ie, the removal of certain studies from the analysis as suggested by the forest plot) and a priori subgroup analyses. We performed tests of interaction to test for differences between subgroups.17 To avoid double patient evaluation among studies that evaluated multiple detection assays and/or target genes, these parameters were combined when possible to keep a maximum of information. Otherwise, cytokeratins (CKs) were prioritized over alternative tumor-cell markers, and immunohistochemistry was prioritized over RT-PCR assays. Sensitivity analyses (by choosing the alternative study arm) were performed to assess the statistical impact of such prioritization. Funnel plot analyses were used to evaluate publication bias.18

Results

Baseline Study Characteristics

We identified a total of 39 eligible studies that included 4,087 patients (Fig 1).5,1956 The median study sample size was 90 patients (range, 21 to 399 patients). All eligible studies were published between 1994 and 2011 (Table 1). The extracted variables related to the study design of the included studies are summarized in Table 2. The median number of lymph nodes that were retrieved, on average, was 12 (range, 4 to 48 lymph nodes), and the median duration of follow-up across all studies was 60 months (range, 28 to 128 months). The majority of studies used CKs for molecular tumor-cell detection. In five studies, additional markers were analyzed,22,26,35,43,46 and in five studies, only alternative genes/antigens were used.20,24,39,51,54 A polymerase chain reaction–based detection assay was used in seven studies.20,24,35,39,40,51,54 In two studies, lymph nodes were analyzed by RT-PCR and immunohistochemistry.43,46 The remaining studies exclusively used immunohistochemistry for tumor-cell detection. Nine studies that used immunohistochemistry assays applied the UICC definition of ITCs and MMs.21,23,28,32,41,45,49,55,56 One or more oncologic outcome parameters were analyzed on multivariate analysis in 22 studies.5,19,21,23,2528,30,32,35,3841,43,45,46,51,5456 A significant association between molecular tumor-cell detection and the long-term outcome of patients was reported in 33% of studies on OS,21,24,39,42,47 58% of studies on DSS,5,20,23,26,33,39,41,46,53,56 and 33% of studies on DFS.24,28,35,51,52 Thirteen studies were graded with low risk of bias (Data Supplement).25,28,32,35,38,40,41,43,45,46,51,54,56 Funnel plot analyses did not indicate a significant publication bias for analyzed outcomes (Data Supplement).
Fig 1. Flow chart of study selection.
Table 1. Baseline Characteristics of Included Studies
First Author Year Enrollment Period Sample Size Age (years) Stage of Disease Rate of Tumor Cell–Positive Patients (%)a Rate of Patients With Adjuvant Chemotherapy (%) Kind of Adjuvant Chemotherapy Study Type Risk of Biasb
Total Colon Rectum Median Range Mean SD
Adell 1996 1981-1988 100 68 32 69 31-89     Dukes B 39 NR   RSCS High
Belly 2001 1984-1993 38 NR NR NR NR NR NR Stage II 37 NR   RSCS High
Bosch Roig 2008 1991-2000 39 39 0 63 39-76     Stage II 10 100 FU-levamisole 1 year or FU-FA 6 months (Mayo Clinic scheme) RSCS High
Broll 1997 1987 32 NR NR 69c 41-91c     Stages I and II 19 0   RSCS High
Bukholm 2003 1989-1999 156 156 0 70 41-93     Stage II 38 NR   RSCS High
Cagir 1999 1989-1995 21 20 1     68.1 9.5 Stage II 47 14 FU-based protocol SCCS High
Choi 2002 1990-1995 93 61 32 58 37-79     Dukes B 31 NR   RSCS Low
Clarke 2000 1985-1991 100 NR NR 70 32-89     Stage II 25 and 26d 0   RSCS High
Davies 2008 1992-1996 105 NR NR 78 48-100     Dukes A and B 47 0   RSCS High
Faerden 2011 2000-2005 126 126 0 71 32-92     Stages I and II 31 0   PSCS Low
Fisher 2003 1977-NR 399 158 241 NR NR NR NR Dukes A and B 18 38 FU, semustine, vincristine (MOF scheme) MRCTe High
Garcia-Saenz 2006 1992-1999 105 78 27 62 31-82     Stage II 25 67 FU-based protocol RSCS High
Greenson 1994 1984-1987 50 NR NR NR NR NR NR Dukes B 28 NR   RSCS High
Haboubi 1998 1989-1992 25 NR NR 68c 45-90c     Dukes B 60 NR   RSCS High
Haince 2010 1991-2005 123 123 0 71 24-95     Stages I and II 19 0   RSCS Low
Hara 2007 1987-1999 144 0 144 56c 28-78c     Stage II 35 36 FU-based protocol RSCS Low
Isaka 1999 1977-1994 44 0 44 61 and 60f       Dukes B 21 57 FU-based protocol RSCS High
Jeffers 1994 1983-1986 77 52 25 61 and 67f       Dukes B 24 NR   RSCS High
Koyanagi 2008 NR 67 NR NR 74 35-95     Stages I and II 40 NR   PMCSe Low
Kronberg 2004 1989-1999 90 65 25     65 11 Stages I and II 28 0   RSCS High
Laso 2004 1990-1991 21 13 8 67 30-90     Stages I and II 38 NR   RSCS High
Lee 2006 1999 120 NR NR 57 28-80     Stages I and II 50 43 FU-based protocol RSCS Low
Liefers 1998 1990-1992 26 NR NR 68       Stage II 54 NR   RSCS High
Merrie 2003 NR 122 122 0 72c 35-92c     Stage II 35 0   PSCS Low
Messerini 2006 1987-1989 395 252 143 68 41-87     Stage IIA 38 0   RSCS Low
Mukai 2005 1987-1999 124 NR NR NR NR NR NR Dukes B 16 15 FU-based protocol RSCS High
Noura 2002 1988-1996 64 35 29     60 10 Stage II 54g NR   RSCS Low
Oberg 1998 1987-1994 147 96 51 70 39-92     Dukes A and B 32 NR   RSCS High
Oh 2011 2005-2009 124 61 63     61 11 Stage II 26 86 FU-FA or FOLFOX RSCS High
Park 2008 1996-2005 160 160 0 62 24-86     Stages I and II 5 NR   RSCS Low
Reggiani-Bonetti 2011 1989-2004 95 41 54     69h 8h Stage I 72h NR   MCCS Low
                69i 10i   1i        
Rosenberg 2002 1988-1995 85 57 28 61       Stages I and II 52j NR   RSCS Low
Shimoyama 2003 1981-1994 57 0 57 60 24-83     Stages I and II 19 NR   RSCS High
Steinert 2008 1997-2000 90 NR NR NR NR NR NR Stages I and II 45 0   RSCS High
Uribarrena-Amezaga 2010 1995-2000 85 62 23 67 29-88     Dukes A and B 36 0   RSCS High
van Schaik 2009 2000-2002 72 72 0 76 43-89     Dukes A and B 15 NR   RSCS High
Waldmanne 2009 2002-2007 257 222 35 68       Stage I/II 87 22 FU-based protocol PMCS Low
Wang 2006 2001 67 0 67 60c 20-84c     Stages I and II 18 100 Oral fluoropyrimidines for 3-12 months (median, 6 months) RSCS High
Yasuda 2001 1984-1992 42 NR NR     62h 14h Dukes B 76 NR   RSCS High
                65i 11i            
Abbreviations: CEA, carcinoembryonic antigen, carboxylesterase; CK, cytokeratin; FA, folinic acid; FOLFOX, infusional fluorouracil, leucovorin, and oxaliplatin; FU, fluorouracil; IHC, immunohistochemistry; MCCS, multicenter case-control study; MOF, semustine, vincristine, and fluorouracil; MRCT, multicenter randomized controlled trial; NR, not reported; PMCS, prospective multicenter cohort study; PSCS, prospective single-center cohort study; RSCS, retrospective single-center cohort study; RT-PCR, reverse transcriptase polymerase chain reaction; SCCS, single-center case-control study; UICC, International Union Against Cancer.
a
Detection rates are given for any kind of occult disease (i.e. isolated tumor cells and/or micrometastases).
b
Risk of bias was assessed using the modified tool recommended by the Cochrane Collaboration (Data Supplement).
c
Refers to all patients (UICC I-III).
d
In two articles, the authors reported detection rates using IHC for CK (25%) and p53 (26%).
e
Of two articles with highly overlapping study populations, the analysis that provided categorical data on outcome of patients with and without positive lymph nodes on molecular analysis was included.51,57
f
Patients with and without tumor cells, respectively.
g
Authors reported detection rates using RT-PCR for CEA (29%) and IHC for CK (54%).
h
Data reported for patients with disease recurrence/tumor-related death.
i
Data reported for patients without disease recurrence/tumor-related death.
j
In three articles, authors reported detections rates using RT-PCR for CK20 (52%), IHC for CK20 (28%), RT-PCR for CEA (58%) and IHC for CEA (27%).
Table 2. Design Variables of Included Studies
First Author SLN Analysis Definition of Tumor-Cell Positivity Definition of ITCs/MMs No. of Retrieved LN No. of LN Detection Method Target Gene/Antigen Follow-Up (months) Sections (μm) Outcomes Reported Multivariate Analysis
Median Range Mean SD Median Range Mean SD
Adell No MMs or ITCs Single or clustered cells 4 1-18     467 IHC CK 49 4-94     4 RFS Yes
Belly No NA NA > 10       474 Nested PCR K12-rasmutation > 36       50 OS, DSS No
Bosch Roig No MMs; MMs or ITCs ITCs: cells < 0.2 mm; MMs: 0.2-2-mm cell clusters 9 1-27     382 IHC CK (Ab AE1/AE3) 81 21-120     NR OS, DFS Yes
Broll No MMs or ITCs Single or clustered cells NR NR     NR IHC CK (Ab AE1/AE3); Epithelial gycoproteins (Ab Ber-EP4) 84 2-102     3 DFS No
Bukholm No MMs or ITCs ITCs: cells < 0.2 mm MMs: 0.2-2-mm cell clusters 4 1-23     NR IHC CK (Ab CAM 5.2) NR       5 DSS Yes
Cagir No NA NA     18 12 524 RT-PCR GUCYC 67 ± 30       10 OS, DFS No
Choi No MMs or ITCs Single or clustered cells 19       1,808 IHC CK (Ab Dako-CK MNF 116) 66 60-85     4 DSS Yes
Clarke No MMs or ITCs Single or clustered cells 7       NR IHC CK 5, 6, 8, 17 P53a > 60       7 DSS Yes
Davies No MMs or ITCs Single or clustered cells NR NR     898 IHC CK (Ab AE1/AE3; MNF116) 48       NR DFS Yes
Faerden No ITCs; MMs or ITCs ITCs: cells < 0.2 mm MMs: 0.2-2 mm cell clusters 12 1-48     NR IHC CK (Ab AE1/AE3; CK 20; CAM 5.2) 60       3-4 DFS Yes
Fisher No MMs or ITCs Mini MMs: single or groups of tumor cells < 1.0 mm NR NR     NR IHC CK (Ab AE1/AE3) NR       NR OS, RFS No
Garcia-Saenz No MMs or ITCs Single or clustered cells NR NR     665 IHC CK (Ab AE1/AE3) 59       4 DFS Yes
Greenson No MMs or ITCs Single or clustered cells 11       568 IHC CK (Ab AE1/AE3) 60       NR DSS Yes
Haboubi No MMs or ITCs Single or clustered cells 48b 10-200b     2,409b IHC CK (Ab AE1/AE3) 55b 3-91b     3 DSS No
Haince No MMs or ITCs NA 12 1-75     NR RT-qPCR GUCYC 53 12-117     5 DFS, RFS Yes
Hara No ITCs ITCs: cells < 0.2 mm MMs: 0.2-2-mm cell clusters 28       4,035 IHC CK (Ab AE1/AE3) 82 3-171     4 OS Yes
Isaka No MMs or ITCs Single or clustered cells 15       644 IHC CK (Ab AE1/AE3) 62 3-210     3 DSS, RFS No
Jeffers No MMs or ITCs Single or clustered cells 7       559 IHC CK (Ab AE1/AE3) 81 63-120     NR OS No
Koyanagi Yes NA NA NR NR     NR RT-qPCR c-MET, MAGE-A3, GalNAc-T, CK20 34       NR OS; DFS Yes
Kronberg No MMs or ITCs Single or clustered cells 15       NR IHC CK (Ab AE1/AE3; PCK2) 90 11-160     5 DSS No
Laso No MMs or ITCs ≥ 1 metastatic group of cells NR NR     NR IHC CK (Ab AE1/AE3) 57       4 OS No
Lee No MMs or ITCs Single or clustered cells 19 5-54     2,235 IHC CK (Ab Dako-CK MNF 116) 57 11-66     4 DFS Yes
Liefers No NA NA 6       192 Nested RT-PCR CEA 73c       50 (5) OS, DSS, RFS Yes
Merrie No NA NA 18b 1-100b     2,317b RT-PCR CK 20 42 23-75     NR OS Yes
Messerini No MMs; ITCs; MMs or ITCs ITCs: cells < 0.2 mm; MMs: 0.2-2-mm cell clusters 18 15-45     8,266 IHC CK (Ab Ks20.8) 128 122-136     5 DSS, DFSd Yes
Mukai No MMs or ITCs Single or clustered cells ≥ 10       1,240 IHC CK (Ab AE1/AE3) ≥ 60       NR OS, RFS No
Noura No MMs or ITCs Single or clustered cells 5.5 1-24     350 RT-PCR, IHC CEA CK (Ab AE1/AE3) 79 6-134     4 DSS, DFS Yes
Oberg No MMs or ITCs Single or clustered cells 4 1-15     609 IHC CK (Ab CAM 5.2) 28 5-67     4 DSS No
Oh No MMs or ITCs ITCs: cells < 0.2 mm; MMs: 0.2-2-mm cell clusters     19 9 2,379 IHC CK (Ab AE1/AE3) 36 1-62     4 OS, DFS, RFS Yes
Park No MMs ITCs: cells < 0.2 mm; MMs: 0.2-2-mm cell clusters 17       2,852 IHC CK 20 45 1-137     5 DSS, DFS Yes
Reggiani-Bonetti No MMs or ITCs Single cells, clusters, glands < 0.2 mm 8e 1-15e     NR IHC CK (Ab Dako-CK) NR       4 DSS Yes
        8f 1-31f                        
Rosenberg No MMs or ITCs Single or clustered cells 25 6-72     NR RT-PCR, IHC CK 20, CEA 86 41-128     15; 6 DSS, DFS Yes
Shimoyama No MMs or ITCs Single or clustered cells     13 9 892 IHC CK (Ab CAM 5.2) NR       4 OS, RFS No
Steinert No MMs or ITCs Clustered but isolated cells 12       1,108 IHC CK18 61 43-95     4 DFS No
Uribarrena-Amezaga No MMs Deposits of tumor cells < 2 mm     10 6 NR IHC CK (Ab AE1/AE3) ≥ 60       5 DSS No
van Schaik No MMs MMs: 0.2-2-mm cell clusters ≥ 8       NR IHC CK (Ab LU-5) ≥ 41       NR OS, DFS No
Waldmann No NA NA 2 2-159     6,699 RT-qPCR GUCY2C 24g 2-63g     NR RFS, DFS Yes
                      35h 2-62h          
Wang No MMs MMs: 0.2-2-mm cell clusters 10b 1-29b     726b IHC CK 20 56 7-62     4 OS, DFS No
Yasuda No MMs or ITCs Single or clustered cells 18 3-94     1,013 IHC CK (Ab CAM 5.2) NR       3 DSS No
Abbreviations: c-MET, hepatocyte growth factor receptor; DFS, disease-free survival; DSS, disease-specific survival; GalNAc-T, β1→4-N-acetylgalactosaminyltransferase; GUCY(2)C, guanylyl cyclase (2)C; ITC, isolated tumor cell; LN, lymph nodes; MAGE-A3, melanoma antigen gene-A3 family; MM, micrometastasis; NA, not applicable; NR, not reported; OS, overall survival; PCR, polymerase chain reaction; PKR, double-stranded RNA-activated protein kinase; RFS, recurrence-free survival; RT-PCR, reverse transcriptase polymerase chain reaction; RT-qPCR, reverse transcriptase quantitative polymerase chain reaction; UICC, International Union Against Cancer; SLN, sentinel lymph node.
a
Authors used CK and p53 in two separate reports on the same study cohort.
b
Refers to all patients (UICC I-III).
c
Length of follow-up refers to patients who were still alive.
d
Data on DSS are extracted from a further study of the same institution with overlapping study population.
e
Data reported for patients with disease recurrence/tumor-related death.
f
Data reported for patients without disease recurrence/tumor-related death.
g
Follow-up period for patients with positive lymph nodes status for GUCY(2)C.
h
Follow-up period for patients without positive lymph nodes status for GUCY(2)C.
i
Reported outcomes suitable for meta-analyses.

Prognostic Value of Molecular Tumor-Cell Detection in Regional Lymph Nodes

The pooled analysis of all studies on OS showed a significant prognostic value of tumor-cell detection in regional lymph nodes (HR, 2.20; 95% CI, 1.43 to 3.40; n = 15; I2 = 66%; Fig 2A). Sensitivity analyses revealed that heterogeneity was not caused by a certain study. Meta-analyses confirmed the prognostic significance of molecular tumor-cell detection for the outcomes of DSS (HR, 3.37; 95% CI, 2.31 to 4.93; n = 17; I2 = 44%) and DFS (HR, 2.24; 95% CI, 1.57-3.20; n = 18; I2 = 44%) with moderate heterogeneity (Figs 2B and 3). Although the analysis of recurrence-free survival also showed less favorable outcome for the group of tumor cell–positive patients (HR, 2.83; 95% CI, 1.68 to 4.77; n = 9; I2 = 61%), heterogeneity could not be reduced substantially by exclusion of a specific study.
Fig 2. Meta-analyses on the association of molecular tumor cell detection in regional lymph nodes with (A) overall survival and (B) disease-specific survival. For studies that evaluated multiple markers and detection methods, cytokeratins were prioritized over alternative markers and analyses by using immunohistochemistry over reverse transcriptase polymerase chain reaction. Squares and horizontal bars indicate point estimates (hazard ratios) with 95% CIs for the individual studies. Diamonds indicate summary estimates of the hazard ratio. The width of the diamond corresponds to the 95% CI.
Fig 3. Meta-analysis on the association of molecular tumor cell detection in regional lymph nodes and disease-free survival. For studies that evaluated multiple markers and detection methods, cytokeratins were prioritized over alternative markers and analyses by using immunohistochemistry over reverse transcriptase polymerase chain reaction. Squares and horizontal bars indicate point estimates (hazard ratios) with 95% CIs for the individual studies. Diamonds indicate summary estimates of the hazard ratio. The width of the diamond corresponds to the 95% CI.

Subgroup Analyses

Despite the limited number of included studies, the subgroup analyses on the definition of ITCs/MMs showed that detection of MMs according to the UICC definition was associated with poor OS (HR, 3.62; 95% CI, 1.34 to 9.80; n = 3; I2 = 20%), DSS (HR, 2.07; 95% CI, 1.11 to 3.86; n = 3; I2 = 26%) and DFS (HR, 2.81; 95% CI, 1.32 to 5.97; n = 5; I2 = 53%), whereas no pooled analyses on ITC detection could be performed because of the lack of studies. Meta-analyses of studies that did not explicitly use the UICC definition of ITCs/MMs revealed a strong prognostic value of molecular tumor-cell detection regarding the oncological outcomes evaluated.
The prognostic impact of molecular tumor-cell detection was independent of the applied detection methods (ie, immunohistochemistry and RT-PCR), although we observed a more pronounced prognostic value for the analyses of studies using RT-PCR as indicated by the test of interaction on DFS (P < .001). The majority of studies used CKs as molecular targets, and meta-analyses that included these studies revealed a strong prognostic significance with respect to all three outcomes. Despite the low number of studies, there was a significant association of tumor-cell detection in regional lymph nodes by using alternative targets and OS (HR, 3.54; 95% CI, 1.23 to 10.16; n = 3; I2 = 61%), DSS (HR, 3.59; 95% CI, 2.16 to 5.97; n = 5; I2 = 0%), and DFS (HR, 5.13; 95% CI, 2.17 to 12.12; n = 4; I2 = 38%).
Subgroup analyses of studies that enrolled patients with stage II CRC only showed that tumor-cell detection was associated with poor OS, DSS, and DFS. However, analyses of studies with patients with stages I and II CRC favored the group of patients with negative lymph nodes regarding DSS (HR, 2.10; 95% CI,1.17 to 3.76; n = 5; I2 = 27%) and DFS (HR, 2.17; 95% CI,1.42 to 3.33; n = 10; I2 = 13%), whereas the association for OS did not reach statistical significance (HR, 1.58; 95% CI, 0.77 to 3.24; n = 4; I2 = 58%).
Subgroup analyses indicated molecular tumor-cell detection to be associated with poor OS (HR, 2.96; 95% CI, 1.29 to 6.79; n = 7; I2 = 72), DSS (HR, 4.08; 95% CI, 2.20 to 7.56; n = 9; I2 = 66%), and DFS (HR, 2.43; 95% CI, 1.27 to 4.68; n = 4; I2 = 5%) even in studies with an inadequate average lymph node count. In studies with at least 12 resected lymph nodes, there was a similar association of tumor-cell detection with poor outcome in all three end points evaluated.
Finally, meta-analyses of studies with a high risk of bias indicated a prognostic impact of molecular tumor-cell detection regarding OS (HR, 2.72; 95% CI, 1.49 to 4.97; n = 12; I2 = 72%), DSS (HR, 3.05; 95% CI,1.96 to 4.75; n = 13; I2 = 45%), and DFS (HR, 2.45; 95% CI,1.32 to 4.55; n = 9; I2 = 55%). Similarly, in studies with a low risk of bias, there was a significantly worse DSS (HR, 3.51; 95% CI, 1.75 to 7.02; n = 6; I2 = 50%) and DFS (HR, 2.05; 95% CI,1.34 to 3.14; n = 9; I2 = 33%) for patients with a positive molecular analysis of regional lymph nodes.
DFS was the only outcome measure with sufficient data to evaluate the prognostic impact of tumor-cell detection depending on the administration of adjuvant therapy. These subgroup analyses showed tumor cell–positive lymph nodes to be associated with less favorable DFS in patients who did (HR, 3.12; 95% CI,1.48 to 6.59; n = 7; 2 = 64%) and did not (HR, 1.79; 95% CI,1.05 to 3.05; I2 = 37%) receive adjuvant chemotherapy. The results of all subgroup analyses are summarized in Table 3.
Table 3. Subgroup Analyses
  Overall Survival Disease-Specific Survival Disease-Free Survival
HR 95% CI Degree of heterogeneity (I2 statistics; %) P No. of Included Studies HR 95% CI Degree of heterogeneity (I2 statistics; %) P No. of Included Studies HR 95% CI Degree of heterogeneity (I2 statistics; %) P No. of Included Studies
Total 2.20 1.43 to 3.40 66 < .001 15 3.37 2.31 to 4.93 44 < .001 17 2.24 1.57 to 3.20 44 < .001 18
Definition of DTC*                              
    ITCs*         1         0         2
    MMs* 3.62 1.34 to 9.80 20 .01 3 2.07 1.11 to 3.86 26 .02 3 2.81 1.32 to 5.97 53 .007 5
    ITCs or MMs*         2         2 2.10 1.09 to 4.04 45 .03 5
    Single or clustered cells 2.26 1.29 to 3.94 72 .004 10 3.22 2.08 to 4.99 39 < .001 12 2.37 1.47 to 3.81 46 < .001 12
Detection method                              
    IHC 2.02 1.17 to 3.49 69 .01 10 3.34 2.18 to 5.13 50 < .001 15 1.56 1.19 to 2.06 1 .001 13
    RT-PCR 2.54 1.37 to 4.69 38 .003 5 4.26 2.19 to 8.29 0 < .001 4 4.53 2.68 to 7.64 11 < .001 6
Tested gene/antigen                              
    CK 2.01 1.21 to 3.33 67 .007 11 3.34 2.18 to 5.13 50 < .001 15 1.60 1.18 to 2.18 9 .002 12
    Other 3.54 1.23 to 10.16 61 .02 3 3.59 2.16 to 5.97 0 < .001 5 5.16 2.69 to 9.89 18 < .001 5
    CK or other         1     0             2
Stage of disease                              
    I and II 1.58 0.77 to 3.24 58 .21 4 2.10 1.17 to 3.76 27 .01 5 2.17 1.42 to 3.33 13 < .001 10
    II 2.27 1.40 to 3.68 66 < .001 12 3.15 2.08 to 4.78 32 < .001 12 2.44 1.35 to 4.40 64 .003 8
Site of disease                              
    Colon 2.14 0.73 to 6.21 70 .16 4         2 2.78 1.49 to 5.20 12 .001 5
    Rectum 1.62 0.92 to 2.86 59 .09 4         1         0
    Colon and rectum 2.03 1.12 to 3.69 70 .02 9 3.29 2.15 to 5.05 53 < .001 14 1.97 1.28 to 3.04 50 .002 12
No. of retrieved lymph nodes                              
    < 12 2.96 1.29 to 6.79 71 .01 7 4.08 2.20 to 7.56 66 < .001 9 2.43 1.27 to 4.68 5 .008 4
    ≥ 12 2.85 1.15 to, 7.05 68 .02 4 2.69 1.77 to 4.08 0 < .001 8 2.46 1.37 to 4.43 62 .003 10
Duration of follow-up, months                              
    < 60 1.87 1.20 to 2.92 0 .006 7 2.02 1.05 to 3.86 10 .03 3 2.05 1.42 to 2.96 6 < .001 10
    ≥ 60 3.65 1.35 to 9.82 83 .01 6 3.11 1.98 to 4.88 36 < .001 10 2.66 1.24 to 5.69 65 .01 8
Adjuvant chemotherapy                              
    Yes§ 2.84 1.36 to 5.92 77 .005 8         1 3.12 1.48 to 6.59 64 .003 7
    No         1 2.54 1.32 to 4.89 77 .005 4 1.79 1.05 to 3.05 37 .03 6
Risk of bias                              
    Low 1.33 0.92 to 1.93 0 .13 3 3.51 1.75 to 7.02 50 < .001 6 2.05 1.34 to 3.14 33 .001 9
    High 2.72 1.49 to 4.97 72 .001 12 3.31 2.06 to 5.31 45 < .001 11 2.45 1.32 to 4.55 55 .004 9
Abbreviations: DTC, disseminating tumor cell; HR, hazard ratio; ITC, isolated tumor cell; MM, micrometastasis; RT-PCR, reverse transcriptase polymerase chain reaction; UICC, International Union Against Cancer.
*
UICC definition of ITCs and/or MMs.11
Definition of ITCs and/or MMs not according to the UICC. Molecular-analysis positive in case single or clustered cells were detected. Analysis included studies that used RT-qPCR.
Test of interaction, P < .01.
§
Studies in which either a part of or the entire study cohort received adjuvant chemotherapy.
Test of interaction, P < .05.

Discussion

This systematic review and meta-analysis showed that the molecular detection of tumor cells in regional lymph nodes is associated with an increased risk of disease recurrence and poor survival in patients with node-negative CRC.
Despite the amount of studies that have so far been conducted and the well-known risk of patients with apparent lymph node metastases,58 the prognostic value of molecular tumor-cell detection in regional lymph nodes of patients with node-negative CRC has remained highly uncertain because of the inconsistent results of available reports. An interesting finding of this study is that pooled analyses of available data revealed a significant association between molecular tumor-cell detection and the long-term outcome of patients, although this association was found to be statistically significant in a rather low proportion of the primary studies. This discrepancy may in part be explained by the low average sample size in the individual studies. Together with the lesser number of events compared with patients with overt lymph nodes metastases,59 there was a lack of statistical power to detect differences in survival in patients with node-negative disease. These data, however, support the approach to aggregate results of available studies on the prognostic significance of molecular tumor-cell detection in CRC as has recently been performed for various other malignancies.60,61
There is recent evidence that clinical criteria currently used to grade the risk status of patients with stage II CRC may be insufficient to identify the subgroup of patients who benefit from adjuvant chemotherapy.62 These data may favor molecular or cellular biomarkers to tailor adjuvant chemotherapy in patients with node-negative disease. Numerous studies have demonstrated a more accurate prediction of the prognosis of patients by using a molecular analysis of the primary tumor and various compartments such as the regional lymph nodes and systemic circulation.13,46,57,63,64 However, it remains a subject of additional investigation by using comparative analyses to assess the independent prognostic impact of molecular analyses for different compartments within the same populations to answer the concern of which biomarkers provide the most accurate prognostic information.
The concept of intratumor heterogeneity might serve as an additional explanation for the inconsistent prognostic impact of molecular tumor-cell detection across and within the identified studies. The genetic heterogeneity within tumors together with epigenetic and morphologic plasticity may contribute to phenotypic differences between single tumor cells.65 Although the dissemination of tumor cells might occur early during tumor progression,66 there is evidence that genomic instability of individual tumor cells persists after dissemination from the primary tumor,67,68 and genomic alterations in these cells are of a higher prognostic impact compared with those in cells of the primary tumor.69 Thus, one might hypothesize that the detection of certain subpopulations, such as the stem-cell fraction within occult tumor cells, may further increase the prognostic accuracy of tumor-cell detection in regional lymph nodes. The existence of cancer stem cells in CRC has been demonstrated convincingly on a functional level.70,71 In accordance with this hypothesis, it has been shown that the number of tumor cells at the invasive front (budding tumor cells) that express high amounts of nuclear β-catenin, as a sign of aberrant Wnt signaling activation72 associated with ongoing epithelial-mesenchymal transition and stem-cell formation,73 is strongly correlated with metastasis and poor survival in rectal cancer patients.74
Much effort has been made to identify patients with node-negative CRC who are at high risk for disease recurrence and, thus, who might benefit from adjuvant therapy.75,76 Recently, there has been an increasing interest in a more detailed evaluation of the nodal status of patients.77 Besides the detection of occult disease, the number of retrieved lymph nodes has been suggested to more accurately predict the survival of patients and is considered a measure of quality control.78 Although current guidelines recommend a minimum number of 12 evaluated lymph nodes to ensure adequate pathologic assessment,11 our analyses demonstrated the prognostic value of molecular tumor-cell detection of being independent of the lymph node count.
The systematic evaluation of the literature revealed certain aspects to be addressed in the design of future studies to further enhance the utility of their results. We noticed a wide range of applied detection assays, and additional standardization is required before molecular tumor-cell detection can be incorporated into routine staging of patients with CRC. In line with our analyses that showed a prognostic impact of occult disease detected by immunohistochemistry or RT-PCR, with a potential advantage of RT-PCR, there are a couple of comparative analyses that favored RT-PCR and immunohistochemistry-controlled RT-PCR.43,46 Time and cost issues associated with routine sampling for occult metastases in all lymph-nodes of patients with CRC are additional arguments in favor of high-throughput screening methods of lymph nodes such as RT-PCR and/or flow cytometric analyses. Moreover, techniques of sentinel lymph-node analyses might serve as an efficient strategy to improve the staging of patients with node-negative disease.79 The issue of standardization also applies to the selection and definition of end points. As opposed to chemotherapy trials for stage III and metastatic CRC,80,81 validated end points for prognostic biomarkers in CRC are lacking. In this study, four different end points were evaluated to summarize the available evidence. However, uniform and standardized end points should be agreed on and applied in future studies to enhance the comparison and aggregation of the results from different studies. Although the design was consistent in the majority of studies, only a few studies were performed prospectively. By using standardized detection assays and end points, prospective studies are required to confirm the results of the present meta-analysis and, moreover, to evaluate the prognostic value of tumor-cell subpopulations detected in the regional lymph nodes of patients with node-negative CRC.
In conclusion, there is evidence that molecular tumor-cell detection in regional lymph nodes indicates poor prognosis in node-negative CRC. Despite a prognostic impact of tumor-cell detection across the applied detection assays, standardized protocols should be used in future studies that, moreover, should use standardized end points and adhere to the UICC definition of ITCs and MMs. By using such protocols, it has to be clarified within prospective randomized trials whether patients with occult lymph node disease benefit from adjuvant chemotherapy. Furthermore, the findings of this study encourage efforts to identify subpopulations of disseminated cells that put patients at particular risk of disease recurrence and poor survival.

Data Supplements

Authors retain all rights in any data supplements associated with their articles.

The ideas and opinions expressed in this Data Supplement do not necessarily reflect those of the American Society of Clinical Oncology (ASCO). The mention of any product, service, or therapy in this Data Supplement should not be construed as an endorsement of the products mentioned. It is the responsibility of the treating physician or other health care provider, relying on independent experience and knowledge of the patient, to determine drug dosages and the best treatment for the patient. Readers are advised to check the appropriate medical literature and the product information currently provided by the manufacturer of each drug to be administered to verify approved uses, the dosage, method, and duration of administration, or contraindications. Readers are also encouraged to contact the manufacturer with questions about the features or limitations of any products. ASCO and JCO assume no responsibility for any injury or damage to persons or property arising out of or related to any use of the material contained in this publication or to any errors or omissions. Readers should contact the corresponding author with any comments related to Data Supplement materials.

Files in this Data Supplement:

Authors' Disclosures of Potential Conflicts of Interest

The author(s) indicated no potential conflicts of interest.

References

1.
A Jemal, R Siegel, J Xu, etal: Cancer statistics, 2010 CA Cancer J Clin 60: 277– 300,2010
2.
E Van Cutsem, J Oliveira: Primary colon cancer: ESMO clinical recommendations for diagnosis, adjuvant treatment and follow-up Ann Oncol 20: 49– 50,2009 suppl 4
3.
J Weitz, M Koch, J Debus, etal : Colorectal cancer Lancet 365: 153– 165,2005
4.
J Weitz, P Kienle, A Magener, etal : Detection of disseminated colorectal cancer cells in lymph nodes, blood and bone marrow Clin Cancer Res 5: 1830– 1836,1999
5.
JK Greenson, CE Isenhart, R Rice, etal : Identification of occult micrometastases in pericolic lymph nodes of Duke's B colorectal cancer patients using monoclonal antibodies against cytokeratin and CC49. Correlation with long-term survival Cancer 73: 563– 569,1994
6.
MD Jeffers, GM O'Dowd, H Mulcahy, etal : The prognostic significance of immunohistochemically detected lymph node micrometastases in colorectal carcinoma J Pathol 172: 183– 187,1994
7.
R Rosenberg, A Hoos, J Mueller, etal : Prognostic significance of cytokeratin-20 reverse transcriptase polymerase chain reaction in lymph nodes of node-negative colorectal cancer patients J Clin Oncol 20: 1049– 1055,2002
8.
M Sasaki, H Watanabe, JR Jass, etal : Occult lymph node metastases detected by cytokeratin immunohistochemistry predict recurrence in “node-negative” colorectal cancer J Gastroenterol 32: 758– 764,1997
9.
N Hayashi, I Ito, A Yanagisawa, etal : Genetic diagnosis of lymph-node metastasis in colorectal cancer Lancet 345: 1257– 1259,1995
10.
A Liberati, DG Altman, J Tetzlaff, etal: The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration PLoS Med 6: e1000100,2009
11.
LH Sobin, MK Gospodarowicz, C Wittekind: UICC: TNM Classification of Malignant Tumours 2009 New York, NY Wiley & Sons
12.
JP Higgins, S Green: Cochrane Handbook for Systematic Reviews of Interventions Version 5.0.2 [updated September 2009] The Cochrane Collaboration 2009
13.
NN Rahbari, M Aigner, K Thorlund, etal : Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer Gastroenterology 138: 1714– 1726,2010
14.
MK Parmar, V Torri, L Stewart: Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints Stat Med 17: 2815– 2834,1998
15.
JF Tierney, LA Stewart, D Ghersi, etal: Practical methods for incorporating summary time-to-event data into meta-analysis Trials 8: 16,2007
16.
R DerSimonian, N Laird: Meta-analysis in clinical trials Control Clin Trials 7: 177– 188,1986
17.
DG Altman, JM Bland: Interaction revisited: The difference between two estimates BMJ 326: 219,2003
18.
M Egger, G Davey Smith, M Schneider, etal : Bias in meta-analysis detected by a simple, graphical test BMJ 315: 629– 634,1997
19.
G Adell, B Boeryd, B Frånlund, etal : Occurrence and prognostic importance of micrometastases in regional lymph nodes in Dukes' B colorectal carcinoma: An immunohistochemical study Eur J Surg 162: 637– 642,1996
20.
RT Belly, JD Rosenblatt, M Steinmann, etal : Detection of mutated K12-ras in histologically negative lymph nodes as an indicator of poor prognosis in stage II colorectal cancer Clin Colorectal Cancer 1: 110– 116,2001
21.
CE Bosch Roig, E Roselló-Sastre, S Alonso Hernández, etal : Prognostic value of the detection of lymph node micrometastases in colon cancer Clin Transl Oncol 10: 572– 578,2008
22.
R Broll, V Schauer, H Schimmelpenning, etal : Prognostic relevance of occult tumor cells in lymph nodes of colorectal carcinomas: An immunohistochemical study Dis Colon Rectum 40: 1465– 1471,1997
23.
IR Bukholm, J Bondi, P Wiik, etal : Presence of isolated tumour cells in mesenteric lymph nodes predicts poor prognosis in patients with stage II colon cancer Eur J Surg Oncol 29: 862– 866,2003
24.
B Cagir, A Gelmann, J Park, etal : Guanylyl cyclase C messenger RNA is a biomarker for recurrent stage II colorectal cancer Ann Intern Med 131: 805– 812,1999
25.
HJ Choi, YY Choi, SH Hong: Incidence and prognostic implications of isolated tumor cells in lymph nodes from patients with Dukes B colorectal carcinoma Dis Colon Rectum 45: 750– 755,2002 discussion755-756
26.
G Clarke, E Ryan, JC O'Keane, etal : The detection of cytokeratins in lymph nodes of Duke's B colorectal cancer subjects predicts a poor outcome Eur J Gastroenterol Hepatol 12: 549– 552,2000
27.
M Davies, PJ Arumugam, VI Shah, etal : The clinical significance of lymph node micrometastasis in stage I and stage II colorectal cancer Clin Transl Oncol 10: 175– 179,2008
28.
AE Faerden, OH Sjo, IR Bukholm, etal : Lymph node micrometastases and isolated tumor cells influence survival in stage I and II colon cancer Dis Colon Rectum 54: 200– 206,2011
29.
ER Fisher, L Colangelo, S Wieand, etal: Lack of influence of cytokeratin-positive mini micrometastases in “Negative Node” patients with colorectal cancer: Findings from the national surgical adjuvant breast and bowel projects protocols R-01 and C-01 Dis Colon Rectum 46: 1021– 1025,2003 discussion 1025-1026
30.
JA Garcia-Saenz, MC Saenz, L Gonzalez, etal : Significance of the immunohistochemical detection of lymph node micrometastases in stage II colorectal carcinoma Clin Transl Oncol 8: 676– 680,2006
31.
NY Haboubi, SA Abdalla, S Amini, etal : The novel combination of fat clearance and immunohistochemistry improves prediction of the outcome of patients with colorectal carcinomas: A preliminary study Int J Colorectal Dis 13: 99– 102,1998
32.
M Hara, T Hirai, H Nakanishi, etal : Isolated tumor cell in lateral lymph node has no influences on the prognosis of rectal cancer patients Int J Colorectal Dis 22: 911– 917,2007
33.
N Isaka, M Nozue, M Doy, etal : Prognostic significance of perirectal lymph node micrometastases in Dukes' B rectal carcinoma: An immunohistochemical study by CAM5.2 Clin Cancer Res 5: 2065– 2068,1999
34.
Reference deleted
35.
K Koyanagi, AJ Bilchik, S Saha, etal : Prognostic relevance of occult nodal micrometastases and circulating tumor cells in colorectal cancer in a prospective multicenter trial Clin Cancer Res 14: 7391– 7396,2008
36.
U Kronberg, F López-Kostner, G Soto, etal : Detection of lymphatic micrometastases in patients with stages I and II colorectal cancer: Impact on five-year survival Dis Colon Rectum 47: 1151– 1157,2004
37.
CA Laso, JJ González, F Fresno, etal : Prognostic value of micrometastases in esophageal and colorectal carcinoma (a clinical experience) Hepatogastroenterology 51: 964– 967,2004
38.
MR Lee, CW Hong, SN Yoon, etal: Isolated tumor cells in lymph nodes are not a prognostic marker for patients with stage I and stage II colorectal cancer J Surg Oncol 93: 13– 18,2006 discussion 18-19
39.
GJ Liefers, AM Cleton-Jansen, CJ van de Velde, etal : Micrometastases and survival in stage II colorectal cancer N Engl J Med 339: 223– 228,1998
40.
AE Merrie, AM van Rij, ER Dennett, etal : Prognostic significance of occult metastases in colon cancer Dis Colon Rectum 46: 221– 231,2003
41.
L Messerini, F Cianchi, C Cortesini, etal : Incidence and prognostic significance of occult tumor cells in lymph nodes from patients with stage IIA colorectal carcinoma Hum Pathol 37: 1259– 1267,2006
42.
M Mukai, S Sato, H Ninomiya, etal : Recurrence and 5-FU sensitivity of stage II/Dukes' B colorectal cancer with occult neoplastic cells in lymph node sinuses Oncol Rep 14: 1171– 1176,2005
43.
S Noura, H Yamamoto, T Ohnishi, etal : Comparative detection of lymph node micrometastases of stage II colorectal cancer by reverse transcriptase polymerase chain reaction and immunohistochemistry J Clin Oncol 20: 4232– 4241,2002
44.
A Oberg, R Stenling, B Tavelin, etal : Are lymph node micrometastases of any clinical significance in Dukes Stages A and B colorectal cancer? Dis Colon Rectum 41: 1244– 1249,1998
45.
SJ Park, KY Lee, SY Kim: Clinical significance of lymph node micrometastasis in stage I and II colon cancer Cancer Res Treat 40: 75– 80,2008
46.
Reference deleted
47.
M Shimoyama, T Yamazaki, T Suda, etal : Prognostic significance of lateral lymph node micrometastases in lower rectal cancer: An immunohistochemical study with CAM5.2 Dis Colon Rectum 46: 333– 339,2003
48.
R Steinert, M Hantschick, M Vieth, etal : Influence of subclinical tumor spreading on survival after curative surgery for colorectal cancer Arch Surg 143: 122– 128,2008
49.
R Uribarrena-Amezaga, J Ortego, J Fuentes, etal : Prognostic value of lymph node micrometastasis in patients with colorectal cancer in Dukes stages A and B (T1–T4, N0, M0) Rev Esp Enferm Dig 102: 176– 186,2010
50.
PM van Schaik, E Hermans, JC van der Linden, etal : Micro-metastases in stages I and II colon cancer are a predictor of the development of distant metastases and worse disease-free survival Eur J Surg Oncol 35: 492– 496,2009
51.
SA Waldman, T Hyslop, S Schulz, etal : Association of GUCY2C expression in lymph nodes with time to recurrence and disease-free survival in pN0 colorectal cancer JAMA 301: 745– 752,2009
52.
C Wang, ZG Zhou, YY Yu, etal : Patterns of lateral pelvic lymph node metastases and micrometastases for patients with lower rectal cancer Eur J Surg Oncol 33: 463– 467,2007
53.
K Yasuda, Y Adachi, N Shiraishi, etal : Pattern of lymph node micrometastasis and prognosis of patients with colorectal cancer Ann Surg Oncol 8: 300– 304,2001
54.
JF Haince, M Houde, G Beaudry, etal : Comparison of histopathology and RT-qPCR amplification of guanylyl cyclase C for detection of colon cancer metastases in lymph nodes J Clin Pathol 63: 530– 537,2010
55.
TY Oh, SM Moon, US Shin, etal : Impact on prognosis of lymph node micrometastasis and isolated tumor cells in stage II colorectal cancer J Korean Soc Coloproctol 27: 71– 77,2011
56.
L Reggiani Bonetti, C Di Gregorio, C De Gaetani, etal : Lymph node micrometastasis and survival of patients with Stage I (Dukes' A) colorectal carcinoma Scand J Gastroenterol 46: 881– 886,2011
57.
T Hyslop, DS Weinberg, S Schulz, etal : Occult tumor burden predicts disease recurrence in lymph node-negative colorectal cancer Clin Cancer Res 17: 3293– 3303,2011
58.
I Zlobec, A Lugli: Prognostic and predictive factors in colorectal cancer J Clin Pathol 61: 561– 569,2008
59.
JB O'Connell, MA Maggard, CY Ko: Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging J Natl Cancer Inst 96: 1420– 1425,2004
60.
S Mocellin, D Hoon, A Ambrosi, etal : The prognostic value of circulating tumor cells in patients with melanoma: A systematic review and meta-analysis Clin Cancer Res 12: 4605– 4613,2006
61.
S Zhao, Y Liu, Q Zhang, etal: The prognostic role of circulating tumor cells (CTCs) detected by RT-PCR in breast cancer: A meta-analysis of published literature Breast Cancer Res Treat [epub ahead of print February 11, 2011; Erratum: Breast Cancer Res Treat epub ahead of print July 8, 2011]
62.
ES O'Connor, DY Greenblatt, NK Loconte, etal : Adjuvant chemotherapy for stage II colon cancer with poor prognostic features J Clin Oncol 29: 3381– 3388,2011
63.
J Fritzmann, M Morkel, D Besser, etal : A colorectal cancer expression profile that includes transforming growth factor beta inhibitor BAMBI predicts metastatic potential Gastroenterology 137: 165– 175,2009
64.
U Stein, W Walther, F Arlt, etal : MACC1, a newly identified key regulator of HGF-MET signaling, predicts colon cancer metastasis Nat Med 15: 59– 67,2009
65.
A Marusyk, K Polyak: Tumor heterogeneity: Causes and consequences Biochim Biophys Acta 1805: 105– 117,2010
66.
CA Klein: Parallel progression of primary tumours and metastases Nat Rev Cancer 9: 302– 312,2009
67.
PJ Campbell, S Yachida, LJ Mudie, etal : The patterns and dynamics of genomic instability in metastatic pancreatic cancer Nature 467: 1109– 1113,2010
68.
CA Klein, TJ Blankenstein, O Schmidt-Kittler, etal : Genetic heterogeneity of single disseminated tumour cells in minimal residual cancer Lancet 360: 683– 689,2002
69.
NH Stoecklein, SB Hosch, M Bezler, etal : Direct genetic analysis of single disseminated cancer cells for prediction of outcome and therapy selection in esophageal cancer Cancer Cell 13: 441– 453,2008
70.
CA O'Brien, A Pollett, S Gallinger, etal : A human colon cancer cell capable of initiating tumour growth in immunodeficient mice Nature 445: 106– 110,2007
71.
L Ricci-Vitiani, DG Lombardi, E Pilozzi, etal : Identification and expansion of human colon-cancer-initiating cells Nature 445: 111– 115,2007
72.
PJ Morin, AB Sparks, V Korinek, etal : Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC Science 275: 1787– 1790,1997
73.
JM Mariadason, M Bordonaro, F Aslam, etal : Down-regulation of beta-catenin TCF signaling is linked to colonic epithelial cell differentiation Cancer Res 61: 3465– 3471,2001
74.
H Ueno, AB Price, KH Wilkinson, etal : A new prognostic staging system for rectal cancer Ann Surg 240: 832– 839,2004
75.
YH Uen, SR Lin, DC Wu, etal : Prognostic significance of multiple molecular markers for patients with stage II colorectal cancer undergoing curative resection Ann Surg 246: 1040– 1046,2007
76.
HM Quah, JF Chou, M Gonen, etal : Identification of patients with high-risk stage II colon cancer for adjuvant therapy Dis Colon Rectum 51: 503– 507,2008
77.
CC Compton, FL Greene: The staging of colorectal cancer: 2004 and beyond CA Cancer J Clin 54: 295– 308,2004
78.
GJ Chang, MA Rodriguez-Bigas, JM Skibber, etal : Lymph node evaluation and survival after curative resection of colon cancer: Systematic review J Natl Cancer Inst 99: 433– 441,2007
79.
AJ Bilchik, DT Nora, LH Sobin, etal : Effect of lymphatic mapping on the new tumor-node-metastasis classification for colorectal cancer J Clin Oncol 21: 668– 672,2003
80.
PA Tang, SM Bentzen, EX Chen, etal : Surrogate end points for median overall survival in metastatic colorectal cancer: Literature-based analysis from 39 randomized controlled trials of first-line chemotherapy J Clin Oncol 25: 4562– 4568,2007
81.
DJ Sargent, HS Wieand, DG Haller, etal: Disease-free survival versus overall survival as a primary end point for adjuvant colon cancer studies: Individual patient data from 20,898 patients on 18 randomized trials J Clin Oncol 23: 8664– 8670,2005

Information & Authors

Information

Published In

Journal of Clinical Oncology
Pages: 60 - 70
PubMed: 22124103

History

Published online: November 28, 2011
Published in print: January 01, 2012

Permissions

Request permissions for this article.

Authors

Affiliations

Nuh N. Rahbari [email protected]
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Ulrich Bork
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Edith Motschall
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Kristian Thorlund
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Markus W. Büchler
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Moritz Koch
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.
Jürgen Weitz
Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, and Jürgen Weitz, University of Heidelberg, Heidelberg, Germany; Edith Motschall, University of Freiburg, Freiburg, Germany; Nuh N. Rahbari, Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA; and Kristian Thorlund, McMaster University, Hamilton, Canada.

Notes

Both N.N.R. and U.B. contributed equally to this work.
Corresponding author: Nuh N. Rahbari, MD, Department of General, Visceral and Transplant Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; e-mail: [email protected].

Author Contributions

Conception and design: Nuh N. Rahbari, Ulrich Bork, Markus W. Büchler, Moritz Koch, Jürgen Weitz
Financial support: Markus W. Büchler, Jürgen Weitz
Administrative support: Markus W. Büchler, Jürgen Weitz
Collection and assembly of data: Nuh N. Rahbari, Ulrich Bork,Edith Motschall
Data analysis and interpretation: Nuh N. Rahbari, Ulrich Bork, Kristian Thorlund, Markus W. Büchler, Moritz Koch, Jürgen Weitz
Manuscript writing: All authors
Final approval of manuscript: All authors

Disclosures

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

Funding Information

Supported by the Deutsche Forschungsgemeinschaft (Grant No. WE 3548/4-1).

Metrics & Citations

Metrics

Altmetric

Citations

Article Citation

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format





Download article citation data for:
Nuh N. Rahbari, Ulrich Bork, Edith Motschall, Kristian Thorlund, Markus W. Büchler, Moritz Koch, Jürgen Weitz
Journal of Clinical Oncology 2012 30:1, 60-70

View Options

View options

PDF

View PDF

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login

Purchase Options

Purchase this article to get full access to it.

Purchase this Article

Subscribe

Subscribe to this Journal
Renew Your Subscription
Become a Member

Media

Figures

Other

Tables

Share

Share

Share article link

Share