Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81

Warning: fopen(upload/ip_log/ip_log_2024-11.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83

Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84
The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers
Skip Navigation
Skip to contents

J Pathol Transl Med : Journal of Pathology and Translational Medicine

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Pathol Transl Med > Forthcoming articles > Article
Original Article
The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers
Ji-Ae Lee1orcid, Hye Eun Park2orcid, Hye-Yeong Jin3,4, Lingyan Jin3,4, Seung Yeon Yoo5orcid, Nam-Yun Cho4orcid, Jeong Mo Bae3,4orcid, Jung Ho Kim3orcid, Gyeong Hoon Kang,3,4orcid

DOI: https://doi.org/10.4132/jptm.2024.09.26
Published online: October 24, 2024

1Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea

2Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea

3Department of Pathology, Seoul National University College of Medicine, Seoul, Korea

4Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea

5Pathology Center, Seegene Medical Foundation, Seoul, Korea

Corresponding Author: Gyeong Hoon Kang, MD, PhD, Department of Pathology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: +82-2-740-8263, Fax: +82-2-743-5530, E-mail: ghkang@snu.ac.kr
• Received: June 3, 2024   • Revised: August 22, 2024   • Accepted: September 26, 2024

© The Korean Society of Pathologists/The Korean Society for Cytopathology

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 266 Views
  • 50 Download
  • Background
    Colorectal carcinomas (CRCs) with caudal-type homeobox 2 (CDX2) loss are recognized to pursue an aggressive behavior but tend to be accompanied by a high density of tumor-infiltrating lymphocytes (TILs). However, little is known about whether there is an interplay between CDX2 loss and TIL density in the survival of patients with CRC.
  • Methods
    Stage III CRC tissues were assessed for CDX2 loss using immunohistochemistry and analyzed for their densities of CD8 TILs in both intraepithelial (iTILs) and stromal areas using a machine learning-based analytic method.
  • Results
    CDX2 loss was significantly associated with a higher density of CD8 TILs in both intraepithelial and stromal areas. Both CDX2 loss and a high CD8 iTIL density were found to be prognostic parameters and showed hazard ratios of 2.314 (1.050–5.100) and 0.378 (0.175–0.817), respectively, for cancer-specific survival. A subset of CRCs with retained CDX2 expression and a high density of CD8 iTILs showed the best clinical outcome (hazard ratio of 0.138 [0.023–0.826]), whereas a subset with CDX2 loss and a high density of CD8 iTILs exhibited the worst clinical outcome (15.781 [3.939–63.230]).
  • Conclusions
    Altogether, a high density of CD8 iTILs did not make a difference in the survival of patients with CRC with CDX2 loss. The combination of CDX2 expression and intraepithelial CD8 TIL density was an independent prognostic marker in adjuvant chemotherapy-treated patients with stage III CRC.
Colorectal cancer (CRC) ranks third in cancer prevalence and second in cancer mortality worldwide [1]. The prognostication of patients with CRC is globally based on the tumor, node, metastasis (TNM) staging, which reflects the extent of the cancer. However, there is significant variation in survival among patients who have the same cancer stage [2,3]. Fluoropyrimidine-oxaliplatin combination (FOLFOX) therapy is standard care after surgery for patients with stage III colon cancer. However, it is estimated that a considerable portion of patients with stage III colon cancer do not benefit from adjuvant FOLFOX therapy, and some patients suffer from unnecessary toxicity [4-7]. Thus, it is necessary to develop clinicopathological or molecular biomarkers that help to identify whether patents will benefit from adjuvant combination therapy.
Caudal-type homeobox 2 (CDX2) is a major regulator of intestine-specific genes involved in cell growth and differentiation. CDX2 is a gene that is only active in the small and large intestines and is detected in most CRCs (90%) [8-10]. Patients with CRC which has no CDX2 expression tend to have worse outcomes [8,11]. Although CDX2 loss has been shown to be a marker of poor prognosis in stage II colon cancer [12], CDX2 loss has been demonstrated to be a marker of chemotherapy sensitivity in stage II colon cancer patients, implying that adjuvant chemotherapy might be a treatment option for patients with stage II CDX2-deficient colon cancer [13]. However, another previous study has suggested that CDX2 loss may be a possible negative marker of chemotherapy response in CRC patients with metastases [14].
Regarding the cause of CDX2 loss in CRC, promoter CpG island hypermethylation has been suggested [11]. CDX2 loss is more frequent in CRCs with CpG island methylator phenotype (CIMP) than in CRCs without CIMP [8,15] and in CRCs with microsatellite instability (MSI) than in CRCs without MSI [8,11,12,16]. CIMP and MSI have been shown to be associated with enhanced tumor-infiltrating lymphocyte (TIL) density [17,18]. There is not much information about the TIL status in CRCs that lack CDX2 expression [8], but CRCs that lack CDX2 expression are likely to have higher TIL density than CRCs with retained CDX2 expression because of the relationship between CDX2 loss and MSI or CIMP. In terms of prognosis, it remains unclear whether patients with CDX2-deficient CRC show different clinical outcomes depending on TIL density status.
In the present study, we aimed to identify (1) whether CDX2 loss is associated with poor clinical outcome in adjuvant FOLFOX-treated patients with stage III CRC, (2) whether CDX2 loss is associated with an increased density of TILs, and (3) whether there is an interplay between CDX2 expression and TIL status in patient survival. To resolve the above issues, we performed CDX2 immunohistochemistry (IHC) of cancer tissue samples from stage III CRC patients treated with adjuvant FOLFOX and quantified CD3-positive TILs and CD8-positive TILs in both intraepithelial and stromal areas using a machine learning-based analytic method and then correlated CDX2 expression status with clinicopathological and molecular features. To investigate whether an interplay exists between CDX2 loss and TIL density in the survival of patients with CRC, we examined the combined CDX2 expression and TIL density statuses to understand their impact on survival characteristics.
Samples
Archival tissue blocks of the surgical specimens from 505 patients with stage III CRC who received adjuvant FOLFOX after curative surgery (R0) at Seoul National University Hospital (SNUH) between April 2005 and December 2012 were available for construction of a tissue microarray (TMA). Whole-slide immunostaining of CD3 and CD8 was possible in 446 of the 505 patients. Patients were chosen for the present study based on the following criteria: they were over 18 years old, they had adenocarcinoma type of cancer, they had stage III CRC, they had their tumor removed completely with no cancer cells at the edges, and they finished at least six cycles of 5-fluorouracil plus oxaliplatin or four cycles of capecitabine plus oxaliplatin as adjuvant therapy. The present study did not include patients who met any of these criteria: having chemotherapy or radiotherapy before surgery, a genetic condition that causes many polyps in the colon and rectum, an idiopathic inflammatory bowel disease, or a previous diagnosis of any other cancer within 5 years. We reviewed electronic medical records and collected demographic and clinicopathological data, such as age, sex, tumor location, histological differentiation, lymphatic emboli or venous invasion, perineural invasion, and American Joint Committee on Cancer/International Union against Cancer (AJCC/UICC) cancer stage (7th edition).
Immunohistochemistry
One pathologist (S.Y.Y.) selected the paraffin tissue block most representative of the tumor, and whole-slide sections were subjected to IHC with antibodies against CD3 (clone F7.2.38, Dako, Carpenteria, CA, USA) and CD8 (clone SP57, Ventana Medical Systems, Tucson, AZ, USA) [19]. The TMA blocks received cores that were 2 mm across and taken from two separate regions of the tumor. TMA sections of 4-μm thickness were stained with primary antibodies against KRT7 (cytokeratin 7 [CK7], clone OV-TL 12/30, Dako), KRT20 (CK20, clone Ks20.8, Dako), and CDX2 (clone EPR2764Y ready-to-use, Cell Marque, Rocklin, CA, USA). An Aperio AT2 slide scanner (Leica Biosystems, Wetzlar, Germany) was used to scan all the stained slides. To interpret the IHC results, the proportion of tumor cells that expressed KRT7 and KRT20 in their cytoplasm was measured. The threshold was based on scores of 10% and 50% for high KRT7 expression and low KRT20 expression, respectively, following a prior study [8]. To interpret CDX2 IHC results, we used the H score to measure the intensity and extent of nuclear staining. the H score was calculated with this formula: 3×percentage of strongly stained nuclei+2×percentage of moderately stained nuclei+ 1×percentage of weakly stained nuclei. The cutoff value was set at an H score of 20, and an H score of <20 was called loss of expression [20]. The analytic pipeline used the virtual slide files of CD3 and CD8 IHC as input, and its detailed protocol can be found at http://dx.doi.org/10.17504/protocols.io.yqvfvw6 [21]. After a user marked the tumor area on a given image, the algorithm divided the area into tiles of 1 mm×1 mm and calculated the median density (number of cells/mm2) of TILs inside the epithelium (iTILs) and TILs in the stroma (sTILs).
DNA extraction, microsatellite instability analysis, and mutation analysis of KRAS and BRAF
Using a microscope, the parts of the tumor with typical histology and the most tumor cells were marked on glass slides, and the corresponding areas from the unstained tissue slides after deparaffinization were scraped from the glass slides. The tissues that were scraped off were transferred into microtubes with tissue lysis buffer and proteinase K and then left to incubate at 55°C for 24 hours. After centrifugation, the supernatant was transferred to a new tube and kept in a deep freezer. The fluorescent multiplex polymerase chain reaction (PCR) method with five microsatellite markers suggested by National Cancer Institute (BAT25, BAT26, D2S123, D5S346, and D17S250) was used to determine the MSI status of each tumor. Tumors were categorized as MSI-H (MSI-high, with ≥2 unstable markers out of 5) or MSS (microsatellite-stable, with 1 or no unstable marker) (n=503). Real-time PCR-based allelic discrimination was used to examine BRAF mutations at codon 600 (V600E) (n=492). Codons 12 and 13 of KRAS exon 2 were sequenced to determine their mutation statuses (n=486).
Bisulfite modification and methylation analysis
Using an EZ DNA methylation kit (Zymo Research, Orange, CA, USA), genomic DNA underwent bisulfite conversion, and then the MethyLight assay was used to assess the methylation level of these CIMP-specific markers (CACNA1G, CDKN2A (p16), CRABP1, IGF2, MLH1, NEUROG1, RUNX3, and SOCS1) (n=500). The primer sequences and PCR conditions have been previously reported [22,23]. We performed the MethyLight assay three times and used the median value to show the methylation level of each marker. To determine the methylation status of a specific marker, the percentage of methylated reference (PMR) was calculated, and a marker with a median PMR >4 was deemed to be methylated. Each tumor was assessed for CIMP status and grouped into CIMP-H (CIMP-high, ≥ 5 out of 8 methylated markers), CIMP-L (CIMP-low, 1–4 methylated markers), or CIMP-0 (no methylation) as previously reported [17].
Statistical analysis
The normality test was conducted to determine whether TIL density was normally distributed using the Shapiro-Wilks test. The null hypothesis that the TIL density is normally distributed was rejected because the p-value was below .05 for all four types of TILs, including intraepithelial CD3 TILs (CD3 iTILs), stromal CD3 TILs (CD3 sTILs), CD8 iTILs, and CD8 sTILs. Thus, comparison of the TIL density between subsets of CRCs with CDX2 loss and with CDX2 retention was performed using the nonparametric Mann-Whitney U test. Student’s t test was used to compare the age distribution of two groups. For 2×2 contingency tables with a sample size of more than 5, a two-sided chi-square test was performed, while a Kruskal-Wallis test was applied for contingency tables with more than 2×2 dimensions. We measured the time from surgery to death by CRC as cancer-specific survival (CSS) time and the time from surgery to confirmed recurrence as recurrence-free survival (RFS) time. We censored the data from patients who did not die from CRC or relapse by the last follow-up visit for the CSS and RFS analyses, respectively. We used the Kaplan-Meier log-rank test to compare survival across groups. We estimated the hazard ratio with the Cox proportional hazard model and adjusted for baseline characteristics with a backward stepwise model that included the following covariates that were significant in univariate survival analysis: differentiation (high grade vs. low grade), venous invasion (present vs. absent), lymphatic emboli (present vs. absent), T category (T4 vs. T1–3), N category (N2 vs. N1), CK7 (expressed vs. not expressed), KRAS (mutant vs. wild type), CD3 sTIL, and CD8 sTIL.
The follow-up period for the 505 patients was a mean of 68.2 months (range, 4.1 to 134.8 months). Table 1 shows the demographic data. There were 303 males and 202 females. The tumor subsite was distributed as follows: 150 in the right colon, 289 in the left colon, and 66 in the rectum. CIMP-H and MSIH CRCs were present in 5.4% and 5.6% of stage III CRCs, respectively. KRAS and BRAF mutations occurred in 28.8% and 3.5% of patients, respectively.
Relationships between decreased expression of CDX2 and clinicopathological features
Decreased expression of CDX2 (CDX2 loss) was found in 12.5% of stage III CRCs (Fig. 1). CDX2 loss was associated with a younger age of onset (56.1 vs. 59.8 years, Student’s t test, p=.003). CDX2 loss was more frequent in the right colon than in the left colon and rectum, in CRCs with high-grade histological differentiation than in CRCs with low-grade histological differentiation, in CIMP-H CRCs than in CIMP-L,0 CRCs, in MSI-H CRCs than in MSS CRCs, in CRCs with BRAF mutations than in CRCs without BRAF mutation, in CRCs with decreased expression of KRT20 than in CRCs without decreased expression of KRT20, and in CRCs with KRT7 expression than in CRCs without KRT7 expression (Table 1). CRCs with CDX2 loss showed a higher density of CD8 iTILs and sTILs than CRCs without CDX2 loss (Fig. 2). The significance of the difference was higher in the density of CD8 iTILs than in that of CD8 sTILs. However, the densities of CD3 iTILs and sTILs tended to be higher in CRCs with CDX2 loss than in CRCs with retained CDX2 expression, but the difference did not reach statistical significance (Fig. 2).
Association of CDX2 loss with worse clinical outcome
In univariate survival analysis, CDX2 loss was found to be significantly associated with shortened CSS but not RFS (Fig. 3A, B). In addition, several clinicopathological parameters, including tumor differentiation, T category, N category, lymphatic emboli, venous invasion, KRAS mutation, KRT7 expression, CD3 sTILs, CD8 iTILs (Fig. 3C, D), and CD8 sTILs, were found to be significant prognostic factors in the univariate analysis of CSS (Table 2).
Combination of CDX2 expression and CD8 iTIL density status as a prognostic parameter
The combination of CDX2 expression and CD8 iTIL density status generates four subsets of CRCs. In the analysis of CSS using the Kaplan-Meier log-rank test, a subset with retained CDX2 expression and a high density of CD8 iTILs showed the best clinical outcome, whereas subsets with CDX2 loss exhibited worse clinical outcomes, regardless of CD8 iTIL status, than a subset with retained CDX2 expression and a high density of CD8 iTILs (Fig. 4). Statistically, there was no significant survival difference between a subset with CDX2 loss and a high density of CD8 iTILs and a subset with CDX2 loss and a low density of CD8 iTILs (p=.384 and p=.501, CSS and RFS, respectively). In the multivariate analysis, the combination of CDX2 expression and CD8 iTILs was found to be an independent prognostic parameter (Table 3).
In the present study, we analyzed a cohort of adjuvant FOLFOX-treated patients with stage III CRC regarding whether CDX2 loss was associated with a shortened survival, whether CDX2 expression status was associated with the intraepithelial or stromal TIL density, and whether there was an interplay between CDX2 expression and TIL density in patient survival. The findings demonstrated that CDX2 loss was a prognostic factor heralding poor prognosis in patients with stage III CRC treated with adjuvant FOLFOX, CDX2 loss was accompanied by increased infiltration of CD8 iTILs and sTILs, and a combined status of CDX2 expression and CD8 iTIL density was an independent prognostic parameter in FOLFOX-treated patients with stage III CRC.
In the present study, CRCs with CDX2 loss showed higher densities of CD8 iTILs and sTILs than CRCs with retained CDX2 expression. However, in terms of prognosis, CDX2 loss might collide with an increased density of CD8 iTILs because CDX2 loss and an increased density of CD8 iTILs were associated with worse and better survival, respectively (Table 2, Fig. 3). Of the four subsets generated by the combination of CDX2 expression and CD8 iTIL density statuses, a subset of CRCs with retained CDX2 expression and high CD8 iTIL density showed the best clinical outcome, which is in accordance with Derangere et al.’s study [24] in which stage III colon cancers harboring high CDX2 expression and a high CD3 TIL were associated with a good prognosis. However, in the present study, the worst clinical outcome was observed in a subset of CRCs with CDX2 loss and high CD8 iTIL density, in contrast to Derangere et al.’s study [24], which showed the worst prognosis in stage III colon cancers with low CDX2 expression and a low CD3 TIL density. The reason for such a discrepancy might be related to the different cutoff values set for CDX2 expression and TIL density for dichotomization. In Derangere et al.’s study [24], three-fourths of colon cancers were classified into tumors with low CDX2 expression, and two-thirds of colon cancers were classified into tumors with a low CD3 TIL density, whereas in our study, one-eighth of CRCs were classified into tumors with CDX2 loss, and one-half of CRCs were classified into tumors with a low CD8 iTIL density.
Because a high CD8 iTIL density is associated with good prognosis, it was expected that tumors with CDX2 loss and a low CD8 iTIL density would be associated with the worst survival. Interestingly, the density of CD8 iTILs did not significantly impact the survival of patients with tumors exhibiting CDX2 loss. The underlying reason for this lack of effect remains challenging to explain. It can be speculated that CD8 iTILs might be not effective in fighting against cancers with CDX2 loss. However, little information is available to support this assumption, and thus, spatial transcriptomics at the single-cell level might provide clues to the explanation. Our result that the number of CD8 iTILs did not affect the survival of patients with tumors that had CDX2 loss seems to contradict the common belief of a better survival of patients with a high CD8 TIL number [25]. But in human cancer, renal cell carcinoma is the tissue type where many CD8 TILs are strongly related to bad clinicopathologic data and worse patient survival [26-28]. Furthermore, in pancreatic cancer, the role of CD8+ T cells is not directly associated with clinicopathological parameters [28]. Thus, loss of CDX2 expression in CRC may indeed indicate a reduction in intestinal differentiation, in which the prognostic role of CD8 TILs might differ from that in CRCs with retained CDX2 expression.
In our study, CDX2 loss was associated with increased infiltration of intraepithelial and stromal CD8-positive lymphocytes. However, the association between CDX2 loss and increased TIL density might be spurious because CDX2 loss was also associated with CIMP-H and MSI-H, which are known to be accompanied by increased TILs. Thus, to elucidate whether CDX2 loss is associated with increased TILs regardless of CIMP and MSI status, we analyzed the relationship between CDX2 loss and increased TIL density in CRCs which are negative for CIMP-H and MSI-H, namely, CIMP-L,0 and MSS CRCs (n=396). An increased density of CD8 iTILs was found in CIMP-L,0 and MSS CRCs with CDX2 loss (n=34) compared with CIMP-L,0 and MSS CRCs with retained CDX2 expression (n=362) (Supplementary Fig. S1). Such a finding indicates that the increased density of CD8 iTILs might be related to CDX2 loss itself.
In summary, we found that in patients with stage III CRC, CDX2 loss was associated with increased infiltration of CD8 iTILs or sTILs, and the density of CD8 iTILs did not significantly impact the survival of patients with CRC exhibiting CDX2 loss. The combination of CDX2 expression and intraepithelial CD8 TIL density was found to be an independent prognostic marker in adjuvant chemotherapy-treated patients with stage III CRC.
The Data Supplement is available with this article at https://doi.org/10.4132/jptm.2024.09.26.
Fig. 1.
Representative cases of colorectal carcinomas (CRCs) with retained caudal-type homeobox 2 (CDX2) expression and a low CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density (A, C) and with CDX2 loss and a high CD8 iTIL density (B, D).
jptm-2024-09-26f1.jpg
Fig. 2.
Comparison of intraepithelial or stromal tumor-infiltrating lymphocyte (iTIL or sTIL) density between colorectal carcinomas with and without CDX2 loss. CDX2–, expressional loss of CDX2; CDX2+, retained expression of CDX2.
jptm-2024-09-26f2.jpg
Fig. 3.
Kaplan-Meier curves of cancer-specific survival (A, C) and recurrence-free survival (B, D) in adjuvant FOLFOX-treated patients with stage III colorectal carcinoma (CRC) according to caudal-type homeobox 2 (CDX2) expression status (A, B) and CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density status (C, D). CDX2+ (retained expression, n = 442) and CDX2- (loss of expression, n = 63); CD8 iTIL-high (n = 223) and CD8 iTIL-low CRC (n = 223).
jptm-2024-09-26f3.jpg
Fig. 4.
Kaplan-Meier survival curves of cancer-specific survival (A) and recurrence-free survival (B). Kaplan-Meier survival analysis with the log-rank test was performed in adjuvant FOLFOX-treated patients with stage III colorectal carcinoma according to the combination of caudal-type homeobox 2 (CDX2) expression and CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density. CDX2+ (CDX2 retained)/CD8 iTIL-high (n = 188); CDX2+/CD8 iTIL-low (n = 209); CDX2– (CDX2 loss)/CD8 iTIL-high (n = 30); CDX2–/CD8 iTIL-low (n = 19).
jptm-2024-09-26f4.jpg
Table 1.
Association of clinicopathological and molecular features between CRCs with and without CDX2 loss
Parameter No. CRCs with CDX2 loss CRCs with retained CDX2 p-value
Sex .681
 Male 303 36 (42.9) 267 (39.6)
 Female 202 27 (57.1) 175 (60.4)
Location .049
 Right colon 150 26 (41.3) 124 (28.1)
 Left colon 289 33 (52.4) 256 (57.9)
 Rectum 66 4 (6.3) 62 (14.0)
Differentiation < .001
 Low 469 50 (79.4) 419 (94.8)
 High 36 13 (20.6) 23 (5.2)
Venous invasion .831
 Absent 450 57 (90.5) 393 (88.9)
 Present 55 6 (9.5) 49 (11.1)
Lymphatic emboli .172
 Absent 298 32 (50.8) 266 (60.2)
 Present 207 31 (49.2) 176 (39.8)
Perineural invasion .288
 Absent 369 50 (79.4) 319 (72.2)
 Present 136 13 (20.6) 123 (27.8)
T category .018
 T1 10 4 (6.3) 6 (1.4)
 T2 46 6 (9.5) 40 (9.0)
 T3 397 43 (68.3) 354 (80.1)
 T4 52 10 (15.9) 42 (9.5)
N category .313
 N1 348 40 (63.5) 308 (69.7)
 N2 157 23 (36.5) 134 (30.3)
CIMP < .001
 CIMP-L,0 473 51 (82.3) 422 (96.3)
 CIMP-H 27 11 (17.7) 16 (3.7)
MSI .001
 MSS 475 53 (84.1) 422 (95.9)
 MSI-H 28 10 (15.9) 18 (4.1)
KRAS .642
 Wild 346 39 (68.4) 307 (71.6)
 Mutant 129 18 (31.6) 122 (28.4)
BRAF < .001
 Wild 475 51 (85.0) 424 (98.1)
 Mutant 17 9 (15.0) 8 (1.9)
KRT20 < .001
 Decreased 68 35 (55.6) 33 (7.5)
 Retained 437 28 (44.4) 409 (92.5)
KRT7 < .001
 Not expressed 474 52 (82.5) 422 (95.5)
 Expressed 31 11 (17.5) 20 (4.5)

CRC, colorectal carcinoma; CDX2, caudal-type homeobox 2; CIMP, CpG island methylator phenotype; CIMP-L, CIMP-low; CIMP-0, no methylation; CIMP-H, CIMP-high; MSI, microsatellite instability; MSS, microsatellite-stable; MSI-H, MSI-high.

Table 2.
Univariate analysis of cancer-specific survival
Parameter No. HR (95% CI) p-value
Differentiation .014
 Low grade 469 -
 High grade 36 2.997 (1.244–7.221)
N category < .001
 N1 348 -
 N2 157 3.459 (1.759–6.803)
T category .001
 T1-3 453 -
 T4 52 3.050 (1.567–5.936)
Lymphatic emboli < .001
 Absent 298 -
 Present 207 7.586 (3.149–18.272)
Venous invasion .004
 Absent 450 -
 Present 55 3.050 (1.429–6.512)
Perineural invasion .089
 Absent 369 -
 Present 136 1.818 (0.914–3.619)
CIMP .076
 CIMP-L,0 473 -
 CIMP-H 27 2.577 (0.906–7.333)
BRAF .320
 Wild 475 -
 Mutant 17 2.070 (0.494–8.678)
KRAS .007
 Wild 346 -
 Mutant 129 2.724 (1.315–5.644)
KRT7 .008
 Not expressed 474 -
 Expressed 31 3.316 (1.377–7.989)
KRT20 .717
 Retained 437 -
 Decreased 68 0.839 (0.324–2.169)
CDX2 .037
 Retained 442 -
 Loss 63 2.314 (1.050–5.100)
CD3 iTIL .316
 Low 224 -
 High 222 0.697 (0.344–1.412)
CD3 sTIL .026
 Low 216 -
 High 230 0.428 (0.203–0.903)
CD8 iTIL .013
 Low 223 -
 High 223 0.378 (0.175–0.817)
CD8 sTIL .028
 Low 223 -
 High 223 0.433 (0.205–0.915)
Combination of the CDX2 expression and CD8 iTIL density statuses
 CDX2 retained/CD8 iTIL-high 188 -
 CDX2 loss/CD8 iTIL-low 19 7.250 (1.211–43.401) .030
 CDX2 loss/CD8 iTIL-high 30 15.781 (3.939–63.230) <.001
 CDX2 retained/CD8 iTIL-low 209 6.686 (1.994–22.419) .002

HR, hazard ratio; CI, confidence interval; CIMP, CpG island methylator phenotype; CIMP-L, CIMP-low; CIMP-0, no methylation; CIMP-H, CIMP-high; iTIL, intraepithelial tumor-infiltrating lymphocyte; sTIL, stromal tumor-infiltrating lymphocyte; CDX2, caudal-type homeobox 2.

Table 3.
Multivariate survival analysis (cancer-specific survival)
Parameter HR (95% CI) p-value
Differentiation
 High grade vs. low grade 1.733 (0.559–5.368) .341
T category
 T4 vs. T1-3 1.934 (0.769–4.867) .161
N category
 N2 vs. N1 1.943 (0.901–4.187) .090
Lymphatic emboli
 Present vs. absent 4.432 (1.779–11.041) .001
Venous invasion
 Present vs. absent 1.618 (0.646–4.053) .304
KRAS
 Mutant vs. wild 2.318 (1.115–4.819) .024
KRT7
 Expressed vs. not expressed 1.984 (0.734–5.360) .177
CD3 sTIL
 High vs. low 0.438 (0.178–1.077) .072
CD8 sTIL
 High vs. low 0.685 (0.182–2.584) .577
Combination of the CDX2 expression and CD8 iTIL density statuses
 CDX2 retained/CD8 iTIL-high -
 CDX2 loss/CD8 iTIL-low 8.176 (1.137–58.783) .037
 CDX2 loss/CD8 iTIL-high 17.868 (3.429–93.100) .001
 CDX2 retained/CD8 iTIL-low 6.769 (1.542–29.703) .011

HR, hazard ratio; CI, confidence interval; sTIL, stromal tumor-infiltrating lymphocyte; iTIL, intraepithelial tumor-infiltrating lymphocyte.

  • 1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209-49. ArticlePubMedPDF
  • 2. Cunningham D, Atkin W, Lenz HJ, et al. Colorectal cancer. Lancet 2010; 375: 1030-47. ArticlePubMed
  • 3. Bockelman C, Engelmann BE, Kaprio T, Hansen TF, Glimelius B. Risk of recurrence in patients with colon cancer stage II and III: a systematic review and meta-analysis of recent literature. Acta Oncol 2015; 54: 5-16. ArticlePubMed
  • 4. Andre T, Boni C, Mounedji-Boudiaf L, et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med 2004; 350: 2343-51. ArticlePubMed
  • 5. Twelves C, Wong A, Nowacki MP, et al. Capecitabine as adjuvant treatment for stage III colon cancer. N Engl J Med 2005; 352: 2696-704. ArticlePubMed
  • 6. Yothers G, O’Connell MJ, Allegra CJ, et al. Oxaliplatin as adjuvant therapy for colon cancer: updated results of NSABP C-07 trial, including survival and subset analyses. J Clin Oncol 2011; 29: 3768-74. ArticlePubMedPMC
  • 7. Schmoll HJ, Twelves C, Sun W, et al. Effect of adjuvant capecitabine or fluorouracil, with or without oxaliplatin, on survival outcomes in stage III colon cancer and the effect of oxaliplatin on post-relapse survival: a pooled analysis of individual patient data from four randomised controlled trials. Lancet Oncol 2014; 15: 1481-92. ArticlePubMedPMC
  • 8. Bae JM, Lee TH, Cho NY, Kim TY, Kang GH. Loss of CDX2 expression is associated with poor prognosis in colorectal cancer patients. World J Gastroenterol 2015; 21: 1457-67. ArticlePubMedPMC
  • 9. James R, Erler T, Kazenwadel J. Structure of the murine homeobox gene CDX-2: expression in embryonic and adult intestinal epithelium. J Biol Chem 1994; 269: 15229-37. ArticlePubMed
  • 10. Li MK, Folpe AL. CDX-2, a new marker for adenocarcinoma of gastrointestinal origin. Adv Anat Pathol 2004; 11: 101-5. ArticlePubMed
  • 11. Graule J, Uth K, Fischer E, et al. CDX2 in colorectal cancer is an independent prognostic factor and regulated by promoter methylation and histone deacetylation in tumors of the serrated pathway. Clin Epigenetics 2018; 10: 120.ArticlePubMedPMCPDF
  • 12. Hansen TF, Kjaer-Frifeldt S, Eriksen AC, et al. Prognostic impact of CDX2 in stage II colon cancer: results from two nationwide cohorts. Br J Cancer 2018; 119: 1367-73. ArticlePubMedPMCPDF
  • 13. Dalerba P, Sahoo D, Paik S, et al. CDX2 as a prognostic biomarker in stage II and stage III colon cancer. N Engl J Med 2016; 374: 211-22. ArticlePubMedPMC
  • 14. Zhang BY, Jones JC, Briggler AM, et al. Lack of caudal-type homeobox transcription factor 2 expression as a prognostic biomarker in metastatic colorectal cancer. Clin Colorectal Cancer 2017; 16: 124-8. ArticlePubMed
  • 15. Baba Y, Nosho K, Shima K, et al. Relationship of CDX2 loss with molecular features and prognosis in colorectal cancer. Clin Cancer Res 2009; 15: 4665-73. ArticlePubMedPMCPDF
  • 16. Konukiewitz B, Schmitt M, Silva M, et al. Loss of CDX2 in colorectal cancer is associated with histopathologic subtypes and microsatellite instability but is prognostically inferior to hematoxylin-eosin-based morphologic parameters from the WHO classification. Br J Cancer 2021; 125: 1632-46. ArticlePubMedPMCPDF
  • 17. Bae JM, Kim JH, Kwak Y, et al. Distinct clinical outcomes of two CIMP-positive colorectal cancer subtypes based on a revised CIMP classification system. Br J Cancer 2017; 116: 1012-20. ArticlePubMedPMCPDF
  • 18. Greenson JK, Huang SC, Herron C, et al. Pathologic predictors of microsatellite instability in colorectal cancer. Am J Surg Pathol 2009; 33: 126-33. ArticlePubMedPMC
  • 19. Jin HY, Yoo SY, Lee JA, et al. Combinatory statuses of tumor stromal percentage and tumor infiltrating lymphocytes as prognostic factors in stage III colorectal cancers. J Gastroenterol Hepatol 2022; 37: 551-7. ArticlePubMedPDF
  • 20. Lee JA, Seo MK, Yoo SY, et al. Comprehensive clinicopathologic, molecular, and immunologic characterization of colorectal carcinomas with loss of three intestinal markers, CDX2, SATB2, and KRT20. Virchows Arch 2022; 480: 543-55. ArticlePubMedPDF
  • 21. Yoo SY, Park HE, Kim JH, et al. Whole-slide image analysis reveals quantitative landscape of tumor-immune microenvironment in colorectal cancers. Clin Cancer Res 2020; 26: 870-81. ArticlePubMedPDF
  • 22. Weisenberger DJ, Siegmund KD, Campan M, et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 2006; 38: 787-93. ArticlePubMedPDF
  • 23. Ogino S, Cantor M, Kawasaki T, et al. CpG island methylator phenotype (CIMP) of colorectal cancer is best characterised by quantitative DNA methylation analysis and prospective cohort studies. Gut 2006; 55: 1000-6. ArticlePubMedPMC
  • 24. Derangere V, Lecuelle J, Lepage C, et al. Combination of CDX2 H-score quantitative analysis with CD3 AI-guided analysis identifies patients with a good prognosis only in stage III colon cancer. Eur J Cancer 2022; 172: 221-30. ArticlePubMed
  • 25. Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 2011; 105: 93-103. ArticlePubMedPMCPDF
  • 26. Nakano O, Sato M, Naito Y, et al. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res 2001; 61: 5132-6. ArticlePubMed
  • 27. Giraldo NA, Becht E, Pages F, et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin Cancer Res 2015; 21: 3031-40. ArticlePubMedPDF
  • 28. Blessin NC, Li W, Mandelkow T, et al. Prognostic role of proliferating CD8(+) cytotoxic T cells in human cancers. Cell Oncol (Dordr) 2021; 44: 793-803. ArticlePubMedPMCPDF

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite this Article
        Cite this Article
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers
        Close
      • XML DownloadXML Download
      Figure
      • 0
      • 1
      • 2
      • 3
      The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers
      Image Image Image Image
      Fig. 1. Representative cases of colorectal carcinomas (CRCs) with retained caudal-type homeobox 2 (CDX2) expression and a low CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density (A, C) and with CDX2 loss and a high CD8 iTIL density (B, D).
      Fig. 2. Comparison of intraepithelial or stromal tumor-infiltrating lymphocyte (iTIL or sTIL) density between colorectal carcinomas with and without CDX2 loss. CDX2–, expressional loss of CDX2; CDX2+, retained expression of CDX2.
      Fig. 3. Kaplan-Meier curves of cancer-specific survival (A, C) and recurrence-free survival (B, D) in adjuvant FOLFOX-treated patients with stage III colorectal carcinoma (CRC) according to caudal-type homeobox 2 (CDX2) expression status (A, B) and CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density status (C, D). CDX2+ (retained expression, n = 442) and CDX2- (loss of expression, n = 63); CD8 iTIL-high (n = 223) and CD8 iTIL-low CRC (n = 223).
      Fig. 4. Kaplan-Meier survival curves of cancer-specific survival (A) and recurrence-free survival (B). Kaplan-Meier survival analysis with the log-rank test was performed in adjuvant FOLFOX-treated patients with stage III colorectal carcinoma according to the combination of caudal-type homeobox 2 (CDX2) expression and CD8 intraepithelial tumor-infiltrating lymphocyte (iTIL) density. CDX2+ (CDX2 retained)/CD8 iTIL-high (n = 188); CDX2+/CD8 iTIL-low (n = 209); CDX2– (CDX2 loss)/CD8 iTIL-high (n = 30); CDX2–/CD8 iTIL-low (n = 19).
      The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers
      Parameter No. CRCs with CDX2 loss CRCs with retained CDX2 p-value
      Sex .681
       Male 303 36 (42.9) 267 (39.6)
       Female 202 27 (57.1) 175 (60.4)
      Location .049
       Right colon 150 26 (41.3) 124 (28.1)
       Left colon 289 33 (52.4) 256 (57.9)
       Rectum 66 4 (6.3) 62 (14.0)
      Differentiation < .001
       Low 469 50 (79.4) 419 (94.8)
       High 36 13 (20.6) 23 (5.2)
      Venous invasion .831
       Absent 450 57 (90.5) 393 (88.9)
       Present 55 6 (9.5) 49 (11.1)
      Lymphatic emboli .172
       Absent 298 32 (50.8) 266 (60.2)
       Present 207 31 (49.2) 176 (39.8)
      Perineural invasion .288
       Absent 369 50 (79.4) 319 (72.2)
       Present 136 13 (20.6) 123 (27.8)
      T category .018
       T1 10 4 (6.3) 6 (1.4)
       T2 46 6 (9.5) 40 (9.0)
       T3 397 43 (68.3) 354 (80.1)
       T4 52 10 (15.9) 42 (9.5)
      N category .313
       N1 348 40 (63.5) 308 (69.7)
       N2 157 23 (36.5) 134 (30.3)
      CIMP < .001
       CIMP-L,0 473 51 (82.3) 422 (96.3)
       CIMP-H 27 11 (17.7) 16 (3.7)
      MSI .001
       MSS 475 53 (84.1) 422 (95.9)
       MSI-H 28 10 (15.9) 18 (4.1)
      KRAS .642
       Wild 346 39 (68.4) 307 (71.6)
       Mutant 129 18 (31.6) 122 (28.4)
      BRAF < .001
       Wild 475 51 (85.0) 424 (98.1)
       Mutant 17 9 (15.0) 8 (1.9)
      KRT20 < .001
       Decreased 68 35 (55.6) 33 (7.5)
       Retained 437 28 (44.4) 409 (92.5)
      KRT7 < .001
       Not expressed 474 52 (82.5) 422 (95.5)
       Expressed 31 11 (17.5) 20 (4.5)
      Parameter No. HR (95% CI) p-value
      Differentiation .014
       Low grade 469 -
       High grade 36 2.997 (1.244–7.221)
      N category < .001
       N1 348 -
       N2 157 3.459 (1.759–6.803)
      T category .001
       T1-3 453 -
       T4 52 3.050 (1.567–5.936)
      Lymphatic emboli < .001
       Absent 298 -
       Present 207 7.586 (3.149–18.272)
      Venous invasion .004
       Absent 450 -
       Present 55 3.050 (1.429–6.512)
      Perineural invasion .089
       Absent 369 -
       Present 136 1.818 (0.914–3.619)
      CIMP .076
       CIMP-L,0 473 -
       CIMP-H 27 2.577 (0.906–7.333)
      BRAF .320
       Wild 475 -
       Mutant 17 2.070 (0.494–8.678)
      KRAS .007
       Wild 346 -
       Mutant 129 2.724 (1.315–5.644)
      KRT7 .008
       Not expressed 474 -
       Expressed 31 3.316 (1.377–7.989)
      KRT20 .717
       Retained 437 -
       Decreased 68 0.839 (0.324–2.169)
      CDX2 .037
       Retained 442 -
       Loss 63 2.314 (1.050–5.100)
      CD3 iTIL .316
       Low 224 -
       High 222 0.697 (0.344–1.412)
      CD3 sTIL .026
       Low 216 -
       High 230 0.428 (0.203–0.903)
      CD8 iTIL .013
       Low 223 -
       High 223 0.378 (0.175–0.817)
      CD8 sTIL .028
       Low 223 -
       High 223 0.433 (0.205–0.915)
      Combination of the CDX2 expression and CD8 iTIL density statuses
       CDX2 retained/CD8 iTIL-high 188 -
       CDX2 loss/CD8 iTIL-low 19 7.250 (1.211–43.401) .030
       CDX2 loss/CD8 iTIL-high 30 15.781 (3.939–63.230) <.001
       CDX2 retained/CD8 iTIL-low 209 6.686 (1.994–22.419) .002
      Parameter HR (95% CI) p-value
      Differentiation
       High grade vs. low grade 1.733 (0.559–5.368) .341
      T category
       T4 vs. T1-3 1.934 (0.769–4.867) .161
      N category
       N2 vs. N1 1.943 (0.901–4.187) .090
      Lymphatic emboli
       Present vs. absent 4.432 (1.779–11.041) .001
      Venous invasion
       Present vs. absent 1.618 (0.646–4.053) .304
      KRAS
       Mutant vs. wild 2.318 (1.115–4.819) .024
      KRT7
       Expressed vs. not expressed 1.984 (0.734–5.360) .177
      CD3 sTIL
       High vs. low 0.438 (0.178–1.077) .072
      CD8 sTIL
       High vs. low 0.685 (0.182–2.584) .577
      Combination of the CDX2 expression and CD8 iTIL density statuses
       CDX2 retained/CD8 iTIL-high -
       CDX2 loss/CD8 iTIL-low 8.176 (1.137–58.783) .037
       CDX2 loss/CD8 iTIL-high 17.868 (3.429–93.100) .001
       CDX2 retained/CD8 iTIL-low 6.769 (1.542–29.703) .011
      Table 1. Association of clinicopathological and molecular features between CRCs with and without CDX2 loss

      CRC, colorectal carcinoma; CDX2, caudal-type homeobox 2; CIMP, CpG island methylator phenotype; CIMP-L, CIMP-low; CIMP-0, no methylation; CIMP-H, CIMP-high; MSI, microsatellite instability; MSS, microsatellite-stable; MSI-H, MSI-high.

      Table 2. Univariate analysis of cancer-specific survival

      HR, hazard ratio; CI, confidence interval; CIMP, CpG island methylator phenotype; CIMP-L, CIMP-low; CIMP-0, no methylation; CIMP-H, CIMP-high; iTIL, intraepithelial tumor-infiltrating lymphocyte; sTIL, stromal tumor-infiltrating lymphocyte; CDX2, caudal-type homeobox 2.

      Table 3. Multivariate survival analysis (cancer-specific survival)

      HR, hazard ratio; CI, confidence interval; sTIL, stromal tumor-infiltrating lymphocyte; iTIL, intraepithelial tumor-infiltrating lymphocyte.


      J Pathol Transl Med : Journal of Pathology and Translational Medicine
      TOP