Perineural invasion is a prognostic factor in stage II colorectal cancer but not a treatment indicator for traditional chemotherapy: a retrospective cohort study
Original Article

Perineural invasion is a prognostic factor in stage II colorectal cancer but not a treatment indicator for traditional chemotherapy: a retrospective cohort study

Juan Li, Shiwen Mei, Sichen Zhou, Fuqiang Zhao, Qian Liu

Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China

Contributions: (I) Conception and design: Q Liu, J Li, S Mei; (II) Administrative support: Q Liu; (III) Provision of study materials or patients: S Mei, S Zhou; (IV) Collection and assembly of data: J Li, S Mei; (V) Data analysis and interpretation: J Li, F Zhao; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Qian Liu. Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China. Email: fcwpumch@163.com.

Background: Perineural invasion (PNI) is considered a risk factor of survival but does not yet inform treatment decisions, and has not been studied separately in stage II colorectal cancer (CRC) patients whose postoperative traditional chemotherapy is controversial. This cohort study aimed to assess the association of PNI with basic clinicopathological features and patient outcomes after curative resection and the effects of PNI on responses to adjuvant chemotherapy in stage II CRC.

Methods: The clinical data of 371 stage II CRC patients who underwent curative-intent surgery at the National Cancer Center/Cancer Hospital in 2014 were retrospectively reviewed. The adjuvant chemotherapy data were acquired from follow-up information. PNI status was examined, and the overall survival (OS) and disease-free survival (DFS) rates were analyzed.

Results: PNI was detected in 82 of the 371 patients (22.1%) and was closely correlated with preoperative serum carcinoembryonic antigen (CEA) levels (P=0.030), gross tumor type (P=0.010), tumor differentiation (P=0.010), p stage (P<0.001), and extramural vascular invasion (EMVI) (P<0.001). The median follow-up time was 71 months. The 5-year OS was 84.1% and 96.5% (P<0.001), and the 5-year DFS was 75.6% and 91.3% (P<0.001) for PNI-positive (+) and PNI-negative (−) patients, respectively. The multivariate regression analyses identified PNI as an independent negative prognostic factor for DFS [hazard ratio (HR): 2.95; 95% confidence interval (CI), 1.546–5.626; P=0.001] and OS (HR: 3.966; 95% CI, 1.642–9.575; P=0.002). Among PNI (+) patients, DFS and OS were positively correlated with CEA levels (P=0.005 and P=0.004, respectively). Postoperative chemotherapy failed to improve DFS (P=0.480 and P=0.267, respectively) and OS (P=0.940 and P=0.077, respectively) regardless of whether the patients were PNI positive or not.

Conclusions: In stage II CRC patients, PNI was a poor independent predictor for DFS and OS. Among PNI (+) patients, CEA levels were positively correlated with DFS and OS. Traditional postoperative adjuvant chemotherapy does not improve outcomes of PNI (+) patients. Therefore, as to the active role of PNI and vacancy for treatment in allusion to PNI, follow-up of PNI (+) patients with elevated CEA level should be strengthened and further research on drug conducted on PNI deserve to be carried on.

Keywords: Colorectal cancer (CRC); perineural invasion (PNI); postoperative adjuvant chemotherapy


Submitted Mar 14, 2022. Accepted for publication Apr 13, 2022.

doi: 10.21037/jgo-22-277


Introduction

With 1,148,515 newly diagnosed patients and 576,858 deaths worldwide in 2020, colorectal cancer (CRC) was the 3rd most commonly diagnosed cancer and the 2nd leading cause of cancer death (1). In total, 24% of all the newly diagnosed cases and 30% of the cancer-related deaths were reported in China (2). In China, the incidence of CRC has increased rapidly, and CRC represents one of the highest burdens of cancer globally.

Tumor-node-metastasis (TNM) staging is considered the most robust predictor of outcomes and the primary guidance for the subsequent therapy of patients with CRC. Stage II CRC accounts for approximately 25% of all CRC cases (3), and approximately 15–25% of patients relapse or die within 5 years of radical surgery (4). For primary curative CRC, adjuvant chemotherapy is inappropriate for stage I patients and considered the standard treatment for stage III patients under the National Comprehensive Cancer Network guidelines. However, the benefits of adjuvant chemotherapy for stage II patients remain controversial. Thus, supplemental pathological features are indispensable to further stratify the risk of stage II CRC.

The criteria for adjuvant therapy for patients with stage II CRC is the presence of any one of the following high risk factors including poor differentiation, emergency surgery, fewer than 12 examined lymph nodes, the presence of extramural vascular invasion (EMVI), perforation or a pT4 tumor (5). Apart from tumor and nodal stage, carcinoembryonic antigen (CEA) level, tumor regression grade, and surgical margin status, and perineural invasion (PNI) have been reported as prognostic factors for CRC (6). PNI is considered a risk factor of survival and has been included in the TNM Supplement for CRC since 2001 (7). However, PNI does not yet inform treatment decisions. As one of markers of risk factors, PNI plays an important role in tumor growth and progression by multiple signal pathways influencing interactions among tumors and nerves (8). However, people payed not enough attention to it, leading to recurrence of quite a lot of stage II CRC patients. Moreover, the clinicopathological factors associated with PNI and significance of postoperative traditional chemotherapy on PNI have not been studied separately in stage II CRC patients whose postoperative treatment is still controversial.

In this study, we examined the association of PNI with basic clinicopathological features and outcomes in stage II CRC patients after curative resection. We also evaluated the factors positively correlated with DFS and OS in PNI-positive (+) patients and estimated the effects of PNI on patients’ responses to adjuvant chemotherapy. We present the following article in accordance with the STROBE reporting checklist (available at https://jgo.amegroups.com/article/view/10.21037/jgo-22-277/rc).


Methods

Patients

The clinical data of 371 stage II CRC patients who underwent curative-intent surgery at the National Cancer Center/Cancer Hospital in 2014 were retrospectively reviewed. The patients’ clinical information was carefully collected from their medical records. To be eligible for inclusion in this study, the patients had to meet the following inclusion criteria: (I) have TNM stage II CRC as confirmed by a pathological report; (II) have been treated with curative complete mesocolic excision (CME) and total mesorectal excision (TME) surgery; and (III) have no evidence of distant metastasis. Patients were excluded from the study if they met any of the following exclusion criteria: (I) had recurrent disease; (II) had synchronous and metachronous multiple cancers; (III) had multiple primary tumors within the colorectum; (IV) had hereditary non-polyposis CRC or familial adenomatous polyposis; (V) had no PNI records; (VI) had only been treated by local resection; (VII) had received presurgical chemo- or radiation therapy; (VIII) and patients missing clinical data. Our study was approved by the Ethics Committee of the National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (the approval number: 21/13902810). The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). Informed consent was taken from all the patients.

Curative-intent surgery was defined as follows: (I) an absence of a gross residual tumor in the surgical bed; and (II) pathologically negative proximal and distal resection margins in terms of tumor invasion. The surgical approach was uniformly mesenteric-based CME or TME. The surgical pathology specimens were evaluated by at least 3 specialized colorectal pathologists at our Center. Clinicopathological staging was assessed according to the American Joint Commission on Cancer/Union for International Cancer Control TNM staging system (2017, 8th ed.). All patients were proven to suffer from TNM stage II CRC. The clinicopathological features included (I) tumor size, (II) gross type, (III) pathological cell type, (IV) invasion depth, (V) lymph node involvement, (VI) histologic grade, (VII) differentiation, (VIII) resection margin, (IX) EMVI, (X) PNI, and (XI) microsatellite instability (MSI) status. PNI was assessed as positive when tumor cells were observed inside any layer of the nerve sheath or when at least 33% of the nerve periphery was surrounded by tumor cells. EMVI was assessed as positive when tumor cells were observed within an endothelium-lined vessel beyond the muscularis propria. This is a retrospective cohort study with selection bias.

Follow up

The follow-up data were acquired by outpatient reexaminations and telephone interviews. All patients were scheduled to receive follow-up outpatient visits or telephone interviews every 3 months for the first 2 years and then every 6–12 months for the next 3 years. Routine examinations included contrast-enhanced computed tomography imaging of the thorax, abdomen, and pelvis, blood tests of tumor markers every 3 months, and colonoscopies every 6 months. Overall survival (OS) time was defined as the time interval from the day of surgery to the date of last follow-up or death from any cause. Disease-free survival (DFS) time was defined as the time from the day of surgery to the day of local recurrence and/or metastases. The endpoints of this study were 5-year OS and DFS.

Statistical analysis

The relationships between PNI positivity and other clinicopathological variables were assessed using the chi-square test and Student’s t-test (the categorical variables were analyzed using the chi-square test, and the continuous variables were analyzed using Student’s t-test). The effects of PNI on OS and DFS were estimated using the Kaplan-Meier method and compared using a log-rank test. To identify the factors that independently affected OS and DFS, the hazard ratio (HR) with a 95% confidence interval (CI) was computed using a univariate Cox proportional-hazards regression model. Next, the predictive value of PNI was analyzed using the multivariate Cox proportional-hazards regression model, which included all the predictive factors with a P value <0.05 from the univariate analyses. The statistical analyses were performed using SPSS software for Mac (version 26.0; SPSS Inc., Chicago, IL, USA). All the tests were two-sided, and the results were considered statistically significant when the P value was <0.05.


Results

Patient characteristics and pathological variables

The patient characteristics and pathological variables of the 371 patients are summarized in Table 1. The cohort of patients comprised 229 men (61.7%) and 142 women (38.3%), with a median age of 60 (range, 20–83) years at the time of the curative-intent resection. Among the patients, 88 (23.7%), 113 (30.5%), and 170 (45.8%) of the tumors occurred in the right colon, left colon, and rectum, respectively. Using the American Joint Committee on Cancer TNM staging system, 299 (80.6%) patients were classified as pT3, and 72 (19.4%) were classified as pT4. The median tumor size was 4.5 (range, 1.6–14) cm, and PNI was observed in 82 (22.1%) patients. After surgery, 157 (42.3%) patients received adjuvant chemotherapy, and 45 (12.1%) suffered from postoperative morbidity.

Table 1

Clinical histopathological characteristics of PNI stage II CRC patients who underwent curative-intent surgery at the National Cancer Center/Cancer Hospital in 2014

Characteristics Total (n=371)
Age at operation (years), median, range 60, 20–83
   <60, n (%) 179 (48.2)
   ≥60, n (%) 192 (51.8)
Gender, n (%)
   Male 229 (61.7)
   Female 142 (38.3)
Family history, n (%)
   Yes 61 (83.6)
   No 310 (16.4)
Preoperative CEA level (ng/mL), median, range 2.965, 0.47–342.4
Location of the tumor, n (%)
   Right-sided colon 88 (23.7)
   Left-sided colon 113 (30.5)
   Rectum 170 (45.8)
Maximal diameter of the tumor (cm), median, range 4.5, 1.6–14
   <4.5, n (%) 160 (43.1)
   ≥4.5, n (%) 211 (56.9)
Gross type, n (%)
   Protruded 177 (47.7)
   Ulcerative 193 (52.0)
   Unknown 1 (0.3)
Tumor differentiation, n (%)
   Poor/mucinous 95 (25.6)
   Good/moderate 276 (74.4)
Tumor p stage, n (%)
   T3 299 (80.6)
   T4a 66 (17.8)
   T4b 6 (1.6)
PNI, n (%)
   Yes 82 (22.1)
   No 289 (77.9)
EMVI, n (%)
   Yes 42 (11.3)
   No 321 (86.5)
   Unknow 8 (2.2)
MSI, n (%)
   Positive 39 (10.5)
   Negative 332 (89.5)
Postoperative chemotherapy, n (%)
   Yes 157 (42.3)
   No 210 (56.6)
   Unknow 4 (1.1)
Postoperative morbidity, n (%)
   Yes 45 (12.1)
   No 326 (87.9)

PNI, perineural invasion; CRC, colorectal cancer; CEA, carcinoembryonic antigen; EMVI, extramural vascular invasion; MSI, microsatellite instability.

Clinicopathological variables associated with the incidence of PNI among stage II CRC patients

The comparison results for the patients according to their PNI status are set out in Table 2. The incidence of PNI was significantly correlated with preoperative serum CEA levels (P=0.030), gross tumor type (P=0.010), tumor differentiation (P=0.010), p stage (P<0.001), and EMVI (P<0.001). A high CEA level, an ulcerative type of lesion, poor differentiation, T4 stage, and EMVI-negative patients were more frequently identified as PNI (+). Additionally, the incidence of PNI was not associated with age, sex, family history, tumor location, tumor size, or MSI status. After surgery, the PNI (+) patients were more likely to accept adjuvant chemotherapy than PNI-negative (−) patients (P=0.008).

Table 2

Comparison of patients according to whether PNI was positive or not

Characteristics PNI (+) (n=82) PNI (−) (n=289) P value
N % N %
Age at operation (years), median, range 61, 21–78 60, 20–83 25.6 0.695
   <60 38 46.3 141 48.8
   ≥60 44 53.7 148 51.2
Gender 0.874
   Male 50 61 179 61.9
   Female 32 39 110 38.1
Family history 0.861
   Yes 14 17.1 47 16.3
   No 68 82.9 242 83.7
Preoperative CEA level (ng/mL), median, range 4.37, 0.72–342.4 2.77, 0.47–228.2 0.030
Location of the tumor 0.655
   Right-sided colon 17 20.7 71 24.6
   Left-sided colon 24 29.3 89 30.8
   Rectum 41 50 129 44.6
Maximal diameter of the tumor (cm), median, range 4.5, 2.2–10.5 4.5, 1.6–8.4 0.872
   <4.5 36 43.9 124 42.9
   ≥4.5 46 56.1 165 57.1
Gross type 0.010
   Protruded 29 35.4 148 51.4
   Ulcerative 53 64.6 140 48.6
Tumor differentiation 0.010
   Poor/mucinous 30 36.6 65 22.5
   Good/moderate 52 63.4 224 77.5
Tumor p stage <0.001
   T3 52 63.4 247 85.5
   T4a 28 34.1 38 13.1
   T4b 2 2.4 4 1.4
EMVI <0.001
   Yes 55 73.3 266 92.4
   No 20 26.7 22 7.6
MSI 0.509
   Positive 7 8.5 32 11.1
   Negative 75 91.5 257 88.9
Postoperative morbidity <0.001
   Yes 20 24.4 25 8.7
   No 62 75.6 264 91.3
Postoperative chemotherapy 0.008
   Yes 45 55.6 112 39.2
   No 36 44.4 174 60.8

PNI, perineural invasion; PNI (+), PNI-positive; PNI (−), PNI-negative; CEA, carcinoembryonic antigen; EMVI, extramural vascular invasion; MSI, microsatellite instability;

Significance of PNI as a predictor among patients with stage II CRC

The prognostic significance of PNI and the other clinicopathological variables was examined using univariate regression models (see Table 3). PNI and preoperative serum CEA levels were the only two factors that were associated with both OS and DFS. Conversely, age, sex, family history, tumor location, tumor size, differentiation, EMVI and MSI status, and postoperative chemotherapy did not affect patient outcomes.

Table 3

Univariate Cox-regression analyses of potential prognostic factors for OS and DFS in stage II CRC patients who underwent curative-intent surgery

Variables Univariate analysis
OS DFS
HR (95% CI) P HR (95% CI) P
Age, years 1.022 (0.986–1.058) 0.232 1.020 (0.995–1.047) 0.123
Gender 0.957 (0.440–2.083) 0.912 0.706 (0.391–1.274) 0.248
Family history 0.867 (0.300–2.511) 0.793 1.173 (0.589–2.338) 0.650
Preoperative CEA level, ng/mL 1.012 (1.004–1.019) 0.002 1.009 (1.002–1.015) 0.007
Location of the tumor 1.136 (0.712–1.181) 0.593 1.427 (0.992–2.051) 0.055
Maximal diameter of the tumor, cm 0.969 (0.774–1.212) 0.780 0.859 (0.715–1.032) 0.105
Gross type 1.613 (0.744–3.498) 0.226 1.275 (0.739–2.200) 0.383
Tumor differentiation 0.848 (0.373–1.928) 0.693 0.849 (0.467–1.544) 0.592
Tumor p stage 1.286 (0.605–2.734) 0.513 1.017 (0.561–1.844) 0.956
PNI 4.902 (2.291–10.491) <0.001 3.075 (1.785–5.298) <0.001
EMVI 1.097 (0.328–3.668) 0.880 1.926 (0.934–3.971) 0.076
MSI 0.235 (0.031–1.760) 0.159 0.416 (0.128–1.352) 0.145
Postoperative therapy 0.782 (0.358–1.708) 0.537 1.073 (0.621–1.855) 0.801

OS, overall survival; DFS, disease-free survival; CRC, colorectal cancer; HR, hazard ratio; CI, confidence interval; CEA, carcinoembryonic antigen; PNI, perineural invasion; EMVI, extramural vascular invasion; MSI, microsatellite instability.

With a median follow-up period of 71 (range, 2–86) months, the 5-year OS rate of this cohort was 94.5%, and the 5-year DFS rate was 87.9%. In all patients, PNI affected OS [84.1% vs. 96.5% for PNI (+) and PNI (−) patients, respectively, P<0.001; Figure 1A]. Notably, when the patients were divided according to tumor location (right-sided colon, left-sided colon, and rectum), their OS rates (P=0.011, P=0.866, and P<0.001, respectively; Figure 1B-1D) were differentially affected by PNI. DFS was the same as OS, PNI affected DFS [75.6% vs. 91.3% for PNI (+) and PNI (−), respectively, P<0.001; Figure 2A], and for right-sided colon, left-sided colon, and rectum their DFS were also differently affected (P=0.005, P=0.613, and P<0.001, respectively; Figure 2B-2D). In brief, while the occurrence of PNI was not associated with the location of the tumor, the presence of PNI was correlated with the prognosis of CRC at different sites. PNI (+) right-sided colon cancer and rectal cancer patients had lower OS and DFS than PNI (−) patients; however, no such differences were observed in the left-sided colon cancer patients.

Figure 1 Kaplan-Meier analysis of OS in stage II CRC patients according to tumor sites: (A) CRC as a whole, (B) right-sided colon cancer, (C) left-sided colon cancer; and (D) rectal cancer. CRC, colorectal cancer; PNI, perineural invasion; PNI (+), PNI-positive; PNI (−), PNI-negative; OS, overall survival.
Figure 2 Kaplan-Meier analysis of DFS in stage II CRC patients according to tumor sites: (A) CRC as a whole, (B) right-sided colon cancer, (C) left-sided colon cancer, and (D) rectal cancer. CRC, colorectal cancer; PNI, perineural invasion; PNI (+), PNI-positive; PNI (−), PNI-negative; DFS, disease-free survival.

PNI is an independent prognostic factor for outcomes among stage II CRC patients

According to the univariate regression models, PNI and preoperative serum CEA levels were correlated with OS and DFS. The multivariate analysis also revealed that both PNI and preoperative serum CEA levels were independent prognostic factors of DFS (see Table 4).

Table 4

Multivariate Cox-regression analysis of OS and DFS in stage II CRC patients

Variables Multivariate analysis
OS DFS
HR (95% CI) P HR (95% CI) P
Preoperative CEA level, ng/mL 1.011 (1.004–1.019) 0.002 1.009 (1.002–1.015) 0.006
PNI 3.966 (1.642–9.575) 0.002 2.950 (1.546–5.626) 0.001

OS, overall survival; DFS, disease-free survival; CRC, colorectal cancer; HR, hazard ratio; CI, confidence interval; CEA, carcinoembryonic antigen; PNI, perineural invasion.

Clinicopathological variables associated with outcomes of PNI (+) stage II CRC patients

According to the univariate regression models (see Table 5), preoperative serum CEA levels were the only factor correlated with OS and DFS in PNI (+) stage II CRC patients.

Table 5

Univariate Cox-regression analysis of OS and DFS in PNI (+) stage II CRC patients

Variables Univariate analysis
OS DFS
HR (95% CI) P HR (95% CI) P
Age, years 1.011 (0.961–1.064) 0.665 1.025 (0.983–1.068) 0.245
Gender 0.853 (0.286–2.546) 0.775 0.930 (0.385–2.243) 0.871
Family history 0.642 (0.140–2.511) 0.567 0.761 (0.252–2.293) 0.627
Preoperative CEA level, ng/mL 1.012 (1.004–1.020) 0.005 1.012 (1.004–1.020) 0.004
Location of the tumor 1.167 (0.582–2.341) 0.664 1.269 (0.714–2.258) 0.417
Maximal diameter of the tumor, cm 1.040 (0.743–1.456) 0.819 0.936 (0.703–1.245) 0.648
Gross type 0.605 (0.219–1.676) 0.334 0.615 (0.270–1.399) 0.246
Tumor differentiation 1.407 (0.441–4.487) 0.564 1.976 (0.724–5.394) 0.184
Tumor p stage 0.713 (0.278–1.828) 0.481 1.359 (0.701–2.633) 0.363
EMVI 0.518 (0.116–2.317) 0.390 1.008 (0.369–2.754) 0.987
MSI 0.500 (0.060–4.151) 0.521 0.267 (0.034–2.114) 0.211
Postoperative therapy 1.042 (0.361–3.004) 0.940 1.371 (0.568–3.308) 0.483

OS, overall survival; DFS, disease-free survival; PNI, perineural invasion; PNI (+), PNI-positive; CRC, colorectal cancer; HR, hazard ratio; CI, confidence interval; CEA, carcinoembryonic antigen; EMVI, extramural vascular invasion; MSI, microsatellite instability.

Effects of adjuvant chemotherapy on PNI in patients with stage II CRC

Given the importance of PNI, we analyzed whether postoperative adjuvant chemotherapy improved outcomes in the PNI (+) group. The results showed that postoperative adjuvant chemotherapy did not improve OS (P=0.077 and P=0.940, respectively; Figure 3A,3B) or DFS (P=0.267 and P=0.480, respectively; Figure 3C,3D) in either PNI (−) or PNI (+) CRC patients.

Figure 3 Kaplan-Meier analysis of OS and DFS in stage II CRC patients according to postoperative adjuvant chemotherapy status and PNI status. (A) OS of PNI (−) patients, (B) OS of PNI (+) patients, (C) DFS of PNI (−) patients, (D) DFS of PNI (+) patients. PNI, perineural invasion; PNI (−), PNI-negative; CRC, colorectal cancer; PNI (+), PNI-positive; OS, overall survival; DFS, disease-free survival.

Discussion

The most widely accepted definition of PNI is tumor cells within any layer of the nerve sheath or tumor cells in the perineural space that involves at least 1/3 of the nerve circumference (9). The clinical significance of PNI was first described in head and neck carcinomas and was considered to be a risk factor for intracranial extension (10). More recently, PNI has also been identified as a key pathological feature of other solid tumors, including pancreatic cancer (11), prostate cancer (12), biliary tract cancer (13) and gastric cancer (14). Additionally, many studies have shown the significance of PNI in CRC (15,16).

A number of hypotheses have been put forward as to the route for CRC metastasis through PNI. For example, it has been hypothesized that cancer cells within the nerve spread to the celiac ganglia along the superior mesenteric and then to the liver because of the shared preganglionic origin of the sympathetic nerves that innervate the liver and colorectum (17,18). Conversely, others have hypothesized that PNI is an invasion rather than a simple tumor diffusion: PNI induces changes in the perineural microenvironment, alters neurotrophic factors and chemokines, and enhances the ability of tumor cell invasion, inducing cancer aggressiveness and metastasis (19).

In our study, PNI was observed in 22.1% of the patients who underwent surgery for stage II CRC. This PNI prevalence conformed with a systematic review that found a weighted average detection rate for PNI of 17% (range, 8–42%) (7). A previous study revealed that the presence of PNI was a significant independent prognostic factor of DFS in multivariate analyses of stage I–II colon cancer patients treated with curative surgery (20). Regarding rectal cancer, Kinugasa et al. reported that PNI was a significant prognostic factor and that PNI status should be considered for therapy stratification (17). Consistent with previous studies, our study showed that PNI is an independent prognostic factor for outcomes among stage II CRC patients in univariate and multivariate analyses (21,22). To better understand the clinicopathological factors associated with PNI, we first found that the incidence of PNI in stage II CRC patients was significantly correlated with preoperative serum CEA levels, gross tumor type, tumor differentiation, p stage, and EMVI.

Both OS and DFS were impaired by PNI. Notably, we first found that while the occurrence of PNI was not associated with the location of the tumor, the presence of PNI was correlated with the prognosis of CRC in different sites. This result may have been caused by the clinical and molecular heterogeneity of different tumor sites. In the current era of personalized medicine, carcinomas of the right colon, left colon, and rectum are regarded as different tumor entities. From the perspective of the embryonic origin, right-sided colon cancer derives from the embryonic midgut, while left-sided colon cancer and rectal cancer derive from the embryonic hindgut (23). The rectum embryonically belongs to the hindgut; however, rectal cancer differs from colon cancer in its metastatic patterns and therapy-related factors (24,25) and thus is often described separately (26). The splenic flexure is often used to differentiate between the left and right sides of the colon in clinical retrospective report (27). From the perspective of anatomy and blood supply, the superior mesenteric artery supplies the proximal colon, while the inferior mesentery artery supplies the distal colon and rectum because of the different embryological origins (28). From a molecular point of view, right-sided colon cancers have higher rates of MSI and higher v-raf murine sarcoma viral oncogene homolog B1, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha, and transforming growth factor, beta receptor II mutation rates (29), while left-sided colon cancer is more associated with mutations in the adenomatous polyposis coli, Kirsten-ras, SMAD4, and TP53 genes (30). Rectal cancers have higher rates of TOPO1 expression and Her2/neu amplification (31). Additionally, gut microbiota dysbiosis is involved in the progression of CRC (32), and different sites of the colorectum have different microbiota phenotypes (33).

As to the significance of PNI, we further analyzed the clinicopathological variables associated with outcomes of PNI (+) stage II CRC patients. However, only preoperative serum CEA levels were a positive indicator for further treatment. Thus, elevated CEA levels can guide the selection of patients for further treatment.

Several studies have suggested that PNI status can be used to select patients with stage I–II CRC for adjuvant chemotherapy (20,34). Fluorouracil-based adjuvant chemotherapy was shown to reverse the adverse effect of PNI on 5- to 10-year DFS (20). This study failed to find that postoperative adjuvant chemotherapy improved the OS and DFS of stage II CRC patients. However, given the significance of PNI, therapies specifically effective for PNI need to be further explored.

Our study has several limitations. First, it was a single-center and retrospective study and had a small sample size. Second, patients had difficulties recalling their chemotherapy regimens and durations during follow up.


Conclusions

In the present study, we found that PNI is an independent prognostic factor in stage II CRC patients, and the incidence of PNI is significantly correlated with preoperative serum CEA levels, gross tumor type, tumor differentiation, p stage, and EMVI. PNI was an independent poor predictor for DFS and OS. Among the PNI (+) patients, preoperative serum CEA levels were positively correlated with DFS and OS. Traditional postoperative adjuvant chemotherapy did not improve the outcomes of PNI (+) stage II patients. However, given the importance of PNI, further research should be conducted on PNI.


Acknowledgments

Funding: This work was supported by the Medicine and Health Technology Innovation Project of Chinese Academy of Medical Sciences (No. 2017-12M-1-006).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://jgo.amegroups.com/article/view/10.21037/jgo-22-277/rc

Data Sharing Statement: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-22-277/dss

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jgo.amegroups.com/article/view/10.21037/jgo-22-277/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The Ethics Committee of the National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College approved this study (approval number: 21/13902810). The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). Informed consent was taken from all the patients.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  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. [Crossref] [PubMed]
  2. Cao W, Chen HD, Yu YW, et al. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J (Engl) 2021;134:783-91. [Crossref] [PubMed]
  3. Lee JJ, Chu E. Adjuvant Chemotherapy for Stage II Colon Cancer: The Debate Goes On. J Oncol Pract 2017;13:245-6. [Crossref] [PubMed]
  4. Li X, Xiong Z, Xie M, et al. Prognostic value of the ratio of carcinoembryonic antigen concentration to maximum tumor diameter in patients with stage II colorectal cancer. J Gastrointest Oncol 2021;12:1470-81. [Crossref] [PubMed]
  5. Benson AB 3rd, Schrag D, Somerfield MR, et al. American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol 2004;22:3408-19. [Crossref] [PubMed]
  6. Valentini V, van Stiphout RG, Lammering G, et al. Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of European randomized clinical trials. J Clin Oncol 2011;29:3163-72. [Crossref] [PubMed]
  7. van Wyk HC, Going J, Horgan P, et al. The role of perineural invasion in predicting survival in patients with primary operable colorectal cancer: A systematic review. Crit Rev Oncol Hematol 2017;112:11-20. [Crossref] [PubMed]
  8. Nozawa H, Hata K, Ushiku T, et al. Accelerated perineural invasion in colitis-associated cancer: A retrospective cohort study. Medicine (Baltimore) 2019;98:e17570. [Crossref] [PubMed]
  9. Chatzistefanou I, Lubek J, Markou K, et al. The role of perineural invasion in treatment decisions for oral cancer patients: A review of the literature. J Craniomaxillofac Surg 2017;45:821-5. [Crossref] [PubMed]
  10. Song JH, Yu M, Kang KM, et al. Significance of perineural and lymphovascular invasion in locally advanced rectal cancer treated by preoperative chemoradiotherapy and radical surgery: Can perineural invasion be an indication of adjuvant chemotherapy? Radiother Oncol 2019;133:125-31. [Crossref] [PubMed]
  11. Jurcak NR, Rucki AA, Muth S, et al. Axon Guidance Molecules Promote Perineural Invasion and Metastasis of Orthotopic Pancreatic Tumors in Mice. Gastroenterology 2019;157:838-50.e6. [Crossref] [PubMed]
  12. Delahunt B, Murray JD, Steigler A, et al. Perineural invasion by prostate adenocarcinoma in needle biopsies predicts bone metastasis: Ten year data from the TROG 03.04 RADAR Trial. Histopathology 2020;77:284-92. [Crossref] [PubMed]
  13. Maruyama S, Kawaida H, Hosomura N, et al. Indications for extrahepatic bile duct resection due to perineural invasion in patients with gallbladder cancer. World J Surg Oncol 2019;17:200. [Crossref] [PubMed]
  14. España-Ferrufino A, Lino-Silva LS, Salcedo-Hernández RA. Extramural Perineural Invasion in pT3 and pT4 Gastric Carcinomas. J Pathol Transl Med 2018;52:79-84. [Crossref] [PubMed]
  15. Knijn N, Mogk SC, Teerenstra S, et al. Perineural Invasion is a Strong Prognostic Factor in Colorectal Cancer: A Systematic Review. Am J Surg Pathol 2016;40:103-12. [Crossref] [PubMed]
  16. Cao Y, Deng S, Yan L, et al. Perineural invasion is associated with poor prognosis of colorectal cancer: a retrospective cohort study. Int J Colorectal Dis 2020;35:1067-75. [Crossref] [PubMed]
  17. Kinugasa T, Mizobe T, Shiraiwa S, et al. Perineural Invasion Is a Prognostic Factor and Treatment Indicator in Patients with Rectal Cancer Undergoing Curative Surgery: 2000-2011 Data from a Single-center Study. Anticancer Res 2017;37:3961-8. [PubMed]
  18. McCuskey RS. Anatomy of efferent hepatic nerves. Anat Rec A Discov Mol Cell Evol Biol 2004;280:821-6. [Crossref] [PubMed]
  19. Marchesi F, Piemonti L, Mantovani A, et al. Molecular mechanisms of perineural invasion, a forgotten pathway of dissemination and metastasis. Cytokine Growth Factor Rev 2010;21:77-82. [Crossref] [PubMed]
  20. Cienfuegos JA, Martínez P, Baixauli J, et al. Perineural Invasion is a Major Prognostic and Predictive Factor of Response to Adjuvant Chemotherapy in Stage I-II Colon Cancer. Ann Surg Oncol 2017;24:1077-84. [Crossref] [PubMed]
  21. Poeschl EM, Pollheimer MJ, Kornprat P, et al. Perineural invasion: correlation with aggressive phenotype and independent prognostic variable in both colon and rectum cancer. J Clin Oncol 2010;28:e358-60; author reply e361-2. [Crossref] [PubMed]
  22. Liebig C, Ayala G, Wilks J, et al. Perineural invasion is an independent predictor of outcome in colorectal cancer. J Clin Oncol 2009;27:5131-7. [Crossref] [PubMed]
  23. Shen H, Yang J, Huang Q, et al. Different treatment strategies and molecular features between right-sided and left-sided colon cancers. World J Gastroenterol 2015;21:6470-8. [Crossref] [PubMed]
  24. Tamas K, Walenkamp AM, de Vries EG, et al. Rectal and colon cancer: Not just a different anatomic site. Cancer Treat Rev 2015;41:671-9. [Crossref] [PubMed]
  25. Riihimäki M, Hemminki A, Sundquist J, et al. Patterns of metastasis in colon and rectal cancer. Sci Rep 2016;6:29765. [Crossref] [PubMed]
  26. Hemminki K, Försti A, Hemminki A. Survival in colon and rectal cancers in Finland and Sweden through 50 years. BMJ Open Gastroenterol 2021;8:e000644. [Crossref] [PubMed]
  27. Mukund K, Syulyukina N, Ramamoorthy S, et al. Right and left-sided colon cancers - specificity of molecular mechanisms in tumorigenesis and progression. BMC Cancer 2020;20:317. [Crossref] [PubMed]
  28. San Roman AK, Shivdasani RA. Boundaries, junctions and transitions in the gastrointestinal tract. Exp Cell Res 2011;317:2711-8. [Crossref] [PubMed]
  29. Stintzing S, Tejpar S, Gibbs P, et al. Understanding the role of primary tumour localisation in colorectal cancer treatment and outcomes. Eur J Cancer 2017;84:69-80. [Crossref] [PubMed]
  30. Mouradov D, Sloggett C, Jorissen RN, et al. Colorectal cancer cell lines are representative models of the main molecular subtypes of primary cancer. Cancer Res 2014;74:3238-47. [Crossref] [PubMed]
  31. Salem ME, Weinberg BA, Xiu J, et al. Comparative molecular analyses of left-sided colon, right-sided colon, and rectal cancers. Oncotarget 2017;8:86356-68. [Crossref] [PubMed]
  32. Gao Z, Guo B, Gao R, et al. Microbiota disbiosis is associated with colorectal cancer. Front Microbiol 2015;6:20. [Crossref] [PubMed]
  33. Gao R, Kong C, Huang L, et al. Mucosa-associated microbiota signature in colorectal cancer. Eur J Clin Microbiol Infect Dis 2017;36:2073-83. [Crossref] [PubMed]
  34. Quah HM, Chou JF, Gonen M, et al. Identification of patients with high-risk stage II colon cancer for adjuvant therapy. Dis Colon Rectum 2008;51:503-7. [Crossref] [PubMed]

(English Language Editor: L. Huleatt)

Cite this article as: Li J, Mei S, Zhou S, Zhao F, Liu Q. Perineural invasion is a prognostic factor in stage II colorectal cancer but not a treatment indicator for traditional chemotherapy: a retrospective cohort study. J Gastrointest Oncol 2022;13(2):710-721. doi: 10.21037/jgo-22-277

Download Citation