Prognostic and safety analysis of capecitabine monotherapy versus CapeOx (capecitabine plus oxaliplatin) in elderly patients with high-risk stage II and stage III MRD-negative colorectal cancer
Original Article

Prognostic and safety analysis of capecitabine monotherapy versus CapeOx (capecitabine plus oxaliplatin) in elderly patients with high-risk stage II and stage III MRD-negative colorectal cancer

Jiawei Zhai1 ORCID logo, Rongjian Tan1, Wenting Ou2, Zhenhao Quan1, Lijun Sun1, Xianjin Huang1, Jiayuan Wu3, Caijin Zhou1

1Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China; 2Department of Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China; 3Department of Clinical Trials Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China

Contributions: (I) Conception and design: J Zhai, R Tan, W Ou, C Zhou; (II) Administrative support: C Zhou; (III) Provision of study materials or patients: C Zhou, W Ou, X Huang, L Sun; (IV) Collection and assembly of data: J Zhai, L Sun, Z Quan; (V) Data analysis and interpretation: None; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Caijin Zhou, PhD. Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, No. 57, South of Renmin Avenue, Zhanjiang 524000, China. Email: zhoucaipingren@163.com.

Background: Postoperative adjuvant chemotherapy is a cornerstone of treatment for high-risk stage II and stage III colorectal cancer (CRC). However, the optimal regimen for elderly minimal residual disease (MRD)-negative patients remains controversial. This study aimed to compare the efficacy and safety of capecitabine monotherapy versus the CapeOx regimen (capecitabine plus oxaliplatin) in this population.

Methods: We conducted a retrospective analysis of 117 elderly patients (aged 65–85 years) with high-risk stage II and stage III CRC who underwent curative surgery and tested MRD-negative. Patients were divided into two groups based on postoperative chemotherapy: capecitabine monotherapy (n=58) and CapeOx (n=59). Baseline characteristics, disease-free survival (DFS), 3-year DFS rates, and adverse events were compared. Propensity score matching (PSM) was used to balance baseline differences, and Kaplan-Meier analysis was performed to assess survival outcomes.

Results: The median DFS was 31 months in the capecitabine group and 40 months in the CapeOx group (P=0.26). The 3-year DFS rates were comparable (90% vs. 81%, P=0.26). After PSM, the survival outcomes remained similar between the two groups (P=0.73). However, the CapeOx group exhibited significantly higher rates of neurotoxicity (61% vs. 7%, P<0.001), leukopenia (49% vs. 22%, P=0.002), and thrombocytopenia (44% vs. 10%, P<0.001).

Conclusions: Capecitabine monotherapy might provide comparable DFS to CapeOx in elderly MRD-negative CRC patients, with better safety profile. MRD status might guide personalized dose-reduction strategies for elderly patients with high-risk stage II and stage III CRC.

Keywords: Colorectal cancer (CRC); chemotherapy; prognosis; minimal residual disease (MRD); elderly patient


Submitted Jun 25, 2025. Accepted for publication Sep 17, 2025. Published online Oct 29, 2025.

doi: 10.21037/jgo-2025-504


Highlight box

Key findings

• Capecitabine monotherapy might provide comparable disease-free survival to CapeOx (capecitabine plus oxaliplatin) in elderly minimal residual disease (MRD)-negative colorectal cancer (CRC) patients, with better safety. MRD status might guide personalized dose-reduction strategies for elderly patients with high-risk stage II and stage III CRC.

What is known and what is new?

• We already know from MOSAIC study that adding oxaliplatin to those older than 70 is not significantly beneficial. This is also supported by National Comprehensive Cancer Network guidelines.

• The role of MRD status in guiding chemotherapy decisions for elderly patients with high-risk CRC remains underexplored. There are few comparative studies on the prognosis of postoperative CapeOx chemotherapy and monotherapy capecitabine chemotherapy in elderly patients with high-risk stage II and stage III CRC and negative MRD.

What is the implication, and what should change now?

• This study may provide some evidence to support that MRD negativity may have certain guiding significance for postoperative reduced-drug chemotherapy in elderly patients with high-risk stage II and stage III CRC, enabling elderly patients with a low possibility of recurrence to avoid the damage caused by excessive chemotherapy.


Introduction

Background

Colorectal cancer (CRC) is the third most common cancer worldwide and a leading cause of cancer-related mortality (1). For patients with high-risk stage II and stage III CRC, postoperative adjuvant chemotherapy is a critical intervention to reduce recurrence rates and improve survival outcomes. The Chinese Society of Clinical Oncology (CSCO) guidelines recommend 3–6 months of combination chemotherapy (Category I) for high-risk patients, with the CapeOx regimen (capecitabine plus oxaliplatin) being the preferred option (2). However, for elderly patients, who often present with comorbidities and reduced physiological reserve, the toxicity associated with combination chemotherapy can outweigh its benefits, leading to compromised quality of life and treatment discontinuation (3-5).

Rationale and knowledge gap

Minimal residual disease (MRD), as detected by circulating tumor DNA (ctDNA) analysis, has emerged as a promising biomarker for risk stratification in CRC (6-10). MRD negativity indicates the low risk of recurrence, potentially enabling de-escalation of adjuvant therapy (7,11,12). While the CapeOx regimen has demonstrated efficacy in improving survival, its associated toxicities, particularly neurotoxicity and hematological adverse events, are a significant concern in elderly populations (13-18). In contrast, capecitabine monotherapy offers a more favorable safety profile but is often considered less effective in high-risk patients (5). Moreover, the role of MRD status in guiding chemotherapy decisions for elderly patients with high-risk CRC remains underexplored.

Objective

This study aimed to evaluate the efficacy and safety of capecitabine monotherapy versus the CapeOx regimen in elderly patients with high-risk stage II and stage III CRC who were MRD-negative. By comparing disease-free survival (DFS) and adverse event rates between the two regimens, we sought to determine whether MRD negativity could identify a subgroup of patients who could safely forego further intensive combination chemotherapy without compromising survival outcomes. Our study provides valuable insights into personalized treatment strategies for elderly CRC patients, with the potential to optimize therapeutic outcomes while minimizing toxicity. We present this article in accordance with the STROBE reporting checklist (available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-504/rc).


Methods

Study design and population

This retrospective study included 117 elderly patients (aged 65–85 years) with high-risk stage II and stage III CRC who underwent curative surgery and tested negative for MRD between January 2018 and December 2022 at the Affiliated Hospital of Guangdong Medical University (Figure 1). Patients were excluded if they had recurrent tumors, familial adenomatous polyposis, or hereditary non-polyposis CRC.

Figure 1 Flow chart of the study. MRD-negative patients with high-risk stage II and stage III CRC were included. CapeOx, capecitabine plus oxaliplatin; CRC, colorectal cancer; MRD, minimal residual disease.

The study was conducted in accordance with the Declaration of Helsinki and its subsequent amendments. The study was approved by the Human Ethics Committee of the Affiliated Hospital of Guangdong Medical University (No. PJKT2024-063). The requirement for patient approval or informed consent was waived by the Affiliated Hospital of Guangdong Medical University, owing to the retrospective nature of the study and the athe use of de-identified data.

MRD assessment

MRD status was determined within 14–21 days postoperatively. A negative MRD result was defined as the absence of the tumor-derived molecular variant in peripheral blood samples. The tumor-derived molecular variant is the total number of driver genes or other class I/II gene variants (i.e., clinically significant or potentially clinically significant gene variants) detected in blood samples. The classification of gene variation is referred to OncoKB, ClinVar, COSMIC and other databases. The MRD assay was performed on blood samples using a next-generation sequencing (NGS) panel on Illumina’s high-throughput sequencing platform, with reference to the Human Genome UCSC hg19 Feb. 2009. Covered genes include ALK, CD274, EGFR, ERBB2(HER2), FGFR1, FGFR2, FGFR3, MET, NTRK1, NTRK2, NTRK3, PDCD1LG2, PDGFRA, RET, ROS1, DDR2, ERBB4, MAP2K1, NOTCH1, PTEN, STK11, SMAD4, TP53, AKT1, APC, BRAF, CTNNB1, ERBB3, FBXW7, HRAS, KRAS, NRAS, PIK3CA, PPP2R1A, RNF43, POLE.

Treatment regimens

Patients were divided into two groups based on postoperative chemotherapy: (I) capecitabine monotherapy group: patients received capecitabine 1,250 mg/m2 orally twice daily on days 1–14, repeated every 3 weeks for 8 cycles. (II) CapeOx Group: patients received oxaliplatin 130 mg/m2 intravenously on day 1 and capecitabine 1,000 mg/m2 orally twice daily on days 1–14, repeated every 3 weeks for 4–8 cycles.

Data collection and outcomes

Baseline characteristics, including age, sex, body mass index (BMI), tumor stage, and pathological features, were collected from the hospital database. Hemoglobin, preoperative serum biochemical indicators (albumin, anemia status), and tumor markers (CEA, CA19-9, CA125) were categorized based on hospital laboratory standards. All the blood test results were derived from one week before the operation. Pathological features assessed included histological type, tumor differentiation grade, tumor stage, high-risk factors (T4, vascular invasion, neural invasion, number of lymph nodes examined, preoperative intestinal obstruction), P53 expression, and BRAF-V600E expression. Tumor differentiation was classified as well-differentiated, moderately differentiated, or poorly differentiated. Histological types included classic adenocarcinoma and mucinous adenocarcinoma. Mismatch repair (MMR) protein expression (MLH1, PMS2, MSH2, and MSH6) was detected using immunohistochemistry. Negative expression of one or more MMR proteins was considered mismatch repair deficiency (dMMR), while positive expression of all four proteins was considered mismatch repair proficiency (pMMR). Adverse reactions after chemotherapy, including gastrointestinal toxicity, neurotoxicity, hematological toxicity, and overall systemic conditions, were observed during follow-up. Follow-up data were obtained from the institutional follow-up center. The follow-up protocol primarily comprised tumor marker assays and contrast-enhanced computed tomography (CT) imaging. The primary outcome was DFS, defined as the time from surgery to recurrence or death. Secondary outcomes included 3-year DFS rates and chemotherapy-related adverse events, which were graded according to the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0.

Statistical analysis

Categorical variables were expressed as frequencies and percentages and compared using the chi-square test or Fisher’s exact test. Continuous variables were expressed as mean ± standard deviation (SD) or median (interquartile range, IQR) and compared using the t-test or Mann-Whitney U test, as appropriate. Univariate and multivariate Cox regression analyses were used to assess independent risk factors. No attrition adjustment was performed because no patients were lost to long-term follow-up. Survival outcomes were analyzed using the Kaplan-Meier method and compared using the log-rank test. Subgroup analysis was used to evaluate the heterogeneity of the results. Propensity score matching (PSM) was performed to balance baseline differences between the two groups, with a caliper value of 0.1. Standardized mean differences (SMD) were used to evaluate the effect of balance. All statistical analyses were conducted using IBM SPSS Statistics 27.0 (IBM Corp., Armonk, NY, USA) and R 4.4.1 (R Foundation for Statistical Computing, Vienna, Austria), with a two-sided P value <0.05 considered statistically significant.


Results

Baseline characteristics

All datasets were fully documented with no missing records. The study included 117 patients, with 58 in the capecitabine monotherapy group and 59 in the CapeOx group (Figure 1, Table 1). All patients underwent laparoscopic radical resection for CRC and received at least one cycle of chemotherapy. The median age was 69 years (range, 60–85), and the majority of patients were male (58%). The ratio of stage II to III patients was approximately 1:1. Most tumors were moderately differentiated (92%) and pMMR (92%). All patients exhibited postoperative pathological high-risk features, including T4 stage, poor differentiation, vascular invasion, neural invasion, preoperative intestinal obstruction, or fewer than 12 lymph nodes examined. As shown in Table 1, significant differences (all P<0.05) were observed in age, tumor stage, N stage, CA19-9 levels, and hemoglobin (Hb) between the groups. No significant differences (all P>0.05) were found in BMI, sex, T stage, differentiation grade, MMR status, albumin (ALB), or CEA levels. PSM (1:1 nearest-neighbor matching with a caliper of 0.1) was performed using covariates with significant baseline differences (age, tumor stage, N stage, CA19-9, and Hb). After PSM, 29 matched pairs were included in the analysis, with no significant differences in baseline characteristics between the two groups (all P>0.05) (Table 1). Timing of treatment, DFS and overall survival (OS) were illustrated in Figure 2.

Table 1

Clinicopathological characteristics of elderly patients with high-risk stage II and stage III CRC and negative MRD

Characteristics Crude data set Matched data set
All (n=117) CapeOx (n=59) Capecitabine (n=58) P CapeOx (n=29) Capecitabine (n=29) P
Age (years) 69 (63, 76) 67 (62, 74) 73 (65, 78) 0.03* 73 (65.5, 75) 70 (62, 75) 0.60
BMI 0.10 0.09
   Underweight 20 (17) 7 (12) 13 (22) 2 (7) 6 (21)
   Normal 80 (68) 40 (68) 40 (69) 20 (69) 21 (72)
   Overweight 17 (15) 12 (20) 5 (9) 7 (24) 2 (7)
Sex 0.08 0.18
   Male 68 (58) 39 (66) 29 (50) 21 (72) 15 (52)
   Female 49 (42) 20 (34) 29 (50) 8 (28) 14 (48)
Pathology
   Location 0.55 0.85
    Right 20 (17) 8 (14) 12 (21) 6 (21) 8 (28)
    Transverse 4 (3) 2 (3) 2 (3) 1 (3) 2 (7)
    Left 5 (4) 4 (7) 1 (2) 2 (7) 1 (3)
    Sigmoid 30 (26) 17 (29) 13 (22) 6 (21) 7 (24)
    Rectum 58 (50) 29 (47) 30 (52) 14 (48) 11 (38)
   Stage 0.001* >0.99
    II 61 (52) 22 (37) 39 (67) 15 (52) 16 (55)
    III 56 (48) 37 (63) 19 (33) 14 (48) 13 (45)
   T stage 0.24 0.13
    2 13 (11) 7 (12) 6 (10) 4 (14) 4 (14)
    3 72 (62) 32 (54) 40 (69) 12 (41) 19 (66)
    4 32 (27) 20 (34) 12 (20) 13 (45) 6 (20)
   N stage 0.004* 0.75
    0 61 (52) 22 (37) 39 (67) 15 (52) 16 (55)
    1 41 (35) 26 (44) 15 (26) 9 (31) 10 (35)
    2 15 (13) 11 (19) 4 (7) 5 (17) 3 (10)
   Differentiation 0.93 0.80
    Well 2 (2) 1 (2) 1 (2) 1 (3) 1 (3)
    Moderate 107 (92) 53 (90) 54 (93) 25 (87) 26 (90)
    Poor 4 (3) 2 (3) 2 (3) 2 (7) 2 (7)
    Mucinous 4 (3) 3 (5) 1 (2) 1 (3) 0
Immunohistochemistry
   MMR 0.53 0.12
    pMMR 107 (92) 55 (93) 52 (90) 29 (100) 25 (86)
    dMMR 10 (8) 4 (7) 6 (10) 0 4 (14)
   P53 0.60 0.07
    Negative 47 (40) 27 (46) 20 (35) 16 (55) 8 (28)
    Weakly positive 44 (38) 21 (36) 23 (40) 10 (35) 13 (44)
    Positive 3 (3) 1 (2) 2 (3) 3 (10) 0
    Strongly positive 23 (19) 10 (16) 13 (22) 0 8 (28)
   BRAF-V600E 0.12 >0.99
    Negative 114 (97) 59 (100) 55 (95) 29 (100) 28 (97)
    Positive 3 (3) 0 3 (5) 0 1 (3)
High-risk features
   T4 0.11 0.09
    No 85 (73) 39 (66) 46 (79) 16 (55) 23 (79)
    Yes 32 (27) 20 (34) 12 (21) 13 (45) 6 (21)
   Vessel invasion 0.07 0.11
    Negative 93 (80) 43 (73) 50 (86) 20 (69) 26 (90)
    Positive 24 (20) 16 (27) 8 (14) 9 (31) 3 (10)
   Neural invasion 0.07 0.17
    Negative 43 (37) 25 (42) 18 (31) 13 (45) 7 (24)
    Positive 74 (63) 34 (58) 40 (69) 16 (55) 22 (76)
   Number of LN 0.11 >0.99
    <12 6 (5) 1 (2) 5 (9) 0 0
    ≥12 111 (95) 58 (98) 53 (91) 29 (100) 29 (100)
   Preoperative intestinal obstruction >0.99
    No 108 (92) 54 (92) 54 (93) 26 (90) 26 (90)
    Yes 9 (8) 5 (8) 4 (7) 3 (10) 3 (10)
   T4 of stage III 0.33 0.74
    No 43 (23) 30 (81) 13 (68) 10 (71) 10 (77)
    Yes 13 (77) 7 (19) 6 (32) 4 (29) 3 (23)
   N2 of stage III 0.54 0.47
    No 42 (75) 26 (70) 15 (79) 9 (64) 10 (77)
    Yes 14 (25) 11 (30) 4 (21) 5 (36) 3 (23)
Laboratory technology
   CEA 0.23 0.79
    Normal 69 (59) 38 (64) 31 (53) 18 (62) 16 (55)
    Abnormal 48 (41) 21 (36) 27 (47) 11 (38) 13 (45)
   CA19-9 0.03* 0.42
    Normal 101 (86) 47 (80) 54 (93) 24 (83) 27 (93)
    Abnormal 16 (14) 12 (20) 4 (7) 5 (17) 2 (7)
   CA125 0.44 >0.99
    Normal 110 (94) 54 (92) 56 (97) 29 (100) 29 (100)
    Abnormal 7 (6) 5 (8) 2 (3) 0 0
   ALB 0.83 0.42
    Normal 98 (84) 49 (83) 49 (85) 24 (83) 27 (93)
    Abnormal 19 (16) 10 (17) 9 (15) 5 (17) 2 (7)
   Anaemia 0.009* 0.24
    No 67 (57) 40 (68) 27 (47) 19 (66) 17 (59)
    Mild 38 (33) 12 (20) 26 (45) 4 (14) 9 (31)
    Moderate 9 (8) 4 (7) 5 (8) 4 (14) 3 (10)
    Severe 3 (2) 3 (5) 0 2 (6) 0

Data are presented as median (interquartile range) or n (%). *, statistical significance. ALB, albumin; BMI, body mass index; CapeOx, capecitabine plus oxaliplatin; CEA, carcinoembryonic antigen; CRC, colorectal cancer; dMMR, deficient mismatch repair; LN, lymph node; MMR, mismatch repair; MRD, minimal residual disease; pMMR, proficient mismatch repair.

Figure 2 Timing of treatment for each patient. Patients are grouped by the pathological stage and ordered by total months of clinical follow up from surgery. CapeOx, capecitabine plus oxaliplatin.

Independent risk factors

The univariate Cox analysis and multivariate Cox analysis were used. As shown in Table 2, stage III [hazard ratio (HR) =3.176, 95% CI: 1.241–8.126, P=0.016] and vascular invasion (HR =3.167, 95% CI: 1.348–7.442, P=0.008) were the independent risk factors among the elderly patients with high-risk stage II and stage III CRC and negative MRD.

Table 2

Univariate Cox and multivariate Cox analysis of elderly patients with high-risk stage II and stage III CRC and negative MRD

Characteristics Univariate Cox Multivariate Cox
Hazard ratio (95% CI) P Hazard ratio (95% CI) P
Age 0.978 (0.919–1.040) 0.47 0.940 (0.861–1.028) 0.18
BMI
   Normal Ref. Ref.
   Underweight 0.974 (0.280–3.392) 0.97 1.489 (0.330–6.710) 0.60
   Overweight 1.163 (0.385–3.511) 0.79 1.368 (0.254–7.358) 0.72
Sex
   Female Ref. Ref.
   Male 1.345 (0.547–3.310) 0.52 1.227 (0.392–3.842) 0.73
Location
   Right Ref. Ref.
   Transverse 1.535 (0.171–13.804) 0.70 3.989 (0.198–80.557) 0.37
   Left 2.058 (0.375–11.300) 0.40 25.761 (1.661–399.452) 0.02*
   Sigmoid 0.518 (0.116–2.319) 0.39 0.948 (0.114–7.922) 0.96
   Rectum 1.101 (0.354–3.420) 0.87 4.908 (0.706–34.142) 0.11
Stage
   II Ref. Ref.
   III 3.176 (1.241–8.126) 0.01* 65.044 (2.841–1490.389) 0.009*
T stage
   T2 Ref. Ref.
   T3 1.427 (0.323–6.318) 0.64 3.001 (0.459–19.601) 0.25
   T4 1.098 (0.221–5.465) 0.91 1.789 (0.210–15.240) 0.60
N stage
   N0 Ref. Ref.
   N1 2.370 (0.917–6.125) 0.08 0.060 (0.003–1.301) 0.07
   N2 2.687 (0.785–9.194) 0.12 0.051 (0.002–1.584) 0.09
MMR
   pMMR Ref. Ref.
   dMMR 2.216 (0.746–6.589) 0.15 4.639 (0.922–23.341) 0.06
BRAF-V600E
   Negative Ref. Ref.
   Positive 2.952 (0.390–22.330) 0.29 32.516 (1.426–741.423) 0.02*
Vascular invasion
   Negative Ref. Ref.
   Positive 3.167 (1.348–7.442) 0.008* 4.504 (1.194–16.983) 0.02*
Neural invasion
   Negative Ref. Ref.
   Positive 1.158 (0.484–2.769) 0.74 0.791 (0.242–2.592) 0.70
Number of LN
   <12 Ref. Ref.
   ≥12 0.453 (0.104–1.970) 0.29 0.317 (0.053–1.897) 0.21
CEA
   Normal Ref. Ref.
   Abnormal 1.046 (0.409–2.239) 0.92 2.018 (0.619–6.582) 0.24
CA19-9
   Normal Ref. Ref.
   Abnormal 2.259 (0.830–6.146) 0.11 3.699 (0.822–16.637) 0.09
Anaemia
   No Ref. Ref.
   Mild 0.443 (0.148–1.329) 0.15 0.448 (0.111–1.808) 0.26
   Moderate 0.425 (0.056–3.210) 0.41 0.292 (0.019–4.396) 0.37
   Severe 1.331 (0.175–10.109) 0.78 2.064 (0.139–30.637) 0.60

*, statistical significance. BMI, body mass index; CEA, carcinoembryonic antigen; CI, confidence interval; CRC, colorectal cancer; dMMR, deficient mismatch repair; LN, lymph node; MMR, mismatch repair; MRD, minimal residual disease; pMMR, proficient mismatch repair.

Survival outcomes

Patients were followed postoperatively for a median of 35 months (range, 14–81) (Figure 2). The median DFS was 31 months in the capecitabine group and 40 months in the CapeOx group, with no significant difference between the two regimens (P=0.26) (Figure 3). The 3-year DFS rates were comparable (90% vs. 81%, HR 0.565, 95% CI: 0.209–1.528, P=0.26). Subgroup analysis showed no significant heterogeneity in DFS across sex, BMI, or tumor location (all P>0.05) (Table 3). After conducting separate analyses of patients in high-risk stage II and stage III, we found that there was still no significant difference in the 3-year DFS rate between the capecitabine group and the CapeOx group (all P>0.05) (Figure 4). PSM was used to control for confounding factors. As shown in Figure 5, confounding factors such as age, stage III, N stage, CA19-9, and Hb were well balanced (all SMD <0.2). After PSM, the survival outcomes remained similar between the two groups (P=0.73) (Figure 6), with a 3-year DFS rate of 90% in the capecitabine group and 86% in the CapeOx group (HR =0.718, 95% CI: 0.161–3.213, P=0.67).

Figure 3 Prognostic difference in the elderly patients with high-risk stage II and stage III CRC with negative MRD. CapeOx, capecitabine plus oxaliplatin; CRC, colorectal cancer; MRD, minimal residual disease.

Table 3

Subgroup analysis of elderly patients with high-risk stage II and stage III CRC and negative MRD

Subgroup Capecitabine, n CapeOx, n HR (95% CI) P value
Total 58 59 1.68 (0.67–4.18) 0.27
Sex
   Female 29 20 3.71 (0.72–19.13) 0.12
   Male 29 39 0.96 (0.31–2.94) 0.94
BMI
   Underweight 13 7 0.73 (0.06–8.19) 0.80
   Normal 40 40 2.53 (0.80–8.05) 0.12
   Overweight 5 12
Location
   Left 12 8 0.28 (0.03–2.95) 0.29
   Transverse 2 2
   Right 1 4
   Sigmoid 13 17
   Rectum 30 28 1.77 (0.52–6.02) 0.36
Stage
   II 39 22 1.15 (0.22–6.02) 0.87
   III 19 37 1.23 (0.39–3.89) 0.72
T stage
   T2 6 7
   T3 40 32 1.68 (0.54–5.22) 0.37
   T4 12 20 1.04 (0.19–5.71) 0.96
N stage
   N0 39 22 1.71 (0.37–7.94) 0.49
   N1 15 26 1.07 (0.28–4.14) 0.92
   N2 4 11 1.04 (0.11–10.07) 0.97
Differentiation
   Well 1 1
   Moderate 54 53 1.85 (0.70–4.91) 0.22
   Poor 2 2
   Mucinous 1 3
Vascular invasion
   No 50 43 1.53 (0.49–4.78) 0.46
   Yes 8 16 1.23 (0.24–6.24) 0.80
Neural invasion
   No 18 25 1.05 (0.25–4.43) 0.95
   Yes 40 34 2.32 (0.70–7.64) 0.17
MMR
   pMMR 52 55 1.96 (0.68–5.61) 0.21
   dMMR 6 4 1.47 (0.21–10.47) 0.70
P53
   Negative 20 27 1.28 (0.32–5.18) 0.73
   Weakly 23 21 3.83 (0.8–18.34) 0.09
   Positive 2 1
   Strongly 13 10 0.28 (0.02–4.14) 0.36

BMI, body mass index; CapeOx, capecitabine plus oxaliplatin; CI, confidence interval; CRC, colorectal cancer; dMMR, deficient mismatch repair; HR, hazard ratio; MMR, mismatch repair; MRD, minimal residual disease; pMMR, proficient mismatch repair.

Figure 4 Prognostic difference in the elderly patients with negative MRD. (A) Prognostic difference in the elderly patients with high-risk stage II. (B) Prognostic difference in the elderly patients with stage III. CapeOx, capecitabine plus oxaliplatin; MRD, minimal residual disease.
Figure 5 Distribution of concomitant variables. (A) Distributional balance for age. (B) Distributional balance for stage. (C) Distributional balance for N stage. (D) Distributional balance for CA19-9. (E) Distributional balance for Hb. (F) Standardized mean differences of covariate. Hb, hemoglobin.
Figure 6 Prognostic difference in the elderly patients with high-risk stage II and stage III CRC with negative MRD after the propensity match. CapeOx, capecitabine plus oxaliplatin; CRC, colorectal cancer; MRD, minimal residual disease.

Adverse events

As summarized in Table 4, both groups experienced common chemotherapy-related adverse reactions, including appetite loss, nausea/vomiting (54%), leukopenia (36%), thrombocytopenia (28%), and fatigue. However, the CapeOx group exhibited significantly higher rates of neurotoxicity (61% vs. 7%, P<0.001), leukopenia (49% vs. 22%, P=0.002), and thrombocytopenia (44% vs. 10%, P<0.001) compared to the capecitabine group. Moderate-to-severe hematological adverse events were observed exclusively in the CapeOx group, including 6 cases (5%) of moderate leukopenia and 4 cases (6%) of moderate-to-severe thrombocytopenia. In contrast, no moderate or severe hematological adverse events were reported in the capecitabine group.

Table 4

Chemotherapy side effect of elderly patients with high-risk stage II and stage III CRC and negative MRD

Adverse reaction All (n=117) CapeOx (n=59) Capecitabine (n=58) P value
Gastrointestinal reaction 0.23
   No 54 (46) 24 (41) 30 (52)
   Yes 63 (54) 35 (59) 28 (48)
Neurotoxicity <0.001*
   No 77 (66) 23 (39) 54 (93)
   Yes 40 (34) 36 (61) 4 (7)
Leukocytopenia 0.002*
   No 75 (64) 30 (51) 45 (78)
   Mild 36 (31) 23 (39) 13 (22)
   Moderate 6 (5) 6 (10) 0
   Severe 0 0 0
Thrombocytopenia <0.001*
   No 85 (72) 33 (56) 52 (90)
   Mild 28 (24) 22 (38) 6 (10)
   Moderate 2 (2) 2 (3) 0
   Severe 2 (2) 2 (3) 0
Fatigue 0.65
   No 44 (38) 21 (36) 23 (40)
   Yes 73 (62) 38 (64) 35 (60)

*, statistical significance. CapeOx, capecitabine plus oxaliplatin; CRC, colorectal cancer; MRD, minimal residual disease.


Discussion

CRC remains a leading cause of cancer-related mortality worldwide (1). Postoperative adjuvant chemotherapy plays a pivotal role in reducing recurrence and improves survival outcomes, particularly in high-risk stage II and stage III patients. However, the optimal chemotherapy regimen for elderly patients, who often present with comorbidities and reduced physiological reserve, remains a subject of debate (16-18). While the MOSAIC trial raised doubts regarding the OS benefit of oxaliplatin in elderly patients (3), the IDEA study proposed a strategy to optimize its use in this population by shortening the treatment duration (19). However, for the elderly patients with MRD-negative status, our findings suggest that completely omitting oxaliplatin may represent a rational and more profound de-escalation strategy compared to merely shortening the course of therapy. Our study provided compelling evidence that capecitabine monotherapy offers comparable DFS to the CapeOx regimen in elderly patients with high-risk stage II and stage III CRC who are MRD-negative, while significantly reducing treatment-related toxicities.

The comparable DFS between capecitabine monotherapy and CapeOx in our study (median DFS: 31 vs. 40 months, P=0.26) aligns with findings from previous studies. For instance, van Erning et al. (20) reported similar survival outcomes between monotherapy and combination chemotherapy in elderly CRC patients, particularly in those with low recurrence risk. Similarly, Okamoto et al. (21) demonstrated that elderly patients with stage III CRC could achieve favorable outcomes with capecitabine monotherapy, especially when stratified based on molecular markers. Our study extends these findings by incorporating MRD status as a stratification factor, suggesting that MRD negativity may identify a subgroup of patients who can safely forego more intensive combination chemotherapy without compromising survival outcomes. This reflects the growing emphasis on precision oncology and the need to balance efficacy with quality of life in the elderly population (22). On the other hand, stage III and vascular invasion were independent risk factors for high-risk stage II and stage III elderly CRC patients with MRD-negative status in our study, suggesting that postoperative treatment plans may still need to be closely combined with pathological features. For patients with stage III CRC, who generally demonstrate a less favorable prognosis, the CSCO guidelines (2) recommend considering combination therapies, including immunotherapy or targeted agents, in addition to adjuvant chemotherapy to further improve clinical outcomes, particularly for those with dMMR/MSI-H or high-risk pathological characteristics.

The superior safety profile of capecitabine monotherapy observed in our study further supports its use in elderly patients. The CapeOx group exhibited significantly higher rates of neurotoxicity (61% vs. 7%, P<0.001), leukopenia (49% vs. 22%, P=0.002), and thrombocytopenia (44% vs. 10%, P<0.001), consistent with prior studies (3,5,23,24) highlighting the toxicity burden of oxaliplatin-based regimens. Notably, moderate-to-severe hematological adverse events were observed in the CapeOx group, underscoring the potential risks of combination chemotherapy in elderly patients. These findings are significantly relevant given the importance of maintaining quality of life in cancer treatment, especially for elderly patients who may prioritize minimizing treatment-related toxicity over marginal survival benefits.

The integration of MRD status into treatment decision-making represents a significant advancement in personalized oncology. MRD, as assessed by ctDNA analysis, provides a dynamic and sensitive measure of residual disease burden, enabling more precise risk stratification than traditional clinicopathological features alone (25-29). Our study adds to the growing body of evidence that MRD negativity may serve as a biomarker for identifying patients who can benefit from de-escalated therapy. This approach not only reduces unnecessary treatment-related toxicity but also optimizes resource utilization in healthcare systems. However, the mechanisms associated with MRD negativity and its relationship with tumor biology remain incompletely understood. Future studies should explore the molecular characteristics of MRD-negative tumors, such as specific gene mutations or immune microenvironment features, to better understand their low recurrence risk and identify potential therapeutic targets.

Despite these promising findings, our study has several limitations that warrant consideration. First, the definition of ‘high-risk’ for stage III patients was based on pathological features typically applied to stage II disease, rather than the conventional anatomical risk stratification (e.g., T4 and/or N2 disease as per the IDEA criteria). This was done to create a homogeneous cohort with both stage II and III patients sharing aggressive tumor biological features. However, we acknowledge that this may not fully capture the distinct risk profile defined by nodal tumor burden in stage III disease. Second, the retrospective design introduces potential biases, particularly in patient selection and treatment allocation. Although PSM was employed to balance baseline characteristics, residual confounding factors may persist. Third, the sample size, which was sufficient for the initial analysis, may limit the generalizability of our findings. Larger, multicenter studies are needed to validate these results. Fourth, the follow-up duration, with a median of 38 months, may be insufficient to capture long-term survival outcomes and late recurrences. Extended follow-up will be necessary to ascertain the durability of the observed survival benefits. Finally, the study population was limited to elderly patients, and the applicability of these findings to younger populations remains uncertain.

Future research is needed to focus on prospective validation of our findings, particularly through randomized controlled trials (RCTs) comparing capecitabine monotherapy with CapeOx in MRD-negative patients. Additionally, the study of Tie et al. (29), exploring the role of dynamic MRD monitoring, such as serial ctDNA testing during and after chemotherapy, is likely to provide further insights into the optimal timing and duration of adjuvant therapy. The integration of molecular profiling, including genomic and immune markers, is expected to enhance our ability to predict which patients are most likely to benefit from de-escalated therapy.


Conclusions

For elderly patients with high-risk stage II and stage III CRC who are MRD-negative, capecitabine monotherapy might provide comparable survival outcomes with CapeOx while offering a superior safety profile. These findings suggest that MRD status might guide the implementation of personalized dose-reduction strategies in this population.


Acknowledgments

None.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-504/rc

Data Sharing Statement: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-504/dss

Peer Review File: Available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-504/prf

Funding: This work was supported by Clinical Research Project of the Affiliated Hospital of Guangdong Medical University (No. LCYJ2023B005) and Guangdong Provincial Medical Science and Technology Research Fund (No. A2024383).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://jgo.amegroups.com/article/view/10.21037/jgo-2025-504/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. This study was conducted in compliance with the postulates of Declaration of Helsinki and its subsequent amendments and was approved by the Human Ethics Committee of the Affiliated Hospital of Guangdong Medical University (No. PJKT2024-063). The requirement for patient approval or informed consent was waived by the Affiliated Hospital of Guangdong Medical University, owing to the retrospective nature of the study and the athe use of de-identified data.

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. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12-49. [Crossref] [PubMed]
  2. Chinese Society of Clinical Oncology (CSCO). CSCO Colorectal Cancer Diagnosis and Treatment Guidelines 2024. Beijing: People’s Medical Publishing House; 2024.
  3. André T, de Gramont A, Vernerey D, et al. Adjuvant Fluorouracil, Leucovorin, and Oxaliplatin in Stage II to III Colon Cancer: Updated 10-Year Survival and Outcomes According to BRAF Mutation and Mismatch Repair Status of the MOSAIC Study. J Clin Oncol 2015;33:4176-87. [Crossref] [PubMed]
  4. Quaglia A, Tavilla A, Shack L, et al. The cancer survival gap between elderly and middle-aged patients in Europe is widening. Eur J Cancer 2009;45:1006-16. [Crossref] [PubMed]
  5. Kim JH. Chemotherapy for colorectal cancer in the elderly. World J Gastroenterol 2015;21:5158-66. [Crossref] [PubMed]
  6. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med 2016;8:346ra92. [Crossref] [PubMed]
  7. Xu M, Shi T, Xu R, et al. The potential role of minimal/molecular residual disease in colorectal cancer: curative surgery, radiotherapy and beyond. J Natl Cancer Cent 2023;3:203-10. [Crossref] [PubMed]
  8. Henriksen TV, Demuth C, Frydendahl A, et al. Unraveling the potential clinical utility of circulating tumor DNA detection in colorectal cancer-evaluation in a nationwide Danish cohort. Ann Oncol 2024;35:229-39. [Crossref] [PubMed]
  9. Malla M, Loree JM, Kasi PM, et al. Using Circulating Tumor DNA in Colorectal Cancer: Current and Evolving Practices. J Clin Oncol 2022;40:2846-57. [Crossref] [PubMed]
  10. Wullaert L, van Rees JM, Martens JWM, et al. Circulating Tumour DNA as Biomarker for Colorectal Liver Metastases: A Systematic Review and Meta-Analysis. Cells 2023;12:2520. [Crossref] [PubMed]
  11. Chinese Society of Clinical Oncology (CSCO). Colorectal Cancer Diagnosis and Treatment guidelines Beijing: People’s Medical Publishing House; 2023.
  12. Arisi MF, Dotan E, Fernandez SV. Circulating Tumor DNA in Precision Oncology and Its Applications in Colorectal Cancer. Int J Mol Sci 2022;23:4441. [Crossref] [PubMed]
  13. Alzahrani SM, Al Doghaither HA, Al-Ghafari AB, et al. 5 Fluorouracil and capecitabine therapies for the treatment of colorectal cancer Oncol Rep 2023;50:175. (Review). [Crossref] [PubMed]
  14. Baxter NN, Kennedy EB, Bergsland E, et al. Adjuvant Therapy for Stage II Colon Cancer: ASCO Guideline Update. J Clin Oncol 2022;40:892-910. [Crossref] [PubMed]
  15. Iveson TJ, Sobrero AF, Yoshino T, et al. Duration of Adjuvant Doublet Chemotherapy (3 or 6 months) in Patients With High-Risk Stage II Colorectal Cancer. J Clin Oncol 2021;39:631-41. [Crossref] [PubMed]
  16. de Gramont A, Figer A, Seymour M, et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J Clin Oncol 2000;18:2938-47. [Crossref] [PubMed]
  17. Haller DG, Tabernero J, Maroun J, et al. Capecitabine plus oxaliplatin compared with fluorouracil and folinic acid as adjuvant therapy for stage III colon cancer. J Clin Oncol 2011;29:1465-71. [Crossref] [PubMed]
  18. Schmoll HJ, Tabernero J, Maroun J, et al. Capecitabine Plus Oxaliplatin Compared With Fluorouracil/Folinic Acid As Adjuvant Therapy for Stage III Colon Cancer: Final Results of the NO16968 Randomized Controlled Phase III Trial. J Clin Oncol 2015;33:3733-40. [Crossref] [PubMed]
  19. Gallois C, Shi Q, Pederson LD, et al. Oxaliplatin-Based Adjuvant Chemotherapy in Older Patients With Stage III Colon Cancer: An ACCENT/IDEA Pooled Analysis of 12 Trials. J Clin Oncol 2024;42:2295-305. [Crossref] [PubMed]
  20. van Erning FN, Janssen-Heijnen ML, Creemers GJ, et al. Recurrence-free and overall survival among elderly stage III colon cancer patients treated with CAPOX or capecitabine monotherapy. Int J Cancer 2017;140:224-33. [Crossref] [PubMed]
  21. Okamoto K, Nozawa H, Emoto S, et al. Adjuvant Capecitabine and Oxaliplatin for Elderly Patients with Colorectal Cancer. Oncology 2022;100:576-82. [Crossref] [PubMed]
  22. Yang L, Yang J, Kleppe A, et al. Personalizing adjuvant therapy for patients with colorectal cancer. Nat Rev Clin Oncol 2024;21:67-79. [Crossref] [PubMed]
  23. Petrioli R, Pascucci A, Francini E, et al. Neurotoxicity of FOLFOX-4 as adjuvant treatment for patients with colon and gastric cancer: a randomized study of two different schedules of oxaliplatin. Cancer Chemother Pharmacol 2008;61:105-11. [Crossref] [PubMed]
  24. Schrag D, Shi Q, Weiser MR, et al. Preoperative Treatment of Locally Advanced Rectal Cancer. N Engl J Med 2023;389:322-34. [Crossref] [PubMed]
  25. Chidharla A, Rapoport E, Agarwal K, et al. Circulating Tumor DNA as a Minimal Residual Disease Assessment and Recurrence Risk in Patients Undergoing Curative-Intent Resection with or without Adjuvant Chemotherapy in Colorectal Cancer: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023;24:10230. [Crossref] [PubMed]
  26. Pascual J, Attard G, Bidard FC, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022;33:750-68. [Crossref] [PubMed]
  27. Tarazona N, Gimeno-Valiente F, Gambardella V, et al. Targeted next-generation sequencing of circulating-tumor DNA for tracking minimal residual disease in localized colon cancer. Ann Oncol 2019;30:1804-12. [Crossref] [PubMed]
  28. Tie J, Cohen JD, Lahouel K, et al. Circulating Tumor DNA Analysis Guiding Adjuvant Therapy in Stage II Colon Cancer. N Engl J Med 2022;386:2261-72. [Crossref] [PubMed]
  29. Tie J, Wang Y, Cohen J, et al. Circulating tumor DNA dynamics and recurrence risk in patients undergoing curative intent resection of colorectal cancer liver metastases: A prospective cohort study. PLoS Med 2021;18:e1003620. [Crossref] [PubMed]
Cite this article as: Zhai J, Tan R, Ou W, Quan Z, Sun L, Huang X, Wu J, Zhou C. Prognostic and safety analysis of capecitabine monotherapy versus CapeOx (capecitabine plus oxaliplatin) in elderly patients with high-risk stage II and stage III MRD-negative colorectal cancer. J Gastrointest Oncol 2025;16(5):1985-2000. doi: 10.21037/jgo-2025-504

Download Citation