Skip to main content

Safety and efficacy of minimally invasive gastrectomy for older patients with gastric cancer after neoadjuvant chemotherapy and immunotherapy: a propensity score-matched analysis

Abstract

Background

The effect of neoadjuvant immunotherapy on minimally invasive gastrectomy (MIG) in older patients with gastric cancer remains controversial. This study aimed to evaluate the safety, and efficacy of MIG for older patients who underwent neoadjuvant chemotherapy and immunotherapy (NICT).

Methods

The clinical data of 726 older patients aged over 65 years who underwent upfront MIG or MIG after NICT in the Department of General Surgery, Chinese PLA General Hospital First Medical Center between Jan 2020 and Nov 2023 were retrospectively analyzed. Propensity score-matched (PSM) analysis at a ratio of 1:2 was performed to reduce bias from confounding patient-related variables, short- and long-term outcomes were compared between the two groups.

Results

The baseline characteristics were comparable between 61 patients in the NICT-MIG group and 114 patients in the MIG group after PSM (P > 0.05). The major pathological response (MPR) rate and pathological complete response (pCR) rate were 44.2% and 21.3%, respectively, in the NICT-MIG group. Patients in the NICT-MIG group had longer operation times (P = 0.005) and postoperative days (P = 0.030) than those in the MIG group. No significant differences were found in intraoperative bleeding, number of retrieved lymph nodes, first flatus day, R0 resection rate, overall postoperative complication (POC) morbidity, severe POC morbidity, 2-year overall, and recurrence-free survival between the MIG and NICT-MIG groups (P > 0.05). Multivariate logistic analysis revealed that an estimated blood loss > 200 mL (P = 0.010) and a lymphocyte-to-monocyte ratio (LMR) ≤ 3.25 (P = 0.006) were independent risk factors for POCs after MIG in older patients.

Conclusion

The safety, and efficacy of NICT-MIG were comparable to those of upfront MIG in older patients with GC. Patients with an estimated blood loss > 200 mL or an LMR ≤ 3.25 should be carefully evaluated for an increased risk of POCs in older patients who undergo MIG.

Trial registration

Chinese Clinical Trial Registry (Registration Number: ChiCTR2400086827).

Peer Review reports

Background

Gastric cancer (GC) is one of the most common malignant tumors and is the second most common cause of both cancer incidence and cancer-related death in China [1]. As the phenomenon of aging intensifies, the proportion of older patients with GC is gradually increasing [2]. Due to poor physical status and comorbidities, it is essential to conduct comprehensive geriatric assessments and select optimal perioperative treatments for older patients [3].

In recent years, minimally invasive gastrectomy (MIG), represented by laparoscopic and robotic gastrectomy, has been widely applied to cure GC due to its several advantages, including less surgical incision, less blood loss and comparable survival benefits compared with open gastrectomy (OG) [4, 5]. The advantages of MIG have attracted surgeons to focus on its safety and efficacy in older patients. Hikage M et al. conducted a propensity score-matched analysis and demonstrated that patients who underwent MIG achieved comparable short-term outcomes and better overall and relapse-free survival than patients who underwent OG [6]. Li’s study also showed that MIG was a feasible and safe procedure for older patients, in contrast to MIG for nonolder patients and OG for older patients [7]. Moreover, our previous study revealed that MIG with intracorporeal anastomosis for older patients was safe and improved postoperative quality of life [8].

Neoadjuvant therapy before surgery is emerging as a highly regarded treatment for patients with advanced GC. The combination of neoadjuvant chemotherapy and immunotherapy (NICT) is one of the most mainstream therapies, resulting in better tumor regression, increasing the proportion of patients who underwent R0 resection, and providing potential survival benefits [9, 10]. However, NICT might result in treatment-related adverse events (TRAEs), decreasing immune and nutritional status, especially in older patients. Moreover, the edema and fibrillation of peri-gastric tissues caused by NICT also increase surgical difficulty [11]. Although the current study retrospectively demonstrated the safety and feasibility of MIG after NICT [12], it is still uncertain whether MIG can be safely conducted after NICT for older patients. This study aimed to evaluate the therapeutic effects of NICT, safety and efficacy of MIG after NICT compared with those of upfront MIG and to determine the risk factors that affect postoperative complications in older patients who undergo MIG.

Methods

Patients

The inclusion criteria were as follows: (1) aged over 65 years, (2) histologically demonstrated gastric adenocarcinoma based on preoperative gastroscopy, (3) accepted laparoscopic or robotic gastrectomy plus D2 lymphadenectomy, (4) pathological tumor stage of p/ypTNM 0-III based on the International Union Against Cancer/American Joint Committee on Cancer guidelines (8th edition), (5) treatment with simultaneous neoadjuvant chemotherapy and immunotherapy in the NICT-MIG group, (6) Eastern Cooperative Oncology Group (ECOG) score 0–1, and (7) available integrated clinical and pathologic data. Patients with an American Society of Anesthesiologists (ASA) grade > III, a history of remnant gastric cancer or gastrointestinal stromal tumor, combined other organ resection, or who received other preoperative regimens, such as radiotherapy and targeted therapy, were excluded from the study.

According to the criteria mentioned above, we retrospectively collected clinical and pathological data from 726 older patients with GC (65 patients in the NICT-MIG group and 661 patients in the MIG group) from Jan 2020 to Nov 2023 at the Chinese PLA General Hospital First Medical Center. The flow chart is shown in Fig. 1.

Fig. 1
figure 1

Flow chart of this study

Propensity score matching (PSM) analysis

In this study, we employed PSM analysis using R statistical software (version 4.2.2; R Foundation, Vienna, Austria) to minimize baseline significant differences. The propensity score for each patient was determined by a logistic regression model based on clinical indices, including body mass index (BMI), nutritional risk screening-2002 (NRS-2002) score, tumor resection, tumor diameter, and p/ypT, p/ypN and p/ypTNM stages. Patients in the NICT-MIG and MIG groups were matched at a 1:2 ratio using the nearest neighbor matching approach with an optimal caliper width of 0.20 without replacement. The covariate balance and distribution of propensity scores before and after PSM are shown in Figs. 2 and 3. After matching, 61 patients in the NICT-MIG group and 114 patients in the MIG group were included in the final analyses.

Fig. 2
figure 2

Covariate balance before and after PSM

Fig. 3
figure 3

Distribution of propensity scores before and after PSM

Treatment with NICT and evaluation of the therapeutic effect

The regimens of neoadjuvant chemotherapy included SOX (S-1 at 60 mg twice daily from Day 1 to 14 combined with oxaliplatin at 130 mg/m2 on Day 1, with 21 days being a cycle), XELOX (capecitabine at 1,000 mg/m2 twice daily from Day 1 to 14 combined with oxaliplatin at 130 mg/m2 on Day 1, with 21 days being a cycle), AS (S-1 at 60 mg twice daily from Day 1 to 14 combined with nab-paclitaxel at 125 mg/m2 on Day 1 and Day 8), capecitabine (1,000 mg/m2 twice daily from Day 1 to 14, with 21 days being a cycle) or S-1 (60 mg twice daily from Day 1 to 14, with 21 days being a cycle). The regimens used for neoadjuvant immunotherapy in this study were all PD-1 inhibitors, including nivolumab (360 mg), tislelizumab (200 mg), camrelizumab (200 mg) and sintilimab (200 mg), which were administered intravenously every 21 days as a cycle.

The therapeutic effect was evaluated every 2 cycles by a multidisciplinary team using enhanced abdominal CT according to the Response Evaluation Criteria in Solid Tumors (version 1.1) [13]. The severity of the treatment-related adverse events (TRAEs) was estimated by the Common Terminology Criteria for Adverse Events (CTCAE - Version 5.0) [14]. We used Becker’s standard to define the tumor regression grade (TRG) from pathological results, which included the following: (1) TRG1a, no residual tumor cells; (2) TRG1b, < 10% residual tumor cells; (3) TRG2, 10–50% residual tumor cells; and (3) TRG3, > 50% residual tumor cells [15]. Pathologic complete response (pCR) and major pathological response (MPR) were defined as TRG1a and TRG1a/1b, respectively.

Surgical approach

MIG was performed within 4–6 weeks after the last cycle in the NICT-MIG group, while patients in the MIG group underwent surgery without any neoadjuvant therapies. Laparoscopic or robotic radical gastrectomy plus D2 lymphadenectomy was conducted based on the surgeons’ individual selection. The standardized surgical procedures were in accordance with the Japanese Gastric Cancer Treatment Guidelines (version 5) [16], the Chinese Guidelines for Laparoscopic Gastrectomy (2023 Edition) [17], and the Chinese Expert Consensus on Robotic Gastrectomy (2015 Edition) [18]. All surgeons had abundant experience with MIGs and overcome the learning curve.

After the intracorporeal procedure, a longitudinal incision less than 10 cm in the upper abdomen was used to remove the specimen, and then intracorporeal or extracorporeal anastomosis was performed according to the surgeons’ discretion. The brief modalities of anastomosis were as follows: (1) Billroth II combined with Braun or Roux-en-Y anastomosis for distal gastrectomy; (2) esophageal–gastric anastomosis, tubular anastomosis, or double-flap anastomosis for proximal gastrectomy; and (3) Roux-en-Y anastomosis for total gastrectomy.

Perioperative indices

We collected several preoperative laboratory indices that might predict postoperative complications, including the following: (1) platelet/lymphocyte ratio (PLR): platelet count/lymphocyte count, (2) lymphocyte/monocyte ratio (LMR): lymphocyte/monocyte ratio, (3) Onodera’s prognostic nutritional index (PNI) score: 10 × albumin (g/dL) + 0.005 × lymphocyte count/mm3, and (4) Albumin (ALB) level.

To evaluate surgical safety and postoperative recovery, estimated blood loss, operation time, R0 resection, first flatus days, postoperative hospitalization days, postoperative overall complications and severe complication rates were recorded. The Clavien–Dindo classification was used to demonstrate the severity of postoperative complications (POCs) (overall POCs: POCs with a Clavien–Dindo classification ≥ Grade II; severe POCs: POCs with a Clavien–Dindo classification ≥ Grade IIIa) [19].

Follow-up

The follow-up data of all patients were regularly recorded by outpatient service or telephone. For the first three years, follow-up was conducted every six months, and then every year thereafter. The missed follow-up rate was 5.1% (9/175) until May 24th, 2024. The median follow-up periods were 25.0 months (95% CI: 20.19–29.81 months) in the MIG group and 24.0 months (95% CI: 21.06–26.94 months) in the NICT-MIG group.

Statistical analysis

Statistical analyses were conducted using R software, version 4.2.2 (R Foundation for Statistical Computing) and SPSS (version 26.0; SPSS, Chicago, IL, USA). For continuous variables following a normal distribution, the mean ± standard deviation.

was used for representation, and Student’s t test was performed to compare the differences. For continuous data with a skewed distribution, the median (interquartile range) [M(IQR)] was utilized, and statistical analysis was carried out using the Mann‒Whitney U test. Categorical data are expressed as frequencies (percentages), and the chi-square test or Fisher’s exact test was used to evaluate differences between two groups. The log-rank test was used to compare long-term survival, and curves were drawn using the Kaplan–Meier method. Binary logistic regression and Cox analyses were conducted, and factors with P < 0.1 in the univariate analysis were included in the multivariate analysis. A significance level of P < 0.05 was considered statistically significant.

Results

Baseline characteristics before and after PSM

Table 1 presents the patient characteristics of the entire cohort (n = 726) and propensity score-matched cohort (n = 175). After comparing the two groups before PSM, we found that BMI (P = 0.035), NRS-2002 score less than 3 (P = 0.031), extent of tumor resection (P = 0.008), p/ypT stage (P = 0.007), p/ypN stage (P = 0.009), p/ypTNM stage (P = 0.009), and tumor diameter (P = 0.035) were significantly different between the NICT-MIG and MIG groups.

After PSM, no significant differences were found in sex, age, BMI, NRS-2002 score, age-adjusted Charlson comorbidity index (aCCI) score, ASA grade, history of abdominal surgery, surgical approach, extent of tumor resection, p/ypT stage, p/ypN stage, p/ypTNM stage, or tumor diameter between the two groups (P > 0.05). The pathological characteristics were also comparable, with no significant difference after PSM, as shown in Table 2.

Table 1 Baseline characteristics between NICT-MIG and MIG group before and after PSM
Table 2 Pathological characteristics of MIG group and NICT-MIG group after PSM for older patients

Therapeutic effects and TRAEs in NICT-MIG

Table 3 shows the preoperative treatment and tumor response in the NICT-MIG group. Most patients (n = 45, 73.8%) accepted ≤ 4 treatment cycles. A total of 39 (63.9%), 16 (26.2%), 3 (4.9%), and 3 (4.9%) patients received SOX, XELOX, AS and other chemotherapy regimens, respectively. According to the RECIST 1.1 criteria, 5 patients (8.2%) achieved CR, 33 patients (54.1%) achieved PR, 19 patients (31.1%) achieved SD, and 4 patients(6.6%) had PD. The objective response rate (ORR) and disease control rate (DCR) were 62.3% and 93.4%, respectively. Based on Becker’s standard, 13 (21.3%) patients achieved pCR with TRG1a, and 27 (44.2%) patients achieved MPR with TRG1a/1b.

As mentioned in Table 4, the three most common TRAEs affecting therapeutic safety were nausea and vomiting (26.2%), thrombocytopenia (24.6%), and leukopenia (23.0%). The overall rates of TRAEs and severe TRAEs were 83.6% and 27.9%, respectively. No treatment-related deaths occurred in the NICT-MIG group.

Table 3 Preoperative treatment and tumor response in the NICT-MIG group
Table 4 Treatment-related adverse events of NICT-MIG group for older patients

Surgical safety and postoperative recovery

As shown in Table 5, compared with the MIG group, the NICT-MIG group had longer operation times (235.20 ± 53.89 min vs. 210.55 ± 55.07 min, P = 0.005) , more volume of postoperative day 3 drainage [200(100-400)ml vs. 100(75-150)ml, P=0.019], and postoperative hospitalization days [9.0 (7.0–10.0) days vs. 7.0 (6.75-9.0) days, P = 0.030]. No significant differences were found in estimated blood loss [100.0 (50.0-100.0) ml vs. 100.0 (50.0-100.0) ml, P = 0.568], number of retrieved lymph nodes [26.5 (18.75–36.25) vs. 25.0 (20.5–40.0), P = 0.650], first flatus days [3.0 (3.0–4.0) d vs. 4.0 (3.0–4.0), P = 0.724], or the R0 resection rate (100% vs. 95.1%, P = 0.076) between the MIG and NICT-MIG groups. For patients who needed adjuvant therapy, the rate of completion of the total cycle was significantly greater in the NICT-MIG group than in the MIG group (90.1% vs. 72.7%, P = 0.012).

Table 5 Comparison of surgical characteristics and postoperative recovery between MIG and NICT-MIG groups

Postoperative complications and risk factors

There was no significant difference between the MIG cohort and the NICT-MIG cohort regarding the overall complication rate (17.5% vs. 24.6%, P = 0.267) or severe complication rate (1.8% vs. 1.6%, P = 1.000) within 30 days after surgery. The most common POCs were anemia and hypoproteinemia in the MIG and NICT-MIG groups, respectively. One patient died due to heart failure in the MIG group, while another patient died as a result of acute pulmonary embolism in the NICT-MIG group.

Table 6 shows the albumin(ALB) changes in the serum levels before and after surgery. Patients in the NICT-MIG group were more prone to have lower serum ALB levels than were those in the MIG group at postoperative Day 3, even though no significant difference was found in the preoperative serum ALB levels between the two groups (Supplementary Fig. 1). A higher incidence of hypoproteinemia with Clavien-Dindo classification II in older patients in the NICT-MIG group was also reported (9.8% vs. 1.8%, P = 0.039).

Table 6 Changes of albumin level between NICT-MIG and MIG group

Table 7 presents the results of logistic regression, which aimed to explore the risk factors for POCs in older patients with GC receiving MIG. Univariate analysis revealed that non-R0 resection [OR (95% CI): 8.424 (0.741–95.716), P = 0.086], estimated blood loss [OR (95% CI): 4.313 (1.527–12.184), P = 0.006], LMR [OR (95% CI): 3.692 (1.705–7.998), P = 0.001], , and PLR [OR (95% CI):.

1.965 (0.922–4.189), P = 0.080] were associated with POCs (P < 0.1). NICT before MIG was not an independent risk factor [OR (95% CI): 1.533 (0.719–3.266), P = 0.269] for POCs. Multivariate logistic analysis revealed that an estimated blood loss > 200 mL [OR (95% CI): 4.232(1.409–12.708), P = 0.010] and a lymphocyte-to-monocyte ratio (LMR) ≤ 3.25 [OR (95% CI): 3.132(1.397–7.026), P = 0.006] were found to be independent risk factors for POCs after MIG in older patients.

Table 7 Uni- and multivariate logistic analysis for overall postoperative complications after MIG in older patients

Long-term survival

Figure 4 shows the Kaplan‒Meier curves of overall survival (OS) and recurrence-free survival (RFS) between the two groups. We found that the 2-year OS and 2-year RFS rates were both comparable in the MIG and NICT-MIG groups (80.0% vs. 72.4%, log-rank χ2 = 0.179, P = 0.672; 78.7% vs. 71.3%, log-rank χ2 = 1.741, P = 0.187). Subgroup analysis revealed that patients with ypTNM stage III in the NICT-MIG group had poorer RFS than patients in the MIG group (P = 0.043) (Supplementary Fig. 2). Cox analysis revealed that LMR ≤ 3.25 and poor p/ypTNM stage were independent risk factors for OS in older patients who underwent MIG (P = 0.015, P = 0.001). Non-R0 resection and poor p/ypTNM stage were found to be independent risk factors for RFS in older patients who underwent MIG (P = 0.004, P < 0.001) (Supplementary Tables 1 and 2).

Fig. 4
figure 4

Kaplan-Meier curve of overall survival and recurrence-free survival between NICT-MIG and MIG group a: Kaplan-Meier curve of overall survival between two groups; b. Kaplan-Meier curve of recurrence-free survival between two groups

Discussion

This retrospective PSM study mainly focused on the short- and long-term outcomes of older patients who underwent upfront MIG or MIG after NICT, aiming to explore the safety and efficacy of NICT and MIG after NICT for older patients with GC.

NICT has played a pivotal role in treating locally advanced GC in recent years, offering advantages such as a greater probability of tumor regression, downstaging and potential long-term survival benefits. However, older patients typically exhibit a weaker immune system, leading to a reduction in the number of T cells and decreased levels of tumor-infiltrating lymphocytes. This may impact the clinical benefits that older patients derive from NICT. Nishijima TF’s study demonstrated similar survival benefits between younger and older patients who received immune checkpoint inhibitors (ICIs) [20]. Wu Q et al. reported that cancer patients older than 75 years failed to achieve remarkable benefits from ICIs in terms of both overall survival and progression-free survival [21]. Our study revealed that 13 (21.3%) and 27 (44.2%) older patients with GC in the NICT-MIG group achieved pCR and MPR, respectively. These findings indicate a comparable tumor response in older patients, aligning with previous studies conducted across all age populations [22]. We also found the comparable 2-year OS and RFS between two groups, especially for pTNM satege I patients undergoing upfront MIG and ypTNM stage 0-I patients who had better tumor response in the NICT-MIG group. Thus, our results suggest that NICT induces better tumor regression in older patients with GC, potentially leading to improved survival, which was similar with the older patients who underwent upfront MIG with the same pathological TNM stage.

The TRAEs resulting from NICT may compromise the physical condition and impact perioperative safety, particularly in older patients. A study by Manji GA reported that 57.1% of patients experienced severe (grade 3 or higher) TRAEs during NICT, and 3 older patients (over 75 years old) experienced grade V TRAEs [23]. A meta-analysis demonstrated that the incidence rates of total TRAEs and grade 3 to 4 TRAEs were 89.1% and 34.4%, respectively [24]. In our study, the overall rates of TRAEs and severe TRAEs in the NICT-MIG group were 83.6% and 27.9%, respectively, which are similar to findings from previous studies. These results indicate the medical safety considerations associated with NICT for older patients with GC.

Operation time and estimated blood loss are crucial indicators reflecting surgical safety and predicting POCs. We observed that patients in the NICT-MIG group had a longer operation time than did those in the upfront MIG group (235.20 ± 53.89 min vs. 210.55 ± 55.07 min, P = 0.005). Although no significant difference was found in the estimated blood loss between the two groups (P = 0.568), multivariate logistic analysis revealed that an estimated blood loss of > 200 mL [OR (95% CI): 4.232(1.409–12.708), P = 0.010] was an independent risk factor for POCs in older patients who underwent MIG. These results indicate that the adverse effects of NICT, such as increased tissue fragility, severe exudative edema, myelosuppression, and immune system weakening, increase surgical difficulty and pose a potential risk of POCs.

Moreover, the longer postoperative hospitalization days in the NICT-MIG group than in the MIG group also indicate delayed postoperative recovery attributable to NICT. According to our clinical experience and observation, for older patients who undergo NICT-MIG, prolonged operation time (P = 0.005) has a greater impact on cardiopulmonary function, and postoperative recovery may be appropriately extended to ensure the perioperative safety. Moreover, due to obvious exudate and edema in the surgical area after NICT, the postoperative drainage volume (postoperative Day 3) in the NICT-MIG group was significantly greater than that in the direct surgery group (P = 0.019), making it easier for patients to develop related complications such as hypoproteinemia (9.8% vs. 1.8%, P = 0.039), and the time for tube removal was extended, leading to a relatively longer postoperative hospital stay. Therefore, it is imperative for surgeons to conduct comprehensive perioperative evaluations and perform meticulous surgical procedures to ensure the safe application of NICT-MIG in older patients.

The application of MIG in older patients has been widely acknowledged, as evidenced by current studies [25, 26]. However, it is still controversial whether it is equally reliable to conduct MIG after NICT. A meta-analysis revealed that surgical complications were more common with neoadjuvant therapy for GC [27]. A multicenter study demonstrated that the incidence of POCs and the proportion of perioperative textbook outcomes were comparable between patients who underwent laparoscopic gastrectomy after NICT and those who received neoadjuvant chemotherapy alone- [28]. In this study, no significant differences were found between the upfront MIG and NICT-MIG cohorts regarding overall and severe POC rates, indicating the safety and feasibility of NICT-MIG for older patients. Moreover, we also need to be vigilant regarding MIG/NICT-MIG-related deaths, including acute pulmonary embolism and heart failure, which are prone to occur in older patients [29].

The LMR is considered a promising biomarker for predicting therapeutic effects and long-term prognosis in patients with GC. Most studies have demonstrated that a lower LMR might be associated with poor prognosis in GC patients [30, 31]. Shigeo Tokumaru et al. also reported that the LMR could predict the tumor response of GC patients who received nivolumab [32]. With respect to predicting postoperative complications, Selçuk Gülmez et al. demonstrated that the preoperative LMR had the best ability to predict postoperative infections after gastrectomy [33]. Hsu JT’s study showed that GC patients with lower LMR had higher surgical mortality rates [34]. In our study, we selected an LMR of 3.25 as the cutoff value according to the ROC curve. Multivariate analysis revealed that a preoperative LMR ≤ 3.25 was an independent risk factor for both POCs and overall survival in older patients with GC who underwent MIG. This interesting finding led us to hypothesize that the decrease in lymphocyte counts reflects the impairment of physical immunity, while elevated monocyte levels often imply increasing levels of tumor-associated macrophages (TAMs), which synergistically affect postoperative recovery, increase the risk of complications, and cause poor survival, especially in older patients with comorbidities or malnutrition [35].The correlation between nutritional status and long-term prognosis in older patients has been widely studied. The serum ALB level and Onodera’s PNI score are representative indices that can reflect the nutritional status of older patients. In this study, we found that the serum ALB level on postoperative Day 3 was significantly lower in the NICT-MIG group than in the MIG group, even though no significant difference was found in the preoperative serum ALB level between the two groups. A decrease in nutritional status caused by NICT or MIG might affect adjuvant therapy, and increase the likelihood of disease progression and deterioration of immunocompetence. Kanda M et al. demonstrated that the prognostic significance of the PNI was more apparent in patients with stage II GC and in those who received adjuvant chemotherapy [36]. We found that a lower preoperative PNI (PNI ≤ 45) was not significantly associated with more POCs, poor OS, or RFS by univariate analysis for older patients who underwent MIG. We attributed this interesting phenomenon to two aspects: (1) NICT has resulted in a better tumor response and improved the completion rate of total cycles of perioperative treatment in older patients, which may offset the adverse effects of adjuvant chemotherapy caused by poor nutritional status after surgery; and (2) all enrolled patients in this study underwent nutritional assessment and timely supplementation of enteral or parenteral nutrition in accordance with the European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines [37], which weakened the impact of nutritional status on prognosis.

This study has several inherent limitations. First, some potential bias may still exist even though we used PSM analysis to balance the baseline characteristics between the two groups. Second, we lack in-depth analysis of some key positive results due to the limited length and understanding. Based on future large-scale research, we will conduct more in-depth exploration of interesting phenomena found in this article. Third, because of the limited sample size in this retrospective study, we did not standardize the regimen or number of cycles of NICT. Further large-scale prospective studies should be conducted to eliminate the potential effect of different types of NICT and explore the longer prognosis for older patients, providing high-level evidence for the broader application of NICT in older patients with GC.

Conclusion

To the best of our knowledge, this is the first study dedicated to assessing the safety and efficacy of NICT-MIG in older patients with GC. Our findings indicate that older patients who undergo NICT can achieve equally good tumor responses, such as ORR, pCR and MPR, with an acceptable incidence of TRAEs. Despite a longer operation time and longer postoperative hospitalization days in the NICT-MIG group, no significant differences were observed in terms of the R0 resection rate, estimated blood loss, number of retrieved lymph nodes, first flatus day, overall POCs, severe POCs, 2-year OS or RFS compared with those in the upfront MIG group. These findings underscore the safety and efficacy of NICT-MIG for older patients. For patients with estimated blood loss > 200 mL and an LMR ≤ 3.25, it is recommended that surgeons prioritize perioperative management to mitigate the risk of POCs.

Data availability

All datasets generated for this study are included in the article and available from corresponding authors upon reasonable request.

Abbreviations

NICT:

Neoadjuvant immunotherapy combined with chemotherapy

GC:

Gastric cancer

LAGC:

Locally advanced gastric cancer

MIG:

Minimally invasive gastrectomy

PSM analysis:

Propensity-score matched analysis

ASA:

American Association of Anesthesiologists

BMI:

Body mass index; aCCI: Age-adjusted Charlson Comorbidity Index

NRS-2002:

Nutritional risk screening-2002

pCR:

Pathological complete response

MMR:

mismatch repair

MPR:

Major pathological response

CR:

Complete response

PR:

Partial response

SD:

Stable disease

PD:

Progressive disease

ORR:

Objective response rate

DCR:

Disease control rate

TRG:

Tumor regression grade

PLR:

Platelet-lymphocyte ratio

PNI:

Onodera’s prognostic nutritional index

ICI:

Immune checkpoint inhibitor

OR:

Odd ratio

TRAEs:

Treatment-related adverse events

POCs:

Postoperative complications

HR:

Hazard ratio

OS:

Overall survival

RFS:

Recurrence-free survival

ESPEN:

European society for clinical nutrition and metabolism

ALB:

Albumin

References

  1. Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J (Engl). 2021;134(7):783–91.

    Article  PubMed  Google Scholar 

  2. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  3. Paredero-Pérez I, Jimenez-Fonseca P, Cano JM, Arrazubi V, Carmona-Bayonas A, Covela-Rúa M, Fernández-Montes A, Martín-Richard M, Gironés-Sarrió R. State of the scientific evidence and recommendations for the management of older patients with gastric cancer. J Geriatr Oncol.2023;101657.

  4. Yu J, Huang C, Sun Y, Su X, Cao H, Hu J, Wang K, Suo J, Tao K, He X, Wei H, Ying M, Hu W, Du X, Hu Y, Liu H, Zheng C, Li P, Xie J, Liu F, Li Z, Zhao G, Yang K, Liu C, Li H, Chen P, Ji J, Li G, Chinese Laparoscopic Gastrointestinal Surgery Study (CLASS) Group. Effect of laparoscopic vs Open Distal Gastrectomy on 3-Year disease-free survival in patients with locally advanced gastric Cancer: the CLASS-01 Randomized Clinical Trial. JAMA. 2019;321(20):1983–92.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Liu F, Huang C, Xu Z, Su X, Zhao G, Ye J, Du X, Huang H, Hu J, Li G, Yu P, Li Y, Suo J, Zhao N, Zhang W, Li H, He H, Sun Y. Chinese laparoscopic gastrointestinal surgery study (CLASS) Group. Morbidity and mortality of laparoscopic vs Open Total Gastrectomy for Clinical Stage I gastric Cancer: the CLASS02 Multicenter Randomized Clinical Trial. JAMA Oncol. 2020;6(10):1590–7.

    Article  PubMed  Google Scholar 

  6. Hikage M, Fujiya K, Kamiya S, Tanizawa Y, Bando E, Terashima M. Efficacy of minimally invasive distal gastrectomy for elderly patients with clinical stage I/IIA gastric cancer: a propensity-score matched analysis. Surg Endosc. 2021;35(12):7082–93.

    Article  PubMed  Google Scholar 

  7. Li ZY, Chen J, Bai B, Xu S, Song D, Lian B, Li JP, Ji G, Zhao QC. Laparoscopic gastrectomy for elderly gastric-cancer patients: comparisons with laparoscopic gastrectomy in non-elderly patients and open gastrectomy in the elderly. Gastroenterol Rep (Oxf). 2021;9(2):146–53.

    Article  PubMed  Google Scholar 

  8. Zhao RY, Li HH, Zhang KC, Cui H, Deng H, Gao JW, Wei B. Comparison of short-term efficacy between totally laparoscopic gastrectomy and laparoscopic assisted gastrectomy for elderly patients with gastric cancer. World J Gastrointest Surg. 2022;14(9):950–62.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ding XW, Wang XJ, Li B, Wang LG, Guo HH, Shang L, Huang WB, Wu LL, Ke B, Liu Y, Liu N, Wang BG, Cai MZ, Deng JY, Zhang RP, Zhang JC, Chai J, Zhao Q, Li LP, Liang H. PERSIST: a multicenter, randomized phase II trial of perioperative oxaliplatin and S-1 (SOX) with or without sintilimab in resectable locally advanced gastric/gastroesophageal junction cancer (GC/GEJC). J Clin Oncol. 2023;41(4suppl):364–364.

    Article  Google Scholar 

  10. Topalian SL, Forde PM, Emens LA, Yarchoan M, Smith KN, Pardoll DM. Neoadjuvant immune checkpoint blockade: a window of opportunity to advance cancer immunotherapy. Cancer Cell. 2023;41(9):1551–66.

    Article  CAS  PubMed  Google Scholar 

  11. Jiang Q, Liu W, Zeng X, Zhang C, Du Y, Zeng L, Yin Y, Fan J, Yang M, Tao K, Zhang P. Safety and efficacy of tislelizumab plus chemotherapy versus chemotherapy alone as neoadjuvant treatment for patients with locally advanced gastric cancer: real-world experience with a consecutive patient cohort. Front Immunol. 2023;14:1122121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Su J, Guo W, Chen Z, Wang L, Liu H, Zhao L, Lin T, Li F, Mao X, Huang H, Yu J, Li G, Hu Y. Safety and short-term outcomes of laparoscopic surgery for advanced gastric cancer after neoadjuvant immunotherapy: a retrospective cohort study. Front Immunol. 2022;13:1078196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.

    Article  CAS  PubMed  Google Scholar 

  14. Freites-Martinez A, Santana N, Arias-Santiago S, Viera A. Using the common terminology criteria for adverse events (CTCAE - version 5.0) to evaluate the severity of adverse events of Anticancer therapies. Actas Dermosifiliogr (Engl Ed). 2021;112(1):90–2.

    Article  CAS  PubMed  Google Scholar 

  15. Becker K, Mueller JD, Schulmacher C, Ott K, Fink U, Busch R, Böttcher K, Siewert JR, Höfler H. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer. 2003;98(7):1521–30.

    Article  PubMed  Google Scholar 

  16. Japanese gastric cancer treatment guidelines 2018 (5th edition). Gastric Cancer. 2021; 24(1):1–21.

  17. Zang L, Ma JJ, Zhao YL, Zhao X, Zhang LY. Operative guidelines for laparoscopic gastrectomy (2023 Edition). Chin J Practical Surg. 2023;43(04):361–70. (In Chinese).

    Google Scholar 

  18. Specialized Committee on Robotic and Laparoscopic Surgery, Chinese Research Hospital Association. Expert consensus on robotic gastrectomy (2015 edition). Chin J Gastrointest Surg. 2016;15(1):7–11. (In Chinese).

    Google Scholar 

  19. Katayama H, Kurokawa Y, Nakamura K, Ito H, Kanemitsu Y, Masuda N, Tsubosa Y, Satoh T, Yokomizo A, Fukuda H, Sasako M. Extended clavien-dindo classification of surgical complications: Japan Clinical Oncology Group postoperative complications criteria. Surg Today. 2016;46(6):668–85.

    Article  PubMed  Google Scholar 

  20. Nishijima TF, Muss HB, Shachar SS, Moschos SJ. Comparison of efficacy of immune checkpoint inhibitors (ICIs) between younger and older patients: a systematic review and meta-analysis. Cancer Treat Rev. 2016;45:30–7.

    Article  CAS  PubMed  Google Scholar 

  21. Wu Q, Luo S, Xie X. Lack of efficacy of Immune checkpoint inhibitors in Cancer patients older Than 75? J Immunother. 2022;45(7):307–20.

    Article  CAS  PubMed  Google Scholar 

  22. Yuan Z, Cui H, Wang S, Liang W, Cao B, Song L, Liu G, Huang J, Chen L, Wei B. Combining neoadjuvant chemotherapy with PD-1/PD-L1 inhibitors for locally advanced, resectable gastric or gastroesophageal junction adenocarcinoma: a systematic review and meta-analysis. Front Oncol. 2023;13:1103320.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Manji GA, Lee S, Del Portillo A, May M, Ana SS, Alouani E, Sender N, Negri T, Gautier K, Ge L, Fan W, Xie M, Sethi A, Schrope B, Tan AC, Park H, Oberstein PE, Shah MA, Raufi AG. Chemotherapy and Immune Checkpoint Blockade for Gastric and Gastroesophageal Junction Adenocarcinoma. JAMA Oncol.2023; e234423.

  24. Xu H, Li T, Shao G, Wang W, He Z, Xu J, Qian Y, Liu H, Ge H, Wang L, Zhang D, Yang L, Li F, Xu Z. Evaluation of neoadjuvant immunotherapy plus chemotherapy in Chinese surgically resectable gastric cancer: a pilot study by meta-analysis. Front Immunol. 2023;14:1193614.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ueda Y, Shiraishi N, Fujishima H, Kawasaki T, Ninomiya S, Shiroshita H, Etoh T, Inomata M. Technical and oncological safety of laparoscopic gastrectomy for gastric cancer in elderly patients ≥ 80 years old. BMC Geriatr. 2022;22(1):475.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Kwon IG, Cho I, Guner A, Kim HI, Noh SH, Hyung WJ. Minimally invasive surgery as a treatment option for gastric cancer in the elderly: comparison with open surgery for patients 80 years and older. Surg Endosc. 2015;29(8):2321–30.

    Article  PubMed  Google Scholar 

  27. Lim Khai Shin A, Ho Si Ying A, Neo Hui Wen S, Yeo SC, Tay KV. Systematic review and meta-analysis of the outcomes following neoadjuvant therapy in upfront resectable gastric cancers compared to surgery alone in phase III randomised controlled trials. J Gastrointest Surg. 2023;27(6):1261–76.

    Article  PubMed  Google Scholar 

  28. Sun YQ, Zhong Q, Lv CB, Zhu JY, Lin GT, Zhang ZQ, Wu D, Weng CM, Chen QX, Lian MQ, Zeng WM, Zhang YB, Chen QY, Lin JX, Xie JW, Li P, Zheng CH, Lu J, Cai LS, Huang CM. The safety and efficacy of neoadjuvant immunochemotherapy following laparoscopic gastrectomy for gastric cancer: a multicenter Real-world clinical study. Int J Surg. 2024 Apr 23.

  29. Hernandez AF, Whellan DJ, Stroud S, Sun JL, O’Connor CM, Jollis JG. Outcomes in heart failure patients after major noncardiac surgery. J Am Coll Cardiol. 2004;44(7):1446–53.

    Article  PubMed  Google Scholar 

  30. Xu BB, Xu Y, Lu J, Wu Y, Wang JB, Lin JX, Xie JW, Li P, Zheng CH, Huang AM, Huang CM. Prognostic significance of combined lymphocyte-monocyte ratio and tumor-associated macrophages in Gastric Cancer patients after Radical Resection. J Cancer. 2020;11(17):5078–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schiefer S, Wirsik NM, Kalkum E, Seide SE, Nienhüser H, Müller B, Billeter A, Büchler MW, Schmidt T, Probst P. Systematic review of Prognostic Role of blood cell ratios in patients with gastric Cancer undergoing surgery. Diagnostics (Basel). 2022;12(3):593.

    Article  PubMed  Google Scholar 

  32. Tokumaru S, Koizumi T, Sekino Y, Takeuchi N, Nakata S, Miyagawa Y, Kitazawa M, Muranaka F, Nakamura S, Koyama M, Yamamoto Y, Ehara T, Hondo N, Soejima Y. Lymphocyte-to-monocyte ratio is a predictive biomarker of response to treatment with Nivolumab for gastric Cancer. Oncology. 2021;99(10):632–40.

    Article  CAS  PubMed  Google Scholar 

  33. Gülmez S, Senger A, Uzun O, Ozduman O, Ofluoglu C, Subasi İ, Sen B, Pence M, Duman U, Polat E. Comparative analysis of preoperative ratio based markers in Predicting Postoperative Infectious complications after Gastrectomy. Pol Przegl Chir. 2022;95(4):1–5.

    Article  PubMed  Google Scholar 

  34. Hsu JT, Wang CC, Le PH, Chen TH, Kuo CJ, Lin CJ, Chou WC, Yeh TS. Lymphocyte-to-monocyte ratios predict gastric cancer surgical outcomes. J Surg Res. 2016;202(2):284–90.

    Article  CAS  PubMed  Google Scholar 

  35. Warny M, Helby J, Nordestgaard BG, Birgens H, Bojesen SE. Lymphopenia and risk of infection and infection-related death in 98,344 individuals from a prospective Danish population-based study. PLoS Med. 2018;15(11):e1002685.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Kanda M, Mizuno A, Tanaka C, Kobayashi D, Fujiwara M, Iwata N, Hayashi M, Yamada S, Nakayama G, Fujii T, Sugimoto H, Koike M, Takami H, Niwa Y, Murotani K, Kodera Y. Nutritional predictors for postoperative short-term and long-term outcomes of patients with gastric cancer. Med (Baltim). 2016;95(24):e3781.

    Article  CAS  Google Scholar 

  37. Volkert D, Beck AM, Cederholm T, Cruz-Jentoft A, Goisser S, Hooper L, Kiesswetter E, Maggio M, Raynaud-Simon A, Sieber CC, Sobotka L, van Asselt D, Wirth R, Bischoff SC. ESPEN guideline on clinical nutrition and hydration in geriatrics. Clin Nutr. 2019;38(1):10–47.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by National Natural Science Foundation of China (82073192, 82273231), and Beijing Science and Technology Program (Z221100007422125). All above-mentioned foundations provided financial support on data collection and statistical analysis.

Author information

Authors and Affiliations

Authors

Contributions

H.C., J.X.C. and B.W. designed the study. H.C. and Z.Y. collected the data. H.C., W.Q.L. and C.B. analyzed and interpreted the data. H.C. prepared the manuscript. B.W., L.C. and J.X.C. provided whole guidance for the paper. All the authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jianxin Cui or Bo Wei.

Ethics declarations

Ethical approval

This study was approved by the Ethics Committee of the Chinese PLA General Hospital (approval no. S2023-776-01), and informed consent was obtained from all patients before treatment.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Supplementary Material 3

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cui, H., Yuan, Z., Liang, W. et al. Safety and efficacy of minimally invasive gastrectomy for older patients with gastric cancer after neoadjuvant chemotherapy and immunotherapy: a propensity score-matched analysis. BMC Geriatr 24, 606 (2024). https://doi.org/10.1186/s12877-024-05193-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12877-024-05193-w

Keywords